Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 82
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 118(43)2021 10 26.
Artículo en Inglés | MEDLINE | ID: mdl-34663724

RESUMEN

Although it is held that proinflammatory changes precede the onset of breast cancer, the underlying mechanisms remain obscure. Here, we demonstrate that FRS2ß, an adaptor protein expressed in a small subset of epithelial cells, triggers the proinflammatory changes that induce stroma in premalignant mammary tissues and is responsible for the disease onset. FRS2ß deficiency in mouse mammary tumor virus (MMTV)-ErbB2 mice markedly attenuated tumorigenesis. Importantly, tumor cells derived from MMTV-ErbB2 mice failed to generate tumors when grafted in the FRS2ß-deficient premalignant tissues. We found that colocalization of FRS2ß and the NEMO subunit of the IκB kinase complex in early endosomes led to activation of nuclear factor-κB (NF-κB), a master regulator of inflammation. Moreover, inhibition of the activities of the NF-κB-induced cytokines, CXC chemokine ligand 12 and insulin-like growth factor 1, abrogated tumorigenesis. Human breast cancer tissues that express higher levels of FRS2ß contain more stroma. The elucidation of the FRS2ß-NF-κB axis uncovers a molecular link between the proinflammatory changes and the disease onset.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Neoplasias de la Mama/etiología , Neoplasias de la Mama/metabolismo , Neoplasias Mamarias Experimentales/etiología , Neoplasias Mamarias Experimentales/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/inmunología , Animales , Neoplasias de la Mama/inmunología , Carcinogénesis , Citocinas/metabolismo , Femenino , Humanos , Inflamación/etiología , Inflamación/metabolismo , Neoplasias Mamarias Experimentales/inmunología , Virus del Tumor Mamario del Ratón , Ratones , Ratones Noqueados , FN-kappa B/metabolismo , Embarazo , Receptor ErbB-2/metabolismo , Infecciones por Retroviridae , Microambiente Tumoral/inmunología , Infecciones Tumorales por Virus
2.
Cancer Sci ; 114(5): 1792-1799, 2023 May.
Artículo en Inglés | MEDLINE | ID: mdl-36704829

RESUMEN

As the incidence of breast cancer continues to increase, it is critical to develop prevention strategies for this disease. Inflammation underlies the onset of the disease, and NF-κB is a master transcription factor for inflammation. Nuclear factor-κB (NF-κB) is activated in a variety of cell types, including normal epithelial cells, cancer cells, cancer-associated fibroblasts (CAFs), and immune cells. Ductal carcinoma in situ (DCIS) is the earliest stage of breast cancer, and not all DCIS lesions develop into invasive breast cancers (IBC). Currently, most patients with DCIS undergo surgery with postoperative therapy, although there is a risk of overtreatment. In BRCA mutants, receptor activator of NF-κB (RANK)-positive progenitors serve as the cell of origin, and treatment using the RANK monoclonal antibody reduces the risk of IBC. There is still an unmet need to diagnose malignant DCIS, which has the potential to progress to IBC, and to establish appropriate prevention strategies. We recently demonstrated novel molecular mechanisms for NF-κB activation in premalignant mammary tissues, which include DCIS, and the resultant cytokine-enriched microenvironment is essential for breast cancer development. On the early endosomes in a few epithelial cells, the adaptor protein FRS2ß, forming a complex with ErbB2, carries the IκB kinase (IKK) complex and leads to the activation of NF-κB, thereby inducing a variety of cytokines. Therefore, the FRS2ß-NFκB axis in the inflammatory premalignant environment could be targetable to prevent IBC. Further analysis of the molecular mechanisms of inflammation in the premalignant microenvironment is necessary to prevent the risk of IBC.


Asunto(s)
Neoplasias de la Mama , Carcinoma Ductal de Mama , Carcinoma Intraductal no Infiltrante , Humanos , Femenino , Neoplasias de la Mama/patología , Carcinoma Intraductal no Infiltrante/patología , FN-kappa B/metabolismo , Citocinas , Quinasa I-kappa B , Inflamación , Carcinoma Ductal de Mama/metabolismo , Microambiente Tumoral
3.
Biochem Biophys Res Commun ; 674: 183-189, 2023 09 24.
Artículo en Inglés | MEDLINE | ID: mdl-37450958

RESUMEN

Mitochondrial one-carbon metabolism is crucial for embryonic development and tumorigenesis, as it supplies one-carbon units necessary for nucleotide synthesis and rapid cell proliferation. However, its contribution to adult tissue homeostasis remains largely unknown. To examine its role in adult tissue homeostasis, we specifically investigated mammary gland development during pregnancy, as it involves heightened cell proliferation. We discovered that MTHFD2, a mitochondrial one-carbon metabolic enzyme, is expressed in both luminal and basal/myoepithelial cell layers, with upregulated expression during pregnancy. Using the mouse mammary tumor virus (MMTV)-Cre recombinase system, we generated mice with a specific mutation of Mthfd2 in mammary epithelial cells. While the mutant mice were capable of properly nurturing their offspring, the pregnancy-induced expansion of mammary glands was significantly delayed. This indicates that MTHFD2 contributes to the rapid development of mammary glands during pregnancy. Our findings shed light on the role of mitochondrial one-carbon metabolism in facilitating rapid cell proliferation, even in the context of the adult tissue homeostasis.


Asunto(s)
Células Epiteliales , Glándulas Mamarias Animales , Metilenotetrahidrofolato Deshidrogenasa (NADP) , Animales , Femenino , Ratones , Embarazo , Proliferación Celular , Células Epiteliales/metabolismo , Hidrolasas/metabolismo , Glándulas Mamarias Animales/metabolismo , Ratones Transgénicos , Metilenotetrahidrofolato Deshidrogenasa (NADP)/metabolismo
4.
Biochem Biophys Res Commun ; 624: 81-88, 2022 10 08.
Artículo en Inglés | MEDLINE | ID: mdl-35940131

RESUMEN

Triple-negative breast cancer (TNBC) is characterized by its high ability of invasiveness and metastasis, namely lacking expression of estrogen receptor (ER), progesterone receptor, and HER2. We previously demonstrated that estrogen responsive finger protein (Efp) plays a tumor-promotive role in ER-positive breast cancer, yet it remains to be addressed whether Efp contributes to TNBC pathophysiology. We here found that Efp mRNA and protein were abundantly expressed in TNBC patient-derived cells and MDA-MB-231 cells. Efp silencing significantly decreased the growth and migration of both TNBC cell models. Cell-cycle profiling showed a decrease in the S phase population upon Efp silencing. Moreover, exogenous Efp expression increased the growth, migration, and the percentages of S phase population of TNBC cells. Transcriptomic analysis in the Efp-silenced TNBC cells identified several candidate Efp targets including cell cycle-related genes CDCA7 and HELLS, whose contribution to cell growth were validated by siRNA-mediated gene silencing. These results suggest that Efp plays a tumor-promotive role in TNBC and can be a potential therapeutic target for the aggressive disease.


Asunto(s)
Neoplasias de la Mama Triple Negativas , Ciclo Celular/genética , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Perfilación de la Expresión Génica , Humanos , Proteínas Nucleares/genética , Factores de Transcripción , Proteínas de Motivos Tripartitos , Neoplasias de la Mama Triple Negativas/patología , Ubiquitina-Proteína Ligasas
5.
Proc Natl Acad Sci U S A ; 116(2): 625-630, 2019 01 08.
Artículo en Inglés | MEDLINE | ID: mdl-30587593

RESUMEN

Cancer stem-like cells (CSCs) are expanded in the CSC niche by increased frequency of symmetric cell divisions at the expense of asymmetric cell divisions. The symmetric division of CSCs is important for the malignant properties of cancer; however, underlying molecular mechanisms remain largely elusive. Here, we show a cytokine, semaphorin 3 (Sema3), produced from the CSC niche, induces symmetric divisions of CSCs to expand the CSC population. Our findings indicate that stimulation with Sema3 induced sphere formation in breast cancer cells through neuropilin 1 (NP1) receptor that was specifically expressed in breast CSCs (BCSCs). Knockdown of MICAL3, a cytoplasmic Sema3 signal transducer, greatly decreased tumor sphere formation and tumor-initiating activity. Mechanistically, Sema3 induced interaction among MICAL3, collapsin response mediator protein 2 (CRMP2), and Numb. It appears that activity of MICAL3 monooxygenase (MO) stimulated by Sema3 is required for tumor sphere formation, interaction between CRMP2 and Numb, and accumulation of Numb protein. We found that knockdown of CRMP2 or Numb significantly decreased tumor sphere formation. Moreover, MICAL3 knockdown significantly decreased Sema3-induced symmetric divisions in NP1/Numb-positive BCSCs and increased asymmetric division that produces NP1/Numb negative cells without stem-like properties. In addition, breast cancer patients with NP1-positive cancer tissues show poor prognosis. Therefore, the niche factor Sema3-stimulated NP1/MICAL3/CRMP2/Numb axis appears to expand CSCs at least partly through increased frequency of MICAL3-mediated symmetric division of CSCs.


Asunto(s)
Neoplasias de la Mama/metabolismo , División Celular , Oxigenasas de Función Mixta/metabolismo , Proteínas de Neoplasias/metabolismo , Células Madre Neoplásicas/metabolismo , Semaforina-3A/metabolismo , Transducción de Señal , Animales , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Técnicas de Silenciamiento del Gen , Humanos , Ratones , Oxigenasas de Función Mixta/genética , Proteínas de Neoplasias/genética , Células Madre Neoplásicas/patología , Semaforina-3A/genética , Esferoides Celulares/metabolismo , Esferoides Celulares/patología
6.
Cancer Sci ; 112(9): 3810-3821, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-34145929

RESUMEN

Epidermal growth factor receptor tyrosine kinase inhibitors (EGFR-TKIs) are effective in patients with non-small-cell lung cancer (NSCLC) harboring EGFR mutations. However, due to acquired resistance to EGFR-TKIs, even patients on third-generation osimertinib have a poor prognosis. Resistance mechanisms are still not fully understood. Here, we demonstrate that the increased expression of MUSASHI-2 (MSI2), an RNA-binding protein, is a novel mechanism for resistance to EGFR-TKIs. We found that after a long-term exposure to gefitinib, the first-generation EGFR-TKI lung cancer cells harboring the EGFR-TKI-sensitive mutations became resistant to both gefitinib and osimertinib. Although other mutations in EGFR were not found, expression levels of Nanog, a stemness core protein, and activities of aldehyde dehydrogenase (ALDH) were increased, suggesting that cancer stem-like properties were increased. Transcriptome analysis revealed that MSI2 was among the stemness-related genes highly upregulated in EGFR-TKI-resistant cells. Knockdown of MSI2 reduced cancer stem-like properties, including the expression levels of Nanog, a core stemness factor. We demonstrated that knockdown of MSI2 restored sensitivity to osimertinib or gefitinib in EGFR-TKI-resistant cells to levels similar to those of parental cells in vitro. An RNA immunoprecipitation (RIP) assay revealed that antibodies against MSI2 were bound to Nanog mRNA, suggesting that MSI2 increases Nanog expression by binding to Nanog mRNA. Moreover, overexpression of MSI2 or Nanog conferred resistance to osimertinib or gefitinib in parental cells. Finally, MSI2 knockdown greatly increased the sensitivity to osimertinib in vivo. Collectively, our findings provide proof of principle that targeting the MSI2-Nanog axis in combination with EGFR-TKIs would effectively prevent the emergence of acquired resistance.


Asunto(s)
Acrilamidas/farmacología , Adenocarcinoma del Pulmón/metabolismo , Compuestos de Anilina/farmacología , Resistencia a Antineoplásicos/efectos de los fármacos , Gefitinib/farmacología , Neoplasias Pulmonares/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Proteínas de Unión al ARN/metabolismo , Regulación hacia Arriba , Células A549 , Acrilamidas/uso terapéutico , Adenocarcinoma del Pulmón/patología , Compuestos de Anilina/uso terapéutico , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Resistencia a Antineoplásicos/genética , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/genética , Gefitinib/uso terapéutico , Perfilación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , Neoplasias Pulmonares/patología , Mutación , Proteína Homeótica Nanog/metabolismo , Inhibidores de Proteínas Quinasas/uso terapéutico , Proteínas de Unión al ARN/genética , Transcriptoma , Transfección
7.
Cancer Sci ; 112(3): 1209-1224, 2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-33340428

RESUMEN

Cancer stem-like cells (CSCs) induce drug resistance and recurrence of tumors when they experience DNA replication stress. However, the mechanisms underlying DNA replication stress in CSCs and its compensation remain unclear. Here, we demonstrate that upregulated c-Myc expression induces stronger DNA replication stress in patient-derived breast CSCs than in differentiated cancer cells. Our results suggest critical roles for mini-chromosome maintenance protein 10 (MCM10), a firing (activating) factor of DNA replication origins, to compensate for DNA replication stress in CSCs. MCM10 expression is upregulated in CSCs and is maintained by c-Myc. c-Myc-dependent collisions between RNA transcription and DNA replication machinery may occur in nuclei, thereby causing DNA replication stress. MCM10 may activate dormant replication origins close to these collisions to ensure the progression of replication. Moreover, patient-derived breast CSCs were found to be dependent on MCM10 for their maintenance, even after enrichment for CSCs that were resistant to paclitaxel, the standard chemotherapeutic agent. Further, MCM10 depletion decreased the growth of cancer cells, but not of normal cells. Therefore, MCM10 may robustly compensate for DNA replication stress and facilitate genome duplication in cancer cells in the S-phase, which is more pronounced in CSCs. Overall, we provide a preclinical rationale to target the c-Myc-MCM10 axis for preventing drug resistance and recurrence of tumors.


Asunto(s)
Neoplasias de la Mama/genética , Proteínas de Mantenimiento de Minicromosoma/metabolismo , Recurrencia Local de Neoplasia/genética , Células Madre Neoplásicas/patología , Proteínas Proto-Oncogénicas c-myc/metabolismo , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Mama/patología , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/patología , Daño del ADN/efectos de los fármacos , Replicación del ADN/efectos de los fármacos , Resistencia a Antineoplásicos/efectos de los fármacos , Resistencia a Antineoplásicos/genética , Femenino , Humanos , Proteínas de Mantenimiento de Minicromosoma/genética , Recurrencia Local de Neoplasia/patología , Recurrencia Local de Neoplasia/prevención & control , Células Madre Neoplásicas/efectos de los fármacos , Cultivo Primario de Células , Proteínas Proto-Oncogénicas c-myc/antagonistas & inhibidores , Esferoides Celulares , Células Tumorales Cultivadas , Regulación hacia Arriba
8.
Biochem Biophys Res Commun ; 584: 7-14, 2021 12 20.
Artículo en Inglés | MEDLINE | ID: mdl-34753066

RESUMEN

Patients with triple-negative breast cancer have a poor prognosis as only a few efficient targeted therapies are available. Cancer cells are characterized by their unregulated proliferation and require large amounts of nucleotides to replicate their DNA. One-carbon metabolism contributes to purine and pyrimidine nucleotide synthesis by supplying one carbon atom. Although mitochondrial one-carbon metabolism has recently been focused on as an important target for cancer treatment, few specific inhibitors have been reported. In this study, we aimed to examine the effects of DS18561882 (DS18), a novel, orally active, specific inhibitor of methylenetetrahydrofolate dehydrogenase (MTHFD2), a mitochondrial enzyme involved in one-carbon metabolism. Treatment with DS18 led to a marked reduction in cancer-cell proliferation; however, it did not induce cell death. Combinatorial treatment with DS18 and inhibitors of checkpoint kinase 1 (Chk1), an activator of the S phase checkpoint pathway, efficiently induced apoptotic cell death in breast cancer cells and suppressed tumorigenesis in a triple-negative breast cancer patient-derived xenograft model. Mechanistically, MTHFD2 inhibition led to cell cycle arrest and slowed nucleotide synthesis. This finding suggests that DNA replication stress occurs due to nucleotide shortage and that the S-phase checkpoint pathway is activated, leading to cell-cycle arrest. Combinatorial treatment with both inhibitors released cell-cycle arrest, but induced accumulation of DNA double-strand breaks, leading to apoptotic cell death. Collectively, a combination of MTHFD2 and Chk1 inhibitors would be a rational treatment option for patients with triple-negative breast cancer.


Asunto(s)
Aminohidrolasas/antagonistas & inhibidores , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1)/antagonistas & inhibidores , Inhibidores Enzimáticos/uso terapéutico , Metilenotetrahidrofolato Deshidrogenasa (NADP)/antagonistas & inhibidores , Enzimas Multifuncionales/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/uso terapéutico , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Administración Oral , Aminohidrolasas/metabolismo , Animales , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1)/metabolismo , Quimioterapia Combinada , Inhibidores Enzimáticos/administración & dosificación , Femenino , Humanos , Metilenotetrahidrofolato Deshidrogenasa (NADP)/metabolismo , Ratones Endogámicos NOD , Ratones Noqueados , Ratones SCID , Enzimas Multifuncionales/metabolismo , Puntos de Control de la Fase S del Ciclo Celular/efectos de los fármacos , Neoplasias de la Mama Triple Negativas/enzimología , Neoplasias de la Mama Triple Negativas/patología , Carga Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto/métodos
9.
Cancer Sci ; 111(12): 4359-4370, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-32976661

RESUMEN

Metastatic progression remains the major cause of death in human breast cancer. Cancer cells with cancer stem cell (CSC) properties drive initiation and growth of metastases at distant sites. We have previously established the breast cancer patient-derived tumor xenograft (PDX) mouse model in which CSC marker CD44+ cancer cells formed spontaneous microscopic metastases in the liver. In this PDX mouse, the expression levels of S100A10 and its family proteins were much higher in the CD44+ cancer cells metastasized to the liver than those at the primary site. Knockdown of S100A10 in breast cancer cells suppressed and overexpression of S100A10 in breast cancer PDX cells enhanced their invasion abilities and 3D organoid formation capacities in vitro. Mechanistically, S100A10 regulated the matrix metalloproteinase activity and the expression levels of stem cell-related genes. Finally, constitutive knockdown of S100A10 significantly reduced their metastatic ability to the liver in vivo. These findings suggest that S100A10 functions as a metastasis promoter of breast CSCs by conferring both invasion ability and CSC properties in breast cancers.


Asunto(s)
Anexina A2/metabolismo , Neoplasias de la Mama/metabolismo , Células Madre Neoplásicas/metabolismo , Proteínas S100/metabolismo , Regulación hacia Arriba , Animales , Anexina A2/genética , Neoplasias de la Mama/patología , Línea Celular Tumoral , Progresión de la Enfermedad , Femenino , Perfilación de la Expresión Génica , Técnicas de Inactivación de Genes , Humanos , Receptores de Hialuranos/metabolismo , Lentivirus/genética , Neoplasias Hepáticas/patología , Neoplasias Hepáticas/secundario , Metaloproteinasas de la Matriz/metabolismo , Ratones , Invasividad Neoplásica , Metástasis de la Neoplasia , Células Madre Neoplásicas/patología , Organoides , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteínas S100/genética
10.
Cancer Sci ; 111(7): 2440-2450, 2020 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-32437068

RESUMEN

Triple-negative breast cancer (TNBC) is an aggressive subtype of breast cancer compared with luminal or epidermal growth factor receptor 2 subtypes, thus effective therapeutic options for TNBC are yet to be developed. Nowadays, oncogenic long noncoding RNAs (lncRNAs) are applied to cancer management as a new class of therapeutic targets. We previously showed that thymopoietin antisense transcript 1 (TMPO-AS1) is a proliferation-associated lncRNA that contributes to hormone-dependent breast cancer progression by stabilizing estrogen receptor-α mRNA. We here showed that TMPO-AS1 is abundantly expressed in basal-like breast cancer subtype based on the transcriptomic data in The Cancer Genome Atlas as well as in TNBC cell lines and patient-derived cells. Small interfering RNA-based loss-of-function analyses showed that TMPO-AS1 knockdown substantially represses the proliferation and migration of TNBC cells. Expression microarray analysis showed that TMPO-AS1 alters gene signatures related to transforming growth factor-ß signaling in addition to proliferative E2F signaling pathways. TMPO-AS1-targeted siRNA treatment through engineered drug delivery systems using cancer-targeted polyion complex micelle or nanoball technology significantly impaired the in vivo growth of primary and metastatic TNBC xenograft tumors. Our findings suggest that TMPO-AS1 plays a key role in TNBC pathophysiology and could be a potential therapeutic target for TNBC.


Asunto(s)
Biomarcadores de Tumor , ARN Largo no Codificante/genética , Neoplasias de la Mama Triple Negativas/genética , Animales , Ciclo Celular , Línea Celular Tumoral , Proliferación Celular/genética , Biología Computacional/métodos , Modelos Animales de Enfermedad , Femenino , Perfilación de la Expresión Génica , Humanos , Ratones , Ratones Noqueados , Terapia Molecular Dirigida , Interferencia de ARN , ARN Interferente Pequeño/genética , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico
11.
PLoS Genet ; 13(10): e1007047, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-29028795

RESUMEN

The sequential use of signaling pathways is essential for the guidance of pluripotent progenitors into diverse cell fates. Here, we show that Shp2 exclusively mediates FGF but not PDGF signaling in the neural crest to control lacrimal gland development. In addition to preventing p53-independent apoptosis and promoting the migration of Sox10-expressing neural crests, Shp2 is also required for expression of the homeodomain transcription factor Alx4, which directly controls Fgf10 expression in the periocular mesenchyme that is necessary for lacrimal gland induction. We show that Alx4 binds an Fgf10 intronic element conserved in terrestrial but not aquatic animals, underlying the evolutionary emergence of the lacrimal gland system in response to an airy environment. Inactivation of ALX4/Alx4 causes lacrimal gland aplasia in both human and mouse. These results reveal a key role of Alx4 in mediating FGF-Shp2-FGF signaling in the neural crest for lacrimal gland development.


Asunto(s)
Factor 10 de Crecimiento de Fibroblastos/genética , Proteínas de Homeodominio/genética , Aparato Lagrimal/crecimiento & desarrollo , Morfogénesis/genética , Cresta Neural/crecimiento & desarrollo , Animales , Apoptosis/genética , Diferenciación Celular/genética , Linaje de la Célula/genética , Regulación del Desarrollo de la Expresión Génica , Humanos , Aparato Lagrimal/metabolismo , Mesodermo/crecimiento & desarrollo , Ratones , Células Madre Pluripotentes/metabolismo , Unión Proteica , Factores de Transcripción SOXE/genética , Transducción de Señal
12.
Genet Mol Biol ; 43(1): e20180269, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31487369

RESUMEN

Triple negative breast cancer (TNBC) is currently the only major breast tumor subtype without effective targeted therapy and, as a consequence, usually presents a poor outcome. Due to its more aggressive phenotype, there is an urgent clinical need to identify novel biomarkers that discriminate individuals with poor prognosis. We hypothesize that miRNAs can be used to this end because they are involved in the initiation and progression of tumors by altering the expression of their target genes. To identify a prognostic biomarker in TNBC, we analyzed the miRNA expression of a cohort composed of 185 patients diagnosed with TNBC using penalized Cox regression models. We identified a four-biomarker signature based on miR-221, miR-1305, miR-4708, and RMDN2 expression levels that allowed for the subdivision of TNBC into high- or low-risk groups (Hazard Ratio - HR = 0.32; 95% Confidence Interval - CI = 0.11-0.91; p = 0.03) and are also statistically associated with survival outcome in subgroups of postmenopausal status (HR = 0.19; 95% CI = 0.04-0.90; p= 0.016), node negative status (HR = 0.12; 95% CI = 0.01-1.04; p = 0.026), and tumors larger than 2cm (HR = 0.21; 95% CI = 0.05-0.81; p = 0.021). This four-biomarker signature was significantly associated with TNBC as an independent prognostic factor for survival.

13.
Cancer Sci ; 108(6): 1210-1222, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28371195

RESUMEN

Epithelial-mesenchymal transition (EMT) and its reverse process, mesenchymal-epithelial transition MET, are crucial in several stages of cancer metastasis. Epithelial-mesenchymal transition allows cancer cells to move to proximal blood vessels for intravasation. However, because EMT and MET processes are dynamic, mesenchymal cancer cells are likely to undergo MET transiently and subsequently re-undergo EMT to restart the metastatic process. Therefore, spatiotemporally coordinated mutual regulation between EMT and MET could occur during metastasis. To elucidate such regulation, we chose HCC38, a human triple-negative breast cancer cell line, because HCC38 is composed of epithelial and mesenchymal populations at a fixed ratio even though mesenchymal cells proliferate significantly more slowly than epithelial cells. We purified epithelial and mesenchymal cells from Venus-labeled and unlabeled HCC38 cells and mixed them at various ratios to follow EMT and MET. Using this system, we found that the efficiency of EMT is approximately an order of magnitude higher than that of MET and that the two populations significantly enhance the transition of cells from the other population to their own. In addition, knockdown of Zinc finger E-box-binding homeobox 1 (ZEB1) or Zinc finger protein SNAI2 (SLUG) significantly suppressed EMT but promoted partial MET, indicating that ZEB1 and SLUG are crucial to EMT and MET. We also show that primary breast cancer cells underwent EMT that correlated with changes in expression profiles of genes determining EMT status and breast cancer subtype. These changes were very similar to those observed in EMT in HCC38 cells. Consequently, we propose HCC38 as a suitable model to analyze EMT-MET dynamics that could affect the development of triple-negative breast cancer.


Asunto(s)
Transición Epitelial-Mesenquimal/genética , Neoplasias de la Mama Triple Negativas/genética , Neoplasias de la Mama Triple Negativas/patología , Línea Celular Tumoral , Proliferación Celular/genética , Femenino , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Células Madre Mesenquimatosas/patología , Homeobox 1 de Unión a la E-Box con Dedos de Zinc/genética
14.
Proc Natl Acad Sci U S A ; 111(43): 15526-31, 2014 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-25313085

RESUMEN

Pyruvate kinase M2 (PKM2) is an alternatively spliced variant of the pyruvate kinase gene that is preferentially expressed during embryonic development and in cancer cells. PKM2 alters the final rate-limiting step of glycolysis, resulting in the cancer-specific Warburg effect (also referred to as aerobic glycolysis). Although previous reports suggest that PKM2 functions in nonmetabolic transcriptional regulation, its significance in cancer biology remains elusive. Here we report that stimulation of epithelial-mesenchymal transition (EMT) results in the nuclear translocation of PKM2 in colon cancer cells, which is pivotal in promoting EMT. Immunoprecipitation and LC-electrospray ionized TOF MS analyses revealed that EMT stimulation causes direct interaction of PKM2 in the nucleus with TGF-ß-induced factor homeobox 2 (TGIF2), a transcriptional cofactor repressor of TGF-ß signaling. The binding of PKM2 with TGIF2 recruits histone deacetylase 3 to the E-cadherin promoter sequence, with subsequent deacetylation of histone H3 and suppression of E-cadherin transcription. This previously unidentified finding of the molecular interaction of PKM2 in the nucleus sheds light on the significance of PKM2 expression in cancer cells.


Asunto(s)
Transición Epitelial-Mesenquimal/genética , Regulación de la Expresión Génica , Piruvato Quinasa/metabolismo , Transcripción Genética , Línea Celular Tumoral , Núcleo Celular/enzimología , Neoplasias Colorrectales/enzimología , Neoplasias Colorrectales/patología , ADN Helicasas/genética , ADN Helicasas/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Histona Desacetilasas/metabolismo , Proteínas de Homeodominio/metabolismo , Humanos , Metástasis de la Neoplasia , Regiones Promotoras Genéticas/genética , Unión Proteica , Transporte de Proteínas , Proteínas Represoras/metabolismo
15.
PLoS Genet ; 10(1): e1004118, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24465223

RESUMEN

Inner ear mechanosensory hair cells transduce sound and balance information. Auditory hair cells emerge from a Sox2-positive sensory patch in the inner ear epithelium, which is progressively restricted during development. This restriction depends on the action of signaling molecules. Fibroblast growth factor (FGF) signalling is important during sensory specification: attenuation of Fgfr1 disrupts cochlear hair cell formation; however, the underlying mechanisms remain unknown. Here we report that in the absence of FGFR1 signaling, the expression of Sox2 within the sensory patch is not maintained. Despite the down-regulation of the prosensory domain markers, p27(Kip1), Hey2, and Hes5, progenitors can still exit the cell cycle to form the zone of non-proliferating cells (ZNPC), however the number of cells that form sensory cells is reduced. Analysis of a mutant Fgfr1 allele, unable to bind to the adaptor protein, Frs2/3, indicates that Sox2 maintenance can be regulated by MAP kinase. We suggest that FGF signaling, through the activation of MAP kinase, is necessary for the maintenance of sensory progenitors and commits precursors to sensory cell differentiation in the mammalian cochlea.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Oído Interno/crecimiento & desarrollo , Células Ciliadas Auditivas Internas/citología , Proteínas de la Membrana/genética , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Ciclo Celular , Diferenciación Celular/genética , Cóclea/crecimiento & desarrollo , Cóclea/metabolismo , Oído Interno/citología , Epitelio/crecimiento & desarrollo , Epitelio/metabolismo , Regulación del Desarrollo de la Expresión Génica , Proteínas de la Membrana/metabolismo , Unión Proteica , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/metabolismo , Factores de Transcripción SOXB1/genética , Transducción de Señal
16.
J Cell Sci ; 127(Pt 3): 571-82, 2014 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-24284065

RESUMEN

Fibroblast growth factor (FGF) signaling requires a plethora of adaptor proteins to elicit downstream responses, but the functional significances of these docking proteins remain controversial. In this study, we used lens development as a model to investigate Frs2α and its structurally related scaffolding proteins, Gab1 and Gab2, in FGF signaling. We show that genetic ablation of Frs2α alone has a modest effect, but additional deletion of tyrosine phosphatase Shp2 causes a complete arrest of lens vesicle development. Biochemical evidence suggests that this Frs2α-Shp2 synergy reflects their epistatic relationship in the FGF signaling cascade, as opposed to compensatory or parallel functions of these two proteins. Genetic interaction experiments further demonstrate that direct binding of Shp2 to Frs2α is necessary for activation of ERK signaling, whereas constitutive activation of either Shp2 or Kras signaling can compensate for the absence of Frs2α in lens development. By contrast, knockout of Gab1 and Gab2 failed to disrupt FGF signaling in vitro and lens development in vivo. These results establish the Frs2α-Shp2 complex as the key mediator of FGF signaling in lens development.


Asunto(s)
Ojo/crecimiento & desarrollo , Factores de Crecimiento de Fibroblastos/metabolismo , Proteínas de la Membrana/metabolismo , Fosfoproteínas/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 11/genética , Proteínas Adaptadoras Transductoras de Señales , Animales , Línea Celular , Quinasas MAP Reguladas por Señal Extracelular , Factores de Crecimiento de Fibroblastos/genética , Regulación del Desarrollo de la Expresión Génica , Cristalino/crecimiento & desarrollo , Cristalino/metabolismo , Proteínas de la Membrana/genética , Ratones , Fosfoproteínas/biosíntesis , Fosforilación , Proteína Tirosina Fosfatasa no Receptora Tipo 11/metabolismo , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Transducción de Señal
17.
Development ; 140(13): 2711-23, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23720040

RESUMEN

FGF signaling plays a pivotal role in eye development. Previous studies using in vitro chick models and systemic zebrafish mutants have suggested that FGF signaling is required for the patterning and specification of the optic vesicle, but due to a lack of genetic models, its role in mammalian retinal development remains elusive. In this study, we show that specific deletion of Fgfr1 and Fgfr2 in the optic vesicle disrupts ERK signaling, which results in optic disc and nerve dysgenesis and, ultimately, ocular coloboma. Defective FGF signaling does not abrogate Shh or BMP signaling, nor does it affect axial patterning of the optic vesicle. Instead, FGF signaling regulates Mitf and Pax2 in coordinating the closure of the optic fissure and optic disc specification, which is necessary for the outgrowth of the optic nerve. Genetic evidence further supports that the formation of an Frs2α-Shp2 complex and its recruitment to FGF receptors are crucial for downstream ERK signaling in this process, whereas constitutively active Ras signaling can rescue ocular coloboma in the FGF signaling mutants. Our results thus reveal a previously unappreciated role of FGF-Frs2α-Shp2-Ras-ERK signaling axis in preventing ocular coloboma. These findings suggest that components of FGF signaling pathway may be novel targets in the diagnosis of and the therapeutic interventions for congenital ocular anomalies.


Asunto(s)
Coloboma/metabolismo , Factores de Crecimiento de Fibroblastos/metabolismo , Nervio Óptico/patología , Transducción de Señal/fisiología , Animales , Coloboma/genética , Factores de Crecimiento de Fibroblastos/genética , Inmunohistoquímica , Hibridación in Situ , Ratones , Ratones Noqueados , Ratones Mutantes , Factor de Transcripción Asociado a Microftalmía/genética , Factor de Transcripción Asociado a Microftalmía/metabolismo , Nervio Óptico/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 11/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 11/metabolismo , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/genética , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/metabolismo , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/genética , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/metabolismo , Transducción de Señal/genética
18.
Nature ; 460(7257): 904-8, 2009 Aug 13.
Artículo en Inglés | MEDLINE | ID: mdl-19620960

RESUMEN

Acquired uniparental disomy (aUPD) is a common feature of cancer genomes, leading to loss of heterozygosity. aUPD is associated not only with loss-of-function mutations of tumour suppressor genes, but also with gain-of-function mutations of proto-oncogenes. Here we show unique gain-of-function mutations of the C-CBL (also known as CBL) tumour suppressor that are tightly associated with aUPD of the 11q arm in myeloid neoplasms showing myeloproliferative features. The C-CBL proto-oncogene, a cellular homologue of v-Cbl, encodes an E3 ubiquitin ligase and negatively regulates signal transduction of tyrosine kinases. Homozygous C-CBL mutations were found in most 11q-aUPD-positive myeloid malignancies. Although the C-CBL mutations were oncogenic in NIH3T3 cells, c-Cbl was shown to functionally and genetically act as a tumour suppressor. C-CBL mutants did not have E3 ubiquitin ligase activity, but inhibited that of wild-type C-CBL and CBL-B (also known as CBLB), leading to prolonged activation of tyrosine kinases after cytokine stimulation. c-Cbl(-/-) haematopoietic stem/progenitor cells (HSPCs) showed enhanced sensitivity to a variety of cytokines compared to c-Cbl(+/+) HSPCs, and transduction of C-CBL mutants into c-Cbl(-/-) HSPCs further augmented their sensitivities to a broader spectrum of cytokines, including stem-cell factor (SCF, also known as KITLG), thrombopoietin (TPO, also known as THPO), IL3 and FLT3 ligand (FLT3LG), indicating the presence of a gain-of-function that could not be attributed to a simple loss-of-function. The gain-of-function effects of C-CBL mutants on cytokine sensitivity of HSPCs largely disappeared in a c-Cbl(+/+) background or by co-transduction of wild-type C-CBL, which suggests the pathogenic importance of loss of wild-type C-CBL alleles found in most cases of C-CBL-mutated myeloid neoplasms. Our findings provide a new insight into a role of gain-of-function mutations of a tumour suppressor associated with aUPD in the pathogenesis of some myeloid cancer subsets.


Asunto(s)
Genes Supresores de Tumor , Leucemia Mieloide/genética , Proteínas Mutantes/metabolismo , Proteínas Proto-Oncogénicas c-cbl/genética , Proteínas Proto-Oncogénicas c-cbl/metabolismo , Desequilibrio Alélico , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Cromosomas Humanos Par 11/genética , Femenino , Humanos , Leucemia Mieloide/metabolismo , Leucemia Mieloide/patología , Masculino , Ratones , Ratones Noqueados , Ratones Desnudos , Modelos Moleculares , Datos de Secuencia Molecular , Proteínas Mutantes/química , Proteínas Mutantes/genética , Mutación , Células 3T3 NIH , Trasplante de Neoplasias , Oncogenes/genética , Fosforilación , Conformación Proteica , Proto-Oncogenes Mas , Proteínas Proto-Oncogénicas c-cbl/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-cbl/química , Proteínas Proto-Oncogénicas c-cbl/deficiencia , Ubiquitinación , Disomía Uniparental/genética , Proteínas ras/genética , Proteínas ras/metabolismo
19.
Proc Natl Acad Sci U S A ; 109(17): 6584-9, 2012 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-22492965

RESUMEN

Breast cancer is one of the most common cancers in humans. However, our understanding of the cellular and molecular mechanisms underlying tumorigenesis in breast tissues is limited. Here, we identified a molecular mechanism that controls the ability of breast cancer cells to form multicellular spheroids (mammospheres). We found that heregulin (HRG), a ligand for ErbB3, induced mammosphere formation of a breast cancer stem cell (BCSC)-enriched population as well as in breast cancer cell lines. HRG-induced mammosphere formation was reduced by treatment with inhibitors for phosphatidyl inositol 3-kinase (PI3K) or NF-κB and by expression of IκBα-Super Repressor (IκBαSR), a dominant-negative inhibitor for NF-κB. Moreover, the overexpression of IκBαSR in breast cancer cells inhibited tumorigenesis in NOD/SCID mice. Furthermore, we found that the expression of IL8, a regulator of self-renewal in BCSC-enriched populations, was induced by HRG through the activation of the PI3K/NF-κB pathway. These findings illustrate that HRG/ErbB3 signaling appears to maintain mammosphere formation through a PI3K/NF-κB pathway in human breast cancer.


Asunto(s)
Neoplasias de la Mama/patología , FN-kappa B/metabolismo , Receptor ErbB-2/metabolismo , Transducción de Señal , Animales , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Femenino , Humanos , Interleucina-8/genética , Ratones , Ratones Endogámicos NOD , Ratones SCID , FN-kappa B/antagonistas & inhibidores , Neurregulina-1/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN Mensajero/genética , Regulación hacia Arriba
20.
Mol Ther ; 21(1): 185-91, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23032975

RESUMEN

Despite the therapeutic potential of nucleic acid drugs, their clinical application has been limited in part by a lack of appropriate delivery systems. Exosomes or microvesicles are small endosomally derived vesicles that are secreted by a variety of cell types and tissues. Here, we show that exosomes can efficiently deliver microRNA (miRNA) to epidermal growth factor receptor (EGFR)-expressing breast cancer cells. Targeting was achieved by engineering the donor cells to express the transmembrane domain of platelet-derived growth factor receptor fused to the GE11 peptide. Intravenously injected exosomes delivered let-7a miRNA to EGFR-expressing xenograft breast cancer tissue in RAG2(-/-) mice. Our results suggest that exosomes can be used therapeutically to target EGFR-expressing cancerous tissues with nucleic acid drugs.


Asunto(s)
Neoplasias de la Mama/terapia , Receptores ErbB/genética , Exosomas/metabolismo , MicroARNs/administración & dosificación , Animales , Secuencia de Bases , Western Blotting , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Técnicas de Cocultivo , Femenino , Humanos , Ratones , Microscopía Inmunoelectrónica , ARN Interferente Pequeño , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA