Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.165
Filtrar
1.
Zhong Nan Da Xue Xue Bao Yi Xue Ban ; 49(1): 113-121, 2024 Jan 28.
Artículo en Inglés, Chino | MEDLINE | ID: mdl-38615172

RESUMEN

Malignant tumors continue to pose a significant threat to human life and safety and their development is primarily due to the activation of proto-oncogenes and the inactivation of suppressor genes. Among these, the activation of proto-oncogenes possesses greater potential to drive the malignant transformation of cells. Targeting oncogenes involved in the malignant transformation of tumor cells has provided a novel approach for the development of current antitumor drugs. Several preclinical and clinical studies have revealed that the development pathway of B cells, and the malignant transformation of mature B cells into tumors have been regulated by oncogenes and their metabolites. Therefore, summarizing the key oncogenes involved in the process of malignant transformation of mature B cells and elucidating the mechanisms of action in tumor development hold significant importance for the clinical treatment of malignant tumors.


Asunto(s)
Linfocitos B , Neoplasias , Humanos , Proto-Oncogenes/genética , Neoplasias/genética
2.
Rinsho Ketsueki ; 64(10): 1258-1265, 2023.
Artículo en Japonés | MEDLINE | ID: mdl-37914237

RESUMEN

In acute myeloid leukemia (AML), EVI1 rearrangement represented by inv(3)(q21q26) or t(3;3)(q21;q26) causes EVI1 overexpression via structural rearrangement of an enhancer, and confers poor prognosis. My colleagues and I performed a mutational analysis of EVI1-rearranged myeloid neoplasms and identified SF3B1, a core RNA splicing factor, as the most commonly co-mutated gene. Indeed, latent leukemia development in transgenic mice bearing the humanized inv(3)(q21q26) allele was significantly accelerated by co-occurrence of Sf3b1 mutation. Intriguingly, we found that this SF3B1 mutant induced mis-splicing of EVI1 itself, which generated an aberrant EVI1 isoform with in-frame insertion of 6 amino acids near the DNA-binding domain of EVI1. This aberrant EVI1 isoform exhibited DNA-binding activity different from wild-type EVI1 and significantly enhanced the self-renewal capacity of murine hematopoietic stem cells. We also identified the cryptic branch point and exonic splicing enhancer required for this EVI1 mis-splicing induced by the SF3B1 mutant. These data provide a basis for further elucidation of the molecular mechanism and potential therapeutic candidates for EVI1-rearranged AML.


Asunto(s)
Leucemia Mieloide Aguda , Trastornos Mieloproliferativos , Ratones , Animales , Humanos , Proteínas de Unión al ADN/genética , Proteína del Locus del Complejo MDS1 y EV11/genética , Proteína del Locus del Complejo MDS1 y EV11/metabolismo , Translocación Genética , Proto-Oncogenes/genética , Factores de Transcripción/genética , Mutación , Trastornos Mieloproliferativos/genética , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/terapia , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , ADN , Cromosomas Humanos Par 3/metabolismo , Factores de Empalme de ARN/genética , Fosfoproteínas/genética
3.
Pathol Res Pract ; 248: 154659, 2023 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-37429176

RESUMEN

Cancer genes are largely categorized into tumor suppressor gene (TSG) and proto-oncogene, but many have dual activities depending on the cellular context. In the present study, we analyzed DYRK1B, ESRP1, MTSS1, ADAMTS1, and INPP5F genes known to possess the dual activities in sporadic colon cancers (CCs). By the mutation analysis, we identified DYRK1B, ESRP1, MTSS1, ADAMTS1, and INPP5F frameshift mutations in 2, 2, 3, 3, and 1 CCs in instability-high (MSI-H) cases (1.1-3.2% of MSI-H CCs), respectively, but not microsatellite stable (MSS) cases. One CC showed regional heterogeneous mutations (RHM) of ESRP1 mutation. Immunohistochemistry identified protein expression of ESRP1, MTSS1, and ADAMTS1 in the CCs, revealing that approximately 30% of CCs lost the protein expression irrespective of the MSI status. Our study showed that dual TSG and proto-oncogene genes DYRK1B, ESRP1, MTSS1, ADAMTS1, and INPP5F harbored low incidences of inactivating mutations, but that the protein losses were frequent in CCs. Our study suggests a possibility that the dual-function genes could be altered mainly at the expression level, which might contribute to CC pathogenesis.


Asunto(s)
Neoplasias del Colon , Neoplasias Colorrectales , Humanos , Neoplasias Colorrectales/patología , Genes Supresores de Tumor , Mutación , Mutación del Sistema de Lectura , Neoplasias del Colon/genética , Inestabilidad de Microsatélites , Proto-Oncogenes/genética , Repeticiones de Microsatélite , Proteínas de Microfilamentos/metabolismo , Proteínas de Neoplasias/metabolismo
4.
Blood ; 141(5): 453-466, 2023 02 02.
Artículo en Inglés | MEDLINE | ID: mdl-36095844

RESUMEN

Chromosomal rearrangements involving the MDS1 and EVI1 complex locus (MECOM) on chromosome 3q26 define an aggressive subtype of acute myeloid leukemia (AML) that is associated with chemotherapy resistance and dismal prognosis. Established treatment regimens commonly fail in these patients, therefore, there is an urgent need for new therapeutic concepts that will require a better understanding of the molecular and cellular functions of the ecotropic viral integration site 1 (EVI1) oncogene. To characterize gene regulatory functions of EVI1 and associated dependencies in AML, we developed experimentally tractable human and murine disease models, investigated the transcriptional consequences of EVI1 withdrawal in vitro and in vivo, and performed the first genome-wide CRISPR screens in EVI1-dependent AML. By integrating conserved transcriptional targets with genetic dependency data, we identified and characterized the ETS transcription factor ERG as a direct transcriptional target of EVI1 that is aberrantly expressed and selectively required in both human and murine EVI1-driven AML. EVI1 controls the expression of ERG and occupies a conserved intragenic enhancer region in AML cell lines and samples from patients with primary AML. Suppression of ERG induces terminal differentiation of EVI1-driven AML cells, whereas ectopic expression of ERG abrogates their dependence on EVI1, indicating that the major oncogenic functions of EVI1 are mediated through aberrant transcriptional activation of ERG. Interfering with this regulatory axis may provide entry points for the development of rational targeted therapies.


Asunto(s)
Proteínas de Unión al ADN , Leucemia Mieloide Aguda , Humanos , Animales , Ratones , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Proteína del Locus del Complejo MDS1 y EV11/genética , Proto-Oncogenes/genética , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Carcinogénesis/genética , Regulador Transcripcional ERG/genética
5.
Blood Cancer J ; 12(11): 149, 2022 11 04.
Artículo en Inglés | MEDLINE | ID: mdl-36329027

RESUMEN

Neoplasms originating from thymic T-cell progenitors and post-thymic mature T-cell subsets account for a minority of lymphoproliferative neoplasms. These T-cell derived neoplasms, while molecularly and genetically heterogeneous, exploit transcription factors and signaling pathways that are critically important in normal T-cell biology, including those implicated in antigen-, costimulatory-, and cytokine-receptor signaling. The transcription factor GATA-3 regulates the growth and proliferation of both immature and mature T cells and has recently been implicated in T-cell neoplasms, including the most common mature T-cell lymphoma observed in much of the Western world. Here we show that GATA-3 is a proto-oncogene across the spectrum of T-cell neoplasms, including those derived from T-cell progenitors and their mature progeny, and further define the transcriptional programs that are GATA-3 dependent, which include therapeutically targetable gene products. The discovery that p300-dependent acetylation regulates GATA-3 mediated transcription by attenuating DNA binding has novel therapeutic implications. As most patients afflicted with GATA-3 driven T-cell neoplasms will succumb to their disease within a few years of diagnosis, these findings suggest opportunities to improve outcomes for these patients.


Asunto(s)
Proteínas de Unión al ADN , Neoplasias , Humanos , Diferenciación Celular , Proteínas de Unión al ADN/genética , Neoplasias/metabolismo , Proto-Oncogenes/genética , Subgrupos de Linfocitos T , Leucemia Linfoide
6.
Blood ; 140(8): 875-888, 2022 08 25.
Artículo en Inglés | MEDLINE | ID: mdl-35709354

RESUMEN

Detailed genomic and epigenomic analyses of MECOM (the MDS1 and EVI1 complex locus) have revealed that inversion or translocation of chromosome 3 drives inv(3)/t(3;3) myeloid leukemias via structural rearrangement of an enhancer that upregulates transcription of EVI1. Here, we identify a novel, previously unannotated oncogenic RNA-splicing derived isoform of EVI1 that is frequently present in inv(3)/t(3;3) acute myeloid leukemia (AML) and directly contributes to leukemic transformation. This EVI1 isoform is generated by oncogenic mutations in the core RNA splicing factor SF3B1, which is mutated in >30% of inv(3)/t(3;3) myeloid neoplasm patients and thereby represents the single most commonly cooccurring genomic alteration in inv(3)/t(3;3) patients. SF3B1 mutations are statistically uniquely enriched in inv(3)/t(3;3) myeloid neoplasm patients and patient-derived cell lines compared with other forms of AML and promote mis-splicing of EVI1 generating an in-frame insertion of 6 amino acids at the 3' end of the second zinc finger domain of EVI1. Expression of this EVI1 splice variant enhanced the self-renewal of hematopoietic stem cells, and introduction of mutant SF3B1 in mice bearing the humanized inv(3)(q21q26) allele resulted in generation of this novel EVI1 isoform in mice and hastened leukemogenesis in vivo. The mutant SF3B1 spliceosome depends upon an exonic splicing enhancer within EVI1 exon 13 to promote usage of a cryptic branch point and aberrant 3' splice site within intron 12 resulting in the generation of this isoform. These data provide a mechanistic basis for the frequent cooccurrence of SF3B1 mutations as well as new insights into the pathogenesis of myeloid leukemias harboring inv(3)/t(3;3).


Asunto(s)
Leucemia Mieloide Aguda , Proto-Oncogenes , Animales , Inversión Cromosómica , Cromosomas Humanos Par 3/metabolismo , Proteínas de Unión al ADN/metabolismo , Humanos , Leucemia Mieloide Aguda/patología , Proteína del Locus del Complejo MDS1 y EV11/genética , Ratones , Proto-Oncogenes/genética , Factores de Transcripción/metabolismo
7.
Int J Mol Sci ; 23(9)2022 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-35563163

RESUMEN

The ETS-related gene (ERG) is proto-oncogene that is classified as a member of the ETS transcription factor family, which has been found to be consistently overexpressed in about half of the patients with clinically significant prostate cancer (PCa). The overexpression of ERG can mostly be attributed to the fusion of the ERG and transmembrane serine protease 2 (TMPRSS2) genes, and this fusion is estimated to represent about 85% of all gene fusions observed in prostate cancer. Clinically, individuals with ERG gene fusion are mostly documented to have advanced tumor stages, increased mortality, and higher rates of metastasis in non-surgical cohorts. In the current review, we elucidate ERG's molecular interaction with downstream genes and the pathways associated with PCa. Studies have documented that ERG plays a central role in PCa progression due to its ability to enhance tumor growth by promoting inflammatory and angiogenic responses. ERG has also been implicated in the epithelial-mesenchymal transition (EMT) in PCa cells, which increases the ability of cancer cells to metastasize. In vivo, research has demonstrated that higher levels of ERG expression are involved with nuclear pleomorphism that prompts hyperplasia and the loss of cell polarity.


Asunto(s)
Proteínas de Fusión Oncogénica , Neoplasias de la Próstata , Carcinogénesis/genética , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Proteínas de Fusión Oncogénica/genética , Neoplasias de la Próstata/metabolismo , Proteínas Proto-Oncogénicas c-ets/genética , Proto-Oncogenes/genética , Regulador Transcripcional ERG/genética , Regulador Transcripcional ERG/metabolismo
8.
Asian Pac J Cancer Prev ; 23(1): 131-142, 2022 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-35092381

RESUMEN

BACKGROUND: Targeted therapy in adenocarcinoma is recommended. The use of immune check point inhibitors for the treatment of Non-small cell lung carcinoma (NSCLC) is used as both first-line and the second-line treatment strategy. The current study was undertaken to assess the frequency of programmed cell death ligand-1 (PD-L1) expression with anaplastic lymphoma kinase (ALK), proto-oncogene 1, receptor tyrosine kinase (ROS), epidermal growth factor receptor (EGFR), Kirsten rat sarcoma (KRAS), and v-Raf murine sarcoma viral oncogene homolog B (BRAF)V600E driver gene mutations in NSCLC adenocarcinoma phenotype. It assesses the frequencies of all markers in the cases where both treatment strategies can be implemented. Expression of the all markers was further compared with demographic, clinical parameters, and overall survival rate. MATERIALS AND METHODS: The formalin-fixed paraffin-embedded (FFPE) tissue blocks were used in immunohistochemistry (IHC) staining and real-time polymerase chain reaction (RT-PCR) for determining the driver genes and PD-L1 expression in the 100 NSCLC-Adenocarcinoma cases. RESULTS: PD-L1 positivity was observed in 26.36% (n=29/110) cases in adenocarcinoma. No significant differences in PD-L1 expression were observed among patients harboring ALK, ROS1, EGFR, KRAS, and BRAF mutations EGFR mutations had significant association with smoking status. (p= 0.008), Thyroid transcription factor 1 (TTF1) (p=0.0005) and Napsin (p=0.002) expression. ALK gene re-arrangement was significantly related to age (p= 0.001), gender (p= 0.009) and smoking status (p= 0.043). The single versus multiple driver mutations were significantly correlated with smoking status (p=0.005). In the survival rate analysis, EGFR (p=0.058), KRAS (p=0.021), and PD-L1 (p=0.039) were significantly high with the positive versus negative group. CONCLUSIONS: The current study is a novel attempt to document the co-expression of multiple driver mutations in the NSCLC-adenocarcinoma phenotype. PD-L1 immunopositivity in NSCLC-adenocarcinoma was higher with EGFR mutation as compared to those of KRAS, ALK, ROS, and BRAF driver genes.


Asunto(s)
Adenocarcinoma/genética , Quinasa de Linfoma Anaplásico/genética , Antígeno B7-H1/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/genética , Neoplasias Pulmonares/genética , Proto-Oncogenes/genética , Animales , Modelos Animales de Enfermedad , Receptores ErbB/genética , Inmunohistoquímica , Ratones , Mutación , Fenotipo , Proteínas Proto-Oncogénicas B-raf/genética , Proteínas Proto-Oncogénicas p21(ras)/genética , Proteínas Tirosina Quinasas Receptoras/genética
9.
Nat Commun ; 12(1): 5679, 2021 09 28.
Artículo en Inglés | MEDLINE | ID: mdl-34584081

RESUMEN

Chromosomal rearrangements are a frequent cause of oncogene deregulation in human malignancies. Overexpression of EVI1 is found in a subgroup of acute myeloid leukemia (AML) with 3q26 chromosomal rearrangements, which is often therapy resistant. In AMLs harboring a t(3;8)(q26;q24), we observed the translocation of a MYC super-enhancer (MYC SE) to the EVI1 locus. We generated an in vitro model mimicking a patient-based t(3;8)(q26;q24) using CRISPR-Cas9 technology and demonstrated hyperactivation of EVI1 by the hijacked MYC SE. This MYC SE contains multiple enhancer modules, of which only one recruits transcription factors active in early hematopoiesis. This enhancer module is critical for EVI1 overexpression as well as enhancer-promoter interaction. Multiple CTCF binding regions in the MYC SE facilitate this enhancer-promoter interaction, which also involves a CTCF binding site upstream of the EVI1 promoter. We hypothesize that this CTCF site acts as an enhancer-docking site in t(3;8) AML. Genomic analyses of other 3q26-rearranged AML patient cells point to a common mechanism by which EVI1 uses this docking site to hijack enhancers active in early hematopoiesis.


Asunto(s)
Factor de Unión a CCCTC/genética , Elementos de Facilitación Genéticos/genética , Leucemia Mieloide/genética , Proteína del Locus del Complejo MDS1 y EV11/genética , Proteínas Proto-Oncogénicas c-myc/genética , Proto-Oncogenes/genética , Enfermedad Aguda , Factor de Unión a CCCTC/metabolismo , Cromosomas Humanos Par 3/genética , Cromosomas Humanos Par 8/genética , Regulación Leucémica de la Expresión Génica , Reordenamiento Génico , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Humanos , Hibridación Fluorescente in Situ/métodos , Células K562 , Cariotipificación , Leucemia Mieloide/metabolismo , Leucemia Mieloide/patología , Regiones Promotoras Genéticas/genética , Unión Proteica , Translocación Genética
10.
Parasit Vectors ; 14(1): 283, 2021 May 26.
Artículo en Inglés | MEDLINE | ID: mdl-34039408

RESUMEN

BACKGROUND: The proliferative stage (tachyzoite) of Toxoplasma gondii (T. gondii) is critical for its transmission and pathogenesis, and a proto-oncogene eukaryotic translation initiation factor (eIF-5A) plays an important role in various cellular processes such as cell multiplication. METHODS: We performed a proteomic study to evaluate the specific roles of eIF-5A involved in invasion and replication of T. gondii, and both in vivo and in vitro trials using eIF-5A-interfered and wild tachyzoites were performed to verify the proteomic results. RESULTS: The results of our study showed that T. gondii eIF-5A affected tachyzoite growth and also participated in the synthesis of proteins through regulation of both ribosomal and splicing pathways. Inhibition of eIF-5A in T. gondii resulted in the downregulated expression of soluble adhesions, such as microneme protein 1 (MIC1) and MIC4, which in turn decreased the parasite population that adhered to the surface of host cells. The reduced attachment, combined with lower expression of some rhoptry proteins (ROPs) and dense granule antigens (GRAs) involved in different stages of T. gondii invasion such as ROP4 and GRA3, ultimately reduce the invasion efficiency. These processes regulated by eIF-5A eventually affect the replication of tachyzoites. CONCLUSIONS: Our findings showed that eIF-5A influenced tachyzoite survival and was also involved in the process of parasite invasion and replication. These results will provide new clues for further development of targeted drugs to control T. gondii infection.


Asunto(s)
Factores de Iniciación de Péptidos/genética , Factores de Iniciación de Péptidos/metabolismo , Proteómica/métodos , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Toxoplasma/genética , Toxoplasma/fisiología , Animales , Sistemas CRISPR-Cas , Replicación del ADN , Femenino , Eliminación de Gen , Ratones Endogámicos BALB C , Mapas de Interacción de Proteínas , Proto-Oncogenes/genética , Proteínas Protozoarias/genética , Proteínas Protozoarias/metabolismo , Ratas , Ratas Sprague-Dawley , Toxoplasmosis/parasitología , Virulencia , Factor 5A Eucariótico de Iniciación de Traducción
11.
J Hematol Oncol ; 14(1): 50, 2021 03 26.
Artículo en Inglés | MEDLINE | ID: mdl-33771190

RESUMEN

The recent approvals by the Food and Drug Administration several tumor-agnostic drugs have resulted in a paradigm shift in cancer treatment from an organ/histology-specific strategy to biomarker-guided approaches. RET gene fusions are oncogenic drivers in multiple tumor types and are known to occur in 1-2% of non-squamous NSCLC patients. RET gene fusions give rise to chimeric, cytosolic proteins with constitutively active RET kinase domain. Standard therapeutic regimens provide limited benefit for NSCLC patients with RET fusion-positive tumors, and the outcomes with immunotherapy in the these patients are generally poor. Selpercatinib (LOXO-292) and pralsetinib (BLU-667) are potent and selective inhibitors that target RET alterations, including fusions and mutations, irrespective of the tissue of origin. Recently, the results from the LIBRETTO-001 and ARROW clinical trials demonstrated significant clinical benefits with selpercatinib and pralsetinib respectively, in NSCLC patients with RET gene fusions, with tolerable toxicity profiles. These studies also demonstrated that these RET-TKIs crossed the blood brain barrier with significant activity. As has been observed with other TKIs, the emergence of acquired resistance may limit long-term efficacy of these agents. Therefore, understanding the mechanisms of resistance is necessary for the development of strategies to overcome them.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/genética , Neoplasias Pulmonares/genética , Proteínas Proto-Oncogénicas c-ret/genética , Proto-Oncogenes/genética , Carcinoma de Pulmón de Células no Pequeñas/patología , Humanos , Neoplasias Pulmonares/patología
12.
Nat Commun ; 12(1): 1622, 2021 03 12.
Artículo en Inglés | MEDLINE | ID: mdl-33712608

RESUMEN

Our mathematical model of integration site data in clinical gene therapy supported the existence of long-term lymphoid progenitors capable of surviving independently from hematopoietic stem cells. To date, no experimental setting has been available to validate this prediction. We here report evidence of a population of lymphoid progenitors capable of independently maintaining T and NK cell production for 15 years in humans. The gene therapy patients of this study lack vector-positive myeloid/B cells indicating absence of engineered stem cells but retain gene marking in both T and NK. Decades after treatment, we can still detect and analyse transduced naïve T cells whose production is likely maintained by a population of long-term lymphoid progenitors. By tracking insertional clonal markers overtime, we suggest that these progenitors can support both T and NK cell production. Identification of these long-term lymphoid progenitors could be utilised for the development of next generation gene- and cancer-immunotherapies.


Asunto(s)
Células Asesinas Naturales/fisiología , Linfocitos/fisiología , Células Progenitoras Linfoides/fisiología , Linfocitos T/fisiología , Linfocitos B , Terapia Genética/métodos , Células Madre Hematopoyéticas , Humanos , Interferón gamma/metabolismo , Mutagénesis , Células Mieloides/fisiología , Proto-Oncogenes/genética , Proto-Oncogenes/fisiología
13.
Genome Biol Evol ; 13(1)2021 01 07.
Artículo en Inglés | MEDLINE | ID: mdl-33196781

RESUMEN

Comparative genomic analyses have enormous potential for identifying key genes central to human health phenotypes, including those that promote cancers. In particular, the successful development of novel therapeutics using model species requires phylogenetic analyses to determine molecular homology. Accordingly, we investigate the evolutionary histories of anaplastic lymphoma kinase (ALK)-which can underlie tumorigenesis in neuroblastoma, nonsmall cell lung cancer, and anaplastic large-cell lymphoma-its close relative leukocyte tyrosine kinase (LTK) and their candidate ligands. Homology of ligands identified in model organisms to those functioning in humans remains unclear. Therefore, we searched for homologs of the human genes across metazoan genomes, finding that the candidate ligands Jeb and Hen-1 were restricted to nonvertebrate species. In contrast, the ligand augmentor (AUG) was only identified in vertebrates. We found two ALK-like and four AUG-like protein-coding genes in lamprey. Of these six genes, only one ALK-like and two AUG-like genes exhibited early embryonic expression that parallels model mammal systems. Two copies of AUG are present in nearly all jawed vertebrates. Our phylogenetic analysis strongly supports the presence of previously unrecognized functional convergences of ALK and LTK between actinopterygians and sarcopterygians-despite contemporaneous, highly conserved synteny of ALK and LTK. These findings provide critical guidance regarding the propriety of fish and mammal models with regard to model organism-based investigation of these medically important genes. In sum, our results provide the phylogenetic context necessary for effective investigations of the functional roles and biology of these critically important receptors.


Asunto(s)
Quinasa de Linfoma Anaplásico/genética , Evolución Biológica , Genómica , Neoplasias Pulmonares/genética , Proto-Oncogenes/genética , Proteínas Tirosina Quinasas Receptoras/genética , Animales , Proteínas de Caenorhabditis elegans/genética , Carcinoma de Pulmón de Células no Pequeñas/genética , Genoma , Humanos , Ligandos , Proteínas del Tejido Nervioso/genética , Filogenia , Proto-Oncogenes Mas , Sintenía , Vertebrados/genética
14.
Clin Chem Lab Med ; 59(2): 353-363, 2020 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-33001846

RESUMEN

Objectives: Plasma free metanephrines are commonly used for diagnosis of pheochromocytoma and paraganglioma (PPGLs), but can also provide other information. This multicenter study prospectively examined whether tumor size, location, and mutations could be predicted by these metabolites. Methods: Predictions of tumor location, size, and mutation type, based on measurements of plasma normetanephrine, metanephrine, and methoxytyramine were made without knowledge of disease in 267 patients subsequently determined to have PPGLs. Results: Predictions of adrenal vs. extra-adrenal locations according to increased plasma concentrations of metanephrine and methoxytyramine were correct in 93 and 97% of the respective 136 and 33 patients in who these predictions were possible. Predicted mean tumor diameters correlated positively (p<0.0001) with measured diameters; predictions agreed well for pheochromocytomas but were overestimated for paragangliomas. Considering only patients with mutations, 51 of the 54 (94%) patients with NF1 or RET mutations were correctly predicted with those mutations according to increased plasma metanephrine, whereas no or minimal increase in metanephrine correctly predicted all 71 patients with either VHL or SDHx mutations; furthermore, among the latter group increases in methoxytyramine correctly predicted SDHx mutations in 93% of the 29 cases for this specific prediction. Conclusions: Extents and patterns of increased plasma O-methylated catecholamine metabolites among patients with PPGLs allow predictions of tumor size, adrenal vs. extra-adrenal locations and general types of mutations. Predictions of tumor location are, however, only possible for patients with clearly increased plasma methoxytyramine or metanephrine. Where possible or clinically relevant the predictions are potentially useful for subsequent clinical decision-making.


Asunto(s)
Neoplasias de las Glándulas Suprarrenales/diagnóstico , Biomarcadores de Tumor/sangre , Metanefrina/sangre , Paraganglioma/diagnóstico , Feocromocitoma/diagnóstico , Adulto , Anciano , Anciano de 80 o más Años , Algoritmos , Dopamina/análogos & derivados , Dopamina/sangre , Femenino , Humanos , Masculino , Persona de Mediana Edad , Mutación , Neurofibromatosis 1/genética , Normetanefrina/sangre , Estudios Prospectivos , Proto-Oncogenes/genética , Factores de Riesgo
15.
Appl. cancer res ; 40: 1-12, Oct. 19, 2020. tab, ilus
Artículo en Inglés | LILACS, Inca | ID: biblio-1281498

RESUMEN

Background: Next-generation sequencing (NGS) based assay for finding an actionable driver in non-small-cell lung cancer is a less used modality in clinical practice. With a long list of actionable targets, limited tissue, arduous single-gene assays, the alternative of NGS for broad testing in one experiment looks attractive. We report here our experience with NGS for biomarker testing in hundred advanced lung cancer patients. Methods: Predictive biomarker testing was performed using the Ion AmpliSeq™ Cancer Hotspot Panel V2 (30 tumors) and Oncomine™ Solid Tumor DNA and Oncomine™ Solid Tumor Fusion Transcript kit (70 tumors) on IonTorrent sequencing platform. Results: One-seventeen distinct aberrations were detected across 29 genes in eighty-six tumors. The most commonly mutated genes were TP53 (43% cases), EGFR (23% cases) and KRAS (17% cases). Thirty-four patients presented an actionable genetic variant for which targeted therapy is presently available, and fifty-two cases harbored non-actionable variants with the possibility of recruitment in clinical trials. NGS results were validated by individual tests for detecting EGFR mutation, ALK1 rearrangement, ROS1 fusion, and c-MET amplification. Compared to single test, NGS exhibited good agreement for detecting EGFR mutations and ALK1 fusion (sensitivity- 88.89%, specificity- 100%, Kappa-score 0.92 and sensitivity- 80%, specificity- 100%, Kappa-score 0.88; respectively). Further, the response of patients harboring tyrosine kinase inhibitor (TKI) sensitizing EGFR mutations was assessed. The progression-free-survival of EGFR positive patients on TKI therapy, harboring a concomitant mutation in PIK3CAmTOR and/or RAS-RAF-MAPK pathway gene and/or TP53 gene was inferior to those with sole-sensitizing EGFR mutation (2 months vs. 9.5 months, P = 0.015). Conclusions: This is the first study from South Asia looking into the analytical validity of NGS and describing the mutational landscape of lung cancer patients to study the impact of co-mutations on cancer biology and treatment outcome. Our study demonstrates the clinical utility of NGS testing for identifying actionable variants and making treatment decisions in advanced lung cancer


Asunto(s)
Humanos , Masculino , Femenino , Proto-Oncogenes/genética , Biomarcadores de Tumor/genética , Carcinoma de Pulmón de Células no Pequeñas/genética , Secuenciación de Nucleótidos de Alto Rendimiento , Neoplasias Pulmonares/genética , Mutación/genética , Reproducibilidad de los Resultados
16.
Gastroenterology ; 159(6): 2203-2220.e14, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32814112

RESUMEN

BACKGROUND AND AIMS: The pattern of genetic alterations in cancer driver genes in patients with hepatocellular carcinoma (HCC) is highly diverse, which partially explains the low efficacy of available therapies. In spite of this, the existing mouse models only recapitulate a small portion of HCC inter-tumor heterogeneity, limiting the understanding of the disease and the nomination of personalized therapies. Here, we aimed at establishing a novel collection of HCC mouse models that captured human HCC diversity. METHODS: By performing hydrodynamic tail-vein injections, we tested the impact of altering a well-established HCC oncogene (either MYC or ß-catenin) in combination with an additional alteration in one of eleven other genes frequently mutated in HCC. Of the 23 unique pairs of genetic alterations that we interrogated, 9 were able to induce HCC. The established HCC mouse models were characterized at histopathological, immune, and transcriptomic level to identify the unique features of each model. Murine HCC cell lines were generated from each tumor model, characterized transcriptionally, and used to identify specific therapies that were validated in vivo. RESULTS: Cooperation between pairs of driver genes produced HCCs with diverse histopathology, immune microenvironments, transcriptomes, and drug responses. Interestingly, MYC expression levels strongly influenced ß-catenin activity, indicating that inter-tumor heterogeneity emerges not only from specific combinations of genetic alterations but also from the acquisition of expression-dependent phenotypes. CONCLUSIONS: This novel collection of murine HCC models and corresponding cell lines establishes the role of driver genes in diverse contexts and enables mechanistic and translational studies.


Asunto(s)
Carcinoma Hepatocelular/genética , Heterogeneidad Genética , Proto-Oncogenes/genética , Animales , Carcinoma Hepatocelular/inmunología , Carcinoma Hepatocelular/patología , Línea Celular Tumoral , Biología Computacional , Modelos Animales de Enfermedad , Resistencia a Antineoplásicos/genética , Femenino , Regulación Neoplásica de la Expresión Génica/inmunología , Humanos , Neoplasias Hepáticas/inmunología , Neoplasias Hepáticas/patología , Masculino , Ratones , Ratones Transgénicos , Escape del Tumor/genética , Microambiente Tumoral/genética , Microambiente Tumoral/inmunología
17.
Mol Biol Rep ; 47(6): 4875-4878, 2020 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-32410140

RESUMEN

Zbtb7a is a transcription factor whose dysfunction is correlated to the development of several types of cancer, including hepatocellular carcinoma (HCC). It generally acts as a repressor of transcription downregulating the expression of several target genes including oncosuppressors ARF and Rb. In this study, Zbtb7a was found to suppress the expression of miR-125a, an oncosuppressive miRNA that is often downregulated in HCC. This effect is mediated by the binding of the transcription factor to a regulatory sequence in the promoter of the transcription unit of miR-125a located 14 bp upstream of the transcription start site. Consistent with this observation, the analysis of 370 HCC samples showed an upregulation of Zbtb7a compared to 50 normal liver tissues and a reverse correlation with miR-125a expression. These data suggest that miR-125a may support the oncogenic potential of Zbtb7a.


Asunto(s)
Carcinoma Hepatocelular/genética , Proteínas de Unión al ADN/genética , MicroARNs/genética , Factores de Transcripción/genética , Línea Celular Tumoral , Proliferación Celular/genética , Proteínas de Unión al ADN/metabolismo , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Neoplasias Hepáticas/genética , MicroARNs/metabolismo , Proto-Oncogenes Mas , Proto-Oncogenes/genética , Factores de Transcripción/metabolismo
18.
Cancer Sci ; 111(8): 2682-2688, 2020 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-32462673

RESUMEN

Deregulation of cytokine signaling is frequently associated with various pathological conditions, including malignancies. In patients with myeloproliferative neoplasms (MPNs), recurrent somatic mutations in the calreticulin (CALR) gene, which encodes a molecular chaperone that resides in the endoplasmic reticulum, have been reported. Studies have defined mutant CALR as an oncogene promoting the development of MPN, and deciphered a novel molecular mechanism by which mutant CALR constitutively activates thrombopoietin receptor MPL and its downstream molecules to induce cellular transformation. The mechanism of interaction and activation of MPL by mutant CALR is unique, not only due to the latter forming a homomultimeric complex through a novel mutant-specific sequence generated by frameshift mutation, but also for its ability to interact with immature asparagine-linked glycan for eventual engagement with immature MPL in the endoplasmic reticulum. The complex formed between mutant CALR and MPL is then transported to the cell surface, where it induces constitutive activation of downstream kinase JAK2 bound to MPL. Refined structural and cell biological studies can provide an in-depth understanding of this unusual mechanism of receptor activation by a mutant molecular chaperone. Mutant CALR is also involved in modulation of the immune response, transcription, and intracellular homeostasis, which could contribute to the development of MPN. In the present article, we comprehensively review the current understanding of the underlying molecular mechanisms for mutant molecular chaperone-induced cellular transformation.


Asunto(s)
Calreticulina/genética , Transformación Celular Neoplásica/genética , Neoplasias Hematológicas/genética , Trastornos Mieloproliferativos/genética , Proto-Oncogenes/genética , Animales , Calreticulina/metabolismo , Modelos Animales de Enfermedad , Retículo Endoplásmico/inmunología , Retículo Endoplásmico/patología , Mutación del Sistema de Lectura , Regulación Neoplásica de la Expresión Génica , Neoplasias Hematológicas/inmunología , Neoplasias Hematológicas/patología , Células Madre Hematopoyéticas/patología , Humanos , Janus Quinasa 2/metabolismo , Trastornos Mieloproliferativos/inmunología , Trastornos Mieloproliferativos/patología , Receptores de Trombopoyetina/metabolismo , Transducción de Señal/genética , Transducción de Señal/inmunología , Transcripción Genética , Microambiente Tumoral/genética , Microambiente Tumoral/inmunología
20.
PLoS Genet ; 16(3): e1008470, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32203506

RESUMEN

Cell invasion allows cells to migrate across compartment boundaries formed by basement membranes. Aberrant cell invasion is a first step during the formation of metastases by malignant cancer cells. Anchor cell (AC) invasion in C. elegans is an excellent in vivo model to study the regulation of cell invasion during development. Here, we have examined the function of egl-43, the homolog of the human Evi1 proto-oncogene (also called MECOM), in the invading AC. egl-43 plays a dual role in this process, firstly by imposing a G1 cell cycle arrest to prevent AC proliferation, and secondly, by activating pro-invasive gene expression. We have identified the AP-1 transcription factor fos-1 and the Notch homolog lin-12 as critical egl-43 targets. A positive feedback loop between fos-1 and egl-43 induces pro-invasive gene expression in the AC, while repression of lin-12 Notch expression by egl-43 ensures the G1 cell cycle arrest necessary for invasion. Reducing lin-12 levels in egl-43 depleted animals restored the G1 arrest, while hyperactivation of lin-12 signaling in the differentiated AC was sufficient to induce proliferation. Taken together, our data have identified egl-43 Evi1 as an important factor coordinating cell invasion with cell cycle arrest.


Asunto(s)
Proteínas de Caenorhabditis elegans/genética , Caenorhabditis elegans/genética , Puntos de Control del Ciclo Celular/genética , Puntos de Control de la Fase G1 del Ciclo Celular/genética , Expresión Génica/genética , Proteína del Locus del Complejo MDS1 y EV11/genética , Proto-Oncogenes/genética , Animales , Membrana Basal/metabolismo , Diferenciación Celular/genética , Proliferación Celular/genética , Proto-Oncogenes Mas , Proteínas Proto-Oncogénicas c-fos/genética , Receptores Notch/genética , Transducción de Señal/genética , Factores de Transcripción/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA