Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Gastroenterology ; 166(3): 466-482, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38065340

RESUMEN

BACKGROUND & AIMS: Although immunotherapy shows substantial advancement in colorectal cancer (CRC) with microsatellite instability high, it has limited efficacy for CRC with microsatellite stability (MSS). Identifying combinations that reverse immune suppression and prime MSS tumors for current immunotherapy approaches remains an urgent need. METHODS: An in vitro CRISPR screen was performed using coculture models of primary tumor cells and autologous immune cells from MSS CRC patients to identify epigenetic targets that could enhance immunotherapy efficacy in MSS tumors. RESULTS: We revealed EHMT2, a histone methyltransferase, as a potential target for MSS CRC. EHMT2 inhibition transformed the immunosuppressive microenvironment of MSS tumors into an immunomodulatory one by altering cytokine expression, leading to T-cell-mediated cytotoxicity activation and improved responsiveness to anti-PD1 treatment. We observed galectin-7 up-regulation upon EHMT2 inhibition, which converted a "cold" MSS tumor environment into a T-cell-inflamed one. Mechanistically, CHD4 repressed galectin-7 expression by recruiting EHMT2 to form a cotranscriptional silencing complex. Galectin-7 administration enhanced anti-PD1 efficacy in MSS CRC, serving as a potent adjunct cytokine therapy. CONCLUSIONS: Our findings suggest that targeting the EHMT2/galectin-7 axis could provide a novel combination strategy for immunotherapy in MSS CRC.


Asunto(s)
Neoplasias Colorrectales , Humanos , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/genética , Inmunoterapia , Citocinas , Galectinas/genética , Repeticiones de Microsatélite , Inestabilidad de Microsatélites , Microambiente Tumoral , Antígenos de Histocompatibilidad , N-Metiltransferasa de Histona-Lisina
2.
EMBO J ; 39(12): e103181, 2020 06 17.
Artículo en Inglés | MEDLINE | ID: mdl-32368828

RESUMEN

N6-methyladenosine (m6 A) is an abundant nucleotide modification in mRNA, known to regulate mRNA stability, splicing, and translation, but it is unclear whether it is also has a physiological role in the intratumoral microenvironment and cancer drug resistance. Here, we find that METTL3, a primary m6 A methyltransferase, is significantly down-regulated in human sorafenib-resistant hepatocellular carcinoma (HCC). Depletion of METTL3 under hypoxia promotes sorafenib resistance and expression of angiogenesis genes in cultured HCC cells and activates autophagy-associated pathways. Mechanistically, we have identified FOXO3 as a key downstream target of METTL3, with m6 A modification of the FOXO3 mRNA 3'-untranslated region increasing its stability through a YTHDF1-dependent mechanism. Analysis of clinical samples furthermore showed that METTL3 and FOXO3 levels are tightly correlated in HCC patients. In mouse xenograft models, METTL3 depletion significantly enhances sorafenib resistance of HCC by abolishing the identified METTL3-mediated FOXO3 mRNA stabilization, and overexpression of FOXO3 restores m6 A-dependent sorafenib sensitivity. Collectively, our work reveals a critical function for METTL3-mediated m6 A modification in the hypoxic tumor microenvironment and identifies FOXO3 as an important target of m6 A modification in the resistance of HCC to sorafenib therapy.


Asunto(s)
Adenosina/análogos & derivados , Autofagia/efectos de los fármacos , Carcinoma Hepatocelular/metabolismo , Resistencia a Antineoplásicos/efectos de los fármacos , Proteína Forkhead Box O3/metabolismo , Neoplasias Hepáticas/metabolismo , Proteínas de Neoplasias/metabolismo , ARN Mensajero/metabolismo , ARN Neoplásico/metabolismo , Sorafenib/farmacología , Adenosina/genética , Adenosina/metabolismo , Animales , Autofagia/genética , Carcinoma Hepatocelular/tratamiento farmacológico , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patología , Proteína Forkhead Box O3/genética , Células HEK293 , Células Hep G2 , Células Endoteliales de la Vena Umbilical Humana , Humanos , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patología , Masculino , Metilación/efectos de los fármacos , Metiltransferasas/genética , Metiltransferasas/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Proteínas de Neoplasias/genética , ARN Mensajero/genética , ARN Neoplásico/genética
3.
Mol Ther ; 31(2): 517-534, 2023 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-36307991

RESUMEN

N6-methyladenosine (m6A) is the most pervasive RNA modification and is recognized as a novel epigenetic regulation in RNA metabolism. Although the m6A modification involves various physiological processes, its roles in drug resistance in colorectal cancer (CRC) still remain unknown. We analyzed the RNA expression profile of m6A/A (%) with MRM mass spectrometry in human 5-fluorouracil (5-FU)-resistant CRC tissues, and used the m6A RNA immunoprecipitation assay to validate the m6A-regulated target. Our results have shown that the m6A demethylase FTO was up-regulated in human primary and 5-FU-resistant CRC. Depletion of FTO decreased cell growth, colony formation and metastasis in 5-FU-resistant CRC cells in vitro and in vivo. Mechanistically, we identified SIVA1, a critical apoptotic gene, as a key downstream target of the FTO-mediated m6A demethylation. The m6A demethylation of SIVA1 at the CDS region induced its mRNA degradation via a YTHDF2-dependent mechanism. The SIVA1 levels were negatively correlated with the FTO levels in clinical CRC tissues. Notably, inhibition of FTO significantly reduced the tolerance of 5-FU in 5-FU-resistant CRC cells via the FTO-SIVA1 axis, whereas SIVA1-depletion could restore the m6A-dependent 5-FU sensitivity in CRC cells. In summary, our findings demonstrate a critical role of FTO as an m6A demethylase enhancing chemo-resistance in CRC cells, and suggest that FTO inhibition may restore the sensitivity of chemo-resistant CRC cells to 5-FU.


Asunto(s)
Neoplasias Colorrectales , Epigénesis Genética , Humanos , ARN , Fluorouracilo/farmacología , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/metabolismo , Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/metabolismo , Dioxigenasa FTO Dependiente de Alfa-Cetoglutarato/genética , Dioxigenasa FTO Dependiente de Alfa-Cetoglutarato/metabolismo
4.
Hepatology ; 74(3): 1461-1479, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-33813748

RESUMEN

BACKGROUND AND AIMS: Hypoxia is a common feature of the tumor microenvironment (TME), which promotes tumor progression, metastasis, and therapeutic drug resistance through a myriad of cell activities in tumor and stroma cells. While targeting hypoxic TME is emerging as a promising strategy for treating solid tumors, preclinical development of this approach is lacking in the study of HCC. APPROACH AND RESULTS: From a genome-wide CRISPR/CRISPR-associated 9 gene knockout screening, we identified aldolase A (ALDOA), a key enzyme in glycolysis and gluconeogenesis, as an essential driver for HCC cell growth under hypoxia. Knockdown of ALDOA in HCC cells leads to lactate depletion and consequently inhibits tumor growth. Supplementation with lactate partly rescues the inhibitory effects mediated by ALDOA knockdown. Upon hypoxia, ALDOA is induced by hypoxia-inducible factor-1α and fat mass and obesity-associated protein-mediated N6 -methyladenosine modification through transcriptional and posttranscriptional regulation, respectively. Analysis of The Cancer Genome Atlas shows that elevated levels of ALDOA are significantly correlated with poor prognosis of patients with HCC. In a screen of Food and Drug Administration-approved drugs based on structured hierarchical virtual platforms, we identified the sulfamonomethoxine derivative compound 5 (cpd-5) as a potential inhibitor to target ALDOA, evidenced by the antitumor activity of cpd-5 in preclinical patient-derived xenograft models of HCC. CONCLUSIONS: Our work identifies ALDOA as an essential driver for HCC cell growth under hypoxia, and we demonstrate that inhibition of ALDOA in the hypoxic TME is a promising therapeutic strategy for treating HCC.


Asunto(s)
Carcinoma Hepatocelular/genética , Fructosa-Bifosfato Aldolasa/genética , Neoplasias Hepáticas/genética , Hipoxia Tumoral/genética , Dioxigenasa FTO Dependiente de Alfa-Cetoglutarato/metabolismo , Animales , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patología , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Fructosa-Bifosfato Aldolasa/metabolismo , Técnicas de Silenciamiento del Gen , Células Hep G2 , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Ácido Láctico/metabolismo , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Mutación con Pérdida de Función , Ratones , Trasplante de Neoplasias , Sulfamonometoxina/análogos & derivados , Microambiente Tumoral , Ensayos Antitumor por Modelo de Xenoinjerto
5.
Gut ; 70(9): 1698-1712, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-33219048

RESUMEN

OBJECTIVE: Dysregulated cellular metabolism is a distinct hallmark of human colorectal cancer (CRC). However, metabolic programme rewiring during tumour progression has yet to be fully understood. DESIGN: We analysed altered gene signatures during colorectal tumour progression, and used a complex of molecular and metabolic assays to study the regulation of metabolism in CRC cell lines, human patient-derived xenograft mouse models and tumour organoid models. RESULTS: We identified a novel RNA-binding protein, RALY (also known as hnRNPCL2), that is highly associated with colorectal tumour aggressiveness. RALY acts as a key regulatory component in the Drosha complex, and promotes the post-transcriptional processing of a specific subset of miRNAs (miR-483, miR-676 and miR-877). These miRNAs systematically downregulate the expression of the metabolism-associated genes (ATP5I, ATP5G1, ATP5G3 and CYC1) and thereby reprogramme mitochondrial metabolism in the cancer cell. Analysis of The Cancer Genome Atlas (TCGA) reveals that increased levels of RALY are associated with poor prognosis in the patients with CRC expressing low levels of mitochondrion-associated genes. Mechanistically, induced processing of these miRNAs is facilitated by their N6-methyladenosine switch under reactive oxygen species (ROS) stress. Inhibition of the m6A methylation abolishes the RALY recognition of the terminal loop of the pri-miRNAs. Knockdown of RALY inhibits colorectal tumour growth and progression in vivo and in organoid models. CONCLUSIONS: Collectively, our results reveal a critical metabolism-centric role of RALY in tumour progression, which may lead to cancer therapeutics targeting RALY for treating CRC.


Asunto(s)
Ribonucleoproteína Heterogénea-Nuclear Grupo C/metabolismo , MicroARNs/metabolismo , Mitocondrias/metabolismo , Animales , Apoptosis , Línea Celular Tumoral , Proliferación Celular , Neoplasias Colorrectales , Regulación Neoplásica de la Expresión Génica , Ratones , Ratones Endogámicos C57BL , Trasplante de Neoplasias , Especies Reactivas de Oxígeno/metabolismo , Ribonucleasa III/metabolismo
6.
Mol Cancer ; 18(1): 46, 2019 03 28.
Artículo en Inglés | MEDLINE | ID: mdl-30922314

RESUMEN

BACKGROUND: N6-methyladenosine (m6A) modification is the most pervasive modification in mRNA, and has been considered as a new layer of epigenetic regulation on mRNA processing, stability and translation. Despite its functional significance in various physiological processes, the role of the m6A modification involved in breast cancer is yet fully understood. METHODS: We used the m6A-RNA immunoprecipitation sequencing to identify the potential targets in breast cancer. To determine the underlying mechanism for the axis of FTO-BNIP3, we performed a series of in vitro and in vivo assays in 3 breast cancer cell lines and 36 primary breast tumor tissues and 12 adjunct tissues. RESULTS: We showed that FTO, a key m6A demethylase, was up-regulated in human breast cancer. High level of FTO was significantly associated with lower survival rates in patients with breast cancer. FTO promoted breast cancer cell proliferation, colony formation and metastasis in vitro and in vivo. We identified BNIP3, a pro-apoptosis gene, as a downstream target of FTO-mediated m6A modification. Epigenetically, FTO mediated m6A demethylation in the 3'UTR of BNIP3 mRNA and induced its degradation via an YTHDF2 independent mechanism. BNIP3 acts as a tumor suppressor and is negatively correlated with FTO expression in clinical breast cancer patients. BNIP3 dramatically alleviated FTO-dependent tumor growth retardation and metastasis. CONCLUSIONS: Our findings demonstrate the functional significance of the m6A modification in breast cancer, and suggest that FTO may serve as a novel potential therapeutic target for breast cancer.


Asunto(s)
Dioxigenasa FTO Dependiente de Alfa-Cetoglutarato/metabolismo , Biomarcadores de Tumor/metabolismo , Neoplasias de la Mama/patología , Epigénesis Genética , Regulación Neoplásica de la Expresión Génica , Proteínas de la Membrana/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Adenosina/análogos & derivados , Adenosina/química , Adenosina/metabolismo , Dioxigenasa FTO Dependiente de Alfa-Cetoglutarato/genética , Animales , Apoptosis , Biomarcadores de Tumor/genética , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Estudios de Casos y Controles , Movimiento Celular , Proliferación Celular , Desmetilación , Progresión de la Enfermedad , Femenino , Estudios de Seguimiento , Humanos , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos NOD , Ratones Desnudos , Ratones SCID , Invasividad Neoplásica , Pronóstico , Proteínas Proto-Oncogénicas/genética , Tasa de Supervivencia , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
7.
Nat Protoc ; 2024 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-38816517

RESUMEN

Numerous toxins threaten humans, but specific antidotes are unavailable for most of them. Although CRISPR screening has aided the discovery of the mechanisms of some toxins, developing targeted antidotes remains a significant challenge. Recently, we established a systematic framework to develop antidotes by combining the identification of novel drug targets by using a genome-wide CRISPR screen with a virtual screen of drugs approved by the US Food and Drug Administration. This approach allows for a comprehensive understanding of toxin mechanisms at the whole-genome level and facilitates the identification of promising antidote drugs targeting specific molecules. Here, we present step-by-step instructions for executing genome-scale CRISPR-Cas9 knockout screens of toxins in HAP1 cells. We also provide detailed guidance for conducting an in silico drug screen and an in vivo drug validation. By using this protocol, it takes ~4 weeks to perform the genome-scale screen, 4 weeks for sequencing and data analysis, 4 weeks to validate candidate genes, 1 week for the virtual screen and 2 weeks for in vitro drug validation. This framework has the potential to accelerate the development of antidotes for a wide range of toxins and can rapidly identify promising drug candidates that are already known to be safe and effective. This could lead to the development of new antidotes much more quickly than traditional methods, protecting lives from diverse toxins and advancing human health.

8.
Acta Pharm Sin B ; 14(1): 223-240, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38261805

RESUMEN

Lenvatinib, a second-generation multi-receptor tyrosine kinase inhibitor approved by the FDA for first-line treatment of advanced liver cancer, facing limitations due to drug resistance. Here, we applied a multidimensional, high-throughput screening platform comprising patient-derived resistant liver tumor cells (PDCs), organoids (PDOs), and xenografts (PDXs) to identify drug susceptibilities for conquering lenvatinib resistance in clinically relevant settings. Expansion and passaging of PDCs and PDOs from resistant patient liver tumors retained functional fidelity to lenvatinib treatment, expediting drug repurposing screens. Pharmacological screening identified romidepsin, YM155, apitolisib, NVP-TAE684 and dasatinib as potential antitumor agents in lenvatinib-resistant PDC and PDO models. Notably, romidepsin treatment enhanced antitumor response in syngeneic mouse models by triggering immunogenic tumor cell death and blocking the EGFR signaling pathway. A combination of romidepsin and immunotherapy achieved robust and synergistic antitumor effects against lenvatinib resistance in humanized immunocompetent PDX models. Collectively, our findings suggest that patient-derived liver cancer models effectively recapitulate lenvatinib resistance observed in clinical settings and expedite drug discovery for advanced liver cancer, providing a feasible multidimensional platform for personalized medicine.

9.
Nat Commun ; 14(1): 2241, 2023 05 16.
Artículo en Inglés | MEDLINE | ID: mdl-37193694

RESUMEN

The "death cap", Amanita phalloides, is the world's most poisonous mushroom, responsible for 90% of mushroom-related fatalities. The most fatal component of the death cap is α-amanitin. Despite its lethal effect, the exact mechanisms of how α-amanitin poisons humans remain unclear, leading to no specific antidote available for treatment. Here we show that STT3B is required for α-amanitin toxicity and its inhibitor, indocyanine green (ICG), can be used as a specific antidote. By combining a genome-wide CRISPR screen with an in silico drug screening and in vivo functional validation, we discover that N-glycan biosynthesis pathway and its key component, STT3B, play a crucial role in α-amanitin toxicity and that ICG is a STT3B inhibitor. Furthermore, we demonstrate that ICG is effective in blocking the toxic effect of α-amanitin in cells, liver organoids, and male mice, resulting in an overall increase in animal survival. Together, by combining a genome-wide CRISPR screen for α-amanitin toxicity with an in silico drug screen and functional validation in vivo, our study highlights ICG as a STT3B inhibitor against the mushroom toxin.


Asunto(s)
Hexosiltransferasas , Micotoxinas , Humanos , Masculino , Animales , Ratones , Alfa-Amanitina/farmacología , Verde de Indocianina/farmacología , Antídotos , Amanita , Proteínas de la Membrana
10.
Comput Struct Biotechnol J ; 21: 4540-4551, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37810279

RESUMEN

Tumor mutation burden (TMB) has emerged as an essential biomarker for assessing the efficacy of cancer immunotherapy. However, due to the inherent complexity of tumors, TMB is not always correlated with the responsiveness of immune checkpoint inhibitors (ICIs). Thus, refining the interpretation and contextualization of TMB is a requisite for enhancing clinical outcomes. In this study, we conducted a comprehensive investigation of the relationship between TMB and multi-omics data across 33 human cancer types. Our analysis revealed distinct biological changes associated with varying TMB statuses in STAD, COAD, and UCEC. While multi-omics data offer an opportunity to dissect the intricacies of tumors, extracting meaningful biological insights from such massive information remains a formidable challenge. To address this, we developed and implemented the PGLCN, a biologically informed graph neural network based on pathway interaction information. This model facilitates the stratification of patients into subgroups with distinct TMB statuses and enables the evaluation of driver biological processes through enhanced interpretability. By integrating multi-omics data for TMB prediction, our PGLCN model outperformed previous traditional machine learning methodologies, demonstrating superior TMB status prediction accuracy (STAD AUC: 0.976 ± 0.007; COAD AUC: 0.994 ± 0.007; UCEC AUC: 0.947 ± 0.023) and enhanced interpretability (BA-House: 1.0; BA-Community: 0.999; BA-Grid: 0.994; Tree-Cycles: 0.917; Tree-Grids: 0.867). Furthermore, the biological interpretability inherent to PGLCN identified the Toll-like receptor family and DNA repair pathways as potential combined biomarkers in conjunction with TMB status in gastric cancer. This finding suggests a potential synergistic targeting strategy with immunotherapy for gastric cancer, thus advancing the field of precision oncology.

11.
Antioxid Redox Signal ; 37(16-18): 1266-1290, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-35369726

RESUMEN

Significance: Immunotherapy, which utilizes the patient's immune system to fight tumor cells, has been approved for the treatment of some types of advanced cancer. Recent Advances: The complexity and diversity of tumor immunity are responsible for the varying response rates toward current immunotherapy strategies and highlight the importance of exploring regulators in tumor immunotherapy. Several genetic factors have proved to be critical regulators of tumor immunotherapy. RNAs, including messenger RNAs and non-coding RNAs, play vital and diverse roles in tumorigenesis, metastasis, drug resistance, and immunotherapy response. RNA modifications, including N6-methyladenosine methylation, are involved in tumor immunity. Critical Issues: A critical issue is the lack of summary of the regulatory RNA molecules and their derivatives in mediating immune activities in human cancers that could provide potential applications for tumor immunotherapeutic strategy. Future Directions: This review summarizes the dual roles (the light and dark sides) of RNA and its derivatives in tumor immunotherapy and discusses the development of RNA-based therapies as novel immunotherapeutic strategies for cancer treatment. Antioxid. Redox Signal. 37, 1266-1290.


Asunto(s)
Adenosina , ARN , Humanos , ARN/genética , ARN Mensajero , Carcinogénesis , Transformación Celular Neoplásica
12.
Front Oncol ; 11: 760765, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34745999

RESUMEN

Management of patients with hepatocellular carcinoma (HCC) largely relies on surgery and other systemic therapies. However, the poor diagnosis of cancer recurrence or metastasis can lead to a high frequency of treatment failure. Thus, factors that can predict disease status and prognosis of patients need to be identified. Circulating tumor cells (CTCs) are known to accurately predict survival of patients. Here, we report a case in which CTCs successfully predicted the progression of metastatic colon polyps after interventional therapy for HCC. A 48-year-old man was diagnosed with HCC with moderate differentiation in 2016 and subsequently underwent orthotopic liver transplantation. Discharge medications were continued with immunosuppressants (tacrolimus) and antiviral drugs (Titin). In 2018, a colon polyp, a type of tubular adenoma, was detected and surgically removed. However, in 2020, the same tubular adenoma recurred. During cancer progression, CTC counts were measured to monitor the status of metastasis, and a positive correlation was noted between the dynamic change in CTC counts and cancer response (metastasis or recurrence). When diagnosing the metastatic adenoma, the number of cytokeratin-positive CTCs was significantly increased; however, it dropped to zero after the polyp was surgically removed. The same change in CTC counts was observed during the second recurrence of the adenoma, and a subgroup of CTCs, cell surface vimentin-positive CTCs, was significantly increased. The CTC count dropped to an undetectable level after the surgery for the first time. In summary, we presented a clinical case in which CTC counts could predict disease progression during HCC metastasis. Thus, CTC counts should be measured after liver transplantation in patients with HCC for diagnosis and clinical decision-making as it is effective in monitoring cancer progression.

13.
J Med Chem ; 63(19): 11286-11301, 2020 10 08.
Artículo en Inglés | MEDLINE | ID: mdl-32844651

RESUMEN

Blockade of immune checkpoint PD-1/PD-L1 facilitates the rescue of immune escapes of tumor cells. Though various monoclonal antibodies have been approved for clinical therapy, the development of small molecular inhibitors lags behind antibodies partially owing to the challenges of protein-protein interaction (PPI) blocker design. In this work, we adopted the skeleton of natural cyclopeptidic antibiotics gramicidin S as the start point for PD-1/PD-L1 inhibitor exploring and discovered a series of novel cyclopeptides that could interfere with the PPI of PD-1/PD-L1 based on several rounds of structural design and optimization. The representative active cyclopeptide 66 can bind two PD-L1 and efficiently block the PD-1/PD-L1 interaction, recruit the immune cells to the tumor cells, enhance their killing against tumor cells by promoting the release of granzyme B and perforin, and display significant CD8+ T cell-dependent tumor suppression activity in vivo.


Asunto(s)
Antígeno B7-H1/antagonistas & inhibidores , Diseño de Fármacos , Inmunoterapia , Neoplasias/tratamiento farmacológico , Péptidos Cíclicos/química , Humanos
14.
Acta Pharm Sin B ; 8(6): 833-843, 2018 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-30505654

RESUMEN

N 6-Methyladenosine (m6A) modification is the most pervasive modification of human mRNA molecules. It is reversible via regulation of m6A modification methyltransferase, demethylase and proteins that preferentially recognize m6A modification as "writers", "erasers" and "readers", respectively. Altered expression levels of the m6A modification key regulators substantially affect their function, leading to significant phenotype changes in the cell and organism. Recent studies have proved that the m6A modification plays significant roles in regulation of metabolism, stem cell self-renewal, and metastasis in a variety of human cancers. In this review, we describe the potential roles of m6A modification in human cancers and summarize their underlying molecular mechanisms. Moreover, we will highlight potential therapeutic approaches by targeting the key m6A modification regulators for cancer drug development.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA