Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
EMBO J ; 42(2): e111673, 2023 01 16.
Artículo en Inglés | MEDLINE | ID: mdl-36514940

RESUMEN

Adenosine N6-methylation (m6A) and N6,2'-O-dimethylation (m6Am) are regulatory modifications of eukaryotic mRNAs. m6Am formation is catalyzed by the methyl transferase phosphorylated CTD-interacting factor 1 (PCIF1); however, the pathophysiological functions of this RNA modification and PCIF1 in cancers are unclear. Here, we show that PCIF1 expression is upregulated in colorectal cancer (CRC) and negatively correlates with patient survival. CRISPR/Cas9-mediated depletion of PCIF1 in human CRC cells leads to loss of cell migration, invasion, and colony formation in vitro and loss of tumor growth in athymic mice. Pcif1 knockout in murine CRC cells inhibits tumor growth in immunocompetent mice and enhances the effects of anti-PD-1 antibody treatment by decreasing intratumoral TGF-ß levels and increasing intratumoral IFN-γ, TNF-α levels, and tumor-infiltrating natural killer cells. We further show that PCIF1 modulates CRC growth and response to anti-PD-1 in a context-dependent mechanism with PCIF1 directly targeting FOS, IFITM3, and STAT1 via m6Am modifications. PCIF1 stabilizes FOS mRNA, which in turn leads to FOS-dependent TGF-ß regulation and tumor growth. While during immunotherapy, Pcif1-Fos-TGF-ß, as well as Pcif1-Stat1/Ifitm3-IFN-γ axes, contributes to the resistance of anti-PD-1 therapy. Collectively, our findings reveal a role of PCIF1 in promoting CRC tumorigenesis and resistance to anti-PD-1 therapy, supporting that the combination of PCIF1 inhibition with anti-PD-1 treatment is a potential therapeutic strategy to enhance CRC response to immunotherapy. Finally, we developed a lipid nanoparticles (LNPs) and chemically modified small interfering RNAs (CMsiRNAs)-based strategy to silence PCIF1 in vivo and found that this treatment significantly reduced tumor growth in mice. Our results therefore provide a proof-of-concept for tumor growth suppression using LNP-CMsiRNA to silence target genes in cancer.


Asunto(s)
Neoplasias Colorrectales , Inmunoterapia , Animales , Humanos , Ratones , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/genética , Proteínas de la Membrana/metabolismo , Metilación , Proteínas Nucleares/metabolismo , ARN Mensajero/genética , Proteínas de Unión al ARN/metabolismo , Factor de Crecimiento Transformador beta/metabolismo
2.
EMBO Rep ; 24(10): e55506, 2023 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-37705505

RESUMEN

N6 -methyladenosine (m6 A), the most abundant internal modification in eukaryotic mRNA, plays important roles in many physiological and pathological processes, including the development and progression of cancer. RNA modification by m6 A is regulated by methyltransferases, demethylases, and m6 A-binding proteins that function in large part by regulating mRNA expression and function. Here, we investigate the expression of m6 A regulatory proteins in breast cancer. We find that expression of KIAA1429/VIRMA, a component of the m6 A methyltransferase complex, is upregulated in breast cancer tissue and correlates positively with poor survival. KIAA1429/VIRMA is mislocalized to the cytosol of breast cancer tissues and cell lines, and shRNA-mediated knockdown inhibits breast cancer cell proliferation, migration, and invasion. Mechanistically, KIAA1429/VIRMA is shown to bind to the m6 A-dependent RNA-binding protein insulin-like growth factor 2 mRNA-binding protein 3 (IGF2BP3), leading to recruitment and stabilization of m6 A-modified hyaluronan synthase 2 (HAS2) mRNA. HAS2 mRNA and KIAA1429/VIRMA mRNA levels correlate positively in breast cancer tissues, suggesting that the KIAA1429/VIRMA-IGF2BP3-HAS2 axis promotes breast cancer growth and contributes to poor prognosis.


Asunto(s)
Neoplasias , Humanos , Citosol , Hialuronano Sintasas , Citoplasma , ARN Mensajero/genética
3.
J Cell Biochem ; 119(11): 8773-8783, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30076753

RESUMEN

MicroRNAs (miRNAs) play an important role in the regulation of human cancers, including breast cancer (BC). In the current study, we examined the expression pattern of the miRNA miR-125a-5p in human BC tissues, tumorigenesis of BC progression. We found that miR-125a-5p was significantly downregulated in human BC tissues. Overexpression of miR-125a-5p in a xenograft mouse model indicated that miR-125a-5p may function as a tumour suppressor during carcinogenesis. To explore the molecular mechanism by which miR-125a-5p contributes to BC progression, we predicted the target genes of miR-125a-5p and identified BC susceptibility gene 1-associated protein 1 (BAP1) as a direct target. Finally, we demonstrated that BAP1 had opposing effects to those of miR-125a-5p on BC cells, suggesting that miR-125a-5p may inhibit cell proliferation and promote cell apoptosis by negatively regulating BAP1. Taken together, our findings provide the first clues regarding the role of miR-125a-5p as a tumour suppressor in BC via the inhibition of BAP1 translation.


Asunto(s)
Neoplasias de la Mama/genética , Regulación hacia Abajo , Genes Supresores de Tumor , MicroARNs/genética , Proteínas Supresoras de Tumor/genética , Ubiquitina Tiolesterasa/genética , Animales , Apoptosis , Neoplasias de la Mama/metabolismo , Carcinogénesis/genética , Carcinogénesis/metabolismo , Proliferación Celular , Supervivencia Celular , Femenino , Silenciador del Gen , Humanos , Células MCF-7 , Ratones , Ratones Desnudos , MicroARNs/química , MicroARNs/metabolismo , Imitación Molecular , Terapia Molecular Dirigida , Transfección , Carga Tumoral , Proteínas Supresoras de Tumor/metabolismo , Ubiquitina Tiolesterasa/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
4.
Cancer Res Commun ; 3(1): 119-129, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36968224

RESUMEN

Although immune checkpoint inhibitors targeting T-cell immunoregulatory proteins have revolutionized cancer treatment, they are effective only in a limited number of patients, and new strategies are needed to enhance tumor responses to immunotherapies. Deletion of protein tyrosine phosphatase non-receptor type 2 (Ptpn2), a regulator of growth factor and cytokine signaling pathways, has been shown to sensitize murine B16F10 melanoma cells to IFNγ and anti-PD-1 immunotherapy. Here, we investigated the potential therapeutic utility of small-molecule PTPN2 inhibitors. Ten inhibitors were synthesized on the basis of in silico modeling and structure-based design and functionally tested in vitro and in vivo. We show that the inhibitors had little effect on B16F10 cells alone, but effectively sensitized the tumor cells to IFNγ treatment in vitro and to anti-PD-1 therapy in vivo. Under both conditions, Ptpn2 inhibitor cotreatment suppressed B16F10 cell growth and enhanced Stat1 phosphorylation and expression of IFNγ response genes. In vivo, PTPN2 inhibitor cotreatment significantly reduced melanoma and colorectal tumor growth and enhanced mouse survival compared with anti-PD-1 treatment alone, and this was accompanied by increased tumor infiltration by granzyme B+ CD8+ T cells. Similar results were obtained with representative murine and human colon cancer and lung cancer cell lines. Collectively, these results demonstrate that small-molecule inhibitors of PTPN2 may have clinical utility as sensitizing agents for immunotherapy-resistant cancers. Significance: To enhance the effectiveness of immunotherapies in resistant or nonresponsive cancers, it is important to develop inhibitors of enzymes that negatively influence the outcome of treatments. We have designed and evaluated small-molecule inhibitors of PTPN2 demonstrating that these compounds may have clinical utility as sensitizing agents for immunotherapy-resistant cancers.


Asunto(s)
Linfocitos T CD8-positivos , Melanoma , Humanos , Ratones , Animales , Linfocitos T CD8-positivos/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 2/genética , Melanoma/tratamiento farmacológico , Interferón gamma , Inmunoterapia/métodos
5.
ANZ J Surg ; 89(7-8): E297-E301, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31297940

RESUMEN

BACKGROUND: The purpose of this study was to evaluate the correlation between serum albumin level change (ΔALB) and post-operative complications in patients with normal preoperative serum albumin after gastrectomy of gastric cancer. METHODS: A total of 193 patients undergoing curative (R0) gastrectomy from September 2015 to May 2017 were enrolled in this study. The risk factors for predicting post-operative complications were identified by univariate and multivariate analysis. The cut-off value and diagnostic accuracy of ΔALB were measured by receiver operating characteristic curves. ΔALB was defined as: (albumin level before surgery - albumin on post-operative day (POD) 1)/albumin level before surgery × 100%. RESULTS: A total of 60 patients (31.0%) had post-operative complications. Our results showed that the cut-off value of ΔALB was 19.0%. Using a cut-off value of 19.0%, multivariate analysis identified that ΔALB was able to predict post-operative complications as an independent factor (odds ratio 13.98, 95% confidence interval 6.048-32.32, P < 0.001). In addition, the area under the curve of ΔALB is higher than C-reactive protein on POD 3 (0.773 versus 0633). Compared with patients with ΔALB <19.0%, patients with ΔALB ≥19.0% have higher risk of post-operative complications suffered (62.3 versus 13.7%, P < 0.001) and longer post-operative stay (22.1 ± 13.5 versus 17.5 ± 4.2, P < 0.001). CONCLUSION: ΔALB acted as an independent predictor in short-term complications for patients with normal preoperative serum albumin and its diagnostic accuracy was higher than C-reactive protein on POD 3. It is promising to be a precise and straight predictor for incidence of post-operative complications to patients with normal preoperative serum albumin.


Asunto(s)
Gastrectomía/efectos adversos , Albúmina Sérica/análisis , Neoplasias Gástricas/cirugía , Anciano , Proteína C-Reactiva/análisis , Estudios de Casos y Controles , Femenino , Gastrectomía/métodos , Humanos , Tiempo de Internación , Masculino , Persona de Mediana Edad , Complicaciones Posoperatorias/epidemiología , Valor Predictivo de las Pruebas , Periodo Preoperatorio , Estudios Retrospectivos , Factores de Riesgo
6.
Cancer Med ; 8(1): 261-275, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30575330

RESUMEN

The nuclear paraspeckle assembly transcript 1 (abbreviated as NEAT1), a nuclear sufficient long noncoding RNA (abbreviated as lncRNA), has aroused a rising concern in recent years. As uncovered by reports, the increase in NEAT1 is related to the deteriorated prognosis of lung cancer, breast cancer, hepatocellular cancer, and colorectal cancer (abbreviated as CRC). Thus far, the mechanism of NEAT1 has not been elucidated by the existing researches. The impact of knockdown of both NEAT1 and its predicted downstream miR-193a-3p in CRC cells was examined here to delve into their interactions and mechanisms. Additionally, the target of miR-193a-3p, Kirsten rat sarcoma viral oncogene homolog (abbreviated as KRAS), was also predicted by bioinformatics algorithms. Small interfering RNA and antisense oligonucleotides that inhibit NEAT1, as well as overexpression or knockdown of miR-193a-3p, were adequately drawn upon to confirm that NEAT1 serves as a miR-193a-3p sponge or competing endogenous RNA, to impact miR-193a-3p's further functions, including modulating KRAS proteins, both in vitro and in vivo. Generally, lncRNA NEAT1/hsa-miR-193a-3p/KRAS axis was substantiated in CRC cells and could provide novel insight into both diagnostic and therapeutic advancement in CRC.


Asunto(s)
Neoplasias Colorrectales/genética , MicroARNs/genética , Proteínas Proto-Oncogénicas p21(ras)/genética , ARN Largo no Codificante/genética , Animales , Apoptosis , Línea Celular Tumoral , Movimiento Celular , Técnicas de Silenciamiento del Gen , Humanos , Ratones Desnudos , ARN Interferente Pequeño/genética
7.
FEBS J ; 286(6): 1136-1153, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30548198

RESUMEN

The protein tyrosine phosphatase PTP1B, which is encoded by PTPN1, is a ubiquitously expressed nonreceptor protein tyrosine phosphatase. PTP1B has long been known to negatively regulate insulin and leptin receptor signalling. Recently, it was reported to be aberrantly expressed in cancer cells and to function as an important oncogene. In this study, we found that PTP1B protein levels are dramatically increased in breast cancer (BC) tissues and that PTP1B promotes the proliferation, and suppresses the apoptosis, of both HER2-positive and triple-negative BC cell lines. Bioinformatics analysis identified that the miRNA, miR-193a-3p, might potentially target PTP1B. We demonstrate that miR-193a-3p regulates PTP1B in BC cells and that it regulates the proliferation and apoptosis of BC cells by targeting PTP1B, both in vitro and in vivo. In conclusion, this study confirms that PTP1B acts as an oncogene in BC and demonstrates that miR-193a-3p can serve as a tumour suppressor gene in BC by targeting PTP1B.


Asunto(s)
Neoplasias de la Mama/patología , Regulación Neoplásica de la Expresión Génica , MicroARNs/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 1/metabolismo , Animales , Apoptosis , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Proliferación Celular , Progresión de la Enfermedad , Femenino , Humanos , Ratones , Ratones Desnudos , Pronóstico , Proteína Tirosina Fosfatasa no Receptora Tipo 1/genética , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
8.
Cell Death Dis ; 9(10): 948, 2018 09 20.
Artículo en Inglés | MEDLINE | ID: mdl-30237423

RESUMEN

Anoikis is a type of programmed cell death induced by detachment from the extracellular matrix. In cancer cells, anoikis resistance is essential for cancer cell survival in blood circulation and distant metastasis. However, the mechanisms behind anoikis resistance of gastric cancer remain largely unknown. Herein, we demonstrate that NADPH oxidase 4 (NOX4) expression and reactive oxygen species (ROS) generation are upregulated in suspension gastric cell cultures compared with adherent cultures. Silencing of NOX4 decreases ROS generation and downregulates EGFR, sensitizing cells to anoikis. NOX4 overexpression upregulates ROS and EGFR levels and promotes anoikis resistance. NOX4 depletion inhibits gastric cancer survival in blood circulation and attenuates distant metastasis. NOX4 expression is correlated with EGFR expression in patients. In conclusion, induction of NOX4 expression by detachment promotes anoikis resistance of gastric cancer through ROS generation and downstream upregulation of EGFR, which is critical for the metastatic progression of gastric cancer.


Asunto(s)
Anoicis/fisiología , Receptores ErbB/metabolismo , NADPH Oxidasa 4/metabolismo , Neoplasias Gástricas/metabolismo , Animales , Anoicis/genética , Apoptosis/genética , Apoptosis/fisiología , Western Blotting , Línea Celular Tumoral , Proliferación Celular/genética , Proliferación Celular/fisiología , Técnica del Anticuerpo Fluorescente , Humanos , Inmunohistoquímica , Lentivirus/genética , Masculino , Ratones , Ratones Desnudos , Microscopía Electrónica de Rastreo , NADPH Oxidasa 4/genética , Especies Reactivas de Oxígeno/metabolismo , Neoplasias Gástricas/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA