Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Mol Ther ; 2024 Sep 06.
Artículo en Inglés | MEDLINE | ID: mdl-39244642

RESUMEN

Chimeric antigen receptor (CAR) T-cell therapy showed preliminary activity in patients with refractory or relapsed T-cell acute lymphoblastic leukemia (r/r T-ALL). However, many obstacles remain, including manufacturing difficulties and risk of infections. This phase I study (NCT04840875) evaluated autologous CD7 CAR T cells manufactured without pre-selection of healthy T cells in r/r T-ALL. Thirty patients (29 children and one adult) with a median of two lines of prior therapy but without detectable peripheral leukemia were enrolled. Excluding three cases of manufacturing failures, a total of 27 (90%) patients received infusions after products were confirmed free of leukemia contamination, including 16 (59%) meeting planned target doses. Common adverse events within 30 days included grade 3-4 cytopenias (100%), grade 1-2 (70%) and 3-4 (7%; including one dose-limiting toxicity) cytokine release syndrome, grade 1 neurotoxicity (7%), grade 2 infection (4%), and grade 2 graft-versus-host disease (4%). Two patients developed grade 2 infections after day 30. At day 30, 96% responded and 85% achieved CR or CRi. 74% underwent transplantation. Twelve-month progression-free survival with and without censoring transplantation was 22% (95%CI 4-100) and 57% (41-81), respectively. These results support that autologous CD7 CAR T-cell therapy without T-cell pre-selection is feasible in patients with r/r T-ALL.

2.
Mol Ther ; 32(6): 1875-1894, 2024 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-38549378

RESUMEN

Chimeric antigen receptor-T (CAR-T) cell has been developed as a promising agent for patients with refractory or relapsed lymphoma and leukemia, but not all the recipients could achieve a long-lasting remission. The limited capacity of in vivo expansion and memory differentiation post activation is one of the major reasons for suboptimal CAR-T therapeutic efficiency. Nitric oxide (NO) plays multifaceted roles in mitochondrial dynamics and T cell activation, but its function on CAR-T cell persistence and anti-tumor efficacy remains unknown. Herein, we found the continuous signaling from CAR not only promotes excessive NO production, but also suppressed S-nitrosoglutathione reductase (GSNOR) expression in T cells, which collectively led to increased protein S-nitrosylation, resulting in impaired mitochondrial fitness and deficiency of T cell stemness. Intriguingly, enforced expression of GSNOR promoted memory differentiation of CAR-T cell after immune activation, rendered CAR-T better resistance to mitochondrial dysfunction, further enhanced CAR-T cell expansion and anti-tumor capacity in vitro and in a mouse tumor model. Thus, we revealed a critical role of NO in restricting CAR-T cell persistence and functionality, and defined that GSNOR overexpression may provide a solution to combat NO stress and render patients with more durable protection from CAR-T therapy.


Asunto(s)
Inmunoterapia Adoptiva , Mitocondrias , Receptores Quiméricos de Antígenos , Animales , Ratones , Mitocondrias/metabolismo , Receptores Quiméricos de Antígenos/metabolismo , Receptores Quiméricos de Antígenos/genética , Humanos , Inmunoterapia Adoptiva/métodos , Aldehído Oxidorreductasas/genética , Aldehído Oxidorreductasas/metabolismo , Linfocitos T/metabolismo , Linfocitos T/inmunología , Óxido Nítrico/metabolismo , Línea Celular Tumoral , Ensayos Antitumor por Modelo de Xenoinjerto , Activación de Linfocitos , Transducción de Señal
3.
Lancet Oncol ; 24(11): 1229-1241, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37863088

RESUMEN

BACKGROUND: Relapses frequently occur following CD19-directed chimeric antigen receptor (CAR) T-cell treatment for relapsed or refractory B-cell acute lymphocytic leukaemia in children. We aimed to assess the activity and safety of sequential CD19-directed and CD22-directed CAR T-cell treatments. METHODS: This single-centre, single-arm, phase 2 trial, done at Beijing GoBroad Boren Hospital, Beijing, China, included patients aged 1-18 years who had relapsed or refractory B-cell acute lymphocytic leukaemia with CD19 and CD22 positivity greater than 95% and an Eastern Cooperative Oncology Group performance status of 0-2. Patients were initially infused with CD19-directed CAR T cells intravenously, followed by CD22-directed CAR T-cell infusion after minimal residual disease-negative complete remission (or complete remission with incomplete haematological recovery) was reached and all adverse events (except haematological adverse events) were grade 2 or better. The target dose for each infusion was 0·5 × 106 to 5·0 × 106 cells per kg. The primary endpoint was objective response rate at 3 months after the first infusion. Secondary endpoints were duration of remission, event-free survival, disease-free survival, overall survival, safety, pharmacokinetics, and B-cell quantification. The prespecified activity analysis included patients who received the target dose and the safety analysis included all treated patients. This study is registered with ClinicalTrials.gov, NCT04340154, and enrolment has ended. FINDINGS: Between May 28, 2020, and Aug 16, 2022, 81 participants were enrolled, of whom 31 (38%) were female and 50 (62%) were male. Median age was 8 years (IQR 6-10), all patients were Asian. All 81 patients received the first infusion and 79 (98%) patients received sequential infusions, CD19-directed CAR T cells at a median dose of 2·7 × 106 per kg (IQR 1·1 × 106 to 3·7 × 106) and CD22-directed CAR T cells at a median dose of 2·2 × 106 per kg (1·1 × 106 to 3·7 × 106), with a median interval of 39 days (37-41) between the two infusions. 62 (77%) patients received the target dose, including two patients who did not receive CD22 CAR T cells. At 3 months, 60 (97%, 95% CI 89-100) of the 62 patients who received the target dose had an objective response. Median follow-up was 17·7 months (IQR 11·4-20·9). 18-month event-free survival for patients who received the target dose was 79% (95% CI 66-91), duration of remission was 80% (68-92), and disease-free survival was 80% (68-92) with transplantation censoring; overall survival was 96% (91-100). Common adverse events of grade 3 or 4 between CD19-directed CAR T-cell infusion and 30 days after CD22-directed CAR T-cell infusion included cytopenias (64 [79%] of 81 patients), cytokine release syndrome (15 [19%]), neurotoxicity (four [5%]), and infections (five [6%]). Non-haematological adverse events of grade 3 or worse more than 30 days after CD22-directed CAR T-cell infusion occurred in six (8%) of 79 patients. No treatment-related deaths occurred. CAR T-cell expansion was observed in all patients, with a median peak at 9 days (IQR 7-14) after CD19-directed and 12 days (10-15) after CD22-directed CAR T-cell infusion. At data cutoff, 35 (45%) of 77 evaluable patients had CAR transgenes and 59 (77%) had B-cell aplasia. INTERPRETATION: This sequential strategy induced deep and sustained responses with an acceptable toxicity profile, and thus potentially provides long-term benefits for children with this condition. FUNDING: The National Key Research & Development Program of China, the CAMS Innovation Fund for Medical Sciences (CIFMS), and the Non-Profit Central Research Institute Fund of Chinese Academy of Medical Sciences. TRANSLATION: For the Chinese translation of the abstract see Supplementary Materials section.


Asunto(s)
Leucemia Linfocítica Crónica de Células B , Leucemia-Linfoma Linfoblástico de Células Precursoras , Receptores Quiméricos de Antígenos , Humanos , Masculino , Niño , Femenino , Receptores Quiméricos de Antígenos/uso terapéutico , Recurrencia Local de Neoplasia/tratamiento farmacológico , Leucemia Linfocítica Crónica de Células B/tratamiento farmacológico , Inmunoterapia Adoptiva/efectos adversos , Leucemia-Linfoma Linfoblástico de Células Precursoras/terapia , Tratamiento Basado en Trasplante de Células y Tejidos , Lectina 2 Similar a Ig de Unión al Ácido Siálico/uso terapéutico
4.
Cell Mol Life Sci ; 80(3): 63, 2023 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-36781473

RESUMEN

Adipose tissue CD11c+ myeloid cell is an independent risk factor associated with obesity and metabolic disorders. However, the underlying molecular basis remains elusive. Here, we demonstrated that liver kinase B1 (Lkb1), a key bioenergetic sensor, is involved in CD11c+ cell-mediated immune responses in diet-induced obesity. Loss of Lkb1 in CD11c+ cells results in obesity resistance but lower glucose tolerance, which accompanies tissue-specific immune abnormalities. The accumulation and CD80's expression of Lkb1 deficient adipose-tissue specific dendritic cells but not macrophages is restrained. Additionally, the balance of IL-17A and IFN-γ remarkably tips towards the latter in fat T cells and CD11c- macrophages. Mechanistically, IFN-γ promotes apoptosis of preadipocytes and inhibits their adipogenesis while IL-17A promotes the adipogenesis in vitro, which might account in part for the fat gain resistant phenotype. In summary, these findings reveal that Lkb1 is essential for fat CD11c+ dendritic cells responding to HFD exposure and provides new insights into the IL-17A/IFN-γ balance in HFD-induced obesity.


Asunto(s)
Proteínas Quinasas Activadas por AMP , Intolerancia a la Glucosa , Resistencia a la Insulina , Obesidad , Animales , Ratones , Tejido Adiposo/metabolismo , Dieta Alta en Grasa/efectos adversos , Intolerancia a la Glucosa/metabolismo , Inflamación/metabolismo , Interleucina-17/genética , Interleucina-17/metabolismo , Macrófagos/metabolismo , Ratones Endogámicos C57BL , Obesidad/complicaciones , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Quinasas Activadas por AMP/genética , Proteínas Quinasas Activadas por AMP/metabolismo , Interferón gamma/metabolismo
5.
Nat Commun ; 12(1): 3913, 2021 06 23.
Artículo en Inglés | MEDLINE | ID: mdl-34162888

RESUMEN

Human FOXP3+ regulatory T (Treg) cells are central to immune tolerance. However, their heterogeneity and differentiation remain incompletely understood. Here we use single-cell RNA and T cell receptor sequencing to resolve Treg cells from healthy individuals and patients with or without acute graft-versus-host disease (aGVHD) who undergo stem cell transplantation. These analyses, combined with functional assays, separate Treg cells into naïve, activated, and effector stages, and resolve the HLA-DRhi, LIMS1hi, highly suppressive FOXP3hi, and highly proliferative MKI67hi effector subsets. Trajectory analysis assembles Treg subsets into two differentiation paths (I/II) with distinctive phenotypic and functional programs, ending with the FOXP3hi and MKI67hi subsets, respectively. Transcription factors FOXP3 and SUB1 contribute to some Path I and Path II phenotypes, respectively. These FOXP3hi and MKI67hi subsets and two differentiation pathways are conserved in transplanted patients, despite having functional and migratory impairments under aGVHD. These findings expand the understanding of Treg cell heterogeneity and differentiation and provide a single-cell atlas for the dissection of Treg complexity in health and disease.


Asunto(s)
Diferenciación Celular/genética , Factores de Transcripción Forkhead/inmunología , Transducción de Señal/genética , Análisis de la Célula Individual/métodos , Linfocitos T Reguladores/inmunología , Transcriptoma/genética , Western Blotting , Ensayo de Inmunoadsorción Enzimática , Factores de Transcripción Forkhead/metabolismo , Enfermedad Injerto contra Huésped/etiología , Enfermedad Injerto contra Huésped/inmunología , Enfermedad Injerto contra Huésped/metabolismo , Trasplante de Células Madre Hematopoyéticas/métodos , Humanos , RNA-Seq/métodos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Linfocitos T Reguladores/metabolismo
6.
Exp Cell Res ; 392(2): 112003, 2020 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-32278689

RESUMEN

Dendritic cells (DCs) play a central role in autoimmunity, immune homeostasis, and presentation of tumor antigens to T cells in order to prime antitumor responses. The number of tumor-infiltrating DCs is associated with survival and prognosis in cancer. Twist1 is a well-known regulator of tumor initiation and promotion, but whether and how DC-derived Twist1 regulates antitumor responses remains poorly understood. Here, we generated a mouse line with Twist1 conditionally depleted in DCs and found that Twist1-deficiency in DCs did not affect the DCs and T cell homeostasis under steady-state conditions; however, in melanoma models, the proportion of conventional DCs (cDCs) in draining lymph nodes (DLNs) was significantly decreased. Accordingly, a decreased ratio and number of tumor-infiltrating cDCs were observed, which reduced the recruitment of tumor-infiltrating T cells. Furthermore, production of IFN-γ, a crucial antitumor factor, by T cells, was dramatically decreased, which can further dampen the T cell antitumor functions. Collectively, our data indicate that Twist1 in DCs regulates antitumor functions by maintain the number of tumor-infiltrating DCs and T cells, and their antitumor activity.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Células Dendríticas/inmunología , Inmunidad Celular/inmunología , Linfocitos Infiltrantes de Tumor/inmunología , Melanoma Experimental/inmunología , Proteína 1 Relacionada con Twist/fisiología , Animales , Antígenos de Neoplasias/inmunología , Células Dendríticas/metabolismo , Células Dendríticas/patología , Melanoma Experimental/metabolismo , Melanoma Experimental/patología , Ratones , Ratones Noqueados
7.
Cell Mol Immunol ; 17(5): 483-495, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-31664223

RESUMEN

Accumulating evidence suggests that a reduction in the number of Foxp3+ regulatory T cells (Tregs) contributes to the pathogenesis of acute graft-versus-host disease (aGVHD), which is a major adverse complication that can occur after allogeneic hematopoietic stem cell transplantation (allo-HSCT). However, the precise features and mechanism underlying the defects in Tregs remain largely unknown. In this study, we demonstrated that Tregs were more dramatically decreased in bone marrow compared with those in peripheral blood from aGVHD patients and that bone marrow Treg defects were negatively associated with hematopoietic reconstitution. Tregs from aGVHD patients exhibited multiple defects, including the instability of Foxp3 expression, especially in response to IL-12, impaired suppressor function, decreased migratory capacity, and increased apoptosis. Transcriptional profiling revealed the downregulation of Lkb1, a previously identified critical regulator of murine Treg identity and metabolism, and murine Lkb1-regulated genes in Tregs from aGVHD patients. Foxp3 expression in human Tregs could be decreased and increased by the knockdown and overexpression of the Lkb1 gene, respectively. Furthermore, a loss-of-function assay in an aGVHD murine model confirmed that Lkb1 deficiency could impair Tregs and aggravate disease severity. These findings reveal that Lkb1 downregulation contributes to multiple defects in Tregs in human aGVHD and highlight the Lkb1-related pathways that could serve as therapeutic targets that may potentially be manipulated to mitigate aGVHD.


Asunto(s)
Trasplante de Médula Ósea/efectos adversos , Enfermedad Injerto contra Huésped/etiología , Enfermedad Injerto contra Huésped/inmunología , Proteínas Serina-Treonina Quinasas/metabolismo , Linfocitos T Reguladores/inmunología , Quinasas de la Proteína-Quinasa Activada por el AMP , Proteínas Quinasas Activadas por AMP , Adolescente , Adulto , Animales , Médula Ósea/efectos de los fármacos , Médula Ósea/metabolismo , Niño , Modelos Animales de Enfermedad , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/genética , Femenino , Factores de Transcripción Forkhead/metabolismo , Eliminación de Gen , Perfilación de la Expresión Génica , Enfermedad Injerto contra Huésped/sangre , Humanos , Masculino , Metilación/efectos de los fármacos , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Persona de Mediana Edad , Linfocitos T Reguladores/efectos de los fármacos , Transcripción Genética/efectos de los fármacos
8.
Biomed Pharmacother ; 120: 109352, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-31586905

RESUMEN

Inflammatory monocyte and macrophage subset accumulation during the inflammatory response that drives atherosclerosis can exacerbate the extent of atherosclerosis. It has been demonstrated that voltage-gated sodium channels (VGSCs) can regulate cell bioactivities in monocytes/macrophages. We hypothesized that blockade of mononuclear phagocyte VGSCs was atheroprotective through monocyte/macrophage subset modulation and macrophage proliferation suppression in atherosclerotic lesions. In this experimental study, when VGSCs were knocked down with RNA interference plasmid transfection in mouse peripheral blood monocytes and monocyte-macrophage lineage RAW264.7 cells in vitro, the biological characteristics of proliferation, phagocytosis, and migration in RAW264.7 cells declined. In addition, suppression of LPS-induced M1 polarization and facilitation of IL-4-induced M2 polarization were also observed. In an in vivo study, ApoE knockout (ApoE-/-) mice were fed a standard chow diet (CD) or a western diet (WD). After feeding with phenytoin (PHT), no significant differences were detected in plasma lipids, and the anti-inflammatory phenotypes of both monocytes and macrophages were elevated and proinflammatory phenotypes declined. The local proliferation of macrophages was also distinctly suppressed, along with a significant reduction in atheromatous plaques. In conclusion, blockade of VGSCs in the mononuclear phagocyte system reduced atherosclerotic lesions, which may occur through altering monocyte/macrophage subsets and suppressing macrophage proliferation in atherosclerotic plaques. Blockage of VGSCs may play an important role in cardiovascular protection.


Asunto(s)
Aterosclerosis/prevención & control , Activación de Macrófagos , Macrófagos/metabolismo , Monocitos/metabolismo , Canal de Sodio Activado por Voltaje NAV1.4/metabolismo , Canal de Sodio Activado por Voltaje NAV1.9/metabolismo , Animales , Aterosclerosis/genética , Aterosclerosis/metabolismo , Aterosclerosis/patología , Proliferación Celular , Modelos Animales de Enfermedad , Regulación hacia Abajo , Macrófagos/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados para ApoE , Monocitos/patología , Canal de Sodio Activado por Voltaje NAV1.4/genética , Canal de Sodio Activado por Voltaje NAV1.9/genética , Fagocitosis , Placa Aterosclerótica , Células RAW 264.7 , Interferencia de ARN , Transducción de Señal
12.
Nat Commun ; 9(1): 5298, 2018 12 13.
Artículo en Inglés | MEDLINE | ID: mdl-30546010

RESUMEN

To balance immunity and tolerance, the endogenous pool of Foxp3+ regulatory T (Treg) cells is tightly controlled, but the underlying mechanisms of this control remain poorly understood. Here we show that the number of Treg cells is negatively regulated by the kinase Lkb1 in dendritic cells (DCs). Conditional knockout of the Lkb1 gene in DCs leads to excessive Treg cell expansion in multiple organs and dampens antigen-specific T cell immunity. Lkb1-deficient DCs are capable of enhancing, compared with wild-type DCs, Treg cell proliferation via cell-cell contact involving the IKK/IKBα-independent activation of the NF-κB/OX40L pathway. Intriguingly, treating wild-type mice with lipopolysaccharide selectively depletes Lkb1 protein in DCs, resulting in Treg cell expansion and suppressed inflammatory injury upon subsequent challenge. Loss of Lkb1 does not obviously upregulate proinflammatory molecules expression on DCs. We thus identify Lkb1 as a regulatory switch in DCs for controlling Treg cell homeostasis, immune response and tolerance.


Asunto(s)
Proliferación Celular/genética , Células Dendríticas/inmunología , Proteínas Serina-Treonina Quinasas/genética , Linfocitos T Reguladores/inmunología , Proteínas Quinasas Activadas por AMP , Animales , Apoptosis/inmunología , Técnicas de Inactivación de Genes , Homeostasis/fisiología , Quinasa I-kappa B/metabolismo , Activación de Linfocitos/inmunología , Glicoproteínas de Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones SCID , Ligando OX40 , Receptores OX40/metabolismo , Factores de Necrosis Tumoral/metabolismo
13.
Am J Transl Res ; 10(3): 901-906, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29636880

RESUMEN

This study reports a case of a 4-year-old boy patient with abnormalities of muscle tone, movement and motor skills, as well as unstable gait leading to frequent falls. The results of the electroencephalogram (EEG) indicate moderately abnormal EEG, accompanied by irregular seizures. Based on these clinical characteristics, the patient was diagnosed with cerebral palsy (CP) in our hospital. In this study, the patient was treated with umbilical cord mesenchymal stem cell (UC-MSC) transplantation therapy. This patient received UC-MSC transplantation 3 times (5.3*107) in total. After three successive cell transplantations, the patient recovered well and showed obvious improvements in EEG and limb strength, motor function, and language expression. However, the improvement in intelligence quotient (IQ) was less obvious. These results indicate that UC-MSC transplantation is a promising treatment for cerebral palsy.

15.
Exp Cell Res ; 363(1): 73-83, 2018 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-29294307

RESUMEN

Dendritic cells (DCs) are pivotal to initiating adaptive immune response. Emerging evidence highlights important roles of tuberous sclerosis complex 1 (Tsc1) in DC development and activation. Our previous study also showed that Tsc1 expression in DCs was required to promote T-cell homeostasis and response partially through inhibiting mammalian target of rapamycin complex1 (mTORC1). However, the molecular mechanism of transcriptional regulation by which Tsc1 control DC homeostasis and function remains largely unknown. Here we globally identified the Tsc1-regulated genes by comparing the transcriptional profiling of Tsc1-deficient DCs with wild-type DCs. It showed that Tsc1 specifically regulated the expression of groups of gene sets critically involved in DC survival, proliferation, metabolism and antigen presentation. The impacts of Tsc1 on DC gene expression were partially dependent on inhibition of mTORC1 signal. Our study thus provides a comprehensive molecular basis for understanding how Tsc1 programs the homeostasis and function of DCs through transcriptional regulation.


Asunto(s)
Células Dendríticas/citología , Homeostasis/fisiología , Diana Mecanicista del Complejo 1 de la Rapamicina/antagonistas & inhibidores , Proteínas Supresoras de Tumor/metabolismo , Animales , Presentación de Antígeno/inmunología , Células Dendríticas/inmunología , Regulación de la Expresión Génica/efectos de los fármacos , Homeostasis/efectos de los fármacos , Ratones Transgénicos , Complejos Multiproteicos/metabolismo , Proteína 1 del Complejo de la Esclerosis Tuberosa , Proteínas Supresoras de Tumor/genética
17.
J Craniofac Surg ; 28(6): 1615-1619, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28863113

RESUMEN

The authors aim to track the distribution of human umbilical cord mesenchymal stem cells (MSCs) in large blood vessel of traumatic brain injury -rats through immunohistochemical method and small animal imaging system. After green fluorescent protein (GFP) gene was transfected into 293T cell, virus was packaged and MSCs were transfected. Mesenchymal stem cells containing GFP were transplanted into brain ventricle of rats when the infection rate reaches 95%. The immunohistochemical and small animal imaging system was used to detect the distribution of MSCs in large blood vessels of rats. Mesenchymal stem cells could be observed in large vessels with positive GFP expression 10 days after transplantation, while control groups (normal group and traumatic brain injury group) have negative GFP expression. The vascular endothelial growth factor in transplantation group was higher than that in control groups. The in vivo imaging showed obvious distribution of MSCs in the blood vessels of rats, while no MSCs could be seen in control groups. The intravascular migration and homing of MSCs could be seen in rats received MSCs transplantation, and new angiogenesis could be seen in MSCs-transplanted blood vessels.


Asunto(s)
Lesiones Traumáticas del Encéfalo/terapia , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas , Cordón Umbilical/citología , Animales , Vasos Sanguíneos/química , Vasos Sanguíneos/citología , Vasos Sanguíneos/metabolismo , Proteínas Fluorescentes Verdes/química , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Células Madre Mesenquimatosas/química , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Ratas , Proyectos de Investigación
18.
Nat Commun ; 8: 15876, 2017 06 16.
Artículo en Inglés | MEDLINE | ID: mdl-28621313

RESUMEN

Regulatory T (Treg) cells are a distinct T-cell lineage characterized by sustained Foxp3 expression and potent suppressor function, but the upstream dominant factors that preserve Treg lineage-specific features are mostly unknown. Here, we show that Lkb1 maintains Treg cell lineage identity by stabilizing Foxp3 expression and enforcing suppressor function. Upon T-cell receptor (TCR) stimulation Lkb1 protein expression is upregulated in Treg cells but not in conventional T cells. Mice with Treg cell-specific deletion of Lkb1 develop a fatal early-onset autoimmune disease, with no Foxp3 expression in most Treg cells. Lkb1 stabilizes Foxp3 expression by preventing STAT4-mediated methylation of the conserved noncoding sequence 2 (CNS2) in the Foxp3 locus. Independent of maintaining Foxp3 expression, Lkb1 programs the expression of a wide spectrum of immunosuppressive genes, through mechanisms involving the augmentation of TGF-ß signalling. These findings identify a critical function of Lkb1 in maintaining Treg cell lineage identity.


Asunto(s)
Proteínas Serina-Treonina Quinasas/metabolismo , Linfocitos T Reguladores/citología , Proteínas Quinasas Activadas por AMP/metabolismo , Animales , Enfermedades Autoinmunes/genética , Linaje de la Célula , Metilación de ADN , Femenino , Factores de Transcripción Forkhead/genética , Regulación de la Expresión Génica , Ratones Endogámicos C57BL , Ratones Transgénicos , FN-kappa B/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Receptores de Antígenos de Linfocitos T/metabolismo , Factor de Transcripción STAT4/genética , Factor de Transcripción STAT4/metabolismo , Linfocitos T Reguladores/fisiología
19.
Cell Death Dis ; 8(1): e2553, 2017 01 12.
Artículo en Inglés | MEDLINE | ID: mdl-28079897

RESUMEN

Dendritic cells (DCs) are pivotal to the induction of adaptive T-cell immune responses. Recent evidence highlights a critical role of tuberous sclerosis complex 1 (Tsc1), a primarily upstream negative regulator of mammalian target of rapamycin (mTOR), in DC development, but whether and how Tsc1 directly regulate mature DC function in vivo remains elusive. Here we show that selective disruption of Tsc1 in DCs results in a lymphoproliferative disorder with the spontaneous activation of T cells. Tsc1 deficiency results in the activation of mTORC1-PPARγ pathway, which leads to the upregulation of neuropilin-1 (Nrp1) expression on DCs to stimulate naive T-cell proliferation. However, Tsc1-deficient DCs have defects in the ability to induce antigen-specific T-cell responses in vitro and in vivo owing to impaired survival during antigen transportation and presentation. Indeed, Tsc1 promotes DC survival through restraining independent mTORC1 and ROS-Bim pathways. Our study identifies Tsc1 as a crucial signaling checkpoint in DCs essential for preserving T-cell homeostasis and response.


Asunto(s)
Células Dendríticas/metabolismo , Trastornos Linfoproliferativos/genética , Linfocitos T/metabolismo , Proteínas Supresoras de Tumor/biosíntesis , Animales , Diferenciación Celular/genética , Células Dendríticas/inmunología , Células Dendríticas/patología , Homeostasis , Humanos , Trastornos Linfoproliferativos/inmunología , Trastornos Linfoproliferativos/patología , Diana Mecanicista del Complejo 1 de la Rapamicina , Ratones , Complejos Multiproteicos/genética , Transducción de Señal/genética , Linfocitos T/inmunología , Linfocitos T/patología , Serina-Treonina Quinasas TOR/biosíntesis , Serina-Treonina Quinasas TOR/genética , Proteína 1 del Complejo de la Esclerosis Tuberosa , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/inmunología
20.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 32(3): 289-93, 2016 Mar.
Artículo en Chino | MEDLINE | ID: mdl-26927543

RESUMEN

OBJECTIVE: To investigate the effect of blocking p38 mitogen-activated protein kinase (p38MAPK) pathway on the proliferation of OT-II cells mediated by splenic dendritic cells (DCs). METHODS: Splenic DCs of C57BL/6 mice were purified with anti-CD11c immunomagnetic beads, and OT-II cells were isolated from the splenic of CD4(+)-ovalbumin transgenic mice (OT-IItransgenic mice) by mouse CD4 T cell isolation kits. After being pretreated with SB203580, an inhibitor of p38MAPK, DCs were stimulated with lipopolysaccharides (LPS). Then the expression levels of co-stimulatory molecules (CD80, CD86) and MHCII in DCs, and antigen-presenting ability of DCs treated with the 52-68 fragment of the E alpha-chain of I-E class II molecules (Eα52-68 peptide) were detected by flow cytometry. The protein levels of tumor necrosis factor α (TNF-α), interleukin 1α (IL-1α), interleukin 6 (IL-6) and transforming growth factor ß (TGF-ß) in the culture supernatants were measured by ELISA. The proliferation of OT-II cells which were co-cultured with OVA323-339-treated DCs was analyzed by flow cytometry. RESULTS: The purity of both DCs and OT-II cells reached over 90% after isolation. SB203580 downregulated the expressions of CD80, CD86 and MHC II, and suppressed the antigen-presenting ability of DCs. The expressions of TNF-α, IL-1α and IL-6 were downregulated, while the expression of TGF-ß was raised. Finally, SB203580 inhibited DCs-mediated proliferation of OT-II cells. CONCLUSION: Blocking p38MAPK pathway with SB203580 could inhibit DCs-mediated proliferation of OT-II cells, which might be involved in modulating the expressions of CD80, CD86 and MHC II, the antigen-presenting ability, as well as the expressions of pro-inflammatory and anti-inflammatory cytokines.


Asunto(s)
Linfocitos T CD4-Positivos/metabolismo , Proliferación Celular/fisiología , Células Dendríticas/metabolismo , Sistema de Señalización de MAP Quinasas/fisiología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Animales , Presentación de Antígeno/efectos de los fármacos , Presentación de Antígeno/inmunología , Antígeno B7-1/metabolismo , Antígeno B7-2/metabolismo , Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/inmunología , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Técnicas de Cocultivo , Citocinas/metabolismo , Células Dendríticas/citología , Células Dendríticas/inmunología , Inhibidores Enzimáticos/farmacología , Ensayo de Inmunoadsorción Enzimática , Femenino , Citometría de Flujo , Antígenos de Histocompatibilidad Clase II/metabolismo , Imidazoles/farmacología , Mediadores de Inflamación/metabolismo , Lipopolisacáridos/farmacología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Ratones Endogámicos C57BL , Ratones Transgénicos , Fragmentos de Péptidos/farmacología , Piridinas/farmacología , Receptores de Antígenos de Linfocitos T , Bazo/citología , Bazo/inmunología , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA