Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
2.
Front Oncol ; 12: 825284, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35402280

RESUMO

Tumor cells are eliminated by the immune system, including T lymphocytes and natural killer cells; however, many types of tumor cells acquire the immune tolerance by inhibiting T-cell activation and functions via immune checkpoint molecules. Immunotherapy targeting immune checkpoint molecules such as Programmed death receptor 1 (PD-1)/Programmed death ligand 1 (PD-L1) and cytotoxic T lymphocyte associated protein 4 (CTLA-4) have shown successful outcomes for multiple cancer treatments, however some patients show the lack of durable responses. Thus, discovering the chemical compounds or drugs manipulating the expression or function of immune checkpoint molecules are anticipated to overcome the drug resistance of immune checkpoint inhibitors. Function of inhibitory immune checkpoint molecules is often dysregulated by the transcriptional and post-translational levels in tumors. Here, this review focuses on the post-translational modification of intrinsic PD-L1 functions and regulators for PD-L1 transcription.

3.
Nat Cell Biol ; 22(9): 1064-1075, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32839551

RESUMO

Immunotherapies that target programmed cell death protein 1 (PD-1) and its ligand PD-L1 as well as cytotoxic T-lymphocyte-associated protein 4 (CTLA4) have shown impressive clinical outcomes for multiple tumours. However, only a subset of patients achieves durable responses, suggesting that the mechanisms of the immune checkpoint pathways are not completely understood. Here, we report that PD-L1 translocates from the plasma membrane into the nucleus through interactions with components of the endocytosis and nucleocytoplasmic transport pathways, regulated by p300-mediated acetylation and HDAC2-dependent deacetylation of PD-L1. Moreover, PD-L1 deficiency leads to compromised expression of multiple immune-response-related genes. Genetically or pharmacologically modulating PD-L1 acetylation blocks its nuclear translocation, reprograms the expression of immune-response-related genes and, as a consequence, enhances the anti-tumour response to PD-1 blockade. Thus, our results reveal an acetylation-dependent regulation of PD-L1 nuclear localization that governs immune-response gene expression, and thereby advocate targeting PD-L1 translocation to enhance the efficacy of PD-1/PD-L1 blockade.


Assuntos
Antígeno B7-H1/metabolismo , Núcleo Celular/metabolismo , Receptor de Morte Celular Programada 1/metabolismo , Acetilação , Animais , Linhagem Celular , Linhagem Celular Tumoral , Proteína p300 Associada a E1A/metabolismo , Expressão Gênica/fisiologia , Células HEK293 , Humanos , Imunoterapia/métodos , Células MCF-7 , Camundongos , Neoplasias/metabolismo , Processamento de Proteína Pós-Traducional/fisiologia , Células RAW 264.7
4.
Nature ; 571(7766): E10, 2019 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-31270456

RESUMO

An Amendment to this paper has been published and can be accessed via a link at the top of the paper. The original Letter has not been corrected.

5.
Cell Signal ; 46: 15-22, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29474981

RESUMO

FBW7 is one of the most well characterized F-box proteins that serve as substrate recognition subunits of SCF (Skp1-Cullin 1-F-box proteins) E3 ubiquitin ligase complexes. SCFFBW7 plays key roles in regulating cell cycle progression, differentiation, and stem cell maintenance largely through targeting a broad range of oncogenic substrates for proteasome-dependent degradation. The identification of an increasing number of FBW7 substrates for ubiquitination, and intensive in vitro and in vivo studies have revealed a network of signaling components controlled by FBW7 that contributes to metabolic regulation as well as its tumor suppressor role. Here we mainly focus on recent findings that highlight a critical role for FBW7 in cancer and metabolism.


Assuntos
Proteína 7 com Repetições F-Box-WD/fisiologia , Neoplasias/metabolismo , Ubiquitinação , Animais , Ciclo Celular , Diferenciação Celular , Linhagem Celular Tumoral , Proteína 7 com Repetições F-Box-WD/genética , Humanos , Camundongos , Fosforilação , Transdução de Sinais , Proteínas Supressoras de Tumor/fisiologia
6.
Nature ; 553(7686): 91-95, 2018 01 04.
Artigo em Inglês | MEDLINE | ID: mdl-29160310

RESUMO

Treatments that target immune checkpoints, such as the one mediated by programmed cell death protein 1 (PD-1) and its ligand PD-L1, have been approved for treating human cancers with durable clinical benefit. However, many patients with cancer fail to respond to compounds that target the PD-1 and PD-L1 interaction, and the underlying mechanism(s) is not well understood. Recent studies revealed that response to PD-1-PD-L1 blockade might correlate with PD-L1 expression levels in tumour cells. Hence, it is important to understand the mechanistic pathways that control PD-L1 protein expression and stability, which can offer a molecular basis to improve the clinical response rate and efficacy of PD-1-PD-L1 blockade in patients with cancer. Here we show that PD-L1 protein abundance is regulated by cyclin D-CDK4 and the cullin 3-SPOP E3 ligase via proteasome-mediated degradation. Inhibition of CDK4 and CDK6 (hereafter CDK4/6) in vivo increases PD-L1 protein levels by impeding cyclin D-CDK4-mediated phosphorylation of speckle-type POZ protein (SPOP) and thereby promoting SPOP degradation by the anaphase-promoting complex activator FZR1. Loss-of-function mutations in SPOP compromise ubiquitination-mediated PD-L1 degradation, leading to increased PD-L1 levels and reduced numbers of tumour-infiltrating lymphocytes in mouse tumours and in primary human prostate cancer specimens. Notably, combining CDK4/6 inhibitor treatment with anti-PD-1 immunotherapy enhances tumour regression and markedly improves overall survival rates in mouse tumour models. Our study uncovers a novel molecular mechanism for regulating PD-L1 protein stability by a cell cycle kinase and reveals the potential for using combination treatment with CDK4/6 inhibitors and PD-1-PD-L1 immune checkpoint blockade to enhance therapeutic efficacy for human cancers.


Assuntos
Antígeno B7-H1/metabolismo , Proteínas Culina/metabolismo , Ciclina D/metabolismo , Quinase 4 Dependente de Ciclina/metabolismo , Vigilância Imunológica , Neoplasias/imunologia , Proteínas Nucleares/metabolismo , Proteínas Repressoras/metabolismo , Evasão Tumoral/imunologia , Proteínas 14-3-3/metabolismo , Animais , Antígeno B7-H1/biossíntese , Proteínas Cdh1/metabolismo , Ciclo Celular , Linhagem Celular , Quinase 4 Dependente de Ciclina/antagonistas & inibidores , Quinase 6 Dependente de Ciclina/antagonistas & inibidores , Feminino , Humanos , Linfócitos do Interstício Tumoral/citologia , Linfócitos do Interstício Tumoral/imunologia , Masculino , Camundongos , Proteínas Nucleares/química , Fosforilação , Receptor de Morte Celular Programada 1/metabolismo , Neoplasias da Próstata/imunologia , Complexo de Endopeptidases do Proteassoma/metabolismo , Estabilidade Proteica , Proteólise , Proteínas Repressoras/química
7.
Nat Cell Biol ; 19(3): 177-188, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-28192421

RESUMO

Progression of mammalian cells through the G1 and S phases of the cell cycle is driven by the D-type and E-type cyclins. According to the current models, at least one of these cyclin families must be present to allow cell proliferation. Here, we show that several cell types can proliferate in the absence of all G1 cyclins. However, following ablation of G1 cyclins, embryonic stem (ES) cells attenuated their pluripotent characteristics, with the majority of cells acquiring the trophectodermal cell fate. We established that G1 cyclins, together with their associated cyclin-dependent kinases (CDKs), phosphorylate and stabilize the core pluripotency factors Nanog, Sox2 and Oct4. Treatment of murine ES cells, patient-derived glioblastoma tumour-initiating cells, or triple-negative breast cancer cells with a CDK inhibitor strongly decreased Sox2 and Oct4 levels. Our findings suggest that CDK inhibition might represent an attractive therapeutic strategy by targeting glioblastoma tumour-initiating cells, which depend on Sox2 to maintain their tumorigenic potential.


Assuntos
Diferenciação Celular , Ciclina G1/metabolismo , Células-Tronco Embrionárias Murinas/citologia , Células-Tronco Embrionárias Murinas/metabolismo , Animais , Biomarcadores/metabolismo , Ciclo Celular , Proliferação de Células , Separação Celular , DNA/análise , Embrião de Mamíferos/citologia , Epigênese Genética , Feminino , Citometria de Fluxo , Perfilação da Expressão Gênica , Histonas/metabolismo , Hormônios/farmacologia , Imageamento Tridimensional , Lisina/metabolismo , Glândulas Mamárias Animais/citologia , Glândulas Mamárias Animais/embriologia , Metilação , Camundongos , Camundongos Endogâmicos C57BL , RNA/análise , Receptores Acoplados a Proteínas G/metabolismo , Esteroides/farmacologia , Tetraspaninas/metabolismo
8.
Sci Signal ; 10(460)2017 01 03.
Artigo em Inglês | MEDLINE | ID: mdl-28049764

RESUMO

The SCFß-TRCP E3 ubiquitin ligase complex plays pivotal roles in normal cellular physiology and in pathophysiological conditions. Identification of ß-transducin repeat-containing protein (ß-TRCP) substrates is therefore critical to understand SCFß-TRCP biology and function. We used a ß-TRCP-phosphodegron motif-specific antibody in a ß-TRCP substrate screen coupled with tandem mass spectrometry and identified multiple ß-TRCP substrates. One of these substrates was Lipin1, an enzyme and suppressor of the family of sterol regulatory element-binding protein (SREBP) transcription factors, which activate genes encoding lipogenic factors. We showed that SCFß-TRCP specifically interacted with and promoted the polyubiquitination of Lipin1 in a manner that required phosphorylation of Lipin1 by mechanistic target of rapamycin 1 (mTORC1) and casein kinase I (CKI). ß-TRCP depletion in HepG2 hepatocellular carcinoma cells resulted in increased Lipin1 protein abundance, suppression of SREBP-dependent gene expression, and attenuation of triglyceride synthesis. Moreover, ß-TRCP1 knockout mice showed increased Lipin1 protein abundance and were protected from hepatic steatosis induced by a high-fat diet. Together, these data reveal a critical physiological function of ß-TRCP in regulating hepatic lipid metabolic homeostasis in part through modulating Lipin1 stability.


Assuntos
Lipogênese , Fígado/metabolismo , Proteínas Nucleares/metabolismo , Fosfatidato Fosfatase/metabolismo , Proteínas Ligases SKP Culina F-Box/metabolismo , Animais , Linhagem Celular Tumoral , Regulação da Expressão Gênica , Células HEK293 , Células HeLa , Células Hep G2 , Humanos , Immunoblotting , Camundongos , Camundongos Knockout , Células NIH 3T3 , Proteínas Nucleares/genética , Fosfatidato Fosfatase/genética , Fosforilação , Ligação Proteica , Proteólise , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Proteínas Ligases SKP Culina F-Box/genética , Especificidade por Substrato , Ubiquitinação
9.
Cell Cycle ; 14(6): 802-7, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25603354

RESUMO

Dysregulation of cell cycle machinery causes abnormal cell division, leading to cancer development. To drive cell cycle properly, expression levels of cell cycle regulators are tightly regulated through the cell cycle. Dual specificity tyrosine phosphorylation-regulated kinase 2 (DYRK2) is a Ser/Thr kinase, and its intracellular functions had not been elucidated for decades. Recent studies have shown that DYRK2 down-regulates key molecules on cell cycle control. This review mainly highlights the DYRK2 function during cell division. In addition, we summarize tumor suppressive role of DYRK2 in cancer cells and discuss future research directions for DYRK2 toward the novel cancer therapies.


Assuntos
Divisão Celular , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Tirosina Quinases/metabolismo , Animais , Humanos , Modelos Biológicos , Mutação/genética , Neoplasias/enzimologia , Neoplasias/genética , Neoplasias/patologia , Ubiquitina-Proteína Ligases/metabolismo , Quinases Dyrk
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...