Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
1.
Cell ; 184(5): 1245-1261.e21, 2021 03 04.
Artículo en Inglés | MEDLINE | ID: mdl-33636132

RESUMEN

How early events in effector T cell (TEFF) subsets tune memory T cell (TMEM) responses remains incompletely understood. Here, we systematically investigated metabolic factors in fate determination of TEFF and TMEM cells using in vivo pooled CRISPR screening, focusing on negative regulators of TMEM responses. We found that amino acid transporters Slc7a1 and Slc38a2 dampened the magnitude of TMEM differentiation, in part through modulating mTORC1 signaling. By integrating genetic and systems approaches, we identified cellular and metabolic heterogeneity among TEFF cells, with terminal effector differentiation associated with establishment of metabolic quiescence and exit from the cell cycle. Importantly, Pofut1 (protein-O-fucosyltransferase-1) linked GDP-fucose availability to downstream Notch-Rbpj signaling, and perturbation of this nutrient signaling axis blocked terminal effector differentiation but drove context-dependent TEFF proliferation and TMEM development. Our study establishes that nutrient uptake and signaling are key determinants of T cell fate and shape the quantity and quality of TMEM responses.


Asunto(s)
Aminoácidos/metabolismo , Linfocitos T CD8-positivos/citología , Memoria Inmunológica , Transducción de Señal , Sistemas de Transporte de Aminoácidos/metabolismo , Animales , Linfocitos T CD8-positivos/inmunología , Sistemas CRISPR-Cas , Ciclo Celular , Diferenciación Celular , Modelos Animales de Enfermedad , Femenino , Técnicas de Sustitución del Gen , Coriomeningitis Linfocítica/inmunología , Masculino , Ratones , Ratones Transgénicos , Células Precursoras de Linfocitos T/citología
2.
Nat Immunol ; 17(3): 277-85, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26808230

RESUMEN

Regulatory T (Treg) cells respond to immune and inflammatory signals to mediate immunosuppression, but how the functional integrity of Treg cells is maintained under activating environments is unclear. Here we show that autophagy is active in Treg cells and supports their lineage stability and survival fitness. Treg cell-specific deletion of Atg7 or Atg5, two essential genes in autophagy, leads to loss of Treg cells, greater tumor resistance and development of inflammatory disorders. Atg7-deficient Treg cells show increased apoptosis and readily lose expression of the transcription factor Foxp3, especially after activation. Mechanistically, autophagy deficiency upregulates metabolic regulators mTORC1 and c-Myc and glycolysis, which contribute to defective Treg function. Therefore, autophagy couples environmental signals and metabolic homeostasis to protect lineage and survival integrity of Treg cells in activating contexts.


Asunto(s)
Apoptosis/genética , Autofagia/genética , Factores de Transcripción Forkhead/genética , Proteínas Asociadas a Microtúbulos/genética , Complejos Multiproteicos/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Linfocitos T Reguladores/inmunología , Serina-Treonina Quinasas TOR/metabolismo , Adenocarcinoma/inmunología , Traslado Adoptivo , Animales , Apoptosis/inmunología , Autofagia/inmunología , Proteína 5 Relacionada con la Autofagia , Proteína 7 Relacionada con la Autofagia , Línea Celular Tumoral , Neoplasias del Colon/inmunología , Metilación de ADN , Citometría de Flujo , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Glucólisis , Homeostasis , Immunoblotting , Activación de Linfocitos/inmunología , Linfocitos Infiltrantes de Tumor/inmunología , Diana Mecanicista del Complejo 1 de la Rapamicina , Ratones , Ratones Noqueados , Trasplante de Neoplasias , Reacción en Cadena en Tiempo Real de la Polimerasa , Regulación hacia Arriba
3.
Immunity ; 51(6): 1012-1027.e7, 2019 12 17.
Artículo en Inglés | MEDLINE | ID: mdl-31668641

RESUMEN

Regulatory T (Treg) cells are critical mediators of immune tolerance whose activity depends upon T cell receptor (TCR) and mTORC1 kinase signaling, but the mechanisms that dictate functional activation of these pathways are incompletely understood. Here, we showed that amino acids license Treg cell function by priming and sustaining TCR-induced mTORC1 activity. mTORC1 activation was induced by amino acids, especially arginine and leucine, accompanied by the dynamic lysosomal localization of the mTOR and Tsc complexes. Rag and Rheb GTPases were central regulators of amino acid-dependent mTORC1 activation in effector Treg (eTreg) cells. Mice bearing RagA-RagB- or Rheb1-Rheb2-deficient Treg cells developed a fatal autoimmune disease and had reduced eTreg cell accumulation and function. RagA-RagB regulated mitochondrial and lysosomal fitness, while Rheb1-Rheb2 enforced eTreg cell suppressive gene signature. Together, these findings reveal a crucial requirement of amino acid signaling for licensing and sustaining mTORC1 activation and functional programming of Treg cells.


Asunto(s)
Arginina/metabolismo , Leucina/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Proteínas de Unión al GTP Monoméricas/metabolismo , Proteína Homóloga de Ras Enriquecida en el Cerebro/metabolismo , Linfocitos T Reguladores/inmunología , Animales , Ciclo Celular , Diferenciación Celular/fisiología , Línea Celular , Humanos , Tolerancia Inmunológica/inmunología , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas de Unión al GTP Monoméricas/genética , Proteína Homóloga de Ras Enriquecida en el Cerebro/genética , Receptores de Antígenos de Linfocitos T/inmunología , Linfocitos T Reguladores/citología
4.
Nature ; 591(7849): 306-311, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33627871

RESUMEN

Regulatory T cells (Treg cells) are essential for immune tolerance1, but also drive immunosuppression in the tumour microenvironment2. Therapeutic targeting of Treg cells in cancer will therefore require the identification of context-specific mechanisms that affect their function. Here we show that inhibiting lipid synthesis and metabolic signalling that are dependent on sterol-regulatory-element-binding proteins (SREBPs) in Treg cells unleashes effective antitumour immune responses without autoimmune toxicity. We find that the activity of SREBPs is upregulated in intratumoral Treg cells. Moreover, deletion of SREBP-cleavage-activating protein (SCAP)-a factor required for SREBP activity-in these cells inhibits tumour growth and boosts immunotherapy that is triggered by targeting the immune-checkpoint protein PD-1. These effects of SCAP deletion are associated with uncontrolled production of interferon-γ and impaired function of intratumoral Treg cells. Mechanistically, signalling through SCAP and SREBPs coordinates cellular programs for lipid synthesis and inhibitory receptor signalling in these cells. First, de novo fatty-acid synthesis mediated by fatty-acid synthase (FASN) contributes to functional maturation of Treg cells, and loss of FASN from Treg cells inhibits tumour growth. Second, Treg cells in tumours show enhanced expression of the PD-1 gene, through a process that depends on SREBP activity and signals via mevalonate metabolism to protein geranylgeranylation. Blocking PD-1 or SREBP signalling results in dysregulated activation of phosphatidylinositol-3-kinase in intratumoral Treg cells. Our findings show that metabolic reprogramming enforces the functional specialization of Treg cells in tumours, pointing to new ways of targeting these cells for cancer therapy.


Asunto(s)
Metabolismo de los Lípidos , Neoplasias/inmunología , Neoplasias/metabolismo , Transducción de Señal , Linfocitos T Reguladores/citología , Linfocitos T Reguladores/inmunología , Animales , Colesterol/metabolismo , Ácido Graso Sintasas/metabolismo , Ácidos Grasos/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Masculino , Proteínas de la Membrana/metabolismo , Ácido Mevalónico/metabolismo , Ratones , Fosfatidilinositol 3-Quinasa/metabolismo , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Receptor de Muerte Celular Programada 1/metabolismo , Proteínas de Unión a los Elementos Reguladores de Esteroles/antagonistas & inhibidores , Proteínas de Unión a los Elementos Reguladores de Esteroles/metabolismo , Linfocitos T Reguladores/enzimología , Regulación hacia Arriba
5.
Nature ; 595(7869): 724-729, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34234346

RESUMEN

T follicular helper (TFH) cells are crucial for B cell-mediated humoral immunity1. Although transcription factors such as BCL6 drive the differentiation of TFH cells2,3, it is unclear whether and how post-transcriptional and metabolic programs enforce TFH cell programming. Here we show that the cytidine diphosphate (CDP)-ethanolamine pathway co-ordinates the expression and localization of CXCR5 with the responses of TFH cells and humoral immunity. Using in vivo CRISPR-Cas9 screening and functional validation in mice, we identify ETNK1, PCYT2, and SELENOI-enzymes in the CDP-ethanolamine pathway for de novo synthesis of phosphatidylethanolamine (PE)-as selective post-transcriptional regulators of TFH cell differentiation that act by promoting the surface expression and functional effects of CXCR5. TFH cells exhibit unique lipid metabolic programs and PE is distributed to the outer layer of the plasma membrane, where it colocalizes with CXCR5. De novo synthesis of PE through the CDP-ethanolamine pathway co-ordinates these events to prevent the internalization and degradation of CXCR5. Genetic deletion of Pcyt2, but not of Pcyt1a (which mediates the CDP-choline pathway), in activated T cells impairs the differentiation of TFH cells, and this is associated with reduced humoral immune responses. Surface levels of PE and CXCR5 expression on B cells also depend on Pcyt2. Our results reveal that phospholipid metabolism orchestrates post-transcriptional mechanisms for TFH cell differentiation and humoral immunity, highlighting the metabolic control of context-dependent immune signalling and effector programs.


Asunto(s)
Inmunidad Humoral , Fosfatidiletanolaminas/metabolismo , Receptores CXCR5/inmunología , Linfocitos T Colaboradores-Inductores/inmunología , Animales , Linfocitos B/inmunología , Sistemas CRISPR-Cas , Diferenciación Celular , Citidina Difosfato , Femenino , Regulación de la Expresión Génica , Humanos , Leucocitos Mononucleares/inmunología , Activación de Linfocitos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Fosfotransferasas (Aceptor de Grupo Alcohol) , ARN Nucleotidiltransferasas , Transducción de Señal
6.
Nature ; 600(7888): 308-313, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34795452

RESUMEN

Nutrients are emerging regulators of adaptive immunity1. Selective nutrients interplay with immunological signals to activate mechanistic target of rapamycin complex 1 (mTORC1), a key driver of cell metabolism2-4, but how these environmental signals are integrated for immune regulation remains unclear. Here we use genome-wide CRISPR screening combined with protein-protein interaction networks to identify regulatory modules that mediate immune receptor- and nutrient-dependent signalling to mTORC1 in mouse regulatory T (Treg) cells. SEC31A is identified to promote mTORC1 activation by interacting with the GATOR2 component SEC13 to protect it from SKP1-dependent proteasomal degradation. Accordingly, loss of SEC31A impairs T cell priming and Treg suppressive function in mice. In addition, the SWI/SNF complex restricts expression of the amino acid sensor CASTOR1, thereby enhancing mTORC1 activation. Moreover, we reveal that the CCDC101-associated SAGA complex is a potent inhibitor of mTORC1, which limits the expression of glucose and amino acid transporters and maintains T cell quiescence in vivo. Specific deletion of Ccdc101 in mouse Treg cells results in uncontrolled inflammation but improved antitumour immunity. Collectively, our results establish epigenetic and post-translational mechanisms that underpin how nutrient transporters, sensors and transducers interplay with immune signals for three-tiered regulation of mTORC1 activity and identify their pivotal roles in licensing T cell immunity and immune tolerance.


Asunto(s)
Sistemas CRISPR-Cas , Edición Génica , Nutrientes , Mapas de Interacción de Proteínas , Linfocitos T Reguladores , Animales , Femenino , Masculino , Ratones , Proteínas Portadoras/metabolismo , Sistemas CRISPR-Cas/genética , Factores de Transcripción Forkhead/metabolismo , Genoma/genética , Homeostasis , Tolerancia Inmunológica , Inflamación/patología , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Neoplasias/inmunología , Proteínas Nucleares/metabolismo , Nutrientes/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteolisis , Proteínas Quinasas Asociadas a Fase-S/metabolismo , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Transactivadores/metabolismo
7.
Nature ; 576(7787): 471-476, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31827283

RESUMEN

Adoptive cell therapy represents a new paradigm in cancer immunotherapy, but it can be limited by the poor persistence and function of transferred T cells1. Here we use an in vivo pooled CRISPR-Cas9 mutagenesis screening approach to demonstrate that, by targeting REGNASE-1, CD8+ T cells are reprogrammed to long-lived effector cells with extensive accumulation, better persistence and robust effector function in tumours. REGNASE-1-deficient CD8+ T cells show markedly improved therapeutic efficacy against mouse models of melanoma and leukaemia. By using a secondary genome-scale CRISPR-Cas9 screening, we identify BATF as the key target of REGNASE-1 and as a rheostat that shapes antitumour responses. Loss of BATF suppresses the increased accumulation and mitochondrial fitness of REGNASE-1-deficient CD8+ T cells. By contrast, the targeting of additional signalling factors-including PTPN2 and SOCS1-improves the therapeutic efficacy of REGNASE-1-deficient CD8+ T cells. Our findings suggest that T cell persistence and effector function can be coordinated in tumour immunity and point to avenues for improving the efficacy of adoptive cell therapy for cancer.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Inmunoterapia Adoptiva/métodos , Leucemia/inmunología , Leucemia/terapia , Melanoma/inmunología , Melanoma/terapia , Terapia Molecular Dirigida , Ribonucleasas/metabolismo , Animales , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/deficiencia , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/metabolismo , Linfocitos T CD8-positivos/citología , Sistemas CRISPR-Cas/genética , Modelos Animales de Enfermedad , Femenino , Eliminación de Gen , Humanos , Leucemia/genética , Leucemia/metabolismo , Linfocitos Infiltrantes de Tumor/inmunología , Linfocitos Infiltrantes de Tumor/metabolismo , Melanoma/genética , Melanoma/metabolismo , Ratones , Mitocondrias/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 2/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 2/metabolismo , Reproducibilidad de los Resultados , Ribonucleasas/deficiencia , Ribonucleasas/genética , Ribonucleasas/inmunología , Proteína 1 Supresora de la Señalización de Citocinas/genética , Proteína 1 Supresora de la Señalización de Citocinas/metabolismo , Microambiente Tumoral/inmunología
8.
Immunol Rev ; 295(1): 15-38, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32212344

RESUMEN

The evolutionarily conserved serine/threonine kinase mTOR (mechanistic target of rapamycin) forms the distinct protein complexes mTORC1 and mTORC2 and integrates signals from the environment to coordinate downstream signaling events and various cellular processes. T cells rely on mTOR activity for their development and to establish their homeostasis and functional fitness. Here, we review recent progress in our understanding of the upstream signaling and downstream targets of mTOR. We also provide an updated overview of the roles of mTOR in T-cell development, homeostasis, activation, and effector-cell fate decisions, as well as its important impacts on the suppressive activity of regulatory T cells. Moreover, we summarize the emerging roles of mTOR in T-cell exhaustion and transdifferentiation. A better understanding of the contribution of mTOR to T-cell fate decisions will ultimately aid in the therapeutic targeting of mTOR in human disease.


Asunto(s)
Transducción de Señal , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Animales , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Transdiferenciación Celular/inmunología , Citocinas/metabolismo , Citoesqueleto/metabolismo , Metabolismo Energético , Humanos , Memoria Inmunológica , Activación de Linfocitos/inmunología , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Diana Mecanicista del Complejo 2 de la Rapamicina/metabolismo , Receptores de Antígenos de Linfocitos T/metabolismo
9.
Blood ; 138(2): 122-135, 2021 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-33690816

RESUMEN

Chimeric antigen receptor (CAR)-T-cell therapeutic efficacy is associated with long-term T-cell persistence and acquisition of memory. Memory-subset formation requires T-cell factor 1 (TCF-1), a master transcription factor for which few regulators have been identified. Here, we demonstrate using an immune-competent mouse model of B-cell acute lymphoblastic leukemia (ALL; B-ALL) that Regnase-1 deficiency promotes TCF-1 expression to enhance CAR-T-cell expansion and memory-like cell formation. This leads to improved CAR-T-mediated tumor clearance, sustained remissions, and protection against secondary tumor challenge. Phenotypic, transcriptional, and epigenetic profiling identified increased tumor-dependent programming of Regnase-1-deficient CAR-T cells into TCF-1+ precursor exhausted T cells (TPEX) characterized by upregulation of both memory and exhaustion markers. Regnase-1 directly targets Tcf7 messenger RNA (mRNA); its deficiency augments TCF-1 expression leading to the formation of TPEX that support long-term CAR-T-cell persistence and function. Regnase-1 deficiency also reduces exhaustion and enhances the activity of TCF-1- CAR-T cells. We further validate these findings in human CAR-T cells, where Regnase-1 deficiency mediates enhanced tumor clearance in a xenograft B-ALL model. This is associated with increased persistence and expansion of a TCF-1+ CAR-T-cell population. Our findings demonstrate the pivotal roles of TPEX, Regnase-1, and TCF-1 in mediating CAR-T-cell persistence and recall responses, and identify Regnase-1 as a modulator of human CAR-T-cell longevity and potency that may be manipulated for improved therapeutic efficacy.


Asunto(s)
Inmunoterapia Adoptiva , Leucemia-Linfoma Linfoblástico de Células Precursoras/inmunología , Leucemia-Linfoma Linfoblástico de Células Precursoras/terapia , Ribonucleasas/metabolismo , Factor 1 de Transcripción de Linfocitos T/metabolismo , Linfocitos T/inmunología , Animales , Antígenos CD19/metabolismo , Línea Celular Tumoral , Reprogramación Celular , Modelos Animales de Enfermedad , Epigénesis Genética , Humanos , Inmunocompetencia/inmunología , Memoria Inmunológica , Ratones Endogámicos C57BL , Ratones Transgénicos , Fenotipo , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/patología
10.
Immunity ; 40(4): 515-29, 2014 Apr 17.
Artículo en Inglés | MEDLINE | ID: mdl-24726876

RESUMEN

The transcription factor IRF3 is a central regulator of type I interferon (IFN) signaling. The mechanisms underlying deactivation of IRF3 are poorly understood although many studies suggest that IRF3 activity is terminated through degradation after viral infection. Here we report that IRF3 is deactivated via dephosphorylation mediated by the serine and threonine phosphatase PP2A and its adaptor protein RACK1. The PP2A-RACK1 complex negatively regulated the IRF3 pathway after LPS or poly(I:C) stimulation or Sendai virus (SeV) infection. After challenge with LPS, poly(I:C), or low-titer SeV, activated IRF3 was dephosphorylated and returned to resting state without being degraded, although high-titer SeV infection triggered the degradation of IRF3. Furthermore, PP2A-deficient macrophages showed enhanced type I IFN signaling upon LPS, poly(I:C), and SeV challenge and protected mice from lethal vesicular stomatitis virus infection. Therefore, dephosphorylation of IRF3 is a deactivation mechanism that contributes to termination of IRF3-type I IFN signaling.


Asunto(s)
Factor 3 Regulador del Interferón/metabolismo , Interferón Tipo I/metabolismo , Neuropéptidos/metabolismo , Proteína Fosfatasa 2/metabolismo , Receptor Toll-Like 3/metabolismo , Receptor Toll-Like 4/metabolismo , Animales , Células HeLa , Humanos , Proteínas de la Membrana/metabolismo , Ratones , Ratones Transgénicos , Proteínas del Tejido Nervioso/metabolismo , Fosforilación , Unión Proteica , Proteína Fosfatasa 2/genética , Receptores de Cinasa C Activada , Receptores de Superficie Celular , Virus Sendai/inmunología , Transducción de Señal , Receptor Toll-Like 3/genética , Receptor Toll-Like 4/genética , Transgenes/genética , Estomatitis Vesicular/inmunología , Virus de la Estomatitis Vesicular Indiana/inmunología
11.
Nature ; 548(7669): 602-606, 2017 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-28847007

RESUMEN

Regulatory T cells (Treg cells) have a pivotal role in the establishment and maintenance of immunological self-tolerance and homeostasis. Transcriptional programming of regulatory mechanisms facilitates the functional activation of Treg cells in the prevention of diverse types of inflammatory responses. It remains unclear how Treg cells orchestrate their homeostasis and interplay with environmental signals. Here we show that liver kinase B1 (LKB1) programs the metabolic and functional fitness of Treg cells in the control of immune tolerance and homeostasis. Mice with a Treg-specific deletion of LKB1 developed a fatal inflammatory disease characterized by excessive TH2-type-dominant responses. LKB1 deficiency disrupted Treg cell survival and mitochondrial fitness and metabolism, but also induced aberrant expression of immune regulatory molecules including the negative co-receptor PD-1 and the TNF receptor superfamily proteins GITR and OX40. Unexpectedly, LKB1 function in Treg cells was independent of conventional AMPK signalling or the mTORC1-HIF-1α axis, but contributed to the activation of ß-catenin signalling for the control of PD-1 and TNF receptor proteins. Blockade of PD-1 activity reinvigorated the ability of LKB1-deficient Treg cells to suppress TH2 responses and the interplay with dendritic cells primed by thymic stromal lymphopoietin. Thus, Treg cells use LKB1 signalling to coordinate their metabolic and immunological homeostasis and to prevent apoptotic and functional exhaustion, thereby orchestrating the balance between immunity and tolerance.


Asunto(s)
Homeostasis , Tolerancia Inmunológica , Proteínas Serina-Treonina Quinasas/metabolismo , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Proteínas Quinasas Activadas por AMP , Animales , Apoptosis , Supervivencia Celular/genética , Citocinas/metabolismo , Células Dendríticas/inmunología , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Proteína Relacionada con TNFR Inducida por Glucocorticoide/metabolismo , Ratones , Mitocondrias/metabolismo , Mitocondrias/patología , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Receptor de Muerte Celular Programada 1/biosíntesis , Receptor de Muerte Celular Programada 1/metabolismo , Proteínas Serina-Treonina Quinasas/deficiencia , Proteínas Serina-Treonina Quinasas/genética , Receptores OX40/metabolismo , Receptores del Factor de Necrosis Tumoral/metabolismo , Transducción de Señal , Linfocitos T Reguladores/citología , Células Th2/inmunología , beta Catenina/metabolismo , Linfopoyetina del Estroma Tímico
12.
Plant Dis ; 2020 Oct 19.
Artículo en Inglés | MEDLINE | ID: mdl-33074071

RESUMEN

Sisal (Agave sisalana Perrine) is an important hard fiber crop that is widely planted in Guangxi, Guangdong, Hainan, Yunnan, and Fujian provinces, China. In July 2019, a new leaf disease of sisal with a disease incident of about 36% was found in Guangxi (Fig.1a~d). The oval or circular black lesions were 2.3 cm to 15.9 cm in length and 1.6 cm to 5.5 cm in width on both sides of the diseased leaves. The central part of the lesions was slightly hollow. The lesions continuously enlarged and ultimately penetrated the leaves. Reddish brown and dark mucus was secreted from the lesions. The junction of lesions and healthy parts was reddish brown to yellow. The diseased leaf fiber and mesophyll tissues were reddish brown and necrotic. Fresh leaf yield was reduced about 30% by the disease, and fiber quality was significantly compromised every year in Guangxi. Six kinds of fungi distinguished by their morphology, size and color of the colonies were isolated from diseased leaf tissues of 60 sisal plants sampled from five different farms in Guangxi. Isolate JMHB1 was isolated at a rate of 95.67%. The isolate JMHB1 was initially white with dense and hairy aerial mycelium, gradually turning dark grey to olive green on PDA (Fig. 2). Conidia, arthrospores, and chlamydospores were observed on PDA in culture (Fig. 3). The conidia formed arthric chains, disarticulating, cylindrical-truncate, oblong-obtuse to doliiform, colorless and transparent, zero- to one-septate, and averaging 4.4 to 13.8 µm × 2.2 to 5.6 µm (n=100). Arthrospores were short columnar, pigmented and transparent, single or formed arthric chains, averaging 5.5 to 17.9 µm × 2.1 to 3.5 µm (n=100). Chlamydospores were dark brown, round or oval, averaging 4.5 to 9.6 µm × 4.5 to 8.6 µm (n=100). Pathogenicity testing was conducted by inoculating 3-year-old healthy sisal plants with PDA plugs (5 × 5 mm) on which the fungus had grown for 5 days. Nine healthy plants were wounded on the leaves with a sterile needle, and mycelial plugs were placed on the wounds, covered with sterile moist cotton, and wrapped with parafilm. Nine control plants were wounded and treated with PDA plugs as the negative control. The test was repeated three times. All treated plants were kept in a greenhouse at ~28 ℃ and 40% RH. After 5 days, only leaves inoculated with isolate JMHB1 showed lesions similar to symptoms observed in the field (Fig.1e~f). The fungus was re-isolated from all nine diseased plants, and no symptoms were observed on the leaves of control plants. Molecular identification of the fungus was made by PCR amplification of the internal transcribed spacer (ITS) region of rDNA, EF1-α gene and ß-tubulin gene using primers ITS1/ITS4 (White et al. 1990), EFl-728F/EF1-986R (Carbone and Kohn 1999), TUB2Fd/TUB4Rd (Aveskamp et al. 2009) respectively. The ITS (MT705646), EF1-α (MT733516) and ß-tubulin (MT773603) sequences of JMHB1 were similar to the ITS (AY819727), EF1-α (EU144063) and ß-tubulin (KF531800) sequences of the epitype of Neoscytalidium dimidiatum (CBS 499.66) with 100%, 99.65% and 99.02% identity, respectively. Based on pathogenicity testing, morphological characteristics, and molecular identification, the pathogen of sisal causing black spot was identified as N. dimidiatum (Penz.) Crous & Slippers (Crous et al. 2006). To our knowledge, this is the first report of black spot caused by N. dimidiatum on sisal in China. Sisal is the main economic crop in arid and semi-arid areas that is widely planted in several provinces of southern China. The serious occurrence of the disease caused by N. dimidiatum has greatly affected the development of sisal industry and local economic income in China. Identification of the pathogen of the disease is of great significance to guide disease control, increase farmers' income and promote the development of sisal industry. References: Aveskamp, M. M., et al. 2009. Mycologia, 101: 363. https://doi.org/10.3852/08-199. Carbone, I., and Kohn, L. M. 1999. Mycologia, 91:553. https://doi.org/10.1080/00275514.1999. 12061051. Crous, P. W., et al. 2006. Stud. Mycol. 55:235. https://doi.org/10.3114/sim.55.1.235. White, T. J., et al. 1990. PCR Protocols: A Guide to Methods and Applications. Academic Press, San Diego, Page 315. doi.org/10.1002/mrd.1080280418. Supplemental photographs: Fig. 1 Symptoms of sisal black spot disease a, b, c, d showed symptoms in the field, e and f were symptoms after inoculating Neoscytalidium dimidiatum JMHB1. a, c, and e were the front of the lesions, b, d, and f were the back of the lesions. Fig. 2 Primary colony (a) and old colony (b) of Neoscytalidium dimidiatum JMHB1 Fig. 3 Arthrospores (a), conidia and chlamydospores (b) of Neoscytalidium dimidiatum JMHB1.

13.
J Biol Chem ; 292(3): 1112-1121, 2017 01 20.
Artículo en Inglés | MEDLINE | ID: mdl-27986811

RESUMEN

Eph receptors, the largest subfamily of transmembrane tyrosine kinase receptors, have been increasingly implicated in various physiologic and pathologic processes, and the roles of the Eph family members during tumorigenesis have recently attracted growing attentions. In the present study, we explored the function of EphB3, one member of Eph family, in papillary thyroid cancer (PTC). We found that the expression of EphB3 was significantly elevated in PTC. Either overexpression of EphB3 or activation of EphB3 by EfnB1-Fc/EfnB2-Fc stimulated in vitro migration of PTC cells. In contrast, siRNA-mediated knockdown of EphB3 or EphB3-Fc treatment, which only blocked EphB3-mediated forward signaling, inhibited migration and metastasis of PTC cells. A mechanism study revealed that EphB3 knockdown led to suppressed activity of Rac1 and enhanced activity of RhoA. Moreover, we found that Vav2, an important regulator of Rho family GTPases, was activated by EphB3 in a kinase-dependent manner. Altogether, our work suggested that EphB3 acted as a tumor promoter in PTC by increasing the in vitro migration as well as the in vivo metastasis of PTC cells through regulating the activities of Vav2 and Rho GTPases in a kinase-dependent manner.


Asunto(s)
Carcinoma/metabolismo , Movimiento Celular , Regulación Neoplásica de la Expresión Génica , Proteínas Proto-Oncogénicas c-vav/metabolismo , Receptor EphB3/metabolismo , Neoplasias de la Tiroides/metabolismo , Proteína de Unión al GTP rac1/metabolismo , Carcinoma/genética , Carcinoma/patología , Carcinoma Papilar , Línea Celular Tumoral , Femenino , Humanos , Masculino , Metástasis de la Neoplasia , Proteínas Proto-Oncogénicas c-vav/genética , Receptor EphB3/genética , Transducción de Señal/genética , Cáncer Papilar Tiroideo , Neoplasias de la Tiroides/genética , Neoplasias de la Tiroides/patología , Proteína de Unión al GTP rac1/genética
14.
Plant Cell ; 27(5): 1409-27, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25944101

RESUMEN

Phytochromes (phys) are red and far-red photoreceptors that control plant development and growth by promoting the proteolysis of a family of antagonistically acting basic helix-loop-helix transcription factors, the PHYTOCHROME-INTERACTING FACTORs (PIFs). We have previously shown that the degradation of PIF1 and PIF3 requires HEMERA (HMR). However, the biochemical function of HMR and the mechanism by which it mediates PIF degradation remain unclear. Here, we provide genetic evidence that HMR acts upstream of PIFs in regulating hypocotyl growth. Surprisingly, genome-wide analysis of HMR- and PIF-dependent genes reveals that HMR is also required for the transactivation of a subset of PIF direct-target genes. We show that HMR interacts with all PIFs. The HMR-PIF interaction is mediated mainly by HMR's N-terminal half and PIFs' conserved active-phytochrome B binding motif. In addition, HMR possesses an acidic nine-amino-acid transcriptional activation domain (9aaTAD) and a loss-of-function mutation in this 9aaTAD impairs the expression of PIF target genes and the destruction of PIF1 and PIF3. Together, these in vivo results support a regulatory mechanism for PIFs in which HMR is a transcriptional coactivator binding directly to PIFs and the 9aaTAD of HMR couples the degradation of PIF1 and PIF3 with the transactivation of PIF target genes.


Asunto(s)
Proteínas de Arabidopsis/metabolismo , Arabidopsis/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Activación Transcripcional , Secuencias de Aminoácidos , Arabidopsis/crecimiento & desarrollo , Arabidopsis/metabolismo , Proteínas de Arabidopsis/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Perfilación de la Expresión Génica , Regulación de la Expresión Génica de las Plantas , Hipocótilo/genética , Hipocótilo/crecimiento & desarrollo , Hipocótilo/metabolismo , Análisis por Micromatrices , Mutación , Fitocromo/genética , Fitocromo/metabolismo , Proteolisis , Proteínas Recombinantes de Fusión , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Técnicas del Sistema de Dos Híbridos
15.
J Hepatol ; 65(1): 137-145, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27013087

RESUMEN

BACKGROUND & AIMS: Iron is an essential metal for fundamental metabolic processes, but little is known regarding the involvement of iron in other nutritional disorders. In the present study, we investigated disordered iron metabolism in a murine model of hereditary tyrosinemia type I (HT1), a disease of the tyrosine degradation pathway. METHODS: We analysed the status of iron accumulation following NTBC withdrawal from Fah(-/-) mice, a murine model for HT1. Liver histology and serum parameters were used to assess the extent of liver injury and iron deposition. To determine the physiological significance of iron accumulation, mice were subjected to a low-iron food intake to reduce the iron accumulation. Mechanistic studies were performed on tissues and cells using immunoblotting, qRT-PCR, adenovirus transfection and other assays. RESULTS: Severe iron overload was observed in the murine model of HT1 with dramatically elevated hepatic and serum iron levels. Mechanistic studies revealed that downregulation and dysfunction of Tfr2 decreased hepcidin, leading to iron overload. The Fah(-/-) hepatocytes lost the ability of transferrin-sensitive induction of hepcidin. Forced expression of Tfr2 in the murine liver reduced the iron accumulation. Moreover, transcription factor Sp1 was downregulated and identified as a new regulator of Tfr2 here. Additionally, low-iron food intake effectively reduced the iron deposits, protected the liver and prolonged the survival in these mice. CONCLUSIONS: Iron was severely overloaded in the HT1 mice via the Sp1/Tfr2/Hepcidin axis. The iron overload induced liver injury in the HT1 mice, and reduction of the iron accumulation ameliorated liver injury. LAY SUMMARY: Primary and secondary iron overload is an abnormal status affecting millions of people worldwide. Here, we reported severe iron overload in a murine model of HT1, a disease of the tyrosine degradation pathway, and elucidated the mechanistic basis and the physiological significance of iron overload in HT1. These studies are of general interest not only with respect to secondary iron-induced liver injury in HT1 but also are important to elucidate the crosstalk between the two metabolic pathways.


Asunto(s)
Hígado/lesiones , Tirosinemias , Animales , Hepcidinas , Hierro , Sobrecarga de Hierro , Ratones
16.
Hepatology ; 62(6): 1791-803, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26257239

RESUMEN

UNLABELLED: Sorafenib is a specific adenosine triphosphate-competitive RAF inhibitor used as a first-line treatment of advanced hepatocellular carcinoma (HCC). However, the responses are variable, reflecting heterogeneity of the disease, while the resistance mechanism remains poorly understood. Here, we report that sorafenib treatment can exacerbate disease progression in both patient-derived xenografts and cell line-derived xenografts and that the therapeutic effect of the drug inversely covaries to the ratio of epithelial cell adhesion molecule-positive cells, which may be tumor initiating cells in HCC. The TSC2-AKT cascade mediates this sorafenib resistance. In response to sorafenib treatment, formation of the TSC1/2 complex is enhanced, causing increased phosphorylation of AKT, which contributes to up-regulation of "stemness"-related genes in epithelial cell adhesion molecule-positive cells and enhancement of tumorigenicity. The expression of TSC2 negatively correlated with prognosis in clinical sorafenib therapy. Furthermore, all-trans retinoic acid decreased AKT activity, reduced the epithelial cell adhesion molecule-positive cell population enriched by sorafenib, and potentiated the therapeutic effect of sorafenib in the patient-derived xenograft model. CONCLUSION: Our findings suggest that a subtype of HCC is not suitable for sorafenib therapy; this resistance to sorafenib can be predicted by the status of TSC2, and agents inducing differentiation of tumor initiating cells (e.g., all-trans retinoic acid) should improve the prognosis of this subtype of HCC.


Asunto(s)
Antígenos de Neoplasias/efectos de los fármacos , Antineoplásicos/efectos adversos , Carcinoma Hepatocelular/inducido químicamente , Moléculas de Adhesión Celular/efectos de los fármacos , Neoplasias Hepáticas/inducido químicamente , Células Madre Neoplásicas/efectos de los fármacos , Niacinamida/análogos & derivados , Proteína Oncogénica v-akt/fisiología , Compuestos de Fenilurea/efectos adversos , Proteínas Supresoras de Tumor/fisiología , Animales , Carcinoma Hepatocelular/clasificación , Progresión de la Enfermedad , Molécula de Adhesión Celular Epitelial , Humanos , Neoplasias Hepáticas/clasificación , Ratones , Niacinamida/efectos adversos , Sorafenib , Proteína 2 del Complejo de la Esclerosis Tuberosa
17.
J Cell Biol ; 223(2)2024 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-38095639

RESUMEN

Metastasis is the main cause of colorectal cancer (CRC)-related death, and the 5-year relative survival rate for CRC patients with distant metastasis is only 14%. X-linked inhibitor of apoptosis (XIAP)-associated factor 1 (XAF1) is a zinc-rich protein belonging to the interferon (IFN)-induced gene family. Here, we report a metastasis-promoting role of XAF1 in CRC by acting as a novel adaptor of valosin-containing protein (VCP). XAF1 facilitates VCP-mediated deubiquitination of the E3 ligase RING finger protein 114 (RNF114), which promotes K48-linked ubiquitination and subsequent degradation of junction plakoglobin (JUP). The XAF1-VCP-RNF114-JUP axis is critical for the migration and metastasis of CRC cells. Moreover, we observe correlations between the protein levels of XAF1, RNF114, and JUP in clinical samples. Collectively, our findings reveal an oncogenic function of XAF1 in mCRC and suggest that the XAF1-VCP-RNF114-JUP axis is a potential therapeutic target for CRC treatment.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Proteínas Reguladoras de la Apoptosis , Neoplasias Colorrectales , Péptidos y Proteínas de Señalización Intracelular , Humanos , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Apoptosis , Proteínas Reguladoras de la Apoptosis/metabolismo , Neoplasias Colorrectales/genética , gamma Catenina/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas de Neoplasias/metabolismo , Proteína que Contiene Valosina/genética , Proteína que Contiene Valosina/metabolismo
18.
Gastroenterology ; 142(4): 812-823.e15, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22240482

RESUMEN

BACKGROUND & AIMS: Dysregulation of Wnt signaling has been involved in gastric tumorigenesis by mechanisms that are not fully understood. The receptor for activated protein kinase C (RACK1, GNB2L1) is involved in development of different tumor types, but its expression and function have not been investigated in gastric tumors. METHODS: We analyzed expression of RACK1 in gastric tumor samples and their matched normal tissues from 116 patients using immunohistochemistry. Effects of knockdown with small interfering RNAs or overexpression of RACK1 in gastric cancer cell lines were evaluated in cell growth and tumor xenograft. RACK1 signaling pathways were investigated in cells and zebrafish embryos using immunoblot, immunoprecipitation, microinjection, and in situ hybridization assays. RESULTS: Expression of RACK1 was reduced in gastric tumor samples and correlated with depth of tumor infiltration and poor differentiation. Knockdown of RACK1 in gastric cancer cells accelerated their anchorage-independent proliferation in soft agar, whereas overexpression of RACK1 reduced their tumorigenicity in nude mice. RACK1 formed a complex with glycogen synthase kinase Gsk3ß and Axin to promote the interaction between Gsk3ß and ß-catenin and thereby stabilized the ß-catenin destruction complex. On stimulation of Wnt3a, RACK1 repressed Wnt signaling by inhibiting recruitment of Axin by Dishevelled 2 (Dvl2). Moreover, there was an inverse correlation between expression of RACK1 and localization of ß-catenin to the cytoplasm/nucleus in human gastric tumor samples. CONCLUSIONS: RACK1 negatively regulates Wnt signaling pathway by stabilizing the ß-catenin destruction complex and act as a tumor suppressor in gastric cancer cells.


Asunto(s)
Complejo de Señalización de la Axina/metabolismo , Proteínas de Unión al GTP/metabolismo , Proteínas de Neoplasias/metabolismo , Receptores de Superficie Celular/metabolismo , Neoplasias Gástricas/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Vía de Señalización Wnt , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Complejo de Señalización de la Axina/genética , Estudios de Casos y Controles , Diferenciación Celular , Línea Celular Tumoral , Proliferación Celular , Proteínas Dishevelled , Femenino , Proteínas de Unión al GTP/genética , Regulación Neoplásica de la Expresión Génica , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Células HEK293 , Humanos , Inmunohistoquímica , Masculino , Ratones , Ratones Desnudos , Persona de Mediana Edad , Invasividad Neoplásica , Proteínas de Neoplasias/genética , Trasplante de Neoplasias , Fosfoproteínas/metabolismo , Interferencia de ARN , Receptores de Cinasa C Activada , Receptores de Superficie Celular/genética , Neoplasias Gástricas/genética , Neoplasias Gástricas/patología , Neoplasias Gástricas/prevención & control , Factores de Tiempo , Transfección , Proteínas Supresoras de Tumor/genética , Vía de Señalización Wnt/genética , Proteína Wnt3A/metabolismo , Pez Cebra/embriología , Pez Cebra/genética , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo , beta Catenina/metabolismo
19.
Front Pharmacol ; 14: 1246761, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38035004

RESUMEN

The importance of adequate sleep for good health cannot be overstated. Excessive light exposure at night disrupts sleep, therefore, it is important to find more healthy drinks that can promote sleep under sleep-disturbed conditions. The present study investigated the use of A. sinensis (Lour.) Spreng leaf tea, a natural product, to reduce the adverse effects of nighttime light on sleep. Here, Aquilaria sinensis leaf tea at 1.0 and 1.5 g/L significantly increased sleep time in zebrafish larvae (5-7 dpf) with light-induced sleep disturbance. Transcriptome sequencing and qRT-PCR analysis revealed a decrease in the immune-related genes, such as nfkbiab, tnfrsf1a, nfkbiaa, il1b, traf3, and cd40 in the 1.5 g/L Aquilaria sinensis leaf tea treatment group. In addition, a gene associated with sleep, bhlhe41, showed a significant decrease. Moreover, Aquilaria sinensis leaf tea suppressed the increase in neutrophils of Tg(mpo:GFP) zebrafish under sleep-disturbed conditions, indicating its ability to improve the immune response. Widely targeted metabolic profiling of the Aquilaria sinensis tea using ultra-performance liquid chromatography coupled with electrospray tandem mass spectrometry (UPLC-ESI-MS/MS) revealed flavonoids as the predominant component. Network pharmacological and molecular docking analyses suggested that the flavonoids quercetin and eupatilin in Aquilaria sinensis leaf tea improved the sleep of zebrafish by interacting with il1b and cd40 genes under light exposure at night. Therefore, the results of the study provide evidence supporting the notion that Aquilaria sinensis leaf tea has a positive impact on sleep patterns in zebrafish subjected to disrupted sleep due to nighttime light exposure. This suggests that the utilization of Aquilaria sinensis leaf tea as a potential therapeutic intervention for sleep disturbances induced by light may yield advantageous outcomes.

20.
Cancer Cell ; 41(11): 1852-1870.e9, 2023 11 13.
Artículo en Inglés | MEDLINE | ID: mdl-37832554

RESUMEN

Neoadjuvant immune checkpoint blockade (ICB) demonstrates promise in operable esophageal squamous cell carcinoma (ESCC), but lacks available efficacy biomarkers. Here, we perform single-cell RNA-sequencing of tumors from patients with ESCC undergoing neoadjuvant ICB, revealing a subset of exhausted CD8+ T cells expressing SPRY1 (CD8+ Tex-SPRY1) that displays a progenitor exhausted T cell (Tpex) phenotype and correlates with complete response to ICB. We validate CD8+ Tex-SPRY1 cells as an ICB-specific predictor of improved response and survival using independent ICB-/non-ICB cohorts and demonstrate that expression of SPRY1 in CD8+ T cells enforces Tpex phenotype and enhances ICB efficacy. Additionally, CD8+ Tex-SPRY1 cells contribute to proinflammatory phenotype of macrophages and functional state of B cells, which thereby promotes antitumor immunity by enhancing CD8+ T cell effector functions. Overall, our findings unravel progenitor-like CD8+ Tex-SPRY1 cells' role in effective responses to ICB for ESCC and inform mechanistic biomarkers for future individualized immunotherapy.


Asunto(s)
Neoplasias Esofágicas , Carcinoma de Células Escamosas de Esófago , Humanos , Carcinoma de Células Escamosas de Esófago/genética , Carcinoma de Células Escamosas de Esófago/patología , Linfocitos T CD8-positivos , Receptor de Muerte Celular Programada 1 , Neoplasias Esofágicas/tratamiento farmacológico , Neoplasias Esofágicas/genética , Terapia Neoadyuvante , Biomarcadores , Microambiente Tumoral , Proteínas de la Membrana/genética , Fosfoproteínas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA