Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 55
Filtrar
1.
Proc Natl Acad Sci U S A ; 121(40): e2406837121, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39312663

RESUMEN

Cancers develop resistance to inhibitors of oncogenes mainly due to target-centric mechanisms such as mutations and splicing. While inhibitors or antagonists force targets to unnatural conformation contributing to protein instability and resistance, activating tumor suppressors may maintain the protein in an agonistic conformation to elicit sustainable growth inhibition. Due to the lack of tumor suppressor agonists, this hypothesis and the mechanisms underlying resistance are not understood. In estrogen receptor (ER)-positive breast cancer (BC), androgen receptor (AR) is a druggable tumor suppressor offering a promising avenue for this investigation. Spatial genomics suggests that the molecular portrait of AR-expressing BC cells in tumor microenvironment corresponds to better overall patient survival, clinically confirming AR's role as a tumor suppressor. Ligand activation of AR in ER-positive BC xenografts reprograms cistromes, inhibits oncogenic pathways, and promotes cellular elasticity toward a more differentiated state. Sustained AR activation results in cistrome rearrangement toward transcription factor PROP paired-like homeobox 1, transformation of AR into oncogene, and activation of the Janus kinase/signal transducer (JAK/STAT) pathway, all culminating in lineage plasticity to an aggressive resistant subtype. While the molecular profile of AR agonist-sensitive tumors corresponds to better patient survival, the profile represented in the resistant phenotype corresponds to shorter survival. Inhibition of activated oncogenes in resistant tumors reduces growth and resensitizes them to AR agonists. These findings indicate that persistent activation of a context-dependent tumor suppressor may lead to resistance through lineage plasticity-driven tumor metamorphosis. Our work provides a framework to explore the above phenomenon across multiple cancer types and underscores the importance of factoring sensitization of tumor suppressor targets while developing agonist-like drugs.


Asunto(s)
Neoplasias de la Mama , Receptores Androgénicos , Receptores de Estrógenos , Factores de Transcripción STAT , Humanos , Receptores Androgénicos/metabolismo , Receptores Androgénicos/genética , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Femenino , Factores de Transcripción STAT/metabolismo , Factores de Transcripción STAT/genética , Animales , Receptores de Estrógenos/metabolismo , Receptores de Estrógenos/genética , Oncogenes , Quinasas Janus/metabolismo , Ratones , Transducción de Señal , Línea Celular Tumoral , Microambiente Tumoral , Regulación Neoplásica de la Expresión Génica
2.
Cell Rep ; 42(12): 113461, 2023 12 26.
Artículo en Inglés | MEDLINE | ID: mdl-37979170

RESUMEN

Triple-negative breast cancer (TNBC) is an aggressive subtype with no targeted therapeutics. The luminal androgen receptor (LAR) subtype constitutes 15% of TNBC and is enriched for androgen receptor (AR) and AR target genes. Here, we show that a cohort of TNBC not only expresses AR at a much higher rate (∼80%) but also expresses AR splice variants (AR-SVs) (∼20%), further subclassifying LAR-TNBC. Higher AR and AR-SV expression and corresponding aggressive phenotypes are observed predominantly in specimens obtained from African American women. LAR TNBC specimens are enriched for interferon, Janus kinase (JAK)-signal activator and transducer (STAT), and androgen signaling pathways, which are exclusive to AR-expressing epithelial cancer cells. AR- and AR-SV-expressing TNBC cell proliferation and xenograft and patient-tumor explant growth are inhibited by AR N-terminal domain-binding selective AR degrader or by a JAK inhibitor. Biochemical analysis suggests that STAT1 is an AR coactivator. Collectively, our work identifies pharmacologically targetable TNBC subtypes and identifies growth-promoting interaction between AR and JAK-STAT signaling.


Asunto(s)
Neoplasias de la Mama Triple Negativas , Humanos , Femenino , Neoplasias de la Mama Triple Negativas/metabolismo , Receptores Androgénicos/genética , Receptores Androgénicos/metabolismo , Transducción de Señal/genética , Proliferación Celular/genética , Regulación Neoplásica de la Expresión Génica
3.
Behav Pharmacol ; 32(1): 21-31, 2021 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-33079734

RESUMEN

There are sex differences in the development of cocaine addiction. For example, the time that it takes for women from initial use to addiction is significantly shorter than for men. Thus, understanding why females are more vulnerable to cocaine addiction will provide insights into sex differences in the mechanisms underlying cocaine addiction. This study aimed to determine how cocaine demand intensity and elasticity might differ between sexes. In addition, the impact of estrous cycle and cocaine intake on demand was investigated. Male and female rats were trained to self-administer 0.125 mg of cocaine intravenously under a chained schedule in daily 2-h sessions for 2 weeks, and then, the cocaine demand function was determined with a modified within-session threshold procedure. Following the test, the rats began to self-administer a higher dose of cocaine (0.25 mg) to increase the cocaine intake. The demand function was then similarly determined in the same rats after 2 weeks of cocaine self-administration of the higher dose. No sex differences were found in either demand intensity or elasticity. Neither did the level of cocaine intake have an impact on demand. The demand elasticity, but not intensity, was significantly lower during proestrus/estrus compared with diestrus. These data suggest that the faster transition to cocaine addiction in women cannot be explained by sex differences in the demand for cocaine and such a demand may change during different phases of estrus cycle.


Asunto(s)
Conducta Adictiva/fisiopatología , Trastornos Relacionados con Cocaína/fisiopatología , Cocaína/administración & dosificación , Autoadministración , Animales , Conducta Animal , Relación Dosis-Respuesta a Droga , Economía del Comportamiento , Ciclo Estral/fisiología , Femenino , Masculino , Ratas , Ratas Wistar , Factores Sexuales , Factores de Tiempo
4.
Cancers (Basel) ; 14(1)2021 Dec 22.
Artículo en Inglés | MEDLINE | ID: mdl-35008190

RESUMEN

The oxygen-responsive hypoxia inducible factor (HIF)-1 promotes several steps of the metastatic cascade. A hypoxic gene signature is enriched in triple-negative breast cancers (TNBCs) and is correlated with poor patient survival. Inhibiting the HIF transcription factors with small molecules is challenging; therefore, we sought to identify genes downstream of HIF-1 that could be targeted to block invasion and metastasis. Creatine kinase brain isoform (CKB) was identified as a highly differentially expressed gene in a screen of HIF-1 wild type and knockout mammary tumor cells derived from a transgenic model of metastatic breast cancer. CKB is a cytosolic enzyme that reversibly catalyzes the phosphorylation of creatine, generating phosphocreatine (PCr) in the forward reaction, and regenerating ATP in the reverse reaction. Creatine kinase activity is inhibited by the creatine analog cyclocreatine (cCr). Loss- and gain-of-function genetic approaches were used in combination with cCr therapy to define the contribution of CKB expression or creatine kinase activity to cell proliferation, migration, invasion, and metastasis in ER-negative breast cancers. CKB was necessary for cell invasion in vitro and strongly promoted tumor growth and lung metastasis in vivo. Similarly, cyclocreatine therapy repressed cell migration, cell invasion, the formation of invadopodia and lung metastasis. Moreover, in common TNBC cell line models, the addition of cCr to conventional cytotoxic chemotherapy agents was either additive or synergistic to repress tumor cell growth.

5.
Nat Commun ; 11(1): 3965, 2020 08 07.
Artículo en Inglés | MEDLINE | ID: mdl-32770022

RESUMEN

Dysregulated Wnt/ß-catenin activation plays a critical role in cancer progression, metastasis, and drug resistance. Genotoxic agents such as radiation and chemotherapeutics have been shown to activate the Wnt/ß-catenin signaling although the underlying mechanism remains incompletely understood. Here, we show that genotoxic agent-activated Wnt/ß-catenin signaling is independent of the FZD/LRP heterodimeric receptors and Wnt ligands. OTULIN, a linear linkage-specific deubiquitinase, is essential for the DNA damage-induced ß-catenin activation. OTULIN inhibits linear ubiquitination of ß-catenin, which attenuates its Lys48-linked ubiquitination and proteasomal degradation upon DNA damage. The association with ß-catenin is enhanced by OTULIN Tyr56 phosphorylation, which depends on genotoxic stress-activated ABL1/c-Abl. Inhibiting OTULIN or Wnt/ß-catenin sensitizes triple-negative breast cancer xenograft tumors to chemotherapeutics and reduces metastasis. Increased OTULIN levels are associated with aggressive molecular subtypes and poor survival in breast cancer patients. Thus, OTULIN-mediated Wnt/ß-catenin activation upon genotoxic treatments promotes drug resistance and metastasis in breast cancers.


Asunto(s)
Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Daño del ADN , Resistencia a Antineoplásicos , Endopeptidasas/metabolismo , Proteínas Proto-Oncogénicas c-abl/metabolismo , Vía de Señalización Wnt , Animales , Línea Celular Tumoral , Supervivencia Celular , Femenino , Células HEK293 , Humanos , Ratones Endogámicos NOD , Ratones SCID , Modelos Biológicos , Metástasis de la Neoplasia , Fosforilación , Fosfotirosina/metabolismo , Ubiquitinación , beta Catenina/metabolismo
8.
Cancer Res ; 79(11): 2909-2922, 2019 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-30737234

RESUMEN

Solar ultraviolet radiation (UVR) suppresses skin immunity, which facilitates initiation of skin lesions and establishment of tumors by promoting immune evasion. It is unclear whether immune checkpoints are involved in the modulation of skin immunity by UVR. Here, we report that UVR exposure significantly increased expression of immune checkpoint molecule PD-L1 in melanoma cells. The damage-associated molecular patterns molecule HMGB1 was secreted by melanocytes and keratinocytes upon UVR, which subsequently activated the receptor for advanced glycation endproducts (RAGE) receptor to promote NF-κB- and IRF3-dependent transcription of PD-L1 in melanocytes. UVR exposure significantly reduced the susceptibility of melanoma cells to CD8+ T-cell-dependent cytotoxicity, which was mitigated by inhibiting the HMGB1/TBK1/IRF3/NF-κB cascade or by blocking the PD-1/PD-L1 checkpoint. Taken together, our findings demonstrate that UVR-induced upregulation of PD-L1 contributes to immune suppression in the skin microenvironment, which may promote immune evasion of oncogenic cells and drive melanoma initiation and progression. SIGNIFICANCE: These findings identify PD-L1 as a critical component of UV-induced immune suppression in the skin, which facilitates immunoevasion of oncogenic melanocytes and development of melanoma.See related commentary by Sahu, p. 2805.


Asunto(s)
Proteína HMGB1 , Rayos Ultravioleta , Antígeno B7-H1/genética , FN-kappa B , Receptor para Productos Finales de Glicación Avanzada , Regulación hacia Arriba
10.
Breast Cancer Res ; 20(1): 117, 2018 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-30285805

RESUMEN

BACKGROUND: Metastasis is responsible for a significant number of breast cancer-related deaths. Hypoxia, a primary driving force of cancer metastasis, induces the expression of BHLHE40, a transcription regulator. This study aimed to elucidate the function of BHLHE40 in the metastatic process of breast cancer cells. METHODS: To define the role of BHLHE40 in breast cancer, BHLHE40 expression was knocked down by a lentiviral construct expressing a short hairpin RNA against BHLHE40 or knocked out by the CRISPR/Cas9 editing system. Orthotopic xenograft and experimental metastasis (tail vein injection) mouse models were used to analyze the role of BHLHE40 in lung metastasis of breast cancer. Global gene expression analysis and public database mining were performed to identify signaling pathways regulated by BHLHE40 in breast cancer. The action mechanism of BHLHE40 was examined by chromatin immunoprecipitation (ChIP), co-immunoprecipitation (CoIP), exosome analysis, and cell-based assays for metastatic potential. RESULTS: BHLHE40 knockdown significantly reduced primary tumor growth and lung metastasis in orthotopic xenograft and experimental metastasis models of breast cancer. Gene expression analysis implicated a role of BHLHE40 in transcriptional activation of heparin-binding epidermal growth factor (HBEGF). ChIP and CoIP assays revealed that BHLHE40 induces HBEGF transcription by blocking DNA binding of histone deacetylases (HDAC)1 and HDAC2. Cell-based assays showed that HBEGF is secreted through exosomes and acts to promote cell survival and migration. Public databases provided evidence linking high expression of BHLHE40 and HBEGF to poor prognosis of triple-negative breast cancer. CONCLUSION: This study reveals a novel role of BHLHE40 in promoting tumor cell survival and migration by regulating HBEGF secretion.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Neoplasias de la Mama/genética , Regulación Neoplásica de la Expresión Génica , Factor de Crecimiento Similar a EGF de Unión a Heparina/genética , Proteínas de Homeodominio/genética , Neoplasias Pulmonares/genética , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Neoplasias de la Mama/patología , Neoplasias de la Mama/terapia , Línea Celular Tumoral , Femenino , Factor de Crecimiento Similar a EGF de Unión a Heparina/metabolismo , Proteínas de Homeodominio/metabolismo , Humanos , Estimación de Kaplan-Meier , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/secundario , Células MCF-7 , Ratones Endogámicos NOD , Ratones Noqueados , Ratones SCID , Fenotipo , Interferencia de ARN , Tratamiento con ARN de Interferencia/métodos , Ensayos Antitumor por Modelo de Xenoinjerto/métodos
11.
Stem Cells ; 36(12): 1804-1815, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30171737

RESUMEN

Glioblastoma multiforme (GBM) is a highly aggressive and malignant brain tumor that is refractory to existing therapeutic regimens, which reflects the presence of stem-like cells, termed glioma-initiating cells (GICs). The complex interactions between different signaling pathways and epigenetic regulation of key genes may be critical in the maintaining GICs in their stem-like state. Although several signaling pathways have been identified as being dysregulated in GBM, the prognosis of GBM patients remains miserable despite improvements in targeted therapies. In this report, we identified that BRG1, the catalytic subunit of the SWI/SNF chromatin remodeling complex, plays a fundamental role in maintaining GICs in their stem-like state. In addition, we identified a novel mechanism by which BRG1 regulates glycolysis genes critical for GICs. BRG1 downregulates the expression of TXNIP, a negative regulator of glycolysis. BRG1 knockdown also triggered the STAT3 pathway, which led to TXNIP activation. We further identified that TXNIP is an STAT3-regulated gene. Moreover, BRG1 suppressed the expression of interferon-stimulated genes, which are negatively regulated by STAT3 and regulate tumorigenesis. We further demonstrate that BRG1 plays a critical role in the drug resistance of GICs and in GIC-induced tumorigenesis. By genetic and pharmacological means, we found that inhibiting BRG1 can sensitize GICs to chemotherapeutic drugs, temozolomide and carmustine. Our studies suggest that BRG1 may be a novel therapeutic target in GBM. The identification of the critical role that BRG1 plays in GIC stemness and chemosensitivity will inform the development of better targeted therapies in GBM and possibly other cancers. Stem Cells 2018;36:1806-12.


Asunto(s)
Cromatina/metabolismo , ADN Helicasas/genética , Glioma/metabolismo , Células Madre Neoplásicas/metabolismo , Proteínas Nucleares/genética , Factores de Transcripción/genética , Animales , Glioma/patología , Humanos , Ratones , Células Madre Neoplásicas/patología
12.
Psychopharmacology (Berl) ; 235(8): 2395-2405, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29947917

RESUMEN

RATIONALE: Compulsive cocaine use is a key feature of cocaine addiction and understanding the factors that promote the development of such a behavior will provide important insights into the mechanism of cocaine addiction and is essential for the clinical management of the disorder. OBJECTIVES: This study aimed to determine how the preexisting compulsive reward-seeking behavior is related to the development of compulsive cocaine-seeking behavior in male and female rats and the potential impact of the reward value and estrous cycle on such behaviors. METHODS: Adult male and female Wistar rats were first trained to self-administer sucrose pellets under a chained schedule, and then, the intensity-response effects of footshock punishment on sucrose SA reinforced by different values of sucrose were measured. Subsequently, the same rats went on to self-administer intravenous cocaine and the punishment intensity-response effects on cocaine SA reinforced by different doses of cocaine were similarly determined. For the female rats, the measurements were made during different phases of the estrous cycle. RESULTS: The rats showed a wide range of levels of the compulsive behaviors despite the similar training history. Surprisingly, the compulsive sucrose-seeking behavior did not predict the compulsive cocaine-seeking behavior in either sex. Increasing cocaine dose significantly increased the compulsive cocaine-seeking behavior in the female but not male rats. Estrous cycle did not have impact on the compulsive behaviors. CONCLUSION: Preexisting differences in compulsive sucrose-seeking behavior do not predict compulsive cocaine-seeking behavior. Compulsive cocaine-seeking behavior is influenced by cocaine dose but not estrous cycle in the female rats.


Asunto(s)
Cocaína/administración & dosificación , Conducta Compulsiva/psicología , Castigo/psicología , Recompensa , Sacarosa/administración & dosificación , Animales , Conducta Adictiva/psicología , Trastornos Relacionados con Cocaína/psicología , Relación Dosis-Respuesta a Droga , Ciclo Estral/efectos de los fármacos , Ciclo Estral/fisiología , Femenino , Masculino , Ratas , Ratas Wistar , Refuerzo en Psicología , Autoadministración
13.
Oncotarget ; 9(31): 22095-22112, 2018 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-29774125

RESUMEN

Glioma-Initiating Cells (GICs) are thought to be responsible for tumor initiation, progression and recurrence in glioblastoma (GBM). In previous studies, we reported the constitutive phosphorylation of the STAT3 transcription factor in GICs derived from GBM patient-derived xenografts, and that STAT3 played a critical role in GBM tumorigenesis. In this study, we show that CRISPR/Cas9-mediated deletion of STAT3 in an established GBM cell line markedly inhibited tumorigenesis by intracranial injection but had little effect on cell proliferation in vitro. Tumorigenesis was rescued by the enforced expression of wild-type STAT3 in cells lacking STAT3. In contrast, GICs were highly addicted to STAT3 and upon STAT3 deletion GICs were non-viable. Moreover, we found that STAT3 was constitutively activated in GICs by phosphorylation on both tyrosine (Y705) and serine (S727) residues. Therefore, to study STAT3 function in GICs we established an inducible system to knockdown STAT3 expression (iSTAT3-KD). Using this approach, we demonstrated that Y705-STAT3 phosphorylation was critical and indispensable for GIC-induced tumor formation. Both phosphorylation sites in STAT3 promoted GIC proliferation in vitro. We further showed that S727-STAT3 phosphorylation was Y705-dependent. Targeted microarray and RNA sequencing revealed that STAT3 activated cell-cycle regulator genes, and downregulated genes involved in the interferon response, the hypoxia response, the TGFß pathway, and remodeling of the extracellular matrix. Since STAT3 is an important oncogenic driver of GBM, the identification of these STAT3 regulated pathways in GICs will inform the development of better targeted therapies against STAT3 in GBM and other cancers.

14.
PLoS Pathog ; 13(10): e1006713, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-29084253

RESUMEN

The activation of interferon (IFN)-regulatory factor-3 (IRF3), characterized by phosphorylation and nuclear translocation of the latent transcription factor, is central to initiating innate antiviral responses. Whereas much has been learned about the upstream pathways and signaling mechanisms leading to IRF3 activation, how activated IRF3 operates in the nucleus to control transcription of IFNs remains obscure. Here we identify EAP30 (a.k.a, SNF8/VPS22), an endosomal sorting complex required for transport (ESCRT)-II subunit, as an essential factor controlling IRF3-dependent antiviral defense. Depletion of EAP30, but not other ESCRT-II subunits, compromised IRF3-dependent induction of type I and III IFNs, IFN-stimulated genes (ISGs) and chemokines by double-stranded RNA or viruses. EAP30, however, was dispensable for the induction of inflammatory mediators of strict NF-κB target. Significantly, knockdown of EAP30 also impaired the establishment of an antiviral state against vesicular stomatitis virus and hepatitis C virus, which are of distinct viral families. Mechanistically, EAP30 was not required for IRF3 activation but rather acted at a downstream step. Specifically, a fraction of EAP30 localized within the nucleus, where it formed a complex with IRF3 and its transcriptional co-activator, CREB-binding protein (CBP), in a virus-inducible manner. These interactions promoted IRF3 binding to target gene promoters such as IFN-ß, IFN-λ1 and ISG56. Together, our data describe an unappreciated role for EAP30 in IRF3-dependent innate antiviral response in the nucleus.


Asunto(s)
Complejos de Clasificación Endosomal Requeridos para el Transporte/inmunología , Hepacivirus/inmunología , Hepatitis C/inmunología , Inmunidad Innata , Factor 3 Regulador del Interferón/inmunología , Proteínas Adaptadoras Transductoras de Señales , Animales , Proteína de Unión a CREB/genética , Proteína de Unión a CREB/inmunología , Línea Celular Tumoral , Chlorocebus aethiops , Complejos de Clasificación Endosomal Requeridos para el Transporte/genética , Técnicas de Silenciamiento del Gen , Hepacivirus/genética , Hepatitis C/genética , Humanos , Factor 3 Regulador del Interferón/genética , Interferón beta/genética , Interferón beta/inmunología , Interferones , Interleucinas/genética , Interleucinas/inmunología , Proteínas de Unión al ARN , Factores de Transcripción/genética , Factores de Transcripción/inmunología , Células Vero
15.
Biochem Biophys Res Commun ; 490(3): 739-745, 2017 08 26.
Artículo en Inglés | MEDLINE | ID: mdl-28642132

RESUMEN

Type I interferon (IFNα/ß) induces antiviral and antiproliferative responses in cells through the induction of IFN-stimulated genes (ISGs). Although the roles of IFN-activated STAT1 and STAT2 in the IFN response are well described, the function of STAT3 is poorly characterized. We investigated the role of STAT3 in the biological response to IFNα/ß in mouse embryonic fibroblasts (MEFs) with a germ line deletion of STAT3. These STAT3 knockout (STAT3-KO) MEFs were reconstituted with STAT3 or the F705-STAT3 mutant (unphosphorylated STAT3) where the canonical Y705 tyrosine phosphorylation site was mutated. We show that both STAT3 and unphosphorylated STAT3 expression enhance the sensitivity of MEFs to the antiviral, antiproliferative and gene-inducing actions of IFN. By chromatin immunoprecipitation assays, unphosphorylated STAT3 appears to bind, albeit weakly, to select gene promoters to enhance their expression. These results suggest that unphosphorylated STAT3 plays an important role in the IFN response pathway.


Asunto(s)
Antivirales/farmacología , Proliferación Celular/efectos de los fármacos , Fibroblastos/efectos de los fármacos , Interferón Tipo I/farmacología , Factor de Transcripción STAT3/metabolismo , Activación Transcripcional/efectos de los fármacos , Animales , Línea Celular , Fibroblastos/citología , Fibroblastos/metabolismo , Técnicas de Inactivación de Genes , Ratones , Fosforilación/efectos de los fármacos , Regiones Promotoras Genéticas/efectos de los fármacos , Unión Proteica/efectos de los fármacos , Factor de Transcripción STAT3/genética
16.
Breast Cancer Res ; 18(1): 81, 2016 08 05.
Artículo en Inglés | MEDLINE | ID: mdl-27495308

RESUMEN

BACKGROUND: While aberrant activation of the chromatin-remodeling SWI/SNF complexes has been associated with cancer development and progression, the role of each subunit in tumor cells is poorly defined. This study is aimed to characterize the role of SMARCE1/BAF57 in regulating metastasis of breast cancer cells. METHODS: Genetic approaches and chemical inhibitors were used to manipulate the activities of SMARCE1 and its downstream targets in multiple breast cancer cell lines. Xenograft mouse models were used to analyze the role of SMARCE1 in lung metastasis in vivo. Nonadherent culture conditions were used to elucidate the role of SMARCE1 in regulating anoikis. Chromatin immunoprecipitation (ChIP), immunoprecipitation, and immunoblotting assays were designed to dissect the mechanism of action of SMARCE1. Public databases were used to investigate the relationship between SMARCE1 deregulation and breast cancer prognosis. RESULTS: SMARCE1 knockdown reduced lung metastasis of breast cancer cells and sensitized tumor cells to anoikis. In response to loss of attachment, SMARCE1 interacted with and potentiated transcriptional activity of HIF1A, resulting in rapid PTK2 activation. Both HIF1A and PTK2 were indispensable for SMARCE1-mediated protection against anoikis by promoting activation of ERK and AKT pathways while suppressing the expression of pro-apoptotic BIM protein. Expression data analysis of a large cohort of human breast tumors revealed that high expression of SMARCE1 or PTK2 is associated with poor prognosis and tumor relapse, and PTK2 expression is positively correlated with SMARCE1 expression in basal-like and luminal B subtypes of breast tumors. CONCLUSIONS: SMARCE1 plays an essential role in breast cancer metastasis by protecting cells against anoikis through the HIF1A/PTK2 pathway. SMARCE1-mediated PTK2 activation likely plays a key role in promoting metastasis of basal-like and luminal B subtype of breast tumors.


Asunto(s)
Neoplasias de la Mama/metabolismo , Proteínas Cromosómicas no Histona/fisiología , Proteínas de Unión al ADN/fisiología , Quinasa 1 de Adhesión Focal/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Neoplasias Pulmonares/metabolismo , Animales , Anoicis , Secuencia de Bases , Sitios de Unión , Neoplasias de la Mama/mortalidad , Neoplasias de la Mama/patología , Carcinogénesis/metabolismo , Línea Celular Tumoral , Ensamble y Desensamble de Cromatina , Femenino , Quinasa 1 de Adhesión Focal/genética , Expresión Génica , Regulación Neoplásica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , Estimación de Kaplan-Meier , Neoplasias Pulmonares/mortalidad , Neoplasias Pulmonares/secundario , Ratones Endogámicos NOD , Ratones SCID , Trasplante de Neoplasias , Regiones Promotoras Genéticas , Transducción de Señal , Activación Transcripcional
17.
Mol Cancer ; 15: 26, 2016 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-27001172

RESUMEN

BACKGROUND: Hypoxia-inducible factors (HIFs) are well-established mediators of tumor growth, the epithelial to mesenchymal transition (EMT) and metastasis. In several types of solid tumors, including breast cancers, the HIFs play a critical role in maintaining cancer stem cell (CSC) activity. Thus, we hypothesized that HIFs may also regulate transcription of markers of breast CSC activity. One approach to enrich for breast cells with stem-like phenotypes is FACS sorting, in which sub-populations of live cells are gated based on the expression of cell surface antigens, including various integrin subunits. Integrin alpha 6 (ITGA6; CD49f) is routinely used in combination with other integrin subunits to enrich for breast stem cells by FACS. Integrins not only mediate interactions with the extracellular matrix (ECM), but also drive intracellular signaling events that communicate from the tumor microenvironment to inside of the tumor cell to alter phenotypes including migration and invasion. METHODS: We used two models of metastatic breast cancer (MBC), polyoma middle T (MMTV-PyMT) and MDA-MB-231 cells, to compare the expression of ITGA6 in wild type and knockout (KO) or knockdown cells. Chromatin immunoprecipitation (ChIP) and luciferase reporter assays verified that ITGA6 is a direct HIF transcriptional target. We also used FACS sorting to enrich for CD49f (+) cells to compare tumorsphere formation, tumor initiating cell activity, invasion and HIF activity relative to CD49f(neg or low) cells. Knockdown of ITGA6 significantly reduced invasion, whereas re-expression of ITGA6 in the context of HIF knockdown partially rescued invasion. A search of public databases also revealed that ITGA6 expression is an independent prognostic factor of survival in breast cancer patients. RESULTS: We report that ITGA6 is a HIF-dependent target gene and that high ITGA6 expression enhances invasion and tumor-initiating cell activities in models of MBC. Moreover, cells that express high levels of ITGA6 are enriched for HIF-1α expression and the expression of HIF-dependent target genes. CONCLUSIONS: Our data suggest that HIF-dependent regulation of ITGA6 is one mechanism by which sorting for CD49f (+) cells enhances CSC and metastatic phenotypes in breast cancers. Our results are particularly relevant to basal-like breast cancers which express higher levels of the HIFα subunits, core HIF-dependent target genes and ITGA6 relative to other molecular subtypes.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Integrina alfa6/genética , Modelos Biológicos , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Línea Celular Tumoral , Supervivencia sin Enfermedad , Regulación hacia Abajo/genética , Femenino , Eliminación de Gen , Regulación Neoplásica de la Expresión Génica , Técnicas de Inactivación de Genes , Humanos , Integrina alfa6/metabolismo , Invasividad Neoplásica , Metástasis de la Neoplasia , ARN Mensajero/genética , ARN Mensajero/metabolismo , Esferoides Celulares/metabolismo , Esferoides Celulares/patología , Transcripción Genética
18.
Methods Mol Biol ; 1406: 81-8, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26820947

RESUMEN

MicroRNA (miRNA) analysis has evolved over the past two decades to become a highly specialized field with broad-reaching applications across a multitude of diseases and cellular processes. The choice of an applicable approach for miRNA quantification will depend on a variety of factors such as cost, time constraints, and throughput. Here, we describe the methods of total RNA isolation, AGO2-bound RNA isolation, miRNA polyadenylation, miRNA-cDNA synthesis, and quantitative real-time polymerase chain reaction for the detection of known miRNAs in cultured cells or xenograft tissues of breast cancer.


Asunto(s)
Neoplasias de la Mama/patología , Transformación Celular Neoplásica , MicroARNs/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa/métodos , Animales , Proteínas Argonautas/metabolismo , Células Cultivadas , ADN Complementario/biosíntesis , Humanos , MicroARNs/genética , MicroARNs/aislamiento & purificación , Poliadenilación
19.
Oncogene ; 35(10): 1302-1313, 2016 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-26028030

RESUMEN

Acquired therapeutic resistance is the major drawback to effective systemic therapies for cancers. Aggressive triple-negative breast cancers (TNBC) develop resistance to chemotherapies rapidly, whereas the underlying mechanisms are not completely understood. Here we show that genotoxic treatments significantly increased the expression of miR-181a in TNBC cells, which enhanced TNBC cell survival and metastasis upon Doxorubicin treatment. Consistently, high miR-181a level associated with poor disease free survival and overall survival after treatments in breast cancer patients. The upregulation of miR-181a was orchestrated by transcription factor STAT3 whose activation depended on NF-κB-mediated IL-6 induction in TNBC cells upon genotoxic treatment. Intriguingly, activated STAT3 not only directly bound to MIR181A1 promoter to drive transcription but also facilitated the recruitment of MSK1 to the same region where MSK1 promoted a local active chromatin state by phosphorylating histone H3. We further identified BAX as a direct functional target of miR-181a, whose suppression decreased apoptosis and increased invasion of TNBC cells upon Dox treatment. These results were further confirmed by evidence that suppression of miR-181a significantly enhanced therapeutic response and reduced lung metastasis in a TNBC orthotopic model. Collectively, our data suggested that miR-181a induction had a critical role in promoting therapeutic resistance and aggressive behavior of TNBC cells upon genotoxic treatment. Antagonizing miR-181a may serve as a promising strategy to sensitize TNBC cells to chemotherapy and mitigate metastasis.


Asunto(s)
Resistencia a Antineoplásicos/genética , MicroARNs/genética , Mutágenos/toxicidad , Neoplasias de la Mama Triple Negativas/patología , Animales , Secuencia de Bases , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Daño del ADN , Doxorrubicina/farmacología , Femenino , Humanos , Neoplasias Pulmonares/secundario , Ratones , FN-kappa B/metabolismo , Invasividad Neoplásica , Metástasis de la Neoplasia , Pronóstico , Regiones Promotoras Genéticas/genética , Transporte de Proteínas/efectos de los fármacos , Transporte de Proteínas/genética , Proteínas Quinasas S6 Ribosómicas 90-kDa/metabolismo , Factor de Transcripción STAT3/metabolismo , Neoplasias de la Mama Triple Negativas/diagnóstico , Neoplasias de la Mama Triple Negativas/genética , Proteína X Asociada a bcl-2/genética , Proteína X Asociada a bcl-2/metabolismo
20.
J Clin Med ; 4(12): 2012-27, 2015 Dec 17.
Artículo en Inglés | MEDLINE | ID: mdl-26694476

RESUMEN

MicroRNAs (miRNAs) are a class of 22-25 nucleotide RNAs that control gene expression at the post-transcriptional level. MiRNAs have potential as cancer biomarkers. Melanoma is a highly aggressive form of skin cancer accounting for almost 4% of cancers among men and women, and ~80% of skin cancer-related deaths in the US. In the present study we analyzed plasma-derived exosomal miRNAs from clinically affected and unaffected familial melanoma patients (CDKN2A/p16 gene carriers) and compared them with affected (nonfamilial melanoma) and unaffected control subjects in order to identify novel risk biomarkers for melanoma. Intact miRNAs can be isolated from the circulation because of their presence in exosomes. A number of differentially regulated miRNAs identified by NanoString human V2 miRNA array were validated by quantitative PCR. Significantly, miR-17, miR-19a, miR-21, miR-126, and miR-149 were expressed at higher levels in patients with metastatic sporadic melanoma as compared with familial melanoma patients or unaffected control subjects. Surprisingly, no substantial differences in miRNA expression were detected between familial melanoma patients (all inclusive) and unaffected control subjects. The miRNAs differentially expressed in the different patient cohorts, especially in patients with metastatic melanoma, may play important roles in tumor progression and metastasis, and may be used as predictive biomarkers to monitor remission as well as relapse following therapeutic intervention.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA