Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Nat Immunol ; 24(2): 255-266, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36658237

RESUMEN

Despite tumor-associated macrophages (TAMs) playing a key role in shaping the tumor microenvironment (TME), the mechanisms by which TAMs influence the TME and contribute to cancer progression remain unclear. Here, we show that the N6-methyladenosine reader YTHDF2 regulates the antitumor functions of TAMs. YTHDF2 deficiency in TAMs suppressed tumor growth by reprogramming TAMs toward an antitumoral phenotype and increasing their antigen cross-presentation ability, which in turn enhanced CD8+ T cell-mediated antitumor immunity. YTHDF2 deficiency facilitated the reprogramming of TAMs by targeting interferon-γ-STAT1 signaling. The expression of YTHDF2 in TAMs was regulated by interleukin-10-STAT3 signaling. Selectively targeting YTHDF2 in TAMs using a Toll-like receptor 9 agonist-conjugated small interfering RNA reprogrammed TAMs toward an antitumoral phenotype, restrained tumor growth and enhanced the efficacy of PD-L1 antibody therapy. Collectively, our findings describe the role of YTHDF2 in orchestrating TAMs and suggest that YTHDF2 inhibition is an effective approach to enhance cancer immunotherapy.


Asunto(s)
Linfocitos T CD8-positivos , Neoplasias , Humanos , Macrófagos , Macrófagos Asociados a Tumores , Neoplasias/metabolismo , Inmunoterapia , Microambiente Tumoral , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo
2.
Cell Chem Biol ; 29(7): 1200-1208.e6, 2022 07 21.
Artículo en Inglés | MEDLINE | ID: mdl-35429459

RESUMEN

Environmental stresses, including hypoxia or detachment for anchorage independence, or attenuation of mitochondrial respiration through inhibition of electron transport chain induce reductive carboxylation in cells with an enhanced fraction of citrate arising through reductive metabolism of glutamine. This metabolic process contributes to redox homeostasis and sustains biosynthesis of lipids. Reductive carboxylation is often dependent on cytosolic isocitrate dehydrogenase 1 (IDH1). However, whether diverse cellular signals induce reductive carboxylation differentially or through a common signaling converging node remains unclear. We found that induction of reductive carboxylation commonly requires enhanced tyrosine phosphorylation and activation of IDH1, which, surprisingly, is achieved by attenuation of a cytosolic protein tyrosine phosphatase, Src homology region 2 domain-containing phosphatase-2 (SHP-2). Mechanistically, diverse signals induce reductive carboxylation by converging at upregulation of NADPH oxidase 2, leading to elevated cytosolic reactive oxygen species that consequently inhibit SHP-2. Together, our work elucidates the signaling basis underlying reductive carboxylation in cancer cells.


Asunto(s)
Isocitrato Deshidrogenasa , Neoplasias , Línea Celular Tumoral , Ciclo del Ácido Cítrico , Glutamina/metabolismo , Isocitrato Deshidrogenasa/metabolismo , Oxidación-Reducción , Fosforilación , Proteína Tirosina Fosfatasa no Receptora Tipo 11/metabolismo
3.
Mol Cell ; 82(11): 1992-2005.e9, 2022 06 02.
Artículo en Inglés | MEDLINE | ID: mdl-35417664

RESUMEN

Phospholipase A2, group VII (PLA2G7) is widely recognized as a secreted, lipoprotein-associated PLA2 in plasma that converts phospholipid platelet-activating factor (PAF) to a biologically inactive product Lyso-PAF during inflammatory response. We report that intracellular PLA2G7 is selectively important for cell proliferation and tumor growth potential of melanoma cells expressing mutant NRAS, but not cells expressing BRAF V600E. Mechanistically, PLA2G7 signals through its product Lyso-PAF to contribute to RAF1 activation by mutant NRAS, which is bypassed by BRAF V600E. Intracellular Lyso-PAF promotes p21-activated kinase 2 (PAK2) activation by binding to its catalytic domain and altering ATP kinetics, while PAK2 significantly contributes to S338-phosphorylation of RAF1 in addition to PAK1. Furthermore, the PLA2G7-PAK2 axis is also required for full activation of RAF1 in cells stimulated by epidermal growth factor (EGF) or cancer cells expressing mutant KRAS. Thus, PLA2G7 and Lyso-PAF exhibit intracellular signaling functions as key elements of RAS-RAF1 signaling.


Asunto(s)
Fosfolípidos , Proteínas Proto-Oncogénicas B-raf , Fosfolipasas A2 , Factor de Activación Plaquetaria/análogos & derivados , Factor de Activación Plaquetaria/metabolismo
4.
Cancer Cell ; 39(7): 945-957.e10, 2021 07 12.
Artículo en Inglés | MEDLINE | ID: mdl-34019807

RESUMEN

Tumor-associated macrophages (TAMs) can dampen the antitumor activity of T cells, yet the underlying mechanism remains incompletely understood. Here, we show that C1q+ TAMs are regulated by an RNA N6-methyladenosine (m6A) program and modulate tumor-infiltrating CD8+ T cells by expressing multiple immunomodulatory ligands. Macrophage-specific knockout of an m6A methyltransferase Mettl14 drives CD8+ T cell differentiation along a dysfunctional trajectory, impairing CD8+ T cells to eliminate tumors. Mettl14-deficient C1q+ TAMs show a decreased m6A abundance on and a higher level of transcripts of Ebi3, a cytokine subunit. In addition, neutralization of EBI3 leads to reinvigoration of dysfunctional CD8+ T cells and overcomes immunosuppressive impact in mice. We show that the METTL14-m6A levels are negatively correlated with dysfunctional T cell levels in patients with colorectal cancer, supporting the clinical relevance of this regulatory pathway. Thus, our study demonstrates how an m6A methyltransferase in TAMs promotes CD8+ T cell dysfunction and tumor progression.


Asunto(s)
Adenosina/análogos & derivados , Linfocitos T CD8-positivos/inmunología , Activación de Linfocitos/inmunología , Metiltransferasas/metabolismo , Metiltransferasas/fisiología , Neoplasias/patología , Macrófagos Asociados a Tumores/metabolismo , Adenosina/química , Animales , Carcinoma Pulmonar de Lewis/inmunología , Carcinoma Pulmonar de Lewis/metabolismo , Carcinoma Pulmonar de Lewis/patología , Neoplasias Colorrectales/inmunología , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Citocinas/metabolismo , Femenino , Humanos , Melanoma Experimental/inmunología , Melanoma Experimental/metabolismo , Melanoma Experimental/patología , Metiltransferasas/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Antígenos de Histocompatibilidad Menor/metabolismo , Neoplasias/inmunología , Neoplasias/metabolismo , Receptores de Citocinas/metabolismo , Microambiente Tumoral , Macrófagos Asociados a Tumores/patología
5.
Mol Cell ; 80(4): 633-647.e7, 2020 11 19.
Artículo en Inglés | MEDLINE | ID: mdl-33217317

RESUMEN

N6-methyladenosine (m6A) is the most abundant mRNA modification and is installed by the METTL3-METTL14-WTAP methyltransferase complex. Although the importance of m6A methylation in mRNA metabolism has been well documented recently, regulation of the m6A machinery remains obscure. Through a genome-wide CRISPR screen, we identify the ERK pathway and USP5 as positive regulators of the m6A deposition. We find that ERK phosphorylates METTL3 at S43/S50/S525 and WTAP at S306/S341, followed by deubiquitination by USP5, resulting in stabilization of the m6A methyltransferase complex. Lack of METTL3/WTAP phosphorylation reduces decay of m6A-labeled pluripotent factor transcripts and traps mouse embryonic stem cells in the pluripotent state. The same phosphorylation can also be found in ERK-activated human cancer cells and contribute to tumorigenesis. Our study reveals an unrecognized function of ERK in regulating m6A methylation.


Asunto(s)
Adenina/análogos & derivados , Carcinogénesis/patología , Endopeptidasas/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Melanoma/patología , Metiltransferasas/química , Adenina/química , Animales , Carcinogénesis/genética , Carcinogénesis/metabolismo , Embrión de Mamíferos/citología , Embrión de Mamíferos/metabolismo , Endopeptidasas/genética , Quinasas MAP Reguladas por Señal Extracelular/genética , Fibroblastos/citología , Fibroblastos/metabolismo , Humanos , Melanoma/genética , Melanoma/metabolismo , Metilación , Metiltransferasas/genética , Metiltransferasas/metabolismo , Metiltransferasas/fisiología , Ratones , Ratones Noqueados , Fosforilación , Estabilidad Proteica , Procesamiento Postranscripcional del ARN
6.
Cell Res ; 29(7): 599, 2019 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-31110249

RESUMEN

In the initial published version of this article, there was a mistake in the P value for the correlation between gene-expression changes and 5 hmC changes in tumors. The correct P value should be same as the P value shown in Fig. S6A: 9.8 × 10-6 (mistakenly shown as "9.8 × 106" in the main text). This correction does not affect the description of results or the conclusions of this study, since the range of P value is between 0 and 1.

7.
Nature ; 568(7751): E3, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-30911170

RESUMEN

In this Letter, a citation to 'Fig. 1e' has been corrected to 'Fig. 1d' in the sentence starting "By contrast, the anti-tumour response…". This has been corrected online.

8.
Nature ; 566(7743): 270-274, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30728504

RESUMEN

There is growing evidence that tumour neoantigens have important roles in generating spontaneous antitumour immune responses and predicting clinical responses to immunotherapies1,2. Despite the presence of numerous neoantigens in patients, complete tumour elimination is rare, owing to failures in mounting a sufficient and lasting antitumour immune response3,4. Here we show that durable neoantigen-specific immunity is regulated by mRNA N6-methyadenosine (m6A) methylation through the m6A-binding protein YTHDF15. In contrast to wild-type mice, Ythdf1-deficient mice show an elevated antigen-specific CD8+ T cell antitumour response. Loss of YTHDF1 in classical dendritic cells enhanced the cross-presentation of tumour antigens and the cross-priming of CD8+ T cells in vivo. Mechanistically, transcripts encoding lysosomal proteases are marked by m6A and recognized by YTHDF1. Binding of YTHDF1 to these transcripts increases the translation of lysosomal cathepsins in dendritic cells, and inhibition of cathepsins markedly enhances cross-presentation of wild-type dendritic cells. Furthermore, the therapeutic efficacy of PD-L1 checkpoint blockade is enhanced in Ythdf1-/- mice, implicating YTHDF1 as a potential therapeutic target in anticancer immunotherapy.


Asunto(s)
Adenosina/análogos & derivados , Adenosina/metabolismo , Células Dendríticas/inmunología , Neoplasias/inmunología , ARN Mensajero/metabolismo , Proteínas de Unión al ARN/metabolismo , Animales , Presentación de Antígeno/inmunología , Antígenos de Neoplasias/inmunología , Antígenos de Neoplasias/metabolismo , Antígeno B7-H1/metabolismo , Sitios de Unión , Linfocitos T CD8-positivos/inmunología , Catepsinas/antagonistas & inhibidores , Catepsinas/biosíntesis , Catepsinas/genética , Reactividad Cruzada/inmunología , Células Dendríticas/enzimología , Femenino , Humanos , Metilación , Ratones , Ratones Endogámicos C57BL , Neoplasias/terapia , Biosíntesis de Proteínas , Proteínas/genética , ARN Mensajero/química , Proteínas de Unión al ARN/genética , Transcriptoma/genética
9.
Cancer Chemother Pharmacol ; 82(2): 309-317, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29948021

RESUMEN

BACKGROUND: Sirolimus is a mammalian target of rapamycin (mTOR) inhibitor. Metformin may potentiate mTOR inhibition by sirolimus while mitigating its adverse effects. We conducted a pilot study to test this hypothesis. METHODS: Patients with advanced solid tumor were treated with sirolimus for 7 days followed by randomization to either sirolimus with metformin (Arm A) or sirolimus (Arm B) until day 21. From day 22 onwards, all patients received sirolimus and metformin. The primary aim was to compare the change in phospho-p70S6K (pp70S6K) in peripheral blood mononuclear cells (PBMC) from day 8 to day 22 using a two-sample t test. Secondary aims were objective response rate, toxicity, and other serum pharmacodynamic biomarkers (e.g., fasting glucose, triglycerides, insulin, C-peptide, IGF-1, IGF-1R, IGF-BP, and leptin). RESULTS: 24 patients were enrolled, with 18 evaluable for the primary endpoint. There was no significant difference in mean change in pp70S6K in arm A vs. arm B (- 0.12 vs. - 0.16; P = 0.64). Similarly, there were no significant differences in other serum pharmacodynamic biomarkers. There were no partial responses. There were no dose-limiting or unexpected toxicities. CONCLUSIONS: Adding metformin to sirolimus, although well tolerated, was not associated with significant changes in pp70S6K in PBMC or other serum pharmacodynamic biomarkers. IMPACT: Combining metformin with sirolimus did not improve mTOR inhibition.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Metformina/farmacología , Neoplasias/tratamiento farmacológico , Sirolimus/farmacología , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Biomarcadores de Tumor/sangre , Sinergismo Farmacológico , Femenino , Humanos , Leucocitos Mononucleares/efectos de los fármacos , Leucocitos Mononucleares/metabolismo , Masculino , Metformina/administración & dosificación , Persona de Mediana Edad , Neoplasias/sangre , Fosforilación , Proyectos Piloto , Proteínas Quinasas S6 Ribosómicas 70-kDa/sangre , Sirolimus/administración & dosificación , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Serina-Treonina Quinasas TOR/sangre , Adulto Joven
10.
Cell Res ; 27(10): 1243-1257, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28925386

RESUMEN

DNA modifications such as 5-methylcytosine (5mC) and 5-hydroxymethylcytosine (5hmC) are epigenetic marks known to affect global gene expression in mammals. Given their prevalence in the human genome, close correlation with gene expression and high chemical stability, these DNA epigenetic marks could serve as ideal biomarkers for cancer diagnosis. Taking advantage of a highly sensitive and selective chemical labeling technology, we report here the genome-wide profiling of 5hmC in circulating cell-free DNA (cfDNA) and in genomic DNA (gDNA) of paired tumor and adjacent tissues collected from a cohort of 260 patients recently diagnosed with colorectal, gastric, pancreatic, liver or thyroid cancer and normal tissues from 90 healthy individuals. 5hmC was mainly distributed in transcriptionally active regions coincident with open chromatin and permissive histone modifications. Robust cancer-associated 5hmC signatures were identified in cfDNA that were characteristic for specific cancer types. 5hmC-based biomarkers of circulating cfDNA were highly predictive of colorectal and gastric cancers and were superior to conventional biomarkers and comparable to 5hmC biomarkers from tissue biopsies. Thus, this new strategy could lead to the development of effective, minimally invasive methods for diagnosis and prognosis of cancer from the analyses of blood samples.


Asunto(s)
5-Metilcitosina/análogos & derivados , Biomarcadores de Tumor/sangre , ADN Tumoral Circulante/sangre , Neoplasias/sangre , 5-Metilcitosina/sangre , Adolescente , Adulto , Anciano , Ácidos Nucleicos Libres de Células/sangre , Ácidos Nucleicos Libres de Células/genética , Metilación de ADN/genética , Epigenómica , Femenino , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Biopsia Líquida , Masculino , Persona de Mediana Edad , Neoplasias/clasificación , Neoplasias/genética , Neoplasias/patología , Adulto Joven
12.
Sci Rep ; 5: 17568, 2015 Dec 03.
Artículo en Inglés | MEDLINE | ID: mdl-26631571

RESUMEN

The formation of differentiated cell types from pluripotent progenitors involves epigenetic regulation of gene expression. DNA hydroxymethylation results from the enzymatic oxidation of 5-methylcytosine (5-mC) to 5-hydroxymethylcytosine (5-hmC) by the ten-eleven translocation (TET) 5-mC dioxygenase enzymes. Previous work has mapped changes in 5-mC during differentiation of intestinal stem cells. However, whether or not 5-hmC regulates colonocyte differentiation is unknown. Here we show that 5-hmC regulates gene expression during colonocyte differentiation and controls gene expression in human colon cancers. Genome-wide profiling of 5-hmC during in vitro colonic differentiation demonstrated that 5-hmC is gained at highly expressed and induced genes and is associated with intestinal transcription factor binding sites, including those for HNF4A and CDX2. TET1 induction occurred during differentiation, and TET1 knockdown altered gene expression and inhibited barrier formation of colonocytes. We find that the 5-hmC distribution in primary human colonocytes parallels the distribution found in differentiated cells in vitro, and that gene-specific 5-hmC changes in human colon cancers are directly correlated with changes in gene expression. Our results support a model in which 5-hmC regulates differentiation of adult human intestine and 5-hmC alterations contribute to the disrupted gene expression in colon cancer.


Asunto(s)
Adenocarcinoma/genética , Colon/citología , Neoplasias del Colon/genética , Citosina/análogos & derivados , Oxigenasas de Función Mixta/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , 5-Metilcitosina/análogos & derivados , Adenocarcinoma/metabolismo , Adenocarcinoma/patología , Sitios de Unión , Diferenciación Celular/genética , Línea Celular Tumoral , Colon/metabolismo , Neoplasias del Colon/metabolismo , Neoplasias del Colon/patología , Citosina/metabolismo , Regulación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Oxigenasas de Función Mixta/genética , Proteínas Proto-Oncogénicas/genética , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
13.
Cancer ; 121(7): 1071-8, 2015 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-25424411

RESUMEN

BACKGROUND: Emerging evidence from observational studies has suggested that metformin may be beneficial in the primary prevention of colorectal cancer (CRC). However, to the authors' knowledge, none of these studies was conducted in a US population. Because environmental factors such as Western diet and obesity are implicated in the causation of CRC, a large case-control study was performed to assess the effects of metformin on the incidence of CRC in a US population. METHODS: MarketScan databases were used to identify diabetic patients with CRC. A case was defined as having an incident diagnosis of CRC. Up to 2 controls matched for age, sex, and geographical region were selected for each case. Metformin exposure was assessed by prescription tracking within the 12-month period before the index date. Conditional logistic regression was used to adjust for multiple potential confounders and to calculate adjusted odds ratios (AORs). RESULTS: The mean age of the study participants was 55 years and 57 years, respectively, in the control and case groups (P = 1.0). Approximately 60% of the study participants were male and 40% were female in each group. In the multivariable model, any metformin use was associated with a 15% reduction in the odds of CRC (AOR, 0.85; 95% confidence interval, 0.76-0.95 [P = .007]). After adjusting for health care use, the beneficial effect of metformin was reduced to 12% (AOR, 0.88; 95% confidence interval, 0.77-1.00 [P = .05]). The dose-response analyses demonstrated no significant association with metformin dose, duration, or total exposure. CONCLUSIONS: Metformin use appears to be associated with a reduced risk of developing CRC among diabetic patients in the United States.


Asunto(s)
Neoplasias Colorrectales/prevención & control , Diabetes Mellitus/tratamiento farmacológico , Diabetes Mellitus/fisiopatología , Hipoglucemiantes/uso terapéutico , Metformina/uso terapéutico , Adulto , Estudios de Casos y Controles , Neoplasias Colorrectales/epidemiología , Bases de Datos Factuales , Femenino , Estudios de Seguimiento , Humanos , Incidencia , Estudios Longitudinales , Masculino , Persona de Mediana Edad , Prevención Primaria , Pronóstico , Adulto Joven
14.
Am J Chin Med ; 39(6): 1161-71, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22083988

RESUMEN

American ginseng is a commonly used herbal medicine in the United States. When ginseng is taken orally, its active components, ginsenosides, are reportedly biotransformed by intestinal microbiota. Previous pharmacokinetic evaluations of ginseng in humans have focused on its parent constituents. However, the metabolites, especially those transformed by intestinal microbiota, have not been carefully studied. We used an ultra-performance liquid chromatography/time-of-flight mass spectrometry (UPLC/TOF-MS) method to determine 15 ginsenosides and/or metabolites and their bioavailability in humans. Six healthy human subjects received a single oral dose of 10 g of American ginseng root powder, after which samples of their blood were collected at 0, 2, 4, 7, 9 and 12 h for measurement of ginsenoside/metabolite levels in plasma. Ginsenosides Rb1, Rd, Rg2 and compound K (C-K) were detected in human plasma samples at different time points. The Rb1 concentration peak was 19.90 ± 5.43 ng/ml at 4 h. C-K was detected from 7 h to 12 h with 7.32 ± 1.35 ng/ml at 12 h. Since the last time point was at 12 h, C-K peak level was not observed. The areas under the concentration curves (AUC) from 0 to 12 h were 155.0 ± 19.5 ng⋅h/ml for Rb1 and 26.4 ± 6.4 ng⋅h/ml for C-K, respectively. The gradual decrease of Rb1 levels and the delayed increase in levels of C-K observed in human subjects supported previous reports that enteric microbiota played a key role in transforming Rb1 to C-K.


Asunto(s)
Ginsenósidos/sangre , Ginsenósidos/metabolismo , Panax/química , Administración Oral , Cromatografía Líquida de Alta Presión , Femenino , Ginsenósidos/administración & dosificación , Humanos , Mucosa Intestinal/metabolismo , Masculino , Espectrometría de Masas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...