Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Int Immunol ; 31(4): 239-250, 2019 03 28.
Artículo en Inglés | MEDLINE | ID: mdl-30778577

RESUMEN

The intrinsic immunosuppressive properties of regulatory T (Treg) cells can be harnessed for therapeutic approaches aiming at down-modulating harmful immune reactions. In this context, expanded type 1 Treg cells (Tr1 cells) specific for ovalbumin (ova-Tr1 cells) have been tested for clinical efficacy in the treatment of autoimmune disorders such as refractory Crohn's disease (CD). The clinical use of these therapeutic products warrants exploration of their mechanism of action. Here, we identified a relationship between the CD activity index and the expression of lytic molecules by the ova-Tr1 cells administered in the previously reported First-in-Man study [Crohn's And Treg cells Study 1 (CATS1) study]. Accordingly, ova-Tr1 cells were found to carry granules containing high levels of lytic molecules, including multiple granzymes and granulysin. These cells displayed a T-cell receptor (TCR)-independent cytotoxic activity, which was preferentially directed toward myeloid cell lines and monocyte-derived dendritic cells. Upon contact with myeloid cells, ova-Tr1 cells induced their apoptosis via a perforin-independent and a granulysin/granzyme-dependent mechanism. As compared to CD8+ cytotoxic T cells, ova-Tr1 cells required more time to lyse target cells and displayed a more gradual lytic activity over time. Notably, this activity was sustained over days resulting in the control of myeloid cell populations at a relatively low ratio. Our study reveals that ova-Tr1 cells are endowed with a sustained cytotoxic activity that relies on a unique combination of granulysin and granzymes and that preferentially eliminates myeloid target cells in a TCR-independent manner.


Asunto(s)
Antígenos de Diferenciación de Linfocitos T/metabolismo , Linfocitos T CD8-positivos/inmunología , Granzimas/metabolismo , Células Mieloides/inmunología , Linfocitos T Reguladores/inmunología , Antígenos/inmunología , Células Cultivadas , Citotoxicidad Inmunológica , Humanos , Activación de Linfocitos , Ovalbúmina/inmunología , Especificidad del Receptor de Antígeno de Linfocitos T , Células THP-1 , Células U937
2.
Gastroenterology ; 143(5): 1207-1217.e2, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22885333

RESUMEN

BACKGROUND & AIMS: New therapeutic strategies are needed for patients with refractory Crohn's disease (CD). We evaluated data from the Crohn's And Treg Cells Study (CATS1) to determine the safety and efficacy of antigen-specific T-regulatory (Treg) cells for treatment of patients with refractory CD. METHODS: We performed a 12-week, open-label, multicenter, single-injection, escalating-dose, phase 1/2a clinical study in 20 patients with refractory CD. Ovalbumin-specific Treg cells (ova-Tregs) were isolated from patients' peripheral blood mononuclear cells (PBMCs), exposed to ovalbumin, and administrated intravenously. Safety and efficacy were assessed using clinical and laboratory parameters. We evaluated proliferation of PBMCs in response to ovalbumin. RESULTS: Injections of ova-Tregs were well tolerated, with 54 adverse events (2 related to the test reagent) and 11 serious adverse events (3 related to the test reagent, all recovered). Overall, a response, based on a reduction in Crohn's Disease Activity Index (CDAI) of 100 points, was observed in 40% of patients at weeks 5 and 8. Six of the 8 patients (75%) who received doses of 10(6) cells had a response at weeks 5 and 8, with a statistically significant reduction in CDAI. In this group, remission (based on CDAI ≤150) was observed in 3 of 8 patients (38%) at week 5 and 2 of 8 patients (25%) at week 8. CONCLUSIONS: Administration of antigen-specific Tregs to patients with refractory CD (CATS1) was well tolerated and had dose-related efficacy. The ovalbumin-specific immune response correlated with clinical response, supporting immune-suppressive mechanisms of ova-Tregs. The consistency of results among different assessment methods supports the efficacy of ova-Tregs; this immune therapy approach warrants further clinical and mechanistic studies in refractory CD. Eudract, Number: 2006-004712-44.


Asunto(s)
Enfermedad de Crohn/terapia , Inmunoterapia , Linfocitos T Reguladores/trasplante , Adulto , Anciano , Proteína C-Reactiva/metabolismo , Enfermedad de Crohn/sangre , Heces , Femenino , Humanos , Inmunoterapia/efectos adversos , Complejo de Antígeno L1 de Leucocito/metabolismo , Recuento de Linfocitos , Masculino , Persona de Mediana Edad , Ovalbúmina/inmunología , Calidad de Vida , Índice de Severidad de la Enfermedad , Encuestas y Cuestionarios , Linfocitos T Reguladores/inmunología , Adulto Joven
3.
Cells ; 11(18)2022 09 13.
Artículo en Inglés | MEDLINE | ID: mdl-36139429

RESUMEN

Accumulated experimental and clinical evidence supports the development of human allogeneic liver-derived progenitor cells (HALPCs) to treat fibro-inflammatory liver diseases. The aim of the present study was to evaluate their therapeutic effect in a non-alcoholic steatohepatitis (NASH)-STAM mouse model. The immune signaling characteristics of HALPCs were first assessed in vitro. Upon inflammation treatment, HALPCs secreted large amounts of potent bioactive prostaglandin E2 and indoleamine 2,3-dioxygenase, which significantly reduced CD4+ T-lymphocyte proliferation and secretion of proinflammatory cytokines. In vivo, HALPCs were intravenously administered as single or triple shots (of a dose of 12.5 × 106 cells/kg BW) in STAM mice. Transplantation of HALPCs was associated with a significant decrease in the NAFLD activity score at an early stage and in both inflammation and hepatocyte ballooning scores in late-stage NASH. Sirius red staining analyses revealed decreased collagen deposition in the pericentral region at both stages of NASH. Altogether, these findings showed the anti-inflammatory and anti-fibrotic features of HALPCs in an in vivo NASH model, which suggests their potential to reverse the progression of this chronic fibro-inflammatory disease.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas , Enfermedad del Hígado Graso no Alcohólico , Animales , Citocinas , Dinoprostona , Modelos Animales de Enfermedad , Humanos , Indolamina-Pirrol 2,3,-Dioxigenasa , Inflamación/complicaciones , Cirrosis Hepática/complicaciones , Cirrosis Hepática/terapia , Ratones , Ratones Endogámicos C57BL , Enfermedad del Hígado Graso no Alcohólico/complicaciones , Enfermedad del Hígado Graso no Alcohólico/terapia
4.
Stem Cells ; 28(5): 965-73, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20333749

RESUMEN

Several reports showed that hematopoietic stem cells (HSCs) participate in muscle regeneration, raising hope for their therapeutic potential for degenerative muscle diseases. However, proof that HSCs are able to reprogram their fate and enter a myogenic pathway, remains elusive. We demonstrate that murine bone marrow (BM)-derived hematopoietic cells, carrying reporter genes controlled by muscle-specific regulatory elements from the Myf5, myosin light chain (MLC3F), or MCK genes, are induced by myoblasts to activate muscle-specific genes. This potential resides in the more undifferentiated progenitors, expressing surface markers typical of HSCs. Comparative gene expression profiling of CD45(+)/Sca1(+) cells isolated from muscle or BM shows that hematopoietic cells participate to muscle regeneration, by undergoing a profound although incomplete myogenic reprogramming on interaction with the muscle microenviroment. These cells undergo specification and differentiation independently from Pax7 and MyoD, and lack Pax7-associated properties, such as self-renewal and proliferation, distinguishing from satellite cells. Our findings indicate that hematopoietic cells, on seeding in the muscle, become a distinct cell population endowed with myogenic potential.


Asunto(s)
Células de la Médula Ósea/fisiología , Diferenciación Celular/genética , Células Madre Hematopoyéticas/fisiología , Desarrollo de Músculos/genética , Mioblastos/fisiología , Factor de Transcripción PAX7/fisiología , Animales , Células de la Médula Ósea/citología , Linaje de la Célula , Células Cultivadas , Regulación de la Expresión Génica/genética , Células Madre Hematopoyéticas/citología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Proteína MioD/genética , Proteína MioD/metabolismo , Mioblastos/citología , Factor de Transcripción PAX7/deficiencia , Factor de Transcripción PAX7/genética
5.
Stem Cells ; 28(4): 753-64, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20135684

RESUMEN

The differentiation of multipotent cells into undesirable lineages is a significant risk factor when performing cell therapy. In muscular diseases, myofiber loss can be associated with progressive fat accumulation that is one of the primary factors leading to decline of muscular strength. Therefore, to avoid any contribution of injected multipotent cells to fat deposition, we have searched for a highly myogenic but nonadipogenic muscle-derived cell population. We show that the myogenic marker CD56, which is the gold standard for myoblast-based therapy, was unable to separate muscle cells into myogenic and adipogenic fractions. Conversely, using the stem cell marker CD34, we were able to sort two distinct populations, CD34(+) and CD34(-), which have been thoroughly characterized in vitro and in vivo using an immunodeficient Rag2(-/-)gamma(c) (-/-) mouse model of muscle regeneration with or without adipose deposition. Our results demonstrate that both populations have equivalent capacities for in vitro amplification. The CD34(+) cells and CD34(-) cells exhibit equivalent myogenic potential, but only the CD34(-) population fails to differentiate into adipocytes in vitro and in vivo after transplantation into regenerative fat muscle. These data indicate that the muscle-derived cells constitute a heterogeneous population of cells with various differentiation potentials. The simple CD34 sorting allows isolation of myogenic cells with no adipogenic potential and therefore could be of high interest for cell therapy when fat is accumulated in diseased muscle.


Asunto(s)
Adipocitos/citología , Técnicas de Cultivo de Célula/métodos , Adipocitos/inmunología , Adipocitos/metabolismo , Adolescente , Adulto , Animales , Antígenos CD34/inmunología , Diferenciación Celular , Linaje de la Célula , Separación Celular , Niño , Preescolar , Femenino , Regulación de la Expresión Génica , Humanos , Lactante , Masculino , Ratones , Fibras Musculares Esqueléticas/citología , Fibras Musculares Esqueléticas/inmunología
6.
Cells ; 8(8)2019 08 07.
Artículo en Inglés | MEDLINE | ID: mdl-31394759

RESUMEN

The efficacy of mesenchymal stem cell infusion is currently tested in numerous clinical trials. However, therapy-induced thrombotic consequences have been reported in several patients. The aim of this study was to optimize protocols for heterologous human adult liver-derived progenitor cell (HHALPC) infusion, in order to eliminate acute thrombogenesis in liver-based metabolic or acute decompensated cirrhotic (ADC) patients. In rats, thrombotic effects were absent when HHALPCs were infused at low cell dose (5 × 106 cells/kg), or at high cell dose (5 × 107 cells/kg) when combined with anticoagulants. When HHALPCs were exposed to human blood in a whole blood perfusion assay, blocking of the tissue factor (TF) coagulation pathway suppressed fibrin generation and platelet activation. In a Chandler tubing loop model, HHALPCs induced less explosive activation of coagulation with blood from ADC patients, when compared to blood from healthy controls, without alterations in coagulation factor levels other than fibrinogen. These studies confirm a link between TF and thrombogenesis, when TF-expressing cells are exposed to human blood. This phenomenon however, could be controlled using either a low, or a high cell dose combined with anticoagulants. In clinical practice, this points to the suitability of a low HHALPC dose infusion to cirrhotic patients, provided that platelet and fibrinogen levels are monitored.


Asunto(s)
Anticoagulantes/farmacología , Coagulación Sanguínea/efectos de los fármacos , Hígado/citología , Trasplante de Células Madre/métodos , Células Madre/inmunología , Trombosis/prevención & control , Adulto , Animales , Plaquetas/metabolismo , Fibrina/metabolismo , Humanos , Masculino , Ratas , Ratas Wistar , Tromboplastina/metabolismo
7.
Transplantation ; 103(9): 1903-1915, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-30801523

RESUMEN

BACKGROUND: Regenerative medicine using stem cell technology is an emerging field that is currently tested for inborn and acquired liver diseases. OBJECTIVE: This phase I/II prospective, open label, multicenter, randomized trial aimed primarily at evaluating the safety of Heterologous Human Adult Liver-derived Progenitor Cells (HepaStem) in pediatric patients with urea cycle disorders (UCDs) or Crigler-Najjar (CN) syndrome 6 months posttransplantation. The secondary objective included the assessment of safety up to 12 months postinfusion and of preliminary efficacy. METHODS: Fourteen patients with UCDs and 6 with CN syndrome were divided into 3 cohorts by body weight and intraportally infused with 3 doses of HepaStem. Clinical status, portal vein hemodynamics, morphology of the liver, de novo detection of circulating anti-human leukocyte antigen antibodies, and clinically significant adverse events (AEs) and serious adverse events to infusion were evaluated by using an intent-to-treat analysis. RESULTS: The overall safety of HepaStem was confirmed. For the entire study period, patient-month incidence rate was 1.76 for the AEs and 0.21 for the serious adverse events, of which 38% occurred within 1 month postinfusion. There was a trend of higher events in UCD as compared with CN patients. Segmental left portal vein thrombosis occurred in 1 patient and intraluminal local transient thrombus in a second patient. The other AEs were in line with expectations for catheter placement, cell infusion, concomitant medications, age, and underlying diseases. CONCLUSIONS: This study led to European clinical trial authorization for a phase II study in a homogeneous patient cohort, with repeated infusions and intermediate doses.


Asunto(s)
Síndrome de Crigler-Najjar/tratamiento farmacológico , Trasplante de Hígado , Hígado/metabolismo , Trasplante de Células Madre , Trastornos Innatos del Ciclo de la Urea/cirugía , Adolescente , Factores de Edad , Niño , Preescolar , Síndrome de Crigler-Najjar/sangre , Síndrome de Crigler-Najjar/diagnóstico , Síndrome de Crigler-Najjar/fisiopatología , Europa (Continente) , Femenino , Humanos , Lactante , Hígado/patología , Hígado/fisiopatología , Regeneración Hepática , Trasplante de Hígado/efectos adversos , Masculino , Estudios Prospectivos , Trasplante de Células Madre/efectos adversos , Factores de Tiempo , Trasplante Heterólogo , Resultado del Tratamiento , Trastornos Innatos del Ciclo de la Urea/sangre , Trastornos Innatos del Ciclo de la Urea/diagnóstico , Trastornos Innatos del Ciclo de la Urea/fisiopatología
8.
Diabetes ; 54(2): 402-11, 2005 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-15677498

RESUMEN

Hyperplasia of adipose tissue is critical for the development of obesity, but molecular mechanisms governing normal or pathological recruitment of new adipocytes remain unclear. The extracellular signal-regulated kinase (ERK) pathway plays a pivotal role in many essential cellular functions, such as proliferation and differentiation. Using ERK1(-/-) mice, we investigated the role of this isoform in adipose tissue development. Mice lacking ERK1 have decreased adiposity and fewer adipocytes than wild-type animals. Furthermore, ERK1(-/-) mice challenged with high-fat diet are resistant to obesity, are protected from insulin resistance, and have a higher postprandial metabolic rate. To get insights into cellular mechanisms implicated in reduced adiposity in ERK1(-/-) animals, we analyzed adipocyte differentiation in ERK1(-/-) cells. Compared with wild-type control cells, mouse embryo fibroblasts and cultures of adult preadipocytes isolated from ERK1(-/-) adult animals exhibit impaired adipogenesis. An inhibitor of the ERK pathway does not affect the residual adipogenesis of the ERK1(-/-) cells, suggesting that ERK2 is not implicated in adipocyte differentiation. Our results clearly link ERK1 to the regulation of adipocyte differentiation, adiposity, and high-fat diet-induced obesity. This suggests that a therapeutic approach of obesity targeting specifically the ERK1 isoform and not ERK2 would be of particular interest.


Asunto(s)
Adipocitos/fisiología , Tejido Adiposo/fisiología , Grasas de la Dieta/farmacología , Metabolismo Energético/fisiología , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Adipocitos/citología , Tejido Adiposo/embriología , Animales , Glucemia/efectos de los fármacos , Glucemia/metabolismo , Diferenciación Celular , Cruzamientos Genéticos , Embrión de Mamíferos , Prueba de Tolerancia a la Glucosa , Insulina/farmacología , Isoenzimas/deficiencia , Isoenzimas/genética , Isoenzimas/metabolismo , Sistema de Señalización de MAP Quinasas/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína Quinasa 3 Activada por Mitógenos/deficiencia , Proteína Quinasa 3 Activada por Mitógenos/genética , Actividad Motora , Células Madre/citología , Células Madre/fisiología
9.
Invest Ophthalmol Vis Sci ; 56(11): 6456-66, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26447979

RESUMEN

PURPOSE: To evaluate the therapeutic potential of Col-Treg, a collagen II-specific type 1 regulatory T-cell immunotherapy for the treatment of noninfectious uveitis (NIU). METHODS: Col-Treg cells were produced from collagen II-specific T cell receptor (TCR) transgenic mice or peripheral blood of healthy donors. Phenotypic characterization was performed by flow cytometry, and cytokine secretion was evaluated with Flowcytomix or ELISA. In vitro functional characterization included ATP hydrolysis, cytotoxicity, and contact-independent T-cell suppression and plasticity assays. Col-Treg migration was assessed by quantitative PCR specific to Col-Treg TCR. Col-Treg cells were administered intravenously in mice displaying experimental autoimmune uveitis (EAU) induced by interphotoreceptor retinoid-binding protein (IRBP) immunizations. Efficacy of Col-Treg was assessed by ophthalmology, histology, and immunohistochemistry. RESULTS: Mice Col-Treg cells displayed identity features of type 1 Treg cells with expression of CD25, FoxP3, low surface expression of CD127, and cytokine secretion profile (IL-10(high), IL-4(low), IFN-γ(int)). In vitro functional assays demonstrated Col-Treg suppressive capacity via soluble factor-dependent immunosuppression, cytotoxicity, and ATP hydrolysis. Col-Treg cells expressed granzyme B, CD39, and glucocorticoid-induced TNF-related protein (GITR). Administration of Col-Treg in EAU mice inhibited clinical and morphologic signs of uveitis and decreased ocular leukocyte infiltration. Col-Treg cells homed in the ocular tissues 24 hours after intravenous injection. Human Col-Treg cells were comparable to mice Col-Treg cells in identity and function and did not show the capacity to differentiate into Th17 cells in vitro. CONCLUSIONS: These results demonstrate the therapeutic potential of Col-Treg cells as a targeted approach for the treatment of NIU and the feasibility of translating this approach to the human clinical setting.


Asunto(s)
Colágeno Tipo II/administración & dosificación , Inmunidad Celular , Inmunoterapia/métodos , Linfocitos T Reguladores/inmunología , Uveítis/terapia , Animales , Colágeno Tipo II/inmunología , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Inyecciones Intravenosas , Ratones , Ratones Endogámicos DBA , Ratones Transgénicos , Uveítis/inmunología , Uveítis/patología
10.
Arthritis Res Ther ; 16(3): R115, 2014 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-24886976

RESUMEN

INTRODUCTION: Regulatory T (Treg) cells play a crucial role in preventing autoimmune diseases and are an ideal target for the development of therapies designed to suppress inflammation in an antigen-specific manner. Type 1 regulatory T (Tr1) cells are defined by their capacity to produce high levels of interleukin 10 (IL-10), which contributes to their ability to suppress pathological immune responses in several settings. The aim of this study was to evaluate the therapeutic potential of collagen type II-specific Tr1 (Col-Treg) cells in two models of rheumatoid arthritis (RA) in mice. METHODS: Col-Treg clones were isolated and expanded from collagen-specific TCR transgenic mice. Their cytokine secretion profile and phenotype characterization were studied. The therapeutic potential of Col-Treg cells was evaluated after adoptive transfer in collagen-antibody- and collagen-induced arthritis models. The in vivo suppressive mechanism of Col-Treg clones on effector T-cell proliferation was also investigated. RESULTS: Col-Treg clones are characterized by their specific cytokine profile (IL-10(high)IL-4(neg)IFN-γ(int)) and mediate contact-independent immune suppression. They also share with natural Tregs high expression of GITR, CD39 and granzyme B. A single infusion of Col-Treg cells reduced the incidence and clinical symptoms of arthritis in both preventive and curative settings, with a significant impact on collagen type II antibodies. Importantly, injection of antigen-specific Tr1 cells decreased the proliferation of antigen-specific effector T cells in vivo significantly. CONCLUSIONS: Our results demonstrate the therapeutic potential of Col-Treg cells in two models of RA, providing evidence that Col-Treg could be an efficient cell-based therapy for RA patients whose disease is refractory to current treatments.


Asunto(s)
Traslado Adoptivo/métodos , Artritis Experimental/inmunología , Artritis Experimental/terapia , Colágeno Tipo II/inmunología , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/trasplante , Animales , Artritis Reumatoide/inmunología , Artritis Reumatoide/terapia , Proliferación Celular , Células Cultivadas , Citometría de Flujo , Humanos , Inmunofenotipificación , Interferón gamma/inmunología , Interferón gamma/metabolismo , Interleucina-10/inmunología , Interleucina-10/metabolismo , Interleucina-4/inmunología , Interleucina-4/metabolismo , Ratones Congénicos , Ratones Endogámicos BALB C , Ratones Endogámicos DBA , Ratones Transgénicos , Ovalbúmina/inmunología , Fragmentos de Péptidos/inmunología , Linfocitos T Reguladores/metabolismo , Resultado del Tratamiento
11.
Blood ; 106(2): 419-27, 2005 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-15784733

RESUMEN

Bone marrow-derived mesenchymal stem cells (BM-MSCs) are stromal cells with the ability to proliferate and differentiate into many tissues. Although they represent powerful tools for several therapeutic settings, mechanisms regulating their migration to peripheral tissues are still unknown. Here, we report chemokine receptor expression on human BM-MSCs and their role in mediating migration to tissues. A minority of BM-MSCs (2% to 25%) expressed a restricted set of chemokine receptors (CXC receptor 4 [CXCR4], CX3C receptor 1 [CX3CR1], CXCR6, CC chemokine receptor 1 [CCR1], CCR7) and, accordingly, showed appreciable chemotactic migration in response to the chemokines CXC ligand 12 (CXCL12), CX3CL1, CXCL16, CC chemokine ligand 3 (CCL3), and CCL19. Using human pancreatic islets as an in vitro model of peripheral tissue, we showed that islet supernatants released factors able to attract BM-MSCs in vitro, and this attraction was principally mediated by CX3CL1 and CXCL12. Moreover, cells with features of BM-MSCs were detected within the pancreatic islets of mice injected with green fluorescent protein (GFP)-positive BM. A population of bona fide MSCs that also expressed CXCR4, CXCR6, CCR1, and CCR7 could be isolated from normal adult human pancreas. This study defines the chemokine receptor repertoire of human BM-MSCs that determines their migratory activity. Modulation of homing capacity may be instrumental for harnessing the therapeutic potential of BM-MSCs.


Asunto(s)
Células de la Médula Ósea/citología , Células de la Médula Ósea/fisiología , Islotes Pancreáticos/citología , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/fisiología , Receptores de Quimiocina/genética , Receptores de Quimiocina/fisiología , Animales , Secuencia de Bases , Separación Celular , Células Cultivadas , Quimiocinas/genética , Quimiocinas/fisiología , Quimiotaxis/fisiología , ADN/genética , Expresión Génica , Humanos , Técnicas In Vitro , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda