Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Leukemia ; 37(11): 2261-2275, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37670087

RESUMEN

The highly conserved MicroRNA-9 (miR-9) family consists of three members. We discovered that miR-9-1 deletion reduced mature miR-9 expression, causing 43% of the mice to display smaller size and postweaning lethality. MiR-9-1-deficient mice with growth defects experienced severe lymphopenia, but other blood cells were unaffected. The lymphopenia wasn't due to defects in hematopoietic progenitors, as mutant bone marrow (BM) cells underwent normal lymphopoiesis after transplantation into wild-type recipients. Additionally, miR-9-1-deficient mice exhibited impaired osteoblastic bone formation, as mutant mesenchymal stem cells (MSCs) failed to differentiate into osteoblastic cells (OBs). RNA sequencing revealed reduced expression of master transcription factors for osteoblastic differentiation, Runt-related transcription factor 2 (Runx2) and Osterix (Osx), and genes related to collagen formation, extracellular matrix organization, and cell adhesion, in miR-9-1-deficient MSCs. Follistatin (Fst), an antagonist of bone morphogenetic proteins (BMPs), was found to be a direct target of miR-9-1. Its deficiency led to the up-regulation of Fst, inhibiting BMP signaling in MSCs, and reducing IL-7 and IGF-1. Thus, miR-9-1 controls osteoblastic regulation of lymphopoiesis by targeting the Fst/BMP/Smad signaling axis.


Asunto(s)
Linfopenia , MicroARNs , Animales , Ratones , Linfopoyesis/genética , Proteínas Morfogenéticas Óseas/metabolismo , Diferenciación Celular , MicroARNs/genética , MicroARNs/metabolismo , Osteogénesis/genética , Osteoblastos/metabolismo
2.
J Immunol ; 210(9): 1222-1235, 2023 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-36961449

RESUMEN

The caspase recruitment domain family member (CARD)11-Bcl10-Malt1 signalosome controls TGF-ß-activated kinase 1 (TAK1) activation and regulates BCR-induced NF-κB activation. In this study, we discovered that CARD19 interacted with TAK1 and inhibited TAB2-mediated TAK1 ubiquitination and activation. Although CARD19 deficiency in mice did not affect B cell development, it enhanced clonal deletion, receptor editing, and anergy of self-reactive B cells, and it reduced autoantibody production. Mechanistically, CARD19 deficiency increased BCR/TAK1-mediated NF-κB activation, leading to increased expression of transcription factors Egr2/3, as well as the E3 ubiquitin ligases c-Cbl/Cbl-b, which are known inducers of B cell tolerance in self-reactive B cells. RNA sequencing analysis revealed that although CARD19 deficiency did not affect the overall Ag-induced gene expression in naive B cells, it suppressed BCR signaling and increased hyporesponsiveness of self-reactive B cells. As a result, CARD19 deficiency prevented Bm12-induced experimental systemic lupus erythematosus. In summary, CARD19 negatively regulates BCR/TAK1-induced NF-κB activation and its deficiency increases Egr2/3 and c-Cbl/Cbl-b expression in self-reactive B cells, thereby enhancing B cell tolerance.


Asunto(s)
FN-kappa B , Transducción de Señal , Animales , Ratones , FN-kappa B/metabolismo , Transducción de Señal/fisiología , Quinasas Quinasa Quinasa PAM/metabolismo , Ubiquitinación
3.
FASEB J ; 37(4): e22862, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36906291

RESUMEN

The paraspeckle protein NONO is a multifunctional nuclear protein participating in the regulation of transcriptional regulation, mRNA splicing and DNA repair. However, whether NONO plays a role in lymphopoiesis is not known. In this study, we generated mice with global deletion of NONO and bone marrow (BM) chimeric mice in which NONO is deleted in all of mature B cells. We found that the global deletion of NONO in mice did not affect T-cell development but impaired early B-cell development in BM at pro- to pre-B-cell transition stage and B-cell maturation in the spleen. Studies of BM chimeric mice demonstrated that the impaired B-cell development in NONO-deficient mice is B-cell-intrinsic. NONO-deficient B cells displayed normal BCR-induced cell proliferation but increased BCR-induced cell apoptosis. Moreover, we found that NONO deficiency impaired BCR-induced activation of ERK, AKT, and NF-κB pathways in B cells, and altered BCR-induced gene expression profile. Thus, NONO plays a critical role in B-cell development and BCR-induced B-cell activation.


Asunto(s)
FN-kappa B , Transducción de Señal , Ratones , Animales , Ratones Noqueados , FN-kappa B/metabolismo , Linfocitos B/metabolismo , Receptores de Antígenos de Linfocitos B , Proteínas de Unión al ADN/metabolismo , Proteínas de Unión al ARN/metabolismo
4.
Blood ; 141(9): 1060-1069, 2023 03 02.
Artículo en Inglés | MEDLINE | ID: mdl-36493339

RESUMEN

Heparin-induced thrombocytopenia (HIT) is a serious adverse drug reaction characterized by antibodies that recognize platelet factor 4/heparin complexes (PF4/H) and activate platelets to create a prothrombotic state. Although a high percentage of heparin-treated patients produce antibodies to PF4/H, only a subset also makes antibodies that are platelet activating (PA). A close correlation between PA antibodies and the likelihood of experiencing HIT has been demonstrated in clinical studies, but how PA (presumptively pathogenic) and nonactivating (NA) (presumptively benign) antibodies differ from each other at the molecular level is unknown. To address this issue, we cloned 7 PA and 47 NA PF4/H-binding antibodies from 6 patients with HIT and characterized their structural and functional properties. Findings showed that PA clones differed significantly from NA clones in possessing 1 of 2 heavy chain complementarity-determining region 3 (HCDR3) motifs, RX1-2R/KX1-2R/H (RKH) and YYYYY (Y5), in an unusually long complementarity-determining region 3 (≥20 residues). Mutagenic studies showed that modification of either motif in PA clones reduced or abolished their PA activity and that appropriate amino acid substitutions in HCDR3 of NA clones can cause them to become PA. Repertoire sequencing showed that the frequency of peripheral blood IgG+ B cells possessing RKH or Y5 was significantly higher in patients with HIT than in patients without HIT given heparin, indicating expansion of B cells possessing RKH or Y5 in HIT. These findings imply that antibodies possessing RKH or Y5 are relevant to HIT pathogenesis and suggest new approaches to diagnosis and treatment of this condition.


Asunto(s)
Regiones Determinantes de Complementariedad , Trombocitopenia , Humanos , Regiones Determinantes de Complementariedad/genética , Trombocitopenia/inducido químicamente , Trombocitopenia/genética , Heparina , Anticuerpos/efectos adversos , Plaquetas/metabolismo , Factor Plaquetario 4
5.
Front Oncol ; 12: 956593, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36059640

RESUMEN

B7-H3 is over-expressed in multiple types of solid tumors, making it an ideal target for chimeric antigen receptor (CAR)-T therapy. Here, we first report a case of multiple basal cell carcinoma (BCC) patient treated with humanized monoclonal anti-B7-H3 CAR-T cells through direct intratumoral injection. After three dose-escalated injections, the lesion in the abdomen decreased by 40% in volume, shrank from bulging to flat, but was not eradicated completely. The large lesion in the forehead became dry from original ulcer and bleeding. The adverse events observed were itching, myalgia, and redness. Immunohistochemistry analysis demonstrated that B7-H3-positive tumor cells and B7-H3 expression intensity were reduced after injections of CAR-T cells. The number of infiltrating CD3 T cells increased significantly but mainly located outside the tumor region. Subsequently, high levels of TGF-ß in the tumor area were observed, suggesting that solid tumor microenvironment may hinder the infiltration and effect of CAR-T cells. In summary, in this particular case report, intratumoral injection of B7-H3 CAR-T cells partially controls tumor growth in the BCC patient with minor adverse events. The efficacy and safety of B7-H3 CAR-T therapy need to be further investigated with a larger cohort of patients. Although only one clinical case is reported here, the anti-B7-H3 CAR-T cell therapy should be considered as a treatment option for solid tumors in the future. This clinical trial was registered at the Chinese Clinical Trial Registry (www.chictr.org.cn) with registration number ChiCTR2100044386.

6.
Leukemia ; 36(11): 2596-2604, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36151140

RESUMEN

C-type lectin-like molecule-1 (CLL1) is preferentially expressed on acute myeloid leukemia (AML) stem cells and AML blasts, which can be considered as AML-associated antigen. Anti-CLL1-based CAR-T cells exhibited effective tumor-killing capacity in vitro and in AML-bearing mouse model. In this report, eight children with relapsed or refractory AML (R/R-AML) were recruited for a phase 1/2 clinical trial of autologous anti-CLL1 CAR-T cell immunotherapy. The objectives of this clinical trial were to evaluate the safety and the preliminary efficacy of anti-CLL1 CAR-T cell treatment. Patients received one dose of autologous anti-CLL1 CAR-T cells after lymphodepletion conditioning. After CAR-T treatment, patients developed grade 1-2 cytokine release syndrome (CRS) but without any lethal events. 4 out of 8 patients achieved morphologic leukemia-free state (MLFS) and minimal residual disease (MRD) negativity, 1 patient with MLFS and MRD positivity, 1 patient achieved complete remission with incomplete hematologic recovery (CRi) but MRD positivity, 1 patient with partial remission (PR), and 1 patient remained at stable disease (SD) status but had CLL1-positive AML blast clearance. These results suggested that anti-CLL1-based CAR-T cell immunotherapy can be considered as a well-tolerated and effective option for treating children with R/R-AML.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas , Leucemia Mieloide Aguda , Receptores Quiméricos de Antígenos , Ratones , Animales , Inmunoterapia Adoptiva/efectos adversos , Inmunoterapia Adoptiva/métodos , Leucemia Mieloide Aguda/tratamiento farmacológico , Lectinas Tipo C , Síndrome de Liberación de Citoquinas
7.
Leukemia ; 36(8): 2032-2041, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35778533

RESUMEN

Acute myeloid leukemia (AML) is driven by mutations that occur in numerous combinations. A better understanding of how mutations interact with one another to cause disease is critical to developing targeted therapies. Approximately 50% of patients that harbor a common mutation in NPM1 (NPM1cA) also have a mutation in the cohesin complex. As cohesin and Npm1 are known to regulate gene expression, we sought to determine how cohesin mutation alters the transcriptome in the context of NPM1cA. We utilized inducible Npm1cAflox/+ and core cohesin subunit Smc3flox/+ mice to examine AML development. While Npm1cA/+;Smc3Δ/+ mice developed AML with a similar latency and penetrance as Npm1cA/+ mice, RNA-seq suggests that the Npm1cA/+; Smc3Δ/+ mutational combination uniquely alters the transcriptome. We found that the Rac1/2 nucleotide exchange factor Dock1 was specifically upregulated in Npm1cA/+;Smc3Δ/+ HSPCs. Knockdown of Dock1 resulted in decreased growth and adhesion and increased apoptosis only in Npm1cA/+;Smc3Δ/+ AML. Higher Rac activity was also observed in Npm1cA/+;Smc3Δ/+ vs. Npm1cA/+ AMLs. Importantly, the Dock1/Rac pathway is targetable in Npm1cA/+;Smc3Δ/+ AMLs. Our results suggest that Dock1/Rac represents a potential target for the treatment of patients harboring NPM1cA and cohesin mutations and supports the use of combinatorial genetics to identify novel precision oncology targets.


Asunto(s)
Leucemia Mieloide Aguda , Proteínas Nucleares , Animales , Proteínas de Ciclo Celular , Proteínas Cromosómicas no Histona , Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Mieloide Aguda/genética , Ratones , Mutación , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Nucleofosmina , Medicina de Precisión , Factores de Transcripción/genética , Proteínas de Unión al GTP rac , Cohesinas , Proteína RCA2 de Unión a GTP
8.
Leukemia ; 36(4): 946-955, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35022500

RESUMEN

The Switch/Sugar Non-Fermenting (SWI/SNF) nucleosome remodeling complexes play important roles in normal development and in the development of various cancers. Core subunits of the SWI/SNF complexes have been shown to have oncogenic roles in acute myeloid leukemia. However, the roles of the unique targeting subunits, including that of Arid2 and Arid1b, in AML leukemogenesis are not well understood. Here, we used conditional knockout mouse models to elucidate their role in MLL-AF9 leukemogenesis. We uncovered that Arid2 has dual roles; enhancing leukemogenesis when deleted during leukemia initiation and yet is required during leukemia maintenance. Whereas, deleting Arid1b in either phase promotes leukemogenesis. Our integrated analyses of transcriptomics and genomic binding data showed that, globally, Arid2 and Arid1b regulate largely distinct sets of genes at different disease stages, respectively, and in comparison, to each other. Amongst the most highly dysregulated transcription factors upon their loss, Arid2 and Arid1b converged on the regulation of Etv4/Etv5, albeit in an opposing manner while also regulating distinct TFs including Gata2,Tcf4, Six4, Irf4 and Hmgn3. Our data demonstrate the differential roles of SWI/SNF subunits in AML leukemogenesis and emphasize that cellular context and disease stage are key in determining their functions during this process.


Asunto(s)
Leucemia , Factores de Transcripción , Animales , Carcinogénesis/genética , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Humanos , Leucemia/genética , Ratones , Ratones Noqueados , Proteína de la Leucemia Mieloide-Linfoide/genética , Proteínas de Fusión Oncogénica , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
9.
ACS Appl Mater Interfaces ; 14(1): 484-491, 2022 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-34962380

RESUMEN

The practical application of lithium-metal batteries (LMBs) is hindered by the lithium dendrite formation during cycling. In this work, we report a multilayered solid polymer electrolyte (SPE) formed by sandwiching a comb-chain cross-linker-based network SPE (ConSPE) film with a linear poly(ethylene oxide) (PEO) SPE coating. Benefiting from the drastically different lithium dendrite resisting properties of the ConSPE and linear PEO SPE, the lithium dendrite growth in the multilayered SPEs could be tuned, with the linear PEO SPE effectively serving as a sacrificial layer to accommodate the lithium dendrite growth. Symmetrical lithium cells with the multilayered SPE exhibited an extended short-circuit time ∼4.1 times that for the single-layer ConSPE at a high current density of 1.5 mA cm-2. Li/LiFePO4 batteries with multilayered SPEs delivered superior cycling performance at extremely high C-rates of 2C and 10C. Our multilayered SPE architecture, therefore, opens up a new gateway for advancing SPE design for future LMBs.

10.
Blood ; 138(23): 2408-2424, 2021 12 09.
Artículo en Inglés | MEDLINE | ID: mdl-34324649

RESUMEN

Immune thrombocytopenia (ITP) is a platelet disorder. Pediatric and adult ITP have been associated with sialic acid alterations, but the pathophysiology of ITP remains elusive, and ITP is often a diagnosis of exclusion. Our analysis of pediatric ITP plasma samples showed increased anti-Thomsen-Friedenreich antigen (TF antigen) antibody representation, suggesting increased exposure of the typically sialylated and cryptic TF antigen in these patients. The O-glycan sialyltransferase St3gal1 adds sialic acid specifically on the TF antigen. To understand if TF antigen exposure associates with thrombocytopenia, we generated a mouse model with targeted deletion of St3gal1 in megakaryocytes (MK) (St3gal1MK-/-). TF antigen exposure was restricted to MKs and resulted in thrombocytopenia. Deletion of Jak3 in St3gal1MK-/- mice normalized platelet counts implicating involvement of immune cells. Interferon-producing Siglec H-positive bone marrow (BM) immune cells engaged with O-glycan sialic acid moieties to regulate type I interferon secretion and platelet release (thrombopoiesis), as evidenced by partially normalized platelet count following inhibition of interferon and Siglec H receptors. Single-cell RNA-sequencing determined that TF antigen exposure by MKs primed St3gal1MK-/- BM immune cells to release type I interferon. Single-cell RNA-sequencing further revealed a new population of immune cells with a plasmacytoid dendritic cell-like signature and concomitant upregulation of the immunoglobulin rearrangement gene transcripts Igkc and Ighm, suggesting additional immune regulatory mechanisms. Thus, aberrant TF antigen moieties, often found in pathological conditions, regulate immune cells and thrombopoiesis in the BM, leading to reduced platelet count.


Asunto(s)
Megacariocitos/patología , Recuento de Plaquetas , Polisacáridos/análisis , Púrpura Trombocitopénica Idiopática/patología , Adolescente , Animales , Antígenos de Carbohidratos Asociados a Tumores/análisis , Niño , Preescolar , Humanos , Lactante , Ratones Endogámicos C57BL , Sialiltransferasas/análisis , beta-Galactosida alfa-2,3-Sialiltransferasa
11.
Res Sq ; 2021 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-34013243

RESUMEN

Severe COVID-19 is associated with unprecedented thromboembolic complications. We found that hospitalized COVID-19 patients develop immunoglobulin Gs (IgGs) that recognize a complex consisting of platelet factor 4 and heparin similar to those developed in heparin-induced thrombocytopenia and thrombosis (HIT), however, independent of heparin exposure. These antibodies activate platelets in the presence of TLR9 stimuli, stimuli that are prominent in COVID-19. Strikingly, 4 out of 42 antibodies cloned from IgG1+ RBD-binding B cells could activate platelets. These antibodies possessed, in the heavy-chain complementarity-determining region 3, an RKH or Y5 motif that we recently described among platelet-activating antibodies cloned from HIT patients. RKH and Y5 motifs were prevalent among published RBD-specific antibodies, and 3 out of 6 such antibodies tested could activate platelets. Features of platelet activation by these antibodies resemble those by pathogenic HIT antibodies. B cells with an RKH or Y5 motif were robustly expanded in COVID-19 patients. Our study demonstrates that SARS-CoV-2 infection drives the development of a subset of RBD-specific antibodies that can activate platelets and have activation properties and structural features similar to those of the pathogenic HIT antibodies.

12.
Exp Hematol ; 94: 37-46, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33346030

RESUMEN

The switch/sugar nonfermenting (SWI/SNF) family of chromatin remodeling complexes have been implicated in normal hematopoiesis. The ARID2 protein is a component of the polybromo-associated BAF (PBAF), one of the two main SWI/SNF complexes. In the current study, we used a conditional Arid2 knockout mouse model to determine its role in normal hematopoiesis. We found that the loss of Arid2 has no discernable effects on steady-state hematopoiesis, with the exception of a modest effect on erythropoiesis. On bone marrow transplantation, however, the loss of Arid2 affects HSC differentiation in a cell-autonomous manner, resulting in significant decreases in the ability to reconstitute the lymphoid lineage. Gene expression analysis of Arid2 knockout cells revealed enrichment of myeloid-biased multipotent progenitor (MPP) cell signatures, while the lymphoid-biased MPPs are enriched in the wild type, consistent with the observed phenotype. Moreover, Arid2 knockout cells revealed enrichment of inflammatory pathways with upregulation of TLR receptors, as well as downstream signaling cascade genes. Furthermore, under lymphocyte-biased growth conditions in vitro, Arid2 null bone marrow cells have significantly impaired proliferation, which decreased further on lipopolysaccharide stimulation. Overall, these data suggest that the loss of Arid2 impairs HSC differentiation ability, and this effect may be mediated through upregulation of inflammatory pathways.


Asunto(s)
Hematopoyesis , Células Madre Hematopoyéticas/citología , Factores de Transcripción/metabolismo , Animales , Células Cultivadas , Eliminación de Gen , Células Madre Hematopoyéticas/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Factores de Transcripción/genética
13.
Elife ; 92020 12 03.
Artículo en Inglés | MEDLINE | ID: mdl-33270017

RESUMEN

The resistance of synovial sublining macrophages to apoptosis has a crucial role in joint inflammation and destruction in rheumatoid arthritis (RA). However, the underlying mechanism is incompletely understood. Here we report that inactivation of the pro-apoptotic BCL-2 family protein BAD is essential for survival of synovial sublining macrophage in RA. Genetic disruption of Bad leads to more severe joint inflammation and cartilage and bone damage with reduced apoptosis of synovial sublining macrophages in collagen-induced arthritis (CIA) and TNFα transgenic (TNF-Tg) mouse models. Conversely, Bad3SA/3SA mice, in which BAD can no longer be inactivated by phosphorylation, are protected from collagen-induced arthritis. Mechanistically, phosphorylation-mediated inactivation of BAD specifically protects synovial sublining macrophages from apoptosis in highly inflammatory environment of arthritic joints in CIA and TNF-Tg mice, and in patients with RA, thereby contributing to RA pathology. Our findings put forward a model in which inactivation of BAD confers the apoptosis resistance on synovial sublining macrophages, thereby contributing to the development of arthritis, suggesting that BAD may be a potential therapeutic target for RA.


Asunto(s)
Artritis Reumatoide/metabolismo , Macrófagos/fisiología , Osteoartritis/inducido químicamente , Proteína Letal Asociada a bcl/metabolismo , Adulto , Anciano , Animales , Artritis Reumatoide/genética , Trasplante de Médula Ósea , Colágeno/toxicidad , Femenino , Humanos , Masculino , Ratones , Ratones Noqueados , Persona de Mediana Edad , Osteoartritis/metabolismo , Proteína Letal Asociada a bcl/genética
14.
J Immunol ; 205(12): 3480-3490, 2020 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-33158956

RESUMEN

Acute graft-versus-host disease (aGVHD) is one major serious complication that is induced by alloreactive donor T cells recognizing host Ags and limits the success of allogeneic hematopoietic stem cell transplantation. In the current studies, we identified a critical role of Kras in regulating alloreactive T cell function during aGVHD. Kras deletion in donor T cells dramatically reduced aGVHD mortality and severity in an MHC-mismatched allogeneic hematopoietic stem cell transplantation mouse model but largely maintained the antitumor capacity. Kras-deficient CD4 and CD8 T cells exhibited impaired TCR-induced activation of the ERK pathway. Kras deficiency altered TCR-induced gene expression profiles, including the reduced expression of various inflammatory cytokines and chemokines. Moreover, Kras deficiency inhibited IL-6-mediated Th17 cell differentiation and impaired IL-6-induced ERK activation and gene expression in CD4 T cells. These findings support Kras as a novel and effective therapeutic target for aGVHD.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Enfermedad Injerto contra Huésped/inmunología , Efecto Injerto vs Leucemia/inmunología , Trasplante de Células Madre Hematopoyéticas , Proteínas Proto-Oncogénicas p21(ras)/deficiencia , Células Th17/inmunología , Aloinjertos , Animales , Línea Celular Tumoral , Enfermedad Injerto contra Huésped/genética , Efecto Injerto vs Leucemia/genética , Interleucina-6/genética , Interleucina-6/inmunología , Sistema de Señalización de MAP Quinasas/genética , Sistema de Señalización de MAP Quinasas/inmunología , Ratones , Ratones Transgénicos , Proteínas Proto-Oncogénicas p21(ras)/inmunología
15.
Nano Lett ; 20(9): 6914-6921, 2020 Sep 09.
Artículo en Inglés | MEDLINE | ID: mdl-32790318

RESUMEN

Developing solid polymer electrolytes (SPEs) is a promising approach to realize practical dendrite-free lithium metal batteries (LMBs). Tuning the nanoscale polymer network chemsitry is of critical importance for SPE design. In this work, we took lessons from the rubber chemistry and developed a series of comb-chain crosslinker-based SPEs (ConSPEs) using a preformed polymer as the multifunctional crosslinker. The high-functionality crosslinker increased the connectivity of nanosized cross-linked domains, which led to a robust network with dramatically improved toughness and superior lithium dendrite resistance even at a current density of 2 mA cm-2. The uniform and flexile network also dramatically improved the anodic stability to over 5.3 V versus Li/Li+. Additive-free, all-solid-state LMBs with the ConSPE showed high discharge capacity and stable cycling up to 10 C rate, and could be stably cycled at 25 °C. Our results demonstrated that ConSPEs are promising for high-performance and dendrite-free LMBs.

16.
Elife ; 92020 05 14.
Artículo en Inglés | MEDLINE | ID: mdl-32406817

RESUMEN

The transcriptional activation and repression during NK cell ontology are poorly understood. Here, using single-cell RNA-sequencing, we reveal a novel role for T-bet in suppressing the immature gene signature during murine NK cell development. Based on transcriptome, we identified five distinct NK cell clusters and define their relative developmental maturity in the bone marrow. Transcriptome-based machine-learning classifiers revealed that half of the mTORC2-deficient NK cells belongs to the least mature NK cluster. Mechanistically, loss of mTORC2 results in an increased expression of signature genes representing immature NK cells. Since mTORC2 regulates the expression of T-bet through AktS473-FoxO1 axis, we further characterized the T-bet-deficient NK cells and found an augmented immature transcriptomic signature. Moreover, deletion of Foxo1 restores the expression of T-bet and corrects the abnormal expression of immature NK genes. Collectively, our study reveals a novel role for mTORC2-AktS473-FoxO1-T-bet axis in suppressing the transcriptional signature of immature NK cells.


Asunto(s)
Células de la Médula Ósea/metabolismo , Perfilación de la Expresión Génica , Células Asesinas Naturales/metabolismo , Aprendizaje Automático , RNA-Seq , Análisis de la Célula Individual , Proteínas de Dominio T Box/genética , Transcriptoma , Animales , Células de la Médula Ósea/inmunología , Análisis por Conglomerados , Proteína Forkhead Box O1/genética , Proteína Forkhead Box O1/metabolismo , Regulación de la Expresión Génica , Genotipo , Células Asesinas Naturales/inmunología , Diana Mecanicista del Complejo 2 de la Rapamicina/deficiencia , Diana Mecanicista del Complejo 2 de la Rapamicina/genética , Ratones Endogámicos C57BL , Ratones Noqueados , Fenotipo , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteína Asociada al mTOR Insensible a la Rapamicina/deficiencia , Proteína Asociada al mTOR Insensible a la Rapamicina/genética , Proteína Reguladora Asociada a mTOR/deficiencia , Proteína Reguladora Asociada a mTOR/genética , Proteínas de Dominio T Box/metabolismo
17.
Biochem Biophys Res Commun ; 525(2): 469-476, 2020 04 30.
Artículo en Inglés | MEDLINE | ID: mdl-32107003

RESUMEN

AIM: The fragile X mental retardation protein (FMRP), the product of the FMR1 gene, is responsible for the fragile X syndrome (FXS). FMRP regulates miRNA expression and is involved in miRNA-mediated gene silencing. However, the question of whether FMRP is, in turn, regulated by miRNAs remains unanswered. MAIN METHODS: We detected the FMRP expression pattern by in situ hybridization. MiR-315 overexpression and knockout models were generated by germ-line transformation and ends-out homologous recombination, respectively. Western blotting and immunohistochemistry were used to detect Drosophila FMRP (dFMRP) and a Luciferase reporter assay was used to confirm the regulation of dfmr1 mRNA by mir-315. Synaptic structural quantification and electrophysiological methods were used to compare synaptic functions among groups. KEY FINDINGS: Here, we determined that the transcription product of dFMR1, the Drosophila homologue of FMR1, is a direct target of miR-315. MiR-315 is mainly expressed in the nervous system of Drosophila. Flies overexpressing miR-315 showed pupation defects and reduced hatching rates. A homozygous miR-315 knockout status is embryonic lethal in flies. These observations indicate that miR-315 is a key regulator of the Drosophila nervous system. Furthermore, computational prediction and cell-based luciferase and in vivo assays demonstrated that dfmr1 is directly targeted by miR-315. Lastly, using the neuromuscular junction as a model, we found that miR-315 regulates synaptic structure and transmission by targeting dfmr1. SIGNIFICANCE: These findings provide compelling evidence that miR-315 targets dfmr1 in the Drosophila nervous system, acting as a regulatory factor for the fine-tuned modulation of FMRP expression.


Asunto(s)
Proteínas de Drosophila/genética , Drosophila melanogaster/embriología , Drosophila melanogaster/genética , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/genética , Regulación del Desarrollo de la Expresión Génica , Animales , Sistema Nervioso/embriología , Sistema Nervioso/metabolismo , Neurogénesis , Unión Neuromuscular/genética , Sinapsis/genética
18.
Cancer Immunol Res ; 7(10): 1647-1662, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31515257

RESUMEN

Natural killer (NK) cells generate proinflammatory cytokines that are required to contain infections and tumor growth. However, the posttranscriptional mechanisms that regulate NK cell functions are not fully understood. Here, we define the role of the microRNA cluster known as Mirc11 (which includes miRNA-23a, miRNA-24a, and miRNA-27a) in NK cell-mediated proinflammatory responses. Absence of Mirc11 did not alter the development or the antitumor cytotoxicity of NK cells. However, loss of Mirc11 reduced generation of proinflammatory factors in vitro and interferon-γ-dependent clearance of Listeria monocytogenes or B16F10 melanoma in vivo by NK cells. These functional changes resulted from Mirc11 silencing ubiquitin modifiers A20, Cbl-b, and Itch, allowing TRAF6-dependent activation of NF-κB and AP-1. Lack of Mirc11 caused increased translation of A20, Cbl-b, and Itch proteins, resulting in deubiquitylation of scaffolding K63 and addition of degradative K48 moieties on TRAF6. Collectively, our results describe a function of Mirc11 that regulates generation of proinflammatory cytokines from effector lymphocytes.


Asunto(s)
Inflamación/inmunología , Células Asesinas Naturales/inmunología , Melanoma Experimental/inmunología , MicroARNs/genética , Linfocitos T Citotóxicos/inmunología , Factor 6 Asociado a Receptor de TNF/metabolismo , Factor de Transcripción AP-1/metabolismo , Factor de Transcripción ReIA/metabolismo , Animales , Células Cultivadas , Citocinas/metabolismo , Femenino , Humanos , Inflamación/metabolismo , Inflamación/patología , Masculino , Melanoma Experimental/metabolismo , Melanoma Experimental/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , MicroARNs/inmunología , MicroARNs/metabolismo , Transducción de Señal , Ubiquitina/metabolismo , Ubiquitinación
19.
Nat Commun ; 10(1): 4415, 2019 09 27.
Artículo en Inglés | MEDLINE | ID: mdl-31562329

RESUMEN

Many autoimmune diseases are characterized by the production of autoantibodies. The current view is that CD4+ T follicular helper (Tfh) cells are the main subset regulating autoreactive B cells. Here we report a CXCR5+PD1+ Tfh subset of CD8+ T cells whose development and function are negatively modulated by Stat5. These CD8+ Tfh cells regulate the germinal center B cell response and control autoantibody production, as deficiency of Stat5 in CD8 T cells leads to an increase of CD8+ Tfh cells, resulting in the breakdown of B cell tolerance and concomitant autoantibody production. CD8+ Tfh cells share similar gene signatures with CD4+ Tfh, and require CD40L/CD40 and TCR/MHCI interactions to deliver help to B cells. Our study thus highlights the diversity of follicular T cell subsets that contribute to the breakdown of B-cell tolerance.


Asunto(s)
Linfocitos B/inmunología , Linfocitos T CD8-positivos/inmunología , Tolerancia Inmunológica/inmunología , Receptor de Muerte Celular Programada 1/inmunología , Receptores CXCR5/inmunología , Linfocitos T Colaboradores-Inductores/inmunología , Autoanticuerpos/inmunología , Autoanticuerpos/metabolismo , Enfermedades Autoinmunes/genética , Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/metabolismo , Linfocitos B/metabolismo , Antígenos CD40/genética , Antígenos CD40/inmunología , Antígenos CD40/metabolismo , Ligando de CD40/genética , Ligando de CD40/inmunología , Ligando de CD40/metabolismo , Linfocitos T CD8-positivos/metabolismo , Perfilación de la Expresión Génica/métodos , Humanos , Tolerancia Inmunológica/genética , Receptor de Muerte Celular Programada 1/genética , Receptor de Muerte Celular Programada 1/metabolismo , Receptores CXCR5/genética , Receptores CXCR5/metabolismo , Factor de Transcripción STAT5/genética , Factor de Transcripción STAT5/inmunología , Factor de Transcripción STAT5/metabolismo , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Linfocitos T Colaboradores-Inductores/metabolismo
20.
ACS Appl Mater Interfaces ; 11(38): 34904-34912, 2019 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-31474106

RESUMEN

The practical application of lithium metal batteries (LMBs) with high energy density is hindered by uneven lithium deposition during battery cycling. To mitigate this process, in this work, an interpenetrating polymer network solid polymer electrolyte (IPN SPE) is prepared by introducing a polymerized ionic liquid (PIL), poly(diallyldimethylammonium) bis(trifluoromethanesulfonyl)imide to a polyhedral oligomeric silsesquioxane-poly(ethylene glycol)-based network SPE. Compared with the virgin SPE, the newly developed IPN SPE exhibited improved ionic conductivity, excellent lithium dendrite resistance, and superior battery performance, which was attributed to the homogeneous, immobilized ion network provided by the PIL-containing IPN. Furthermore, the effects of ionic conductivity and homogenized ion distribution were quantitatively delineated by galvanostatic cycling and polarization measurements at current densities that were below and above the estimated Sand's critical current density based on the Chazalviel model. Full battery tests also showed excellent discharge capacity, cycle life, and Coulombic efficiency. Our results suggest that the PIL-containing IPN SPEs provide a promising system for stabilizing lithium electrodeposition and fabricating high-performance LMBs.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...