Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Cancer Gene Ther ; 30(10): 1390-1402, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37488294

RESUMEN

Malignant peripheral nerve sheath tumors (MPNSTs) are aggressive soft-tissue sarcomas characterized by poor prognosis and low drug response rates. Traditional chemo/radiotherapies show only mild benefits for patients with MPNSTs, and no targeted therapy is available in the clinic. A better understanding of the molecular background of MPNSTs is critical for the development of effective targeted therapies. Forkhead box M1 (FOXM1) has been implicated in the progression of many human malignancies, though its role in MPNSTs is unclear. In this study, using four Gene Expression Omnibus (GEO) datasets and a tissue microarray, we demonstrated that FOXM1 upregulation was associated with poor prognosis in patients with MPNSTs. FOXM1 overexpression and knockdown regulated the proliferation and colony formation of MPNST cells. Using bioinformatics analysis and luciferase reporter assays, we identified NUF2 as a direct downstream target of FOXM1. Both in vitro and in vivo experiments demonstrated that the induction of MPNST cell proliferation by FOXM1 was dependent on elevated NUF2 expression, as NUF2 knockdown abolished the FOXM1-induced proliferation of MPNST cells. Our study showed that the FOXM1-NUF2 axis mediates human MPNST progression and could be a potential therapeutic target.


Asunto(s)
Proteína Forkhead Box M1 , Neoplasias de la Vaina del Nervio , Neurofibromatosis 1 , Humanos , Proteínas de Ciclo Celular , Proliferación Celular , Proteína Forkhead Box M1/genética , Neoplasias de la Vaina del Nervio/genética , Neoplasias de la Vaina del Nervio/metabolismo , Neoplasias de la Vaina del Nervio/patología , Neurofibromatosis 1/complicaciones , Neurofibromatosis 1/genética , Neurofibromatosis 1/patología , Neurofibrosarcoma/complicaciones
2.
Cell Oncol (Dordr) ; 46(5): 1399-1413, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37086345

RESUMEN

BACKGROUND: Malignant peripheral nerve sheath tumors (MPNSTs) are aggressive sarcomas that typically develop in the setting of neurofibromatosis type 1 (NF1) and cause significant morbidity. Conventional therapies are often ineffective for MPNSTs. Ribonucleotide reductase subunit M2 (RRM2) is involved in DNA synthesis and repair, and is overexpressed in multiple cancers. However, its role in NF1-associated MPNSTs remains unknown. Our objective was to determine the therapeutic and prognostic potential of RRM2 in NF1-associated MPNSTs. METHODS: Identification of hub genes was performed by using NF1-associated MPNST microarray datasets. We detected RRM2 expression by immunochemical staining in an MPNST tissue microarray, and assessed the clinical and prognostic significance of RRM2 in an MPNST cohort. RRM2 knockdown and the RRM2 inhibitor Triapine were used to assess cell proliferation and apoptosis in NF1-associated MPNST cells in vitro and in vivo. The underlying mechanism of RRM2 in NF1-associated MPNST was revealed by transcriptome analysis. RESULTS: RRM2 is a key hub gene and its expression is significantly elevated in NF1-associated MPNST. We revealed that high RRM2 expression accounted for a larger proportion of NF1-associated MPNSTs and confirmed the correlation of high RRM2 expression with poor overall survival. Knockdown of RRM2 inhibited NF1-associated MPNST cell proliferation and promoted apoptosis and S-phase arrest. The RRM2 inhibitor Triapine displayed dose-dependent inhibitory effects in vitro and induced significant tumor growth reduction in vivo in NF1-associated MPNST. Analysis of transcriptomic changes induced by RRM2 knockdown revealed suppression of the AKT-mTOR signaling pathway. Overexpression of RRM2 activates the AKT pathway to promote NF1-associated MPNST cell proliferation. CONCLUSIONS: RRM2 expression is significantly elevated in NF1-associated MPNST and that high RRM2 expression correlates with poorer outcomes. RRM2 acts as an integral part in the promotion of NF1-associated MPNST cell proliferation via the AKT-mTOR signaling pathway. Inhibition of RRM2 may be a promising therapeutic strategy for NF1-associated MPNST.


Asunto(s)
Neurofibromatosis 1 , Neurofibrosarcoma , Humanos , Neurofibromatosis 1/complicaciones , Neurofibromatosis 1/genética , Neurofibromatosis 1/patología , Neurofibrosarcoma/complicaciones , Neurofibrosarcoma/patología , Neurofibrosarcoma/terapia , Proteínas Proto-Oncogénicas c-akt/metabolismo , Pronóstico , Serina-Treonina Quinasas TOR/metabolismo
3.
Cancers (Basel) ; 14(23)2022 Nov 24.
Artículo en Inglés | MEDLINE | ID: mdl-36497280

RESUMEN

(1) Background: malignant peripheral nerve sheath tumours (MPNSTs) are aggressive Schwann cell-derived sarcomas with dismal prognoses. Previous studies have shown that nuclear receptor corepressor 2 (NCOR2) plays a vital role in neurodevelopment and in various tumours. However, the impact of NCOR2 on the progression of MPNST remains unclear. (2) Methods: by GEO database, MPNST tissue microarray, and NF1-related tumour tissues and cell lines were used to explore NCOR2 expression level in the MPNSTs. The role and mechanism of NCOR2 in NF1-derived MPNSTs were explored by experiments in vivo and in vitro and by transcriptome high-throughput sequencing. (3) Results: NCOR2 expression is significantly elevated in NF1-derived MPNSTs and is associated with patient 10-year survival time. Knockdown of NCOR2 suppressed NF1-derived MPNST cell proliferation by blocking the cell cycle in the G0/G1 phase. Moreover, decreased NCOR2 expression could down-regulate MAPK signal activity through the BDNF/TrkB pathway. (4) Conclusions: our findings demonstrated that NCOR2 expression is significantly elevated in NF1-derived MPNSTs. NCOR2 knockdown can inhibit NF1-derived MPNST cell proliferation by weakened BDNF/TrkB/ERK signalling. Targeting NF1-derived MPNSTs with TrkB inhibitors, or in combination with ERK inhibitors, may be a novel therapeutic strategy for clinical trials.

4.
Front Oncol ; 12: 910505, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35965583

RESUMEN

Malignant peripheral nerve sheath tumors (MPNST) are aggressive soft-tissue sarcomas which lack effective drugs. Loss of the RAS GTPase-activating protein NF1 and subsequent overactivation of mitogen-activated protein kinase kinase (MAPK) signaling exist nearly uniformly in MPNST, making MAPK inhibition a promising therapeutic intervention. However, the efficacy of MEK inhibitor (MEKi) monotherapy was limited in MPNST and the relative mechanisms remained largely unexplored. In this study, we generated three MEKi-resistant cell models and investigated the mechanisms of MEKi resistance using high-throughput transcriptomic sequencing. We discovered that cell apoptosis and cell cycle arrest induced by MEKi were rescued in MEKi-resistant cells and the upregulation of LAMA4/ITGB1/FAK/SRC signaling conferred resistance to MEKi. In addition, concurrent inhibition of MAPK signaling and FAK/SRC cascade could sensitize MPNST cells to MEKi. Our findings provide potential solutions to overcome MEKi resistance and effective combination therapeutic strategies for treating MPNSTs.

5.
Front Oncol ; 12: 903177, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35875109

RESUMEN

Malignant peripheral nerve sheath tumors (MPNSTs) are rare soft-tissue sarcomas refractory to standard therapies. Inactivation of NF1 and subsequent upregulation of RAS/RAF/MEK/ERK signaling exist in the majority of MPNSTs. However, the lack of preclinical assessment of MEK inhibitors in MPNSTs hinders the clinical application as well as the development of combination therapy. To guide further clinical studies, we evaluated different MEK inhibitors in terms of efficacy, safety, and mechanism of adaptive response in treating MPNSTs. Using a MPNST tissue microarray, we found that p-ERK could serve as a biomarker for predicting the prognosis of MPNST patients as well as an effective therapeutic target. Through in vitro and in vivo experiments, we identified trametinib as the most potent MEK inhibitor for the treatment of MPNSTs. Mechanistically, reduced reactivation of the MAPK pathway and compensatory activation of the parallel pathways contributed to better efficacy. Our results provide a basis for the further clinical application of MEK inhibitors as single agents or combinational therapies.

6.
Dis Markers ; 2022: 8114731, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35722625

RESUMEN

Cutaneous malignant melanoma (CMM) is the most deadly skin cancer worldwide. Despite advances in the treatments of CMM, its incidence and mortality rates are still increasing. N6-methyladenosine (m6A) is the most common form of RNA modification and has attracted increasing interest in cancer initiation and progression. However, the role of m6A regulators in CMM and their correlation with prognosis remain elusive. Here, we demonstrated that by applying consensus clustering, all CMM patient cases can be divided into two clusters based on overall expression levels of 25 m6A genes. We systematically analyzed the prognostic value of the 25 m6A RNA methylation regulators in CMM and found that ELAVL1, ABCF1, and IGF2BP1 yield the highest scores for predicting the prognosis of CMM. Accordingly, we derived a risk signature consisting of three selected m6A genes as an independent prognostic marker for CMM and validated our findings with data derived from a different CMM cohort. Next, we determined that CNVs in m6A genes had a significant negative impact on patient survival. The mRNA expression levels of m6A genes were correlated with CNV mutation. Moreover, in the selected three risk signature m6A regulators, GSEA analysis showed that they were closely correlated with inflammation and immune pathways. TME analysis proved that m6A gene expressions were negatively correlated with immune cell infiltration. In conclusion, m6A regulators are vital participants in CMM pathology; and ELAVL1, ABCF1, and IGF2BP1 mRNA levels are valuable factors for prognosis prediction and treatment strategy development.


Asunto(s)
Melanoma , Neoplasias Cutáneas , Transportadoras de Casetes de Unión a ATP , Biología Computacional , Humanos , Melanoma/genética , Melanoma/patología , Pronóstico , ARN Mensajero/metabolismo , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/patología , Melanoma Cutáneo Maligno
7.
Pigment Cell Melanoma Res ; 35(3): 356-368, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35218152

RESUMEN

Treatments for giant congenital melanocytic nevi (GCMN) are extremely limited. Thus, there is an urgent need for development of relevant targeted therapies. However, current lack of preclinical cell models restricts progress in GCMN research. In this study, we aimed to establish and characterize an immortalized GCMN cell line. GCMN cells were successfully immortalized by means of lentivirus-mediated simian virus 40 large T transfection. The immortalized GNC cell line (ImGNC) showed lower proliferation rate and higher melanin content than primary melanocytes. Expression levels of the differentiation gene MITF and stemness genes TWIST1, SNAI1, and FOXD3 were elevated in ImGNCs; however, the established ImGNC cell line was immortalized but not transformed. Sanger sequencing detected the heterozygous NRASQ61K mutation in ImGNCs, but not the BRAFV600E mutation. Despite carrying the NRASQ61K allele, ImGNCs demonstrated suppressed MAPK activation and elevated PI3K/Akt activation, as compared with primary melanocytes. Drug sensitivity analysis showed that ImGNCs are more sensitive to PI3K/Akt and Bcl-2 inhibitors than to MEK or ERK inhibitors. Unlike the proliferation-inhibiting effect of PI3K/Akt inhibitors, the Bcl-2 inhibitor navitoclax promptly promoted apoptosis in ImGNCs. Considering the low proliferation characteristics of GCMN in vivo, Bcl-2 may be a potential therapeutic target that warrants further research.


Asunto(s)
Antineoplásicos , Nevo Pigmentado , Línea Celular , Humanos , Nevo Pigmentado/congénito , Nevo Pigmentado/genética , Fosfatidilinositol 3-Quinasas , Inhibidores de las Quinasa Fosfoinosítidos-3 , Inhibidores de Proteínas Quinasas , Proteínas Proto-Oncogénicas c-akt , Proteínas Proto-Oncogénicas c-bcl-2 , Neoplasias Cutáneas
8.
Exp Dermatol ; 31(4): 582-585, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35020224

RESUMEN

The mechanism underlying giant congenital melanocytic nevus (GCMN) formation is not fully understood. According to recent research, NRAS gene mutation is the main driving factor in GCMN. Melanocytic precursor cells proliferate during the embryonic stage after acquiring NRAS mutations. However, why GCMN undergoes intense proliferation in the embryonic stage and then stops postnatally remains unknown. The current theory for this phenomenon is that the GCMN undergoes oncogene-induced senescence. However, there is not enough evidence to indicate that senescence induces growth arrest in GCMN. In this study, we hypothesized that the expression level of the NRAS gene changes dynamically during the development and differentiation of neural crest cells into melanocytes and that the NRAS expression level determines whether the cell proliferates or becomes quiescent.


Asunto(s)
Nevo Pigmentado , Neoplasias Cutáneas , GTP Fosfohidrolasas/genética , GTP Fosfohidrolasas/metabolismo , Humanos , Melanocitos/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Mutación , Nevo Pigmentado/congénito , Nevo Pigmentado/genética , Neoplasias Cutáneas/genética
9.
J Invest Dermatol ; 142(3 Pt A): 613-623.e7, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34534577

RESUMEN

MAPK/extracellular signal-regulated kinase kinase (MEK) 1/2 inhibitors (MEKis) have recently achieved surprising success in treating unresectable plexiform neurofibromas (PNFs). However, few studies have investigated the mechanisms of MEKi resistance in patients with PNF. We determined the efficacy of six different MEKis for treating PNFs, explored drug resistance mechanisms, and identified potential combination therapies to overcome resistance. By screening drug efficacy among six MEKis in human NF1-deficient PNF cell lines, TAK-733 was found to reduce PNF cell viability the most. We then cultured the TAK-733‒resistant cells and explored the potential targets for further treatment. Both high-throughput drug screening and RNA sequencing analyses of MEKi-resistant PNF cells identified cyclin-dependent kinase inhibitors as potential agents for PNFs. Dinaciclib, a cyclin-dependent kinase inhibitor, showed synergistic effects on MEKi-resistant cells. Coadministration of dinaciclib and TAK-733 significantly reduced cell viability and inhibited sphere formation and colony formation. Dinaciclib did not affect MEK signaling but decreased the expression of several prosurvival proteins, including survivin and cyclin-dependent kinase 1, to induce apoptosis and inhibit mitosis. TAK-733/dinaciclib combination therapy induced tumor reduction in PNF patient‒derived xenografts mouse models. Therefore, the combination of MEKi and cyclin-dependent kinase inhibitor may be promising for treating inoperable PNFs, especially when drug resistance exists. Our findings provide evidence for future clinical trials with MEKi-resistant patients with PNF.


Asunto(s)
Neurofibroma Plexiforme , Neurofibromatosis 1 , Animales , Quinasas Ciclina-Dependientes , Quinasas MAP Reguladas por Señal Extracelular , Humanos , Ratones , Quinasas de Proteína Quinasa Activadas por Mitógenos , Neurofibroma Plexiforme/tratamiento farmacológico , Neurofibromatosis 1/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico
10.
PLoS One ; 16(2): e0241821, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33630851

RESUMEN

Real-time quantitative PCR (RT-qPCR) has been widely applied in uncovering disease mechanisms and screening potential biomarkers. Internal reference gene selection determines the accuracy and reproducibility of data analyses. The aim of this study was to identify the optimal reference genes for the relative quantitative analysis of RT-qPCR in fourteen NF1 related cell lines, including non-tumor, benign and malignant Schwann cell lines. The expression characteristics of eleven candidate reference genes (RPS18, ACTB, B2M, GAPDH, PPIA, HPRT1, TBP, UBC, RPLP0, TFRC and RPL32) were screened and analyzed by four software programs: geNorm, NormFinder, BestKeeper and RefFinder. Results showed that GAPDH, the most frequently used internal reference gene, was significantly unstable between various cell lines. The combinational use of two reference genes (PPIA and TBP) was optimal in malignant Schwann cell lines and the use of single reference genes (PPIA or PRLP0) alone or in combination was optimal in benign Schwann cell lines. These recommended internal reference gene selections may improve the accuracy and reproducibility of RT-qPCR in gene expression analyses of NF1 related tumors.


Asunto(s)
Genes de Neurofibromatosis 1 , Neurofibromatosis 1/genética , Reacción en Cadena en Tiempo Real de la Polimerasa/métodos , Células de Schwann/metabolismo , Línea Celular , Línea Celular Tumoral , Humanos , Neurofibromina 1/genética , Reacción en Cadena en Tiempo Real de la Polimerasa/normas , Estándares de Referencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA