Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 128
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Nat Immunol ; 20(7): 835-851, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31160797

RESUMEN

How tumor cells genetically lose antigenicity and evade immune checkpoints remains largely elusive. We report that tissue-specific expression of the human long noncoding RNA LINK-A in mouse mammary glands initiates metastatic mammary gland tumors, which phenotypically resemble human triple-negative breast cancer (TNBC). LINK-A expression facilitated crosstalk between phosphatidylinositol-(3,4,5)-trisphosphate and inhibitory G-protein-coupled receptor (GPCR) pathways, attenuating protein kinase A-mediated phosphorylation of the E3 ubiquitin ligase TRIM71. Consequently, LINK-A expression enhanced K48-polyubiquitination-mediated degradation of the antigen peptide-loading complex (PLC) and intrinsic tumor suppressors Rb and p53. Treatment with LINK-A locked nucleic acids or GPCR antagonists stabilized the PLC components, Rb and p53, and sensitized mammary gland tumors to immune checkpoint blockers. Patients with programmed ccll death protein-1(PD-1) blockade-resistant TNBC exhibited elevated LINK-A levels and downregulated PLC components. Hence we demonstrate lncRNA-dependent downregulation of antigenicity and intrinsic tumor suppression, which provides the basis for developing combinational immunotherapy treatment regimens and early TNBC prevention.


Asunto(s)
Presentación de Antígeno/inmunología , Regulación Neoplásica de la Expresión Génica , Neoplasias/genética , Neoplasias/inmunología , Oncogenes , ARN Largo no Codificante/genética , Escape del Tumor/genética , Escape del Tumor/inmunología , Adenoma/genética , Adenoma/metabolismo , Animales , Línea Celular Tumoral , Proliferación Celular , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Humanos , Ratones , Neoplasias/metabolismo , Neoplasias/patología , Fosforilación , Receptores Acoplados a Proteínas G/antagonistas & inhibidores , Microambiente Tumoral/genética , Microambiente Tumoral/inmunología , Proteína p53 Supresora de Tumor/metabolismo , Ubiquitinación , Ensayos Antitumor por Modelo de Xenoinjerto
2.
Cell ; 161(2): 240-54, 2015 Apr 09.
Artículo en Inglés | MEDLINE | ID: mdl-25860607

RESUMEN

In vitro modeling of human disease has recently become feasible with induced pluripotent stem cell (iPSC) technology. Here, we established patient-derived iPSCs from a Li-Fraumeni syndrome (LFS) family and investigated the role of mutant p53 in the development of osteosarcoma (OS). LFS iPSC-derived osteoblasts (OBs) recapitulated OS features including defective osteoblastic differentiation as well as tumorigenic ability. Systematic analyses revealed that the expression of genes enriched in LFS-derived OBs strongly correlated with decreased time to tumor recurrence and poor patient survival. Furthermore, LFS OBs exhibited impaired upregulation of the imprinted gene H19 during osteogenesis. Restoration of H19 expression in LFS OBs facilitated osteoblastic differentiation and repressed tumorigenic potential. By integrating human imprinted gene network (IGN) into functional genomic analyses, we found that H19 mediates suppression of LFS-associated OS through the IGN component DECORIN (DCN). In summary, these findings demonstrate the feasibility of studying inherited human cancer syndromes with iPSCs.


Asunto(s)
Redes Reguladoras de Genes , Células Madre Pluripotentes Inducidas/citología , Síndrome de Li-Fraumeni/complicaciones , Osteosarcoma/etiología , Adolescente , Adulto , Animales , Niño , Decorina/metabolismo , Femenino , Humanos , Síndrome de Li-Fraumeni/genética , Síndrome de Li-Fraumeni/patología , Masculino , Células Madre Mesenquimatosas/metabolismo , Ratones , Modelos Biológicos , Trasplante de Neoplasias , Osteoblastos/citología , Osteoblastos/metabolismo , Osteogénesis , Osteosarcoma/genética , Osteosarcoma/patología , ARN Largo no Codificante/metabolismo , Trasplante Heterólogo , Proteína p53 Supresora de Tumor/metabolismo
3.
Nature ; 580(7804): 530-535, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32322062

RESUMEN

Cancer cells increase lipogenesis for their proliferation and the activation of sterol regulatory element-binding proteins (SREBPs) has a central role in this process. SREBPs are inhibited by a complex composed of INSIG proteins, SREBP cleavage-activating protein (SCAP) and sterols in the endoplasmic reticulum. Regulation of the interaction between INSIG proteins and SCAP by sterol levels is critical for the dissociation of the SCAP-SREBP complex from the endoplasmic reticulum and the activation of SREBPs1,2. However, whether this protein interaction is regulated by a mechanism other than the abundance of sterol-and in particular, whether oncogenic signalling has a role-is unclear. Here we show that activated AKT in human hepatocellular carcinoma (HCC) cells phosphorylates cytosolic phosphoenolpyruvate carboxykinase 1 (PCK1), the rate-limiting enzyme in gluconeogenesis, at Ser90. Phosphorylated PCK1 translocates to the endoplasmic reticulum, where it uses GTP as a phosphate donor to phosphorylate INSIG1 at Ser207 and INSIG2 at Ser151. This phosphorylation reduces the binding of sterols to INSIG1 and INSIG2 and disrupts the interaction between INSIG proteins and SCAP, leading to the translocation of the SCAP-SREBP complex to the Golgi apparatus, the activation of SREBP proteins (SREBP1 or SREBP2) and the transcription of downstream lipogenesis-related genes, proliferation of tumour cells, and tumorigenesis in mice. In addition, phosphorylation of PCK1 at Ser90, INSIG1 at Ser207 and INSIG2 at Ser151 is not only positively correlated with the nuclear accumulation of SREBP1 in samples from patients with HCC, but also associated with poor HCC prognosis. Our findings highlight the importance of the protein kinase activity of PCK1 in the activation of SREBPs, lipogenesis and the development of HCC.


Asunto(s)
Carcinoma Hepatocelular/metabolismo , Gluconeogénesis , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Lipogénesis , Neoplasias Hepáticas/metabolismo , Proteínas de la Membrana/metabolismo , Fosfoenolpiruvato Carboxiquinasa (GTP)/metabolismo , Animales , Carcinogénesis , Carcinoma Hepatocelular/patología , Proliferación Celular , Modelos Animales de Enfermedad , Retículo Endoplásmico/metabolismo , Aparato de Golgi/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intracelular/química , Neoplasias Hepáticas/patología , Masculino , Proteínas de la Membrana/química , Ratones , Ratones Desnudos , Oxiesteroles/metabolismo , Fosforilación , Pronóstico , Unión Proteica , Transporte de Proteínas , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/metabolismo , Proteína 2 de Unión a Elementos Reguladores de Esteroles/metabolismo
4.
Mol Cell ; 71(4): 606-620.e7, 2018 08 16.
Artículo en Inglés | MEDLINE | ID: mdl-30118680

RESUMEN

Metformin has been reported to possess antitumor activity and maintain high cytotoxic T lymphocyte (CTL) immune surveillance. However, the functions and detailed mechanisms of metformin's role in cancer immunity are not fully understood. Here, we show that metformin increases CTL activity by reducing the stability and membrane localization of programmed death ligand-1 (PD-L1). Furthermore, we discover that AMP-activated protein kinase (AMPK) activated by metformin directly phosphorylates S195 of PD-L1. S195 phosphorylation induces abnormal PD-L1 glycosylation, resulting in its ER accumulation and ER-associated protein degradation (ERAD). Consistently, tumor tissues from metformin-treated breast cancer patients exhibit reduced PD-L1 levels with AMPK activation. Blocking the inhibitory signal of PD-L1 by metformin enhances CTL activity against cancer cells. Our findings identify a new regulatory mechanism of PD-L1 expression through the ERAD pathway and suggest that the metformin-CTLA4 blockade combination has the potential to increase the efficacy of immunotherapy.


Asunto(s)
Antineoplásicos/farmacología , Antígeno B7-H1/genética , Antígeno CTLA-4/genética , Regulación Neoplásica de la Expresión Génica , Hipoglucemiantes/farmacología , Metformina/farmacología , Proteínas Quinasas Activadas por AMP/genética , Proteínas Quinasas Activadas por AMP/inmunología , Animales , Antígeno B7-H1/inmunología , Antígeno CTLA-4/inmunología , Línea Celular Tumoral , Retículo Endoplásmico/efectos de los fármacos , Retículo Endoplásmico/genética , Retículo Endoplásmico/metabolismo , Degradación Asociada con el Retículo Endoplásmico , Células Epiteliales/citología , Células Epiteliales/efectos de los fármacos , Células Epiteliales/inmunología , Femenino , Glicosilación , Humanos , Glándulas Mamarias Humanas/citología , Glándulas Mamarias Humanas/efectos de los fármacos , Glándulas Mamarias Humanas/inmunología , Melanoma Experimental/tratamiento farmacológico , Melanoma Experimental/genética , Melanoma Experimental/inmunología , Melanoma Experimental/patología , Ratones , Ratones Endogámicos NOD , Fosforilación , Serina/metabolismo , Linfocitos T Citotóxicos/citología , Linfocitos T Citotóxicos/efectos de los fármacos , Linfocitos T Citotóxicos/inmunología
5.
Mol Cell ; 69(2): 279-291.e5, 2018 01 18.
Artículo en Inglés | MEDLINE | ID: mdl-29351847

RESUMEN

Sustained energy starvation leads to activation of AMP-activated protein kinase (AMPK), which coordinates energy status with numerous cellular processes including metabolism, protein synthesis, and autophagy. Here, we report that AMPK phosphorylates the histone methyltransferase EZH2 at T311 to disrupt the interaction between EZH2 and SUZ12, another core component of the polycomb repressive complex 2 (PRC2), leading to attenuated PRC2-dependent methylation of histone H3 at Lys27. As such, PRC2 target genes, many of which are known tumor suppressors, were upregulated upon T311-EZH2 phosphorylation, which suppressed tumor cell growth both in cell culture and mouse xenografts. Pathologically, immunohistochemical analyses uncovered a positive correlation between AMPK activity and pT311-EZH2, and higher pT311-EZH2 correlates with better survival in both ovarian and breast cancer patients. Our finding suggests that AMPK agonists might be promising sensitizers for EZH2-targeting cancer therapies.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Proteína Potenciadora del Homólogo Zeste 2/metabolismo , Animales , Carcinogénesis/genética , Ciclo Celular , Línea Celular Tumoral , Proliferación Celular , Metilación de ADN , Proteínas de Unión al ADN/metabolismo , Proteína Potenciadora del Homólogo Zeste 2/genética , Proteína Potenciadora del Homólogo Zeste 2/fisiología , Epigénesis Genética , Femenino , Histonas/metabolismo , Humanos , Ratones , Proteínas de Neoplasias , Proteínas Nucleares/metabolismo , Oncogenes , Neoplasias Ováricas/metabolismo , Fosforilación , Complejo Represivo Polycomb 2/metabolismo , Complejo Represivo Polycomb 2/fisiología , Factores de Transcripción , Regulación hacia Arriba
6.
J Biol Chem ; 298(4): 101821, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35283189

RESUMEN

Antibodies that target immune checkpoint proteins such as programmed cell death protein 1, programmed death ligand 1, and cytotoxic T-lymphocyte-associated antigen 4 in human cancers have achieved impressive clinical success; however, a significant proportion of patients fail to respond to these treatments. Galectin-9 (Gal-9), a ß-galactoside-binding protein, has been shown to induce T-cell death and facilitate immunosuppression in the tumor microenvironment by binding to immunomodulatory receptors such as T-cell immunoglobulin and mucin domain-containing molecule 3 and the innate immune receptor dectin-1, suggesting that it may have potential as a target for cancer immunotherapy. Here, we report the development of two novel Gal-9-neutralizing antibodies that specifically react with the N-carbohydrate-recognition domain of human Gal-9 with high affinity. We also show using cell-based functional assays that these antibodies efficiently protected human T cells from Gal-9-induced cell death. Notably, in a T-cell/tumor cell coculture assay of cytotoxicity, these antibodies significantly promoted T cell-mediated killing of tumor cells. Taken together, our findings demonstrate potent inhibition of human Gal-9 by neutralizing antibodies, which may open new avenues for cancer immunotherapy.


Asunto(s)
Anticuerpos Neutralizantes , Muerte Celular , Galectinas , Linfocitos T , Anticuerpos Neutralizantes/metabolismo , Anticuerpos Neutralizantes/farmacología , Muerte Celular/efectos de los fármacos , Galectinas/metabolismo , Humanos , Neoplasias/metabolismo , Neoplasias/terapia , Linfocitos T/efectos de los fármacos , Linfocitos T/metabolismo , Microambiente Tumoral
7.
J Biol Chem ; 298(4): 101817, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35278434

RESUMEN

Expression of the receptor tyrosine kinase ephrin receptor A10 (EphA10), which is undetectable in most normal tissues except for the male testis, has been shown to correlate with tumor progression and poor prognosis in several malignancies, including triple-negative breast cancer (TNBC). Therefore, EphA10 could be a potential therapeutic target, likely with minimal adverse effects. However, no effective clinical drugs against EphA10 are currently available. Here, we report high expression levels of EphA10 in tumor regions of breast, lung, and ovarian cancers as well as in immunosuppressive myeloid cells in the tumor microenvironment. Furthermore, we developed anti-EphA10 monoclonal antibodies (mAbs) that specifically recognize cell surface EphA10, but not other EphA family isoforms, and target tumor regions precisely in vivo with no apparent accumulation in other organs. In syngeneic TNBC mouse models, we found that anti-EphA10 mAb clone #4 enhanced tumor regression, therapeutic response rate, and T cell-mediated antitumor immunity. Notably, the chimeric antigen receptor T cells derived from clone #4 significantly inhibited TNBC cell viability in vitro and tumor growth in vivo. Together, our findings suggest that targeting EphA10 via EphA10 mAbs and EphA10-specific chimeric antigen receptor-T cell therapy may represent a promising strategy for patients with EphA10-positive tumors.


Asunto(s)
Anticuerpos Monoclonales , Receptores Quiméricos de Antígenos , Receptores de la Familia Eph , Linfocitos T , Neoplasias de la Mama Triple Negativas , Animales , Anticuerpos Monoclonales/metabolismo , Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales/uso terapéutico , Antineoplásicos Inmunológicos/uso terapéutico , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Humanos , Ratones , Receptores de la Familia Eph/inmunología , Linfocitos T/metabolismo , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Ensayos Antitumor por Modelo de Xenoinjerto
8.
J Oral Pathol Med ; 52(6): 554-563, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-36807323

RESUMEN

BACKGROUND: Venous malformation (VM) is a kind of congenital vascular anomaly with a high incidence of recurrence, detailed pathogenesis and standard treatment of VM still lack now. Increasing evidence showed exosomal RNA plays a pivotal role in various diseases. However, the underlying mechanism of VM based on the potential differentially exosomal RNAs remains unclear. METHODS: Comparative high-throughput sequencing with serum exosomes from three VM patients and three healthy donors was used to explore differentially expressed (DE) circRNAs, DE lncRNAs, and DE mRNAs involving the formation of VM. We identified and verified DE circRNAs, DE lncRNAs, and DE mRNAs via qRT-PCR assay. We explored the potential functions of these exosomal DE non-coding RNAs via performing further Gene Ontology and Kyoto Encyclopedia of Genes and Genomes enrichment analysis. Besides, circRNA/lncRNA-miRNA-mRNA linkages were also constructed to find their potential relationships in VM. RESULTS: A total of 121 circRNAs, 53 lncRNAs, and 42 mRNAs (|log2 FC| ≥ 2.0, FDR <0.05, n = 3) were determined to be differentially expressed. QRT-PCR validated that these top-changed DE circRNAs, lncRNAs, and mRNAs had significant expression changes. Functional studies demonstrated that DE circRNAs play a pivotal role in thyroid hormone signaling pathway, DE lncRNAs function as a key regulator in MAPK signaling pathway and DE miRNAs participate in the process of hepatocellular carcinoma mostly. CONCLUSION: Our study comprehensively depicted exosomal DE non-coding RNAs networks related to the pathogenesis of VM which can provide new insight into, a novel target for treating VM.


Asunto(s)
MicroARNs , ARN Largo no Codificante , Humanos , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Circular/genética , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Perfilación de la Expresión Génica , MicroARNs/genética , MicroARNs/metabolismo
9.
J Oral Pathol Med ; 51(10): 911-919, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-35854627

RESUMEN

BACKGROUNDS: Head and neck vascular malformation (HNVM) is a highly complex congenital condition that is difficult to diagnose, monitor and treat. Therefore, it is critical to explore serum cytokines that may be related to its pathology and prognosis. METHODS: An antibody-based microarray was used to examine the expression of 31 angiogenic cytokines in 11 HNVM patients relative to 11 healthy subjects. ELISA was used to verify the results. We performed Gene Ontology and Kyoto Encyclopedia of Genes and Genomes pathway analyses of the differentially expressed cytokines (DECs). Additionally, we explored the function of DECs in human umbilical vein endothelial cells (HUVECs) in vitro via CCK-8, wound healing, transwell and tube formation assays. RESULTS: Expression of interleukin (IL)-10, matrix metallopeptidase-9 (MMP-9) and vascular endothelial growth factor receptor 2 (VEGF-R2) in HNVM patients was significantly higher, whereas levels of IL-12p40 and angiostatin were significantly lower in HNVM patients relative to healthy controls (p < 0.05). However, ELISA only verified that IL-10, MMP-9, VEGF-R2 and IL-12p40 had significant expression changes. Functional enrichment analysis revealed DECs mainly participated in the RAS signalling pathway. Functional studies demonstrated that IL-10, MMP-9 and VEGF-R2 promote cell proliferation, migration, invasion and tube formation, while IL-12p40 inhibited these processes in HUVECs. CONCLUSIONS: The present study not only indicates that IL-10, MMP-9, VEGF-R2 and IL-12p40 may participate in the development of HNVMs but also provides a theoretical basis for the discovery of new targeted molecules in the treatment of HNVMs.


Asunto(s)
Factor A de Crecimiento Endotelial Vascular , Malformaciones Vasculares , Humanos , Factor A de Crecimiento Endotelial Vascular/metabolismo , Interleucina-10/metabolismo , Movimiento Celular , Metaloproteinasa 9 de la Matriz/metabolismo , Subunidad p40 de la Interleucina-12/metabolismo , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Malformaciones Vasculares/metabolismo , Citocinas/metabolismo
10.
J Hepatol ; 74(4): 907-918, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33031845

RESUMEN

BACKGROUND & AIMS: There are currently limited therapeutic options for hepatocellular carcinoma (HCC), particularly when it is diagnosed at advanced stages. Herein, we examined the pathophysiological role of ROS1 and assessed the utility of ROS1-targeted therapy for the treatment of HCC. METHODS: Recombinant ribonucleases (RNases) were purified, and the ligand-receptor relationship between RNase7 and ROS1 was validated in HCC cell lines by Duolink, immunofluorescence, and immunoprecipitation assays. Potential interacting residues between ROS1 and RNase7 were predicted using a protein-protein docking approach. The oncogenic function of RNase7 was analyzed by cell proliferation, migration and invasion assays, and a xenograft mouse model. The efficacy of anti-ROS1 inhibitor treatment was evaluated in patient-derived xenograft (PDX) and orthotopic models. Two independent patient cohorts were analyzed to evaluate the pathological relevance of RNase7/ROS1. RESULTS: RNase7 associated with ROS1's N3-P2 domain and promoted ROS1-mediated oncogenic transformation. Patients with HCC exhibited elevated plasma RNase7 levels compared with healthy individuals. High ROS1 and RNase7 expression were strongly associated with poor prognosis in patients with HCC. In both HCC PDX and orthotopic mouse models, ROS1 inhibitor treatment markedly suppressed RNase7-induced tumorigenesis, leading to decreased plasma RNase7 levels and tumor shrinkage in mice. CONCLUSIONS: RNase7 serves as a high-affinity ligand for ROS1. Plasma RNase7 could be used as a biomarker to identify patients with HCC who may benefit from anti-ROS1 treatment. LAY SUMMARY: Receptor tyrosine kinases are known to be involved in tumorigenesis and have been targeted therapeutically for a number of cancers, including hepatocellular carcinoma. ROS1 is the only such receptor with kinase activity whose ligand has not been identified. Herein, we show that RNase7 acts as a ligand to activate ROS1 signaling. This has important pathophysiological and therapeutic implications. Anti-ROS1 inhibitors could be used to treatment patients with hepatocellular carcinoma and high RNase7 levels.


Asunto(s)
Carcinogénesis , Carcinoma Hepatocelular , Crizotinib/farmacología , Neoplasias Hepáticas , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Tirosina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Tirosina Quinasas Receptoras/metabolismo , Ribonucleasas/metabolismo , Animales , Biomarcadores de Tumor/metabolismo , Carcinogénesis/efectos de los fármacos , Carcinogénesis/metabolismo , Carcinoma Hepatocelular/tratamiento farmacológico , Carcinoma Hepatocelular/metabolismo , Ensayos de Migración Celular/métodos , Proliferación Celular/efectos de los fármacos , Perfilación de la Expresión Génica/métodos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Ligandos , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/metabolismo , Ratones , Proteínas Tirosina Quinasas Receptoras/antagonistas & inhibidores , Ensayos Antitumor por Modelo de Xenoinjerto
11.
Genes Dev ; 27(3): 274-87, 2013 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-23388826

RESUMEN

Steroid receptor coactivator 3 (SRC-3) is an oncogenic nuclear receptor coactivator that plays a significant role in drug resistance. Using a lentiviral cDNA library rescue screening approach, we identified a SRC-3 downstream gene-TRAF4 (tumor necrosis factor [TNF] receptor associated-factor 4)-that functions in cell resistance to cytotoxic stress. TRAF4 expression is positively correlated with SRC-3 expression in human breast cancers. Similar to that observed for SRC-3 overexpression, breast cancer cells overexpressing TRAF4 are more resistant to stress-induced death. Here, we further dissected the underlying molecular mechanism for SRC-3 and TRAF4-mediated resistance to cytotoxic agents. We observed that SRC-3 expression is inversely correlated with the expression of p53-regulated proapoptotic genes in breast cancers and further found that SRC-3 and TRAF4 overexpression diminished cytotoxic stress-induced up-regulation of the tumor suppressor p53 protein. To determine the mechanism, we showed that the TRAF domain of TRAF4 bound to the N-terminal TRAF-like region of the deubiquitinase HAUSP (herpesvirus-associated ubiquitin-specific protease; also named USP7) and blocked the access of p53 to the same region of HAUSP. This TRAF4-mediated inhibition of HAUSP then led to the loss of p53 deubiquitination and its stabilization in response to cellular stress. Consistent with this cellular function, we also found that TRAF4 overexpression in breast cancer patients was associated significantly with poor prognosis. Because of SRC-3's ability to abrogate p53 function, our results suggest that SRC-3 overexpression may be especially important in tumors in which p53 is not mutated.


Asunto(s)
Coactivador 3 de Receptor Nuclear/metabolismo , Estrés Fisiológico/fisiología , Factor 4 Asociado a Receptor de TNF/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Animales , Antineoplásicos/farmacología , Neoplasias de la Mama/mortalidad , Neoplasias de la Mama/patología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Endopeptidasas/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Células HEK293 , Humanos , Ratones , Coactivador 3 de Receptor Nuclear/genética , Unión Proteica , Estabilidad Proteica , Peptidasa Específica de Ubiquitina 7 , Proteasas Ubiquitina-Específicas , Ubiquitinación
12.
Gastroenterology ; 156(6): 1849-1861.e13, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30711629

RESUMEN

BACKGROUND & AIMS: Inhibitors of MET have not produced satisfactory outcomes in trials of patients with liver cancer. We investigated the mechanisms of liver tumor resistance to MET inhibitors in mice. METHODS: We tested the effects of MET inhibitors tivantinib and capmatinib in the mouse hepatocellular carcinoma (HCC) cell line HCA-1 and in immune-competent and immunodeficient mice with subcutaneous tumors grown from this cell line. Tumors were collected from mice and tumor cells were analyzed by time-of-flight mass cytometry. We used short hairpin RNAs to weaken expression of MET in Hep3B, SK-HEP-1, HA59T, and HA22T liver cancer cell lines and analyzed cells by immunoblot, immunofluorescence, and immunoprecipitation assays. Mass spectrometry was used to assess interactions between MET and glycogen synthase kinase 3ß (GSK3B), and GSK3B phosphorylation, in liver cancer cell lines. C57/BL6 mice with orthotopic tumors grown from Hep1-6 cells were given combinations of capmatinib or tivantinib and antibodies against programmed cell death 1 (PDCD1; also called PD1); tumors were collected and analyzed by immunofluorescence. We analyzed 268 HCCsamples in a tissue microarray by immunohistochemistry. RESULTS: Exposure of liver cancer cell lines to MET inhibitors increased their expression of PD ligand 1 (PDL1) and inactivated cocultured T cells. MET phosphorylated and activated GSK3B at tyrosine 56, which decreased the expression of PDL1 by liver cancer cells. In orthotopic tumors grown in immune-competent mice, MET inhibitors decreased the antitumor activity of T cells. However, addition of anti-PD1 decreased orthotopic tumor growth and prolonged survival of mice compared with anti-PD1 or MET inhibitors alone. Tissue microarray analysis of HCC samples showed an inverse correlation between levels of MET and PDL1 and a positive correlation between levels of MET and phosphorylated GSK3B. CONCLUSIONS: In studies of liver cancer cell lines and mice with orthotopic tumors, MET mediated phosphorylation and activated GSK3B, leading to decreased expression of PDL1. Combined with a MET inhibitor, anti-PD1 and anti-PDL1 produced additive effect to slow growth of HCCs in mice.


Asunto(s)
Antígeno B7-H1/metabolismo , Carcinoma Hepatocelular/enzimología , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Neoplasias Hepáticas/enzimología , Proteínas Proto-Oncogénicas c-met/metabolismo , Factor 6 Asociado a Receptor de TNF/metabolismo , Escape del Tumor/efectos de los fármacos , Animales , Anticuerpos Monoclonales/uso terapéutico , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Antígeno B7-H1/antagonistas & inhibidores , Antígeno B7-H1/inmunología , Benzamidas , Carcinoma Hepatocelular/tratamiento farmacológico , Carcinoma Hepatocelular/inmunología , Línea Celular Tumoral , Regulación hacia Abajo , Granzimas/metabolismo , Imidazoles/farmacología , Imidazoles/uso terapéutico , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/inmunología , Masculino , Ratones , Fosforilación , Proteínas Proto-Oncogénicas c-met/antagonistas & inhibidores , Pirrolidinonas/farmacología , Pirrolidinonas/uso terapéutico , Quinolinas/farmacología , Quinolinas/uso terapéutico , Factor 6 Asociado a Receptor de TNF/inmunología , Triazinas/farmacología , Triazinas/uso terapéutico , Ubiquitinación
13.
Mol Cell ; 45(2): 171-84, 2012 Jan 27.
Artículo en Inglés | MEDLINE | ID: mdl-22196886

RESUMEN

Proinflammatory cytokine TNFα plays critical roles in promoting malignant cell proliferation, angiogenesis, and tumor metastasis in many cancers. However, the mechanism of TNFα-mediated tumor development remains unclear. Here, we show that IKKα, an important downstream kinase of TNFα, interacts with and phosphorylates FOXA2 at S107/S111, thereby suppressing FOXA2 transactivation activity and leading to decreased NUMB expression, and further activates the downstream NOTCH pathway and promotes cell proliferation and tumorigenesis. Moreover, we found that levels of IKKα, pFOXA2 (S107/111), and activated NOTCH1 were significantly higher in hepatocellular carcinoma tumors than in normal liver tissues and that pFOXA2 (S107/111) expression was positively correlated with IKKα and activated NOTCH1 expression in tumor tissues. Therefore, dysregulation of NUMB-mediated suppression of NOTCH1 by TNFα/IKKα-associated FOXA2 inhibition likely contributes to inflammation-mediated cancer pathogenesis. Here, we report a TNFα/IKKα/FOXA2/NUMB/NOTCH1 pathway that is critical for inflammation-mediated tumorigenesis and may provide a target for clinical intervention in human cancer.


Asunto(s)
Carcinoma Hepatocelular/metabolismo , Transformación Celular Neoplásica/metabolismo , Factor Nuclear 3-beta del Hepatocito/genética , Quinasa I-kappa B/metabolismo , Neoplasias Hepáticas/metabolismo , Receptor Notch1/metabolismo , Animales , Carcinoma Hepatocelular/patología , Proliferación Celular , Regulación Neoplásica de la Expresión Génica , Factor Nuclear 3-beta del Hepatocito/metabolismo , Humanos , Neoplasias Hepáticas Experimentales/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Modelos Biológicos , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Fosforilación , Receptor Notch1/genética , Transducción de Señal , Factor de Necrosis Tumoral alfa/metabolismo
14.
Nature ; 497(7449): 383-7, 2013 May 16.
Artículo en Inglés | MEDLINE | ID: mdl-23636329

RESUMEN

MicroRNAs (miRNAs) are generated by two-step processing to yield small RNAs that negatively regulate target gene expression at the post-transcriptional level. Deregulation of miRNAs has been linked to diverse pathological processes, including cancer. Recent studies have also implicated miRNAs in the regulation of cellular response to a spectrum of stresses, such as hypoxia, which is frequently encountered in the poorly angiogenic core of a solid tumour. However, the upstream regulators of miRNA biogenesis machineries remain obscure, raising the question of how tumour cells efficiently coordinate and impose specificity on miRNA expression and function in response to stresses. Here we show that epidermal growth factor receptor (EGFR), which is the product of a well-characterized oncogene in human cancers, suppresses the maturation of specific tumour-suppressor-like miRNAs in response to hypoxic stress through phosphorylation of argonaute 2 (AGO2) at Tyr 393. The association between EGFR and AGO2 is enhanced by hypoxia, leading to elevated AGO2-Y393 phosphorylation, which in turn reduces the binding of Dicer to AGO2 and inhibits miRNA processing from precursor miRNAs to mature miRNAs. We also identify a long-loop structure in precursor miRNAs as a critical regulatory element in phospho-Y393-AGO2-mediated miRNA maturation. Furthermore, AGO2-Y393 phosphorylation mediates EGFR-enhanced cell survival and invasiveness under hypoxia, and correlates with poorer overall survival in breast cancer patients. Our study reveals a previously unrecognized function of EGFR in miRNA maturation and demonstrates how EGFR is likely to function as a regulator of AGO2 through novel post-translational modification. These findings suggest that modulation of miRNA biogenesis is important for stress response in tumour cells and has potential clinical implications.


Asunto(s)
Proteínas Argonautas/química , Proteínas Argonautas/metabolismo , Hipoxia de la Célula/fisiología , Receptores ErbB/metabolismo , MicroARNs/metabolismo , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/mortalidad , Neoplasias de la Mama/patología , Hipoxia de la Célula/genética , Línea Celular Tumoral , Supervivencia Celular , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , MicroARNs/biosíntesis , MicroARNs/química , MicroARNs/genética , Invasividad Neoplásica , Conformación de Ácido Nucleico , Fosforilación , Fosfotirosina/metabolismo , Pronóstico , Unión Proteica , Precursores del ARN/química , Precursores del ARN/genética , Precursores del ARN/metabolismo , Ribonucleasa III/metabolismo , Análisis de Supervivencia
16.
Mol Cell ; 36(1): 131-40, 2009 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-19818716

RESUMEN

IkappaB kinase beta (IKKbeta) is involved in tumor development and progression through activation of the nuclear factor (NF)-kappaB pathway. However, the molecular mechanism that regulates IKKbeta degradation remains largely unknown. Here, we show that a Cullin 3 (CUL3)-based ubiquitin ligase, Kelch-like ECH-associated protein 1 (KEAP1), is responsible for IKKbeta ubiquitination. Depletion of KEAP1 led to the accumulation and stabilization of IKKbeta and to upregulation of NF-kappaB-derived tumor angiogenic factors. A systematic analysis of the CUL3, KEAP1, and RBX1 genomic loci revealed a high percentage of genome loss and missense mutations in human cancers that failed to facilitate IKKbeta degradation. Our results suggest that the dysregulation of KEAP1-mediated IKKbeta ubiquitination may contribute to tumorigenesis.


Asunto(s)
Quinasa I-kappa B/metabolismo , Péptidos y Proteínas de Señalización Intracelular/fisiología , FN-kappa B/metabolismo , Transducción de Señal/fisiología , Animales , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Línea Celular , Línea Celular Tumoral , Proteínas Cullin/genética , Proteínas Cullin/metabolismo , Variaciones en el Número de Copia de ADN/genética , Femenino , Expresión Génica/efectos de los fármacos , Expresión Génica/genética , Humanos , Quinasa I-kappa B/genética , Interleucina-8/genética , Estimación de Kaplan-Meier , Proteína 1 Asociada A ECH Tipo Kelch , Ratones , Mutación/fisiología , Neoplasias/genética , Neoplasias/metabolismo , Neovascularización Fisiológica/genética , Unión Proteica/fisiología , Dominios y Motivos de Interacción de Proteínas/fisiología , ARN Interferente Pequeño/genética , Transducción de Señal/efectos de los fármacos , Factor de Transcripción ReIA/metabolismo , Transfección , Factor de Necrosis Tumoral alfa/farmacología , Ubiquitinación/fisiología
17.
Nature ; 468(7326): 927-32, 2010 Dec 16.
Artículo en Inglés | MEDLINE | ID: mdl-21164480

RESUMEN

Recognition of modified histone species by distinct structural domains within 'reader' proteins plays a critical role in the regulation of gene expression. Readers that simultaneously recognize histones with multiple marks allow transduction of complex chromatin modification patterns into specific biological outcomes. Here we report that chromatin regulator tripartite motif-containing 24 (TRIM24) functions in humans as a reader of dual histone marks by means of tandem plant homeodomain (PHD) and bromodomain (Bromo) regions. The three-dimensional structure of the PHD-Bromo region of TRIM24 revealed a single functional unit for combinatorial recognition of unmodified H3K4 (that is, histone H3 unmodified at lysine 4, H3K4me0) and acetylated H3K23 (histone H3 acetylated at lysine 23, H3K23ac) within the same histone tail. TRIM24 binds chromatin and oestrogen receptor to activate oestrogen-dependent genes associated with cellular proliferation and tumour development. Aberrant expression of TRIM24 negatively correlates with survival of breast cancer patients. The PHD-Bromo of TRIM24 provides a structural rationale for chromatin activation through a non-canonical histone signature, establishing a new route by which chromatin readers may influence cancer pathogenesis.


Asunto(s)
Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Proteínas Portadoras/metabolismo , Regulación Neoplásica de la Expresión Génica , Histonas/metabolismo , Acetilación , Neoplasias de la Mama/patología , Proteínas Portadoras/química , Proteínas Portadoras/genética , Línea Celular Tumoral , Cromatina/metabolismo , Ensamble y Desensamble de Cromatina , Cristalografía por Rayos X , Receptor alfa de Estrógeno/metabolismo , Estrógenos/metabolismo , Regulación Neoplásica de la Expresión Génica/genética , Células HEK293 , Histonas/química , Humanos , Metilación , Análisis por Matrices de Proteínas , Unión Proteica , Estructura Terciaria de Proteína , Especificidad por Sustrato , Tasa de Supervivencia
18.
Cancer Cell ; 12(1): 52-65, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17613436

RESUMEN

Pancreatic cancer is an aggressive malignancy with morbidity rates almost equal to mortality rates because of the current lack of effective treatment options. Here, we describe a targeted approach to treating pancreatic cancer with effective therapeutic efficacy and safety in noninvasive imaging models. We developed a versatile expression vector "VISA" (VP16-GAL4-WPRE integrated systemic amplifier) and a CCKAR (cholecystokinin type A receptor) gene-based, pancreatic-cancer-specific promoter VISA (CCKAR-VISA) composite to target transgene expression in pancreatic tumors in vivo. Targeted expression of BikDD, a potent proapoptotic gene driven by CCKAR-VISA, exhibited significant antitumor effects on pancreatic cancer and prolonged survival in multiple xenograft and syngeneic orthotopic mouse models of pancreatic tumors with virtually no toxicity.


Asunto(s)
Modelos Biológicos , Neoplasias Pancreáticas/genética , Animales , Ratones , Ratones Endogámicos C57BL , Receptores de Colecistoquinina/genética , Transgenes
19.
Mol Carcinog ; 53(12): 1011-26, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24000122

RESUMEN

Metastatic dissemination is a multi-step process that depends on cancer cells' ability to respond to microenvironmental cues by adapting adhesion abilities and undergoing cytoskeletal rearrangement. Breast Cancer Metastasis Suppressor 1 (BRMS1) affects several steps of the metastatic cascade: it decreases survival in circulation, increases susceptibility to anoikis, and reduces capacity to colonize secondary organs. In this report, BRMS1 expression is shown to not significantly alter expression levels of integrin monomers, while time-lapse and confocal microscopy revealed that BRMS1-expressing cells exhibited reduced activation of both ß1 integrin and focal adhesion kinase, and decreased localization of these molecules to sites of focal adhesions. Short-term plating of BRMS1-expressing cells onto collagen or fibronectin markedly decreased cytoskeletal reorganization and formation of cellular adhesion projections. Under 3D culture conditions, BRMS1-expressing cells remained rounded and failed to reorganize their cytoskeleton and form invasive colonies. Taken together, BRMS1-expressing breast cancer cells are greatly attenuated in their ability to respond to microenvironment changes. © 2013 Wiley Periodicals, Inc.


Asunto(s)
Neoplasias de la Mama/genética , Adhesión Celular/genética , Metástasis de la Neoplasia/genética , Proteínas de Neoplasias/genética , Línea Celular Tumoral , Citoesqueleto/genética , Femenino , Proteína-Tirosina Quinasas de Adhesión Focal/genética , Humanos , Integrina beta1/genética , Proteínas Represoras
20.
Nat Commun ; 15(1): 1009, 2024 Feb 02.
Artículo en Inglés | MEDLINE | ID: mdl-38307859

RESUMEN

Tumor-secreted factors contribute to the development of a microenvironment that facilitates the escape of cancer cells from immunotherapy. In this study, we conduct a retrospective comparison of the proteins secreted by hepatocellular carcinoma (HCC) cells in responders and non-responders among a cohort of ten patients who received Nivolumab (anti-PD-1 antibody). Our findings indicate that non-responders have a high abundance of secreted RNase1, which is associated with a poor prognosis in various cancer types. Furthermore, mice implanted with HCC cells that overexpress RNase1 exhibit immunosuppressive tumor microenvironments and diminished response to anti-PD-1 therapy. RNase1 induces the polarization of macrophages towards a tumor growth-promoting phenotype through activation of the anaplastic lymphoma kinase (ALK) signaling pathway. Targeting the RNase1/ALK axis reprograms the macrophage polarization, with increased CD8+ T- and Th1- cell recruitment. Moreover, simultaneous targeting of the checkpoint protein PD-1 unleashes cytotoxic CD8+ T-cell responses. Treatment utilizing both an ALK inhibitor and an anti-PD-1 antibody exhibits enhanced tumor regression and facilitates long-term immunity. Our study elucidates the role of RNase1 in mediating tumor resistance to immunotherapy and reveals an RNase1-mediated immunosuppressive tumor microenvironment, highlighting the potential of targeting RNase1 as a promising strategy for cancer immunotherapy in HCC.


Asunto(s)
Carcinoma Hepatocelular , Neoplasias Hepáticas , Animales , Humanos , Ratones , Quinasa de Linfoma Anaplásico , Carcinoma Hepatocelular/metabolismo , Linfocitos T CD8-positivos , Terapia de Inmunosupresión , Neoplasias Hepáticas/metabolismo , Estudios Retrospectivos , Ribonucleasas , Microambiente Tumoral
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA