Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
EMBO J ; 35(6): 668-84, 2016 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-26912724

RESUMEN

A key step of Wnt signaling activation is the recruitment of ß-catenin to the Wnt target-gene promoter in the nucleus, but its mechanisms are largely unknown. Here, we identified FoxM1 as a novel target of Wnt signaling, which is essential for ß-catenin/TCF4 transactivation. GSK3 phosphorylates FoxM1 on serine 474 which induces FoxM1 ubiquitination mediated by FBXW7. Wnt signaling activation inhibits FoxM1 phosphorylation by GSK3-Axin complex and leads to interaction between FoxM1 and deubiquitinating enzyme USP5, thereby deubiquitination and stabilization of FoxM1. FoxM1 accumulation in the nucleus promotes recruitment of ß-catenin to Wnt target-gene promoter and activates the Wnt signaling pathway by protecting the ß-catenin/TCF4 complex from ICAT inhibition. Subsequently, the USP5-FoxM1 axis abolishes the inhibitory effect of ICAT and is required for Wnt-mediated tumor cell proliferation. Therefore, Wnt-induced deubiquitination of FoxM1 represents a novel and critical mechanism for controlling canonical Wnt signaling and cell proliferation.


Asunto(s)
Factores de Transcripción Forkhead/metabolismo , Regulación de la Expresión Génica , Proteínas Wnt/metabolismo , beta Catenina/metabolismo , Línea Celular , Endopeptidasas/metabolismo , Proteína Forkhead Box M1 , Humanos , Activación Transcripcional , Ubiquitinación , Vía de Señalización Wnt
2.
EMBO Rep ; 18(8): 1318-1330, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28623188

RESUMEN

Aberrant activation of the Hedgehog (Hh) signaling pathway drives the tumorigenesis of multiple cancers. In this study, we screened a panel of deubiquitinases that may regulate the Hh pathway. We find that deubiquitinase USP48 activates Gli-dependent transcription by stabilizing Gli1 protein. Mechanistically, USP48 interacts with Gli1 and cleaves its ubiquitin off directly. In glioblastoma cells, knockdown of USP48 inhibits cell proliferation and the expression of Gli1's downstream targets, which leads to repressed glioblastoma tumorigenesis. Importantly, USP48's effect on cell proliferation and tumorigenesis depends to some extent on Gli1. In addition, we find that the Sonic Hedgehog (SHH) pathway induces USP48 expression through Gli1-mediated transcriptional activation, which forms thus a positive feedback loop to regulate Hh signaling. In human glioblastoma specimens, the expression levels of USP48 and Gli1 proteins are clinically relevant, and high expression of USP48 correlates with glioma malignancy. In summary, our study reveals that the USP48-Gli1 regulatory axis is critical for glioma cell proliferation and glioblastoma tumorigenesis.


Asunto(s)
Carcinogénesis , Glioblastoma/metabolismo , Proteasas Ubiquitina-Específicas/metabolismo , Proteína con Dedos de Zinc GLI1/metabolismo , Línea Celular Tumoral , Proliferación Celular/genética , Transformación Celular Neoplásica , Regulación Neoplásica de la Expresión Génica , Glioblastoma/enzimología , Glioblastoma/genética , Glioblastoma/fisiopatología , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Humanos , Transducción de Señal , Factores de Transcripción/metabolismo , Proteasas Ubiquitina-Específicas/genética , Proteína con Dedos de Zinc GLI1/genética
3.
Hum Mol Genet ; 23(12): 3278-88, 2014 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-24488769

RESUMEN

Long non-coding RNAs (lncRNAs) have been recently found to be pervasively transcribed in human genome and link to diverse human diseases. However, the expression patterns and regulatory roles of lncRNAs in hematopoietic malignancies have not been reported. Here, we carried out a genome-wide lncRNA expression study in MLL-rearranged acute lymphoblastic leukemia (MLL-r ALL) and established lncRNA/messenger RNA coexpression networks to gain insight into the biological roles of these dysregulated lncRNAs. We detected a number of lncRNAs that were differentially expressed in MLL-r ALL samples compared with MLL-r wild-type and identified unique lncRNA expression patterns between MLL-r subtypes with different translocations as well as between infant MLL-r ALL with other MLL-r ALL patients, suggesting that they might be served as novel biomarkers for the disease. Importantly, several lncRNAs that correspond with membrane protein genes, including a lysosome-associated membrane protein, were identified. No such link between the membrane proteins and MLL-r leukemia has been reported previously. Impressively, the functional analysis showed that several lncRNAs corresponded to the expression of MLL-fusion protein target genes, including HOXA9, MEIS1, etc., while some other associated with histone-related functions or membrane proteins. Further experiments characterize the effect of some lncRNAs on MLL-r leukemia apoptosis and proliferation as the function of the coexpressed HOXA gene cluster. Finally, a set of lncRNAs epigenetically regulated by H3K79 methylation were also discovered. These findings may provide novel insights into the mechanisms of lncRNAs involved in the initiation of MLL-r leukemia. This is the first study linking lncRNAs to leukemogenesis.


Asunto(s)
Metilación de ADN , Proteína de la Leucemia Mieloide-Linfoide/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , ARN Largo no Codificante/genética , Adolescente , Apoptosis , Biomarcadores de Tumor , Proliferación Celular , Niño , Preescolar , Epigénesis Genética , Femenino , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , N-Metiltransferasa de Histona-Lisina , Histonas/genética , Histonas/metabolismo , Humanos , Lactante , Masculino , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Proteína de la Leucemia Mieloide-Linfoide/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras/patología , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo
4.
Mol Cancer Res ; 22(4): 337-346, 2024 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-38156967

RESUMEN

Cancer stem cells (CSC) play a critical role in metastasis, relapse, and therapy resistance in colorectal cancer. While characterization of the normal lineage of cell development in the intestine has led to the identification of many genes involved in the induction and maintenance of pluripotency, recent studies suggest significant heterogeneity in CSC populations. Moreover, while many canonical colorectal cancer CSC marker genes have been identified, the ability to use these classical markers to annotate stemness at the single-cell level is limited. In this study, we performed single-cell RNA sequencing on a cohort of 6 primary colon, 9 liver metastatic tumors, and 11 normal (nontumor) controls to identify colorectal CSCs at the single-cell level. Finding poor alignment of the 11 genes most used to identify colorectal CSC, we instead extracted a single-cell stemness signature (SCS_sig) that robustly identified "gold-standard" colorectal CSCs that expressed all marker genes. Using this SCS_sig to quantify stemness, we found that while normal epithelial cells show a bimodal distribution, indicating distinct stem and differentiated states, in tumor epithelial cells stemness is a continuum, suggesting greater plasticity in these cells. The SCS_sig score was quite variable between different tumors, reflective of the known transcriptomic heterogeneity of CRC. Notably, patients with higher SCS_sig scores had significantly shorter disease-free survival time after curative intent surgical resection, suggesting stemness is associated with relapse. IMPLICATIONS: This study reveals significant heterogeneity of expression of genes commonly used to identify colorectal CSCs, and identifies a novel stemness signature to identify these cells from scRNA-seq data.


Asunto(s)
Neoplasias Colorrectales , Recurrencia Local de Neoplasia , Humanos , Recurrencia Local de Neoplasia/patología , Perfilación de la Expresión Génica , Neoplasias Colorrectales/patología , Recurrencia , Análisis de Secuencia de ARN , Células Madre Neoplásicas/metabolismo , Línea Celular Tumoral
5.
Mol Cancer ; 12: 164, 2013 Dec 13.
Artículo en Inglés | MEDLINE | ID: mdl-24330809

RESUMEN

BACKGROUND: MicroRNA-133b (miR-133b), which is a muscle-specific microRNA, has been reported to be downregulated in human colorectal carcinoma (CRC) when compared to adjacent non-tumor tissue. However, its diagnostic value and role in CRC have yet to be described. CXC chemokine receptor-4 (CXCR4), which participates in multiple cell processes such as cell invasion-related signaling pathways, was predicted to be a potential target of miR-133b. The aim of this study was to investigate the associations and functions of miR-133b and CXCR4 in CRC initiation and invasion. METHODS: Mature miR-133b and CXCR4 expression levels were detected in 31 tumor samples and their adjacent, non-tumor tissues from patients with CRC, as well as in 6 CRC cell lines, using real-time quantitative RT-PCR (qRT-PCR). Luciferase reporter assays and Western blots were used to validate CXCR4 as a putative target gene of miR-133b. Regulation of CXCR4 expression by miR-133b was assessed using qRT-PCR and Western blot analysis, and the effects of exogenous miR-133b and CXCR4 on cell invasion and migration were evaluated in vitro using the SW-480 and SW-620 CRC cell lines. RESULTS: A significant downregulation of miR-133b was observed in 93.55% of CRC tissues, and the expression of miR-133b was much lower in metastatic tumors (stage C and D, stratified by the Modified Dukes Staging System) than in primary tumors (stage A and B). In contrast, CXCR4 protein expression significantly increased in 52.63% of CRC samples, and increased CXCR4 expression in CRC was associated with advanced tumor stage. CXCR4 was shown to be a direct target of miR-133b by luciferase reporter assays, and transfection of miR-133b mimics inhibited invasion and stimulated apoptosis of SW-480 and SW-620 CRC cells. CONCLUSIONS: Our study demonstrated that downregulated miR-133b contributed to increased cell invasion and migration in CRC by negatively regulating CXCR4. These findings may be significant for the development of therapy target for CRC.


Asunto(s)
Neoplasias Colorrectales/genética , Regulación Neoplásica de la Expresión Génica , MicroARNs/genética , Receptores CXCR4/genética , Adulto , Anciano , Anciano de 80 o más Años , Apoptosis , Células CACO-2 , Línea Celular Tumoral , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Progresión de la Enfermedad , Femenino , Células HEK293 , Humanos , Masculino , MicroARNs/metabolismo , Persona de Mediana Edad , Invasividad Neoplásica , Receptores CXCR4/metabolismo
6.
Hum Mol Genet ; 20(24): 4903-15, 2011 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-21926415

RESUMEN

Relapse is a major challenge in the successful treatment of childhood acute lymphoblastic leukemia (ALL). Despite intensive research efforts, the mechanisms of ALL relapse are still not fully understood. An understanding of the molecular mechanisms underlying treatment outcome, therapy response and the biology of relapse is required. In this study, we carried out a genome-wide microRNA (miRNA) microarray analysis to determine the miRNA expression profiles and relapse-associated miRNA patterns in a panel of matched diagnosis-relapse or diagnosis-complete remission (CR) childhood ALL samples. A set of miRNAs differentially expressed either in relapsed patients or at diagnosis compared with CR was further validated by quantitative real-time polymerase chain reaction in an independent sample set. Analysis of the predicted functions of target genes based on gene ontology 'biological process' categories revealed that the abnormally expressed miRNAs are associated with oncogenesis, classical multidrug resistance pathways and leukemic stem cell self-renewal and differentiation pathways. Several targets of the miRNAs associated with ALL relapse were experimentally validated, including FOXO3, BMI1 and E2F1. We further investigated the association of these dysregulated miRNAs with clinical outcome and confirmed significant associations for miR-708, miR-223 and miR-27a with individual relapse-free survival. Notably, miR-708 was also found to be associated with the in vivo glucocorticoid therapy response and with disease risk stratification. These miRNAs and their targets might be used to optimize anti-leukemic therapy, and serve as novel targets for development of new countermeasures of leukemia. This fundamental study may also contribute to establish the mechanisms of relapse in other cancers.


Asunto(s)
Diferenciación Celular/genética , Resistencia a Antineoplásicos/genética , Glucocorticoides/uso terapéutico , MicroARNs/genética , Células Madre Neoplásicas/patología , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Transducción de Señal/genética , Adolescente , Secuencia de Bases , Diferenciación Celular/efectos de los fármacos , Niño , Preescolar , Análisis por Conglomerados , Progresión de la Enfermedad , Supervivencia sin Enfermedad , Resistencia a Antineoplásicos/efectos de los fármacos , Femenino , Regulación Leucémica de la Expresión Génica/efectos de los fármacos , Redes Reguladoras de Genes/genética , Glucocorticoides/farmacología , Humanos , Lactante , Masculino , MicroARNs/metabolismo , Datos de Secuencia Molecular , Análisis Multivariante , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico , Modelos de Riesgos Proporcionales , Recurrencia , Regulación hacia Arriba/efectos de los fármacos
7.
JCO Precis Oncol ; 7: e2200422, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37487150

RESUMEN

PURPOSE: Activating mutations in KRAS, NRAS, and BRAF are known to cause resistance to anti-epidermal growth factor receptor (EGFR) therapy; however, only approximately 40% of patients with colorectal cancer (CRC) with RASWT tumors respond to anti-EGFR treatment. We sought to discover novel biomarkers to predict response to anti-EGFR antibody treatment in CRC and to understand mechanisms of resistance to anti-EGFR therapy. MATERIALS AND METHODS: Transcriptomic profiles from three clinical and two preclinical cohorts treated with cetuximab were used to assign consensus molecular subtypes (CMS) to each sample and correlated with outcomes. RESULTS: Restricting to RASWT patients, we observed that CMS2 tumors (canonical subtype) had significantly higher response rates relative to other CMS when treated with cetuximab combination with doublet chemotherapy (Okita et al cohort: 92% disease control rate (DCR) for CMS2, chi-square P = .04; CALGB/SWOG 80405 cohort: 90% objective response rate (ORR) for CMS2, chi-square P < .001) and with single-agent cetuximab (68%, chi-square P = .01). CMS2 tumors showed best response among right-sided (ORR = 80%) and left-sided (ORR = 92%) tumors in the CALGB/SWOG 80405 cohort. CMS2 cells lines were most likely to be sensitive to cetuximab (60%) and CMS2 patient-derived xenograft had the highest DCR (84%). We found Myc, E2F, and mammalian target of rapamycin pathways were consistently upregulated in resistant samples (enrichment score >1, false discovery rate <0.25). Inhibitors of these pathways in resistant cell lines exhibited additive effects with cetuximab. CONCLUSION: These data suggest that CRC transcriptional profiles, when used to assign CMS, provide additional ability to predict response to anti-EGFR therapy relative to using tumor sidedness alone. Notably both right-sided and left-sided CMS2 tumors had excellent response, suggesting that anti-EGFR therapy be included as a treatment option for right-sided CMS2 tumors.


Asunto(s)
Neoplasias Colorrectales , Humanos , Cetuximab/farmacología , Cetuximab/uso terapéutico , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Receptores de Factores de Crecimiento/uso terapéutico
8.
J Clin Invest ; 133(5)2023 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-36856111

RESUMEN

Emerging evidence suggests that cryptic translation within long noncoding RNAs (lncRNAs) may produce novel proteins with important developmental/physiological functions. However, the role of this cryptic translation in complex diseases (e.g., cancer) remains elusive. Here, we applied an integrative strategy combining ribosome profiling and CRISPR/Cas9 screening with large-scale analysis of molecular/clinical data for breast cancer (BC) and identified estrogen receptor α-positive (ER+) BC dependency on the cryptic ORFs encoded by lncRNA genes that were upregulated in luminal tumors. We confirmed the in vivo tumor-promoting function of an unannotated protein, GATA3-interacting cryptic protein (GT3-INCP) encoded by LINC00992, the expression of which was associated with poor prognosis in luminal tumors. GTE-INCP was upregulated by estrogen/ER and regulated estrogen-dependent cell growth. Mechanistically, GT3-INCP interacted with GATA3, a master transcription factor key to mammary gland development/BC cell proliferation, and coregulated a gene expression program that involved many BC susceptibility/risk genes and impacted estrogen response/cell proliferation. GT3-INCP/GATA3 bound to common cis regulatory elements and upregulated the expression of the tumor-promoting and estrogen-regulated BC susceptibility/risk genes MYB and PDZK1. Our study indicates that cryptic lncRNA-encoded proteins can be an important integrated component of the master transcriptional regulatory network driving aberrant transcription in cancer, and suggests that the "hidden" lncRNA-encoded proteome might be a new space for therapeutic target discovery.


Asunto(s)
Neoplasias de la Mama , ARN Largo no Codificante , Humanos , Femenino , ARN Largo no Codificante/genética , Sistemas de Lectura Abierta , Sistemas CRISPR-Cas , Neoplasias de la Mama/genética , Estrógenos
9.
Cancer Discov ; 13(12): 2652-2673, 2023 12 12.
Artículo en Inglés | MEDLINE | ID: mdl-37768068

RESUMEN

Oncogenic KRAS (KRAS*) contributes to many cancer hallmarks. In colorectal cancer, KRAS* suppresses antitumor immunity to promote tumor invasion and metastasis. Here, we uncovered that KRAS* transforms the phenotype of carcinoma-associated fibroblasts (CAF) into lipid-laden CAFs, promoting angiogenesis and tumor progression. Mechanistically, KRAS* activates the transcription factor CP2 (TFCP2) that upregulates the expression of the proadipogenic factors BMP4 and WNT5B, triggering the transformation of CAFs into lipid-rich CAFs. These lipid-rich CAFs, in turn, produce VEGFA to spur angiogenesis. In KRAS*-driven colorectal cancer mouse models, genetic or pharmacologic neutralization of TFCP2 reduced lipid-rich CAFs, lessened tumor angiogenesis, and improved overall survival. Correspondingly, in human colorectal cancer, lipid-rich CAF and TFCP2 signatures correlate with worse prognosis. This work unveils a new role for KRAS* in transforming CAFs, driving tumor angiogenesis and disease progression, providing an actionable therapeutic intervention for KRAS*-driven colorectal cancer. SIGNIFICANCE: This study identified a molecular mechanism contributing to KRAS*-driven colorectal cancer progression via fibroblast transformation in the tumor microenvironment to produce VEGFA driving tumor angiogenesis. In preclinical models, targeting the KRAS*-TFCP2-VEGFA axis impaired tumor progression, revealing a potential novel therapeutic option for patients with KRAS*-driven colorectal cancer. This article is featured in Selected Articles from This Issue, p. 2489.


Asunto(s)
Fibroblastos Asociados al Cáncer , Neoplasias del Colon , Proteínas Proto-Oncogénicas p21(ras) , Animales , Humanos , Ratones , Angiogénesis , Fibroblastos Asociados al Cáncer/metabolismo , Neoplasias del Colon/genética , Neoplasias del Colon/patología , Neoplasias Colorrectales/metabolismo , Proteínas de Unión al ADN/metabolismo , Fibroblastos/metabolismo , Lípidos , Proteínas Proto-Oncogénicas p21(ras)/genética , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Factores de Transcripción/metabolismo , Microambiente Tumoral/genética
10.
Nat Struct Mol Biol ; 30(12): 1878-1892, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37932451

RESUMEN

Emerging evidence suggests that cryptic translation beyond the annotated translatome produces proteins with developmental or physiological functions. However, functions of cryptic non-canonical open reading frames (ORFs) in cancer remain largely unknown. To fill this gap and systematically identify colorectal cancer (CRC) dependency on non-canonical ORFs, we apply an integrative multiomic strategy, combining ribosome profiling and a CRISPR-Cas9 knockout screen with large-scale analysis of molecular and clinical data. Many such ORFs are upregulated in CRC compared to normal tissues and are associated with clinically relevant molecular subtypes. We confirm the in vivo tumor-promoting function of the microprotein SMIMP, encoded by a primate-specific, long noncoding RNA, the expression of which is associated with poor prognosis in CRC, is low in normal tissues and is specifically elevated in CRC and several other cancer types. Mechanistically, SMIMP interacts with the ATPase-forming domains of SMC1A, the core subunit of the cohesin complex, and facilitates SMC1A binding to cis-regulatory elements to promote epigenetic repression of the tumor-suppressive cell cycle regulators encoded by CDKN1A and CDKN2B. Thus, our study reveals a cryptic microprotein as an important component of cohesin-mediated gene regulation and suggests that the 'dark' proteome, encoded by cryptic non-canonical ORFs, may contain potential therapeutic or diagnostic targets.


Asunto(s)
Sistemas CRISPR-Cas , Neoplasias , Animales , Humanos , Sistemas de Lectura Abierta/genética , Sistemas CRISPR-Cas/genética , Neoplasias/genética , Proteoma/genética
11.
Mol Pharmacol ; 81(4): 578-86, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22252650

RESUMEN

Camptothecin (CPT) is an effective chemotherapeutic agent for treatment of patients with cancer. The mechanisms underlying CPT-mediated responses in cancer cells are not fully understood. MicroRNA (miRNA) play important roles in tumorigenesis and drug sensitivity. However, the interaction between camptothecin and miRNA has not been previously explored. In this study, we verified that miR-125b was down-regulated in CPT-induced apoptosis in cancer cells and that ectopic expression of miR-125b partially restored cell viability and inhibited cell apoptosis that was induced by CPT. In addition, we demonstrated that CPT induced apoptosis in cancer cells by miR-125b-mediated mitochondrial pathways via targeting to the 3'-untranslated (UTR) regions of Bak1, Mcl1, and p53. A significant increase in Bak1, Mcl1, and p53 protein levels was detected in response to the treatments of CPT. It is noteworthy that the expression levels of Bak1, Mcl1, and p53 increased in a time-dependent manner and negatively correlated with miR-125b expression. It is noteworthy that we revealed that miR-125b directly targeted the 3'UTR regions of multiple genes in a CPT-induced mitochondrial pathway. In addition, most targets of miR-125b were proapoptotic genes, whereas some of the targets were antiapoptotic genes. We hypothesized that miR-125b may mediate the activity of chemotherapeutic agents to induce apoptosis by regulating multiple targets. This is the first report to show that camptothecin induces cancer cell apoptosis via miRNA-mediated mitochondrial pathways. The results suggest that suppression of miR-125b may be a novel approach for the treatment of cancer.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Apoptosis/efectos de los fármacos , Camptotecina/farmacología , MicroARNs/fisiología , Mitocondrias/fisiología , Neoplasias/patología , Células HeLa , Humanos , Proteína 1 de la Secuencia de Leucemia de Células Mieloides , Neoplasias/metabolismo , Reacción en Cadena de la Polimerasa , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Regulación hacia Arriba , Proteína Destructora del Antagonista Homólogo bcl-2/metabolismo
12.
J Biol Chem ; 286(44): 38253-38263, 2011 Nov 04.
Artículo en Inglés | MEDLINE | ID: mdl-21903586

RESUMEN

MicroRNA-125b (miR-125b), a small noncoding RNA molecule, has been found to be deregulated and functions as an oncogene in many cancers including hematopoietic malignancies. However, the mechanisms accounting for miR-125b dysregulation remain to be elucidated. The present study aims to identify the factors that might contribute to up-regulation of miR-125b in human hematopoietic malignancies and its downstream targets for lineage-specific differentiation. We at first reported that CDX2, a homeobox transcription factor, binds to promoter regions of the miR-125b gene and activates transcriptional regulation of miR-125b in malignant myeloid cells. We further revealed that increasing levels of CDX2 in malignant myeloid cells activate miR-125b expression, which in turn inhibits core binding factor ß (CBFß) translation, thereby counteracting myeloid cell differentiation, at least for granulocytic lineage, and promoting leukemogenesis. Interestingly, we found that this novel pathway including CDX2, miR-125b, and CBFß was mediated by undergoing all-trans-retinoic acid induction. Once differentiation ensues with all-trans-retinoic acid treatment, CDX2 activity decreases, leading to a reduction in miR-125b transcription and up-regulation of CBFß in myeloid cells and in patients. The study provides a new mechanism that contributes to hematopoietic malignancies, which could involve deregulation of miR-125b and its up- and downstream factors. As altered expression of miRNAs has been reported in a wide range of malignancies, delineating the underlying molecular mechanisms of aberrant miRNA expression and characterizing the upstream and downstream factors will help to understand important steps in the pathogenesis of these afflictions.


Asunto(s)
Subunidad beta del Factor de Unión al Sitio Principal/metabolismo , Regulación Leucémica de la Expresión Génica , Neoplasias Hematológicas/metabolismo , Proteínas de Homeodominio/metabolismo , MicroARNs/metabolismo , Factor de Transcripción CDX2 , Diferenciación Celular , Línea Celular Tumoral , Células HL-60 , Humanos , Células K562 , Modelos Biológicos , Unión Proteica , ARN Mensajero/metabolismo
13.
Cancers (Basel) ; 13(19)2021 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-34638407

RESUMEN

The implications of intratumor heterogeneity on the four consensus molecular subtypes (CMS) of colorectal cancer (CRC) are not well known. Here, we use single-cell RNA sequencing (scRNASeq) to build an algorithm to assign CMS classification to individual cells, which we use to explore the distributions of CMSs in tumor and non-tumor cells. A dataset of colorectal tumors with bulk RNAseq (n = 3232) was used to identify CMS specific-marker gene sets. These gene sets were then applied to a discovery dataset of scRNASeq profiles (n = 10) to develop an algorithm for single-cell CMS (scCMS) assignment, which recapitulated the intrinsic biology of all four CMSs. The single-cell CMS assignment algorithm was used to explore the scRNASeq profiles of two prospective CRC tumors with mixed CMS via bulk sequencing. We find that every CRC tumor contains individual cells of each scCMS, as well as many individual cells that have enrichment for features of more than one scCMS (called mixed cells). scCMS4 and scCMS1 cells dominate stroma and immune cell clusters, respectively, but account for less than 3% epithelial cells. These data imply that CMS1 and CMS4 are driven by the transcriptomic contribution of immune and stromal cells, respectively, not tumor cells.

14.
Cancer Res ; 79(1): 72-85, 2019 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-30425057

RESUMEN

Aberrant activation of ß-catenin signaling is a critical driver for tumorigenesis, but the mechanism underlying this activation is not completely understood. In this study, we demonstrate a critical role of ß-catenin signaling in stabilization of enhancer of zeste homolog 2 (EZH2) and control of EZH2-mediated gene repression in oncogenesis. ß-Catenin/TCF4 activated the transcription of the deubiquitinase USP1, which then interacted with and deubiquitinated EZH2 directly. USP1-mediated stabilization of EZH2 promoted its recruitment to the promoters of CDKN1B, RUNX3, and HOXA5, resulting in enhanced enrichment of histone H3K27me3 and repression of target gene expression. In human glioma specimens, expression levels of nuclear ß-catenin, USP1, and EZH2 correlated with one another. Depletion of ß-catenin/USP1/EZH2 repressed glioma cell proliferation in vitro and tumor formation in vivo. Our findings indicate that a ß-catenin-USP1-EZH2 axis orchestrates the interplay between dysregulated ß-catenin signaling and EZH2-mediated gene epigenetic silencing during glioma tumorigenesis. SIGNIFICANCE: These findings identify the ß-catenin-USP1-EZH2 signaling axis as a critical mechanism for glioma tumorigenesis that may serve as a new therapeutic target in glioblastoma.


Asunto(s)
Carcinogénesis/patología , Proteína Potenciadora del Homólogo Zeste 2/química , Regulación Neoplásica de la Expresión Génica , Glioma/patología , Proteasas Ubiquitina-Específicas/metabolismo , beta Catenina/metabolismo , Animales , Carcinogénesis/genética , Carcinogénesis/metabolismo , Proliferación Celular , Proteína Potenciadora del Homólogo Zeste 2/genética , Proteína Potenciadora del Homólogo Zeste 2/metabolismo , Epigénesis Genética , Glioma/genética , Glioma/metabolismo , Humanos , Ratones , Ratones Desnudos , Pronóstico , Estabilidad Proteica , Tasa de Supervivencia , Células Tumorales Cultivadas , Proteasas Ubiquitina-Específicas/genética , Ensayos Antitumor por Modelo de Xenoinjerto , beta Catenina/genética
15.
Cancer Cell ; 31(4): 591-606.e6, 2017 04 10.
Artículo en Inglés | MEDLINE | ID: mdl-28344040

RESUMEN

The dynamic and reversible N6-methyladenosine (m6A) RNA modification installed and erased by N6-methyltransferases and demethylases regulates gene expression and cell fate. We show that the m6A demethylase ALKBH5 is highly expressed in glioblastoma stem-like cells (GSCs). Silencing ALKBH5 suppresses the proliferation of patient-derived GSCs. Integrated transcriptome and m6A-seq analyses revealed altered expression of certain ALKBH5 target genes, including the transcription factor FOXM1. ALKBH5 demethylates FOXM1 nascent transcripts, leading to enhanced FOXM1 expression. Furthermore, a long non-coding RNA antisense to FOXM1 (FOXM1-AS) promotes the interaction of ALKBH5 with FOXM1 nascent transcripts. Depleting ALKBH5 and FOXM1-AS disrupted GSC tumorigenesis through the FOXM1 axis. Our work uncovers a critical function for ALKBH5 and provides insight into critical roles of m6A methylation in glioblastoma.


Asunto(s)
Desmetilasa de ARN, Homólogo 5 de AlkB/metabolismo , Neoplasias Encefálicas/patología , Proteína Forkhead Box M1/metabolismo , Glioblastoma/patología , Regiones no Traducidas 3' , Desmetilasa de ARN, Homólogo 5 de AlkB/genética , Animales , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/mortalidad , Línea Celular Tumoral , Proliferación Celular , Proteína 1 Similar a ELAV/genética , Proteína 1 Similar a ELAV/metabolismo , Proteína Forkhead Box M1/genética , Regulación Neoplásica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Glioblastoma/genética , Glioblastoma/mortalidad , Masculino , Ratones Desnudos , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
16.
J Hematol Oncol ; 9: 12, 2016 Feb 18.
Artículo en Inglés | MEDLINE | ID: mdl-26892465

RESUMEN

BACKGROUND: Acute lymphoblastic leukemia (ALL) is an aggressive malignant disorder of lymphoid progenitor cells in both children and adults. Although improvements in contemporary therapy and development of new treatment strategies have led to dramatic increases in the cure rate in children with ALL, the relapse rate remains high and the prognosis of relapsed childhood ALL is poor. Molecularly targeted therapies have emerged as the leading treatments in cancer therapy. Multi-cytotoxic drug regimens have achieved success, yet many studies addressing targeted therapies have focused on only one single agent. In this study, we attempted to investigate whether the effect of the mammalian target of rapamycin (mTOR) inhibitor rapamycin is synergistic with the effect of focal adhesion kinase (FAK) down-regulation in the treatment of ALL. METHODS: The effect of rapamycin combined with FAK down-regulation on cell proliferation, the cell cycle, and apoptosis was investigated in the human precursor B acute lymphoblastic leukemia cells REH and on survival time and leukemia progression in a non-obese diabetic/severe combined immunodeficiency (NOD/SCID) mouse model. RESULTS: When combined with FAK down-regulation, rapamycin-induced suppression of cell proliferation, G0/G1 cell cycle arrest, and apoptosis were significantly enhanced. In addition, REH cell-injected NOD/SCID mice treated with rapamycin and a short-hairpin RNA (shRNA) to down-regulate FAK had significantly longer survival times and slower leukemia progression compared with mice injected with REH-empty vector cells and treated with rapamycin. Moreover, the B-cell CLL/lymphoma-2 (BCL-2) gene family was shown to be involved in the enhancement, by combined treatment, of REH cell apoptosis. CONCLUSIONS: FAK down-regulation enhanced the in vitro and in vivo inhibitory effects of rapamycin on REH cell growth, indicating that the simultaneous targeting of mTOR- and FAK-related pathways might offer a novel and powerful strategy for treating ALL.


Asunto(s)
Quinasa 1 de Adhesión Focal/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/terapia , Interferencia de ARN , Sirolimus/farmacología , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Adolescente , Animales , Apoptosis/efectos de los fármacos , Apoptosis/genética , Western Blotting , Puntos de Control del Ciclo Celular/efectos de los fármacos , Puntos de Control del Ciclo Celular/genética , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Proliferación Celular/genética , Niño , Preescolar , Terapia Combinada , Regulación hacia Abajo/genética , Femenino , Quinasa 1 de Adhesión Focal/metabolismo , Humanos , Masculino , Ratones Endogámicos NOD , Ratones SCID , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Serina-Treonina Quinasas TOR/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
17.
Cancer Res ; 76(14): 4293-304, 2016 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-27246830

RESUMEN

Malignant glioma is an often fatal type of cancer. Aberrant activation of STAT3 leads to glioma tumorigenesis. STAT3-induced transcription of protein-coding genes has been extensively studied; however, little is known about STAT3-regulated miRNA gene transcription in glioma tumorigenesis. In this study, we found that abnormal activation or decreased expression of STAT3 promotes or inhibits the expression of miR-182-5p, respectively. Bioinformatics analyses determined that tumor suppressor protocadherin-8 (PCDH8) is a candidate target gene of miR-182-5p. miR-182-5p negatively regulated PCDH8 expression by directly targeting its 3'-untranslated region. PCDH8 knockdown induced the proliferative and invasive capacities of glioma cells. Silencing of PCDH8 or miR-182-5p mimics could reverse the inhibitory effect of WP1066, a STAT3 inhibitor, or STAT3 knockdown in vitro and in vivo on glioma progression. Clinically, expression levels of PCDH8 were inversely correlated with those of p-STAT3 or miR-182-5p in glioblastoma tissues. These findings reveal that the STAT3/miR-182-5p/PCDH8 axis has a critical role in glioma tumorigenesis and that targeting the axis may provide a new therapeutic approach for human glioma. Cancer Res; 76(14); 4293-304. ©2016 AACR.


Asunto(s)
Neoplasias Encefálicas/etiología , Cadherinas/fisiología , Glioma/etiología , MicroARNs/fisiología , Factor de Transcripción STAT3/fisiología , Animales , Neoplasias Encefálicas/patología , Cadherinas/genética , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Biología Computacional , Glioma/patología , Humanos , Ratones , MicroARNs/análisis , Invasividad Neoplásica , Protocadherinas
18.
Nat Cell Biol ; 18(9): 954-966, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27501329

RESUMEN

Emerging evidence has shown that GSK3ß plays oncogenic roles in multiple tumour types; however, the underlying mechanisms remain largely unknown. Here, we show that nuclear GSK3ß is responsible for the accumulation of the histone demethylase KDM1A and critically regulates histone H3K4 methylation during tumorigenesis. GSK3ß phosphorylates KDM1A Ser683 upon priming phosphorylation of KDM1A Ser687 by CK1α. Phosphorylation of KDM1A induces its binding with and deubiquitylation by USP22, leading to KDM1A stabilization. GSK3ß- and USP22-dependent KDM1A stabilization is required for the demethylation of histone H3K4, thereby repressing BMP2, CDKN1A and GATA6 transcription, which results in cancer stem cell self-renewal and glioblastoma tumorigenesis. In human glioblastoma specimens, KDM1A levels are correlated with nuclear GSK3ß and USP22 levels. Furthermore, a GSK3 inhibitor, tideglusib, sensitizes tumour xenografts to chemotherapy in mice via KDM1A downregulation and improves survival. Our findings demonstrate that nuclear GSK3ß- and USP22-mediated KDM1A stabilization is essential for glioblastoma tumorigenesis.


Asunto(s)
Carcinogénesis/metabolismo , Endopeptidasas/metabolismo , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Histona Demetilasas/metabolismo , Tioléster Hidrolasas/metabolismo , Animales , Carcinogénesis/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Glioblastoma/metabolismo , Histonas/metabolismo , Humanos , Ratones Desnudos , Fosforilación/efectos de los fármacos , Tiadiazoles/farmacología , Ubiquitina Tiolesterasa , Ubiquitinación
19.
Nat Commun ; 6: 6156, 2015 Feb 02.
Artículo en Inglés | MEDLINE | ID: mdl-25639486

RESUMEN

Aberrant activation of ß-catenin in the nucleus has been implicated in a variety of human cancers, but the fate of nuclear ß-catenin is unknown. Here we demonstrate that the tripartite motif-containing protein 33 (TRIM33), acting as an E3 ubiquitin ligase, reduces the abundance of nuclear ß-catenin protein. TRIM33-mediated ß-catenin is destabilized and is GSK-3ß or ß-TrCP independent. TRIM33 interacts with and ubiquitylates nuclear ß-catenin. Moreover, protein kinase Cδ, which directly phosphorylates ß-catenin at Ser715, is required for the TRIM33-ß-catenin interaction. The function of TRIM33 in suppressing tumour cell proliferation and brain tumour development depends on TRIM33-promoted ß-catenin degradation. In human glioblastoma specimens, endogenous TRIM33 levels are inversely correlated with ß-catenin. In summary, our findings identify TRIM33 as a tumour suppressor that can abolish tumour cell proliferation and tumorigenesis by degrading nuclear ß-catenin. This work suggests a new therapeutic strategy against human cancers caused by aberrant activation of ß-catenin.


Asunto(s)
Factores de Transcripción/metabolismo , beta Catenina/metabolismo , Animales , Línea Celular , Línea Celular Tumoral , Proliferación Celular , Humanos , Ratones , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Transcripción/genética , Ubiquitinación , beta Catenina/genética
20.
Autophagy ; 10(10): 1726-37, 2014 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-25126724

RESUMEN

Acute promyelocytic leukemia (APL) is characterized by the t(15;17)-associated PML-RARA fusion gene. We have previously found that MIR125B1 is highly expressed in patients with APL and may be associated with disease pathogenesis; however, the mechanism by which MIR125B1 exerts its oncogenic potential has not been fully elucidated. Here, we demonstrated that MIR125B1 abundance correlates with the PML-RARA status. MIR125B1 overexpression enhanced PML-RARA expression and inhibited the ATRA-induced degradation of the PML-RARA oncoprotein. RNA-seq analysis revealed a direct link between the PML-RARA degradation pathway and MIR125B1-arrested differentiation. We further demonstrated that the MIR125B1-mediated blockade of PML-RARA proteolysis was regulated via an autophagy-lysosomal pathway, contributing to the inhibition of APL differentiation. Furthermore, we identified DRAM2 (DNA-damage regulated autophagy modulator 2), a critical regulator of autophagy, as a novel target that was at least partly responsible for the function of MIR125B1 involved in autophagy. Importantly, the knockdown phenotypes for DRAM2 are similar to the effects of overexpressing MIR125B1 as impairment of PML-RARA degradation, inhibition of autophagy, and myeloid cell differentiation arrest. These effects of MIR125B1 and its target DRAM2 were further confirmed in an APL mouse model. Thus, MIR125B1 dysregulation may interfere with the effectiveness of ATRA-mediated differentiation through an autophagy-dependent pathway, representing a novel potential APL therapeutic target.


Asunto(s)
Autofagia , Leucemia Promielocítica Aguda/genética , Leucemia Promielocítica Aguda/patología , Lisosomas/metabolismo , MicroARNs/metabolismo , Proteínas de Fusión Oncogénica/metabolismo , Proteolisis , Adolescente , Animales , Autofagia/efectos de los fármacos , Autofagia/genética , Secuencia de Bases , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Línea Celular Tumoral , Niño , Preescolar , Modelos Animales de Enfermedad , Femenino , Perfilación de la Expresión Génica , Humanos , Lactante , Recién Nacido , Lisosomas/efectos de los fármacos , Masculino , Proteínas de la Membrana/metabolismo , Ratones , MicroARNs/genética , Datos de Secuencia Molecular , Fagosomas/efectos de los fármacos , Fagosomas/metabolismo , Fagosomas/ultraestructura , Proteolisis/efectos de los fármacos , Tretinoina/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA