Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
1.
iScience ; 27(4): 109366, 2024 Apr 19.
Artículo en Inglés | MEDLINE | ID: mdl-38510127

RESUMEN

Triple-negative breast cancer (TNBC) contributes greatly to mortality of breast cancer, demanding new targetable options. We have shown that TNBC patients have high ΔNp63 expression in tumors. However, the function of ΔNp63 in established TNBC is yet to be explored. In current studies, targeting ΔNp63 with inducible CRISPR knockout and Histone deacetylase inhibitor Quisinostat showed that ΔNp63 is important for tumor progression and metastasis in established tumors by promoting myeloid-derived suppressor cell (MDSC) survival through tumor necrosis factor alpha. Decreasing ΔNp63 levels are associated with decreased CD4+ and FOXP3+ T-cells but increased CD8+ T-cells. RNA sequencing analysis indicates that loss of ΔNp63 alters multiple MDSC properties such as lipid metabolism, chemotaxis, migration, and neutrophil degranulation besides survival. We further demonstrated that targeting ΔNp63 sensitizes chemotherapy. Overall, we showed that ΔNp63 reprograms the MDSC-mediated immunosuppressive functions in TNBC, highlighting the benefit of targeting ΔNp63 in chemotherapy-resistant TNBC.

2.
Cancer Res ; 83(13): 2093-2095, 2023 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-37403627

RESUMEN

Cytokines in the tumor microenvironment can affect tumor growth, progression, and response to therapy, making them compelling therapeutic agents and targets. IFNγ is a pleiotropic cytokine predominantly secreted by immune cells that binds to its receptors IFNGR1 and IFNGR2 on target cells. Multiple clinical trials have investigated the efficacy of IFNγ in combination with other therapies for treating patients with cancer and have shown varying results. Here, we summarize the known effects of IFNγ signaling on tumor cells and explore the possibility of its use in clinical settings.


Asunto(s)
Neoplasias , Humanos , Neoplasias/tratamiento farmacológico , Interferón gamma/metabolismo , Proteínas Portadoras , Microambiente Tumoral
3.
Sci Transl Med ; 15(686): eabl4414, 2023 03 08.
Artículo en Inglés | MEDLINE | ID: mdl-36888695

RESUMEN

Natural killer (NK) cells are cytotoxic lymphocytes that accumulate within the tumor microenvironment and are generally considered to be antitumorigenic. Using single-cell RNA sequencing and functional analysis of multiple triple-negative breast cancer (TNBC) and basal tumor samples, we observed a unique subcluster of Socs3highCD11b-CD27- immature NK cells that were present only in TNBC samples. These tumor-infiltrating NK cells expressed a reduced cytotoxic granzyme signature and, in mice, were responsible for activating cancer stem cells through Wnt signaling. NK cell-mediated activation of these cancer stem cells subsequently enhanced tumor progression in mice, whereas depletion of NK cells or Wnt ligand secretion from NK cells by LGK-974 decreased tumor progression. In addition, NK cell depletion or inhibition of their function improved anti-programmed cell death ligand 1 (PD-L1) antibody or chemotherapy response in mice with TNBC. Furthermore, tumor samples from patients with TNBC and non-TNBC revealed that increased numbers of CD56bright NK cells were present in TNBC tumors and were correlated to poor overall survival in patients with TNBC. Together, our findings identify a population of protumorigenic NK cells that may be exploited for both diagnostic and therapeutic strategies to improve outcomes for patients with TNBC.


Asunto(s)
Antineoplásicos , Neoplasias de la Mama Triple Negativas , Humanos , Animales , Ratones , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Antineoplásicos/uso terapéutico , Células Asesinas Naturales , Antígeno B7-H1/metabolismo , Microambiente Tumoral
6.
Methods Mol Biol ; 2429: 485-500, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35507183

RESUMEN

Evidence is emerging that cancer cells are arranged as a hierarchy that spans from stem cells to lineage-restricted progenitor cells. The recent development of spheroid cultures with several tissue type has provided new opportunities to assess cancer stem cell (CSC) activity by allowing them to propagate under conditions that resemble the microenvironment for growth of tumors. One tissue type widely used for stem cell investigations is mammary tissue, and the sphere formation assay has been used in both normal mammary tissue and in breast cancer. Here, we describe detailed experimental methodology for generating and propagating spheres from normal mammary tissue and primary breast tumors of mice, patient derived xenografts (PDXs) and breast cancer cell lines. We further describe how these sphere cultures can be employed for coculture assays to assess the effect of tumor microenvironment (TME) on self-renewal ability of CSCs in breast cancer.


Asunto(s)
Neoplasias de la Mama , Animales , Neoplasias de la Mama/patología , Línea Celular Tumoral , Femenino , Humanos , Ratones , Células Madre Neoplásicas/metabolismo , Microambiente Tumoral
8.
Nat Commun ; 12(1): 432, 2021 01 18.
Artículo en Inglés | MEDLINE | ID: mdl-33462238

RESUMEN

Development of chemoresistance in breast cancer patients greatly increases mortality. Thus, understanding mechanisms underlying breast cancer resistance to chemotherapy is of paramount importance to overcome this clinical challenge. Although activated Notch receptors have been associated with chemoresistance in cancer, the specific Notch ligands and their molecular mechanisms leading to chemoresistance in breast cancer remain elusive. Using conditional knockout and reporter mouse models, we demonstrate that tumor cells expressing the Notch ligand Dll1 is important for tumor growth and metastasis and bear similarities to tumor-initiating cancer cells (TICs) in breast cancer. RNA-seq and ATAC-seq using reporter models and patient data demonstrated that NF-κB activation is downstream of Dll1 and is associated with a chemoresistant phenotype. Finally, pharmacological blocking of Dll1 or NF-κB pathway completely sensitizes Dll1+ tumors to chemotherapy, highlighting therapeutic avenues for chemotherapy resistant breast cancer patients in the near future.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Proteínas de Unión al Calcio/metabolismo , Resistencia a Antineoplásicos/genética , Proteínas de la Membrana/metabolismo , Subunidad p50 de NF-kappa B/metabolismo , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Benzamidas/farmacología , Benzamidas/uso terapéutico , Mama/patología , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Proteínas de Unión al Calcio/antagonistas & inhibidores , Proteínas de Unión al Calcio/genética , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Proliferación Celular/genética , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Conjuntos de Datos como Asunto , Modelos Animales de Enfermedad , Doxorrubicina/farmacología , Doxorrubicina/uso terapéutico , Resistencia a Antineoplásicos/efectos de los fármacos , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Ratones , Ratones Noqueados , Subunidad p50 de NF-kappa B/antagonistas & inhibidores , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/metabolismo , RNA-Seq , Receptores Notch/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética
9.
Cancer Res ; 81(9): 2399-2414, 2021 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-33514514

RESUMEN

Inflammatory breast cancer (IBC) is a highly metastatic breast carcinoma with high frequency of estrogen receptor α (ERα) negativity. Here we explored the role of the second ER subtype, ERß, and report expression in IBC tumors and its correlation with reduced metastasis. Ablation of ERß in IBC cells promoted cell migration and activated gene networks that control actin reorganization, including G-protein-coupled receptors and downstream effectors that activate Rho GTPases. Analysis of preclinical mouse models of IBC revealed decreased metastasis of IBC tumors when ERß was expressed or activated by chemical agonists. Our findings support a tumor-suppressive role of ERß by demonstrating the ability of the receptor to inhibit dissemination of IBC cells and prevent metastasis. On the basis of these findings, we propose ERß as a potentially novel biomarker and therapeutic target that can inhibit IBC metastasis and reduce its associated mortality. SIGNIFICANCE: These findings demonstrate the capacity of ERß to elicit antimetastatic effects in highly aggressive inflammatory breast cancer and propose ERß and the identified associated genes as potential therapeutic targets in this disease.


Asunto(s)
Actinas/metabolismo , Movimiento Celular/genética , Receptor beta de Estrógeno/metabolismo , Neoplasias Inflamatorias de la Mama/metabolismo , Transducción de Señal/genética , Citoesqueleto de Actina/metabolismo , Animales , Estudios de Cohortes , Receptor alfa de Estrógeno/genética , Receptor alfa de Estrógeno/metabolismo , Receptor beta de Estrógeno/genética , Femenino , Técnicas de Inactivación de Genes , Células HEK293 , Humanos , Neoplasias Inflamatorias de la Mama/genética , Neoplasias Inflamatorias de la Mama/patología , Células MCF-7 , Ratones , Metástasis de la Neoplasia/genética , Transfección , Carga Tumoral/genética , Ensayos Antitumor por Modelo de Xenoinjerto
10.
Genome Res ; 31(2): 337-347, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-33361113

RESUMEN

Understanding the changes in diverse molecular pathways underlying the development of breast tumors is critical for improving diagnosis, treatment, and drug development. Here, we used RNA-profiling of canine mammary tumors (CMTs) coupled with a robust analysis framework to model molecular changes in human breast cancer. Our study leveraged a key advantage of the canine model, the frequent presence of multiple naturally occurring tumors at diagnosis, thus providing samples spanning normal tissue and benign and malignant tumors from each patient. We showed human breast cancer signals, at both expression and mutation level, are evident in CMTs. Profiling multiple tumors per patient enabled by the CMT model allowed us to resolve statistically robust transcription patterns and biological pathways specific to malignant tumors versus those arising in benign tumors or shared with normal tissues. We showed that multiple histological samples per patient is necessary to effectively capture these progression-related signatures, and that carcinoma-specific signatures are predictive of survival for human breast cancer patients. To catalyze and support similar analyses and use of the CMT model by other biomedical researchers, we provide FREYA, a robust data processing pipeline and statistical analyses framework.

11.
Nat Cell Biol ; 22(5): 591-602, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32284542

RESUMEN

Triple-negative breast cancer (TNBC) is characterized by a high degree of immune infiltrate in the tumour microenvironment, which may influence the fate of TNBC cells. We reveal that loss of the tumour suppressive transcription factor Elf5 in TNBC cells activates intrinsic interferon-γ (IFN-γ) signalling, promoting tumour progression and metastasis. Mechanistically, we find that loss of the Elf5-regulated ubiquitin ligase FBXW7 ensures stabilization of its putative protein substrate IFN-γ receptor 1 (IFNGR1) at the protein level in TNBC. Elf5low tumours show enhanced IFN-γ signalling accompanied by an increase of immunosuppressive neutrophils within the tumour microenvironment and increased programmed death ligand 1 expression. Inactivation of either programmed death ligand 1 or IFNGR1 elicited a robust anti-tumour and/or anti-metastatic effect. A positive correlation between ELF5 and FBXW7 expression and a negative correlation between ELF5, FBXW7 and IFNGR1 expression in the tumours of patients with TNBC strongly suggest that this signalling axis could be exploited for patient stratification and immunotherapeutic treatment strategies for Elf5low patients with TNBC.


Asunto(s)
Proliferación Celular/fisiología , Proteínas de Unión al ADN/metabolismo , Proteína 7 que Contiene Repeticiones F-Box-WD/metabolismo , Interferón gamma/metabolismo , Metástasis de la Neoplasia/patología , Receptores de Interferón/metabolismo , Factores de Transcripción/metabolismo , Neoplasias de la Mama Triple Negativas/metabolismo , Animales , Línea Celular , Línea Celular Tumoral , Femenino , Células HEK293 , Humanos , Ratones , Ratones Endogámicos BALB C , Transducción de Señal/fisiología , Microambiente Tumoral/fisiología , Receptor de Interferón gamma
12.
Genes Dev ; 34(21-22): 1422-1438, 2020 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-33872192

RESUMEN

Breast cancer is the second leading cause of cancer-related death in women and is a complex disease with high intratumoral and intertumoral heterogeneity. Such heterogeneity is a major driving force behind failure of current therapies and development of resistance. Due to the limitations of conventional therapies and inevitable emergence of acquired drug resistance (chemo and endocrine) as well as radio resistance, it is essential to design novel therapeutic strategies to improve the prognosis for breast cancer patients. Deregulated Notch signaling within the breast tumor and its tumor microenvironment (TME) is linked to poor clinical outcomes in treatment of resistant breast cancer. Notch receptors and ligands are also important for normal mammary development, suggesting the potential for conserved signaling pathways between normal mammary gland development and breast cancer. In this review, we focus on mechanisms by which Notch receptors and ligands contribute to normal mammary gland development and breast tumor progression. We also discuss how complex interactions between cancer cells and the TME may reduce treatment efficacy and ultimately lead to acquired drug or radio resistance. Potential combinatorial approaches aimed at disrupting Notch- and TME-mediated resistance that may aid in achieving in an improved patient prognosis are also highlighted.


Asunto(s)
Antineoplásicos/uso terapéutico , Neoplasias de la Mama/fisiopatología , Neoplasias de la Mama/terapia , Resistencia a Antineoplásicos/genética , Receptores Notch/metabolismo , Transducción de Señal/fisiología , Antineoplásicos/farmacología , Humanos
13.
FEBS Lett ; 594(6): 973-985, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31794060

RESUMEN

The ∆Np63 isoform of the p53-family transcription factor Trp63 is a key regulator of mammary epithelial stem cells that is involved in breast cancer development. To investigate the role of ∆Np63 at different stages of normal mammary gland development, we generated a ∆Np63-inducible conditional knockout (cKO) mouse model. We demonstrate that the deletion of ∆Np63 at puberty results in depletion of mammary stem cell-enriched basal cells, reduces expression of E-cadherin and ß-catenin, and leads to a closed ductal lumen. RNA-sequencing analysis reveals reduced expression of oxidative phosphorylation (OXPHOS)-associated proteins and desmosomal polarity proteins. Functional assays show reduced numbers of mitochondria in the mammary epithelial cells of ΔNp63 cKO compared to wild-type, supporting the reduced OXPHOS phenotype. These findings identify a novel role for ∆Np63 in cellular metabolism and mammary epithelial cell polarity.


Asunto(s)
Polaridad Celular , Glándulas Mamarias Animales/metabolismo , Maduración Sexual , Células Madre/metabolismo , Transactivadores/metabolismo , Animales , Cadherinas/genética , Cadherinas/metabolismo , Femenino , Ratones , Ratones Noqueados , Transactivadores/genética , beta Catenina/genética , beta Catenina/metabolismo
14.
J Clin Med ; 8(5)2019 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-31075939

RESUMEN

Epithelial-to-mesenchymal transition (EMT) is a process through which epithelial cells lose their epithelial characteristics and cell-cell contact, thus increasing their invasive potential. In addition to its well-known roles in embryonic development, wound healing, and regeneration, EMT plays an important role in tumor progression and metastatic invasion. In breast cancer, EMT both increases the migratory capacity and invasive potential of tumor cells, and initiates protumorigenic alterations in the tumor microenvironment (TME). In particular, recent evidence has linked increased expression of EMT markers such as TWIST1 and MMPs in breast tumors with increased immune infiltration in the TME. These immune cells then provide cues that promote immune evasion by tumor cells, which is associated with enhanced tumor progression and metastasis. In the current review, we will summarize the current knowledge of the role of EMT in the biology of different subtypes of breast cancer. We will further explore the correlation between genetic switches leading to EMT and EMT-induced alterations within the TME that drive tumor growth and metastasis, as well as their possible effect on therapeutic response in breast cancer.

15.
FEBS Lett ; 593(10): 1030-1039, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-31002388

RESUMEN

Elf5 is a transcription factor known to regulate critical developmental processes and has been shown to act as a tumour suppressor in multiple cancers. Elf5 knockout mice are embryonically lethal, limiting in vivo studies pertaining to its function. Moreover, haploinsufficiency of Elf5 limits the use of current mouse models to investigate adult tissue distribution of Elf5. Here, we successfully generated Elf5CreERT2-GFP bacterial artificial chromosome (BAC) transgenic mice and show that Elf5+ cells are present in several adult tissues, where its expression was previously not known. Our study demonstrates the unique distribution of Elf5+ cells in multiple adult organs, which will facilitate future studies investigating the function of Elf5 in these tissues during homeostasis, repair and cancer.


Asunto(s)
Linaje de la Célula , Proteínas de Unión al ADN/genética , Ratones Transgénicos , Modelos Animales , Factores de Transcripción/genética , Animales , Cromosomas Artificiales Bacterianos , Integrasas , Ratones , Ratones Noqueados
16.
Oncogene ; 38(12): 2092-2107, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30442981

RESUMEN

Aberrant Notch signaling is implicated in several cancers, including breast cancer. However, the mechanistic details of the specific receptors and function of ligand-mediated Notch signaling that promote breast cancer remains elusive. In our studies we show that DLL1, a Notch signaling ligand, is significantly overexpressed in ERα+ luminal breast cancer. Intriguingly, DLL1 overexpression correlates with poor prognosis in ERα+ luminal breast cancer, but not in other subtypes of breast cancer. In addition, this effect is specific to DLL1, as other Notch ligands (DLL3, JAGGED1, and JAGGED2) do not influence the clinical outcome of ERα+ patients. Genetic studies show that DLL1-mediated Notch signaling in breast cancer is important for tumor cell proliferation, angiogenesis, and cancer stem cell function. Consistent with prognostic clinical data, we found the tumor-promoting function of DLL1 is exclusive to ERα+ luminal breast cancer, as loss of DLL1 inhibits both tumor growth and lung metastasis of luminal breast cancer. Importantly, we find that estrogen signaling stabilizes DLL1 protein by preventing its proteasomal and lysososmal degradations. Moreover, estrogen inhibits ubiquitination of DLL1. Together, our results highlight an unexpected and novel subtype-specific function of DLL1 in promoting luminal breast cancer that is regulated by estrogen signaling. Our studies also emphasize the critical role of assessing subtype-specific mechanisms driving tumor growth and metastasis to generate effective subtype-specific therapeutics.


Asunto(s)
Neoplasias de la Mama/patología , Estrógenos/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Proteínas de la Membrana/metabolismo , Receptores Notch/metabolismo , Transducción de Señal , Animales , Neoplasias de la Mama/irrigación sanguínea , Neoplasias de la Mama/diagnóstico , Neoplasias de la Mama/metabolismo , Proteínas de Unión al Calcio , Línea Celular Tumoral , Proliferación Celular , Transformación Celular Neoplásica , Progresión de la Enfermedad , Receptor alfa de Estrógeno/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Lisosomas/metabolismo , Ratones , Metástasis de la Neoplasia , Células Madre Neoplásicas/patología , Neovascularización Patológica , Pronóstico , Ubiquitinación
17.
J Clin Invest ; 128(11): 5095-5109, 2018 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-30295647

RESUMEN

Triple-negative breast cancer (TNBC) is particularly aggressive, with enhanced incidence of tumor relapse, resistance to chemotherapy, and metastases. As the mechanistic basis for this aggressive phenotype is unclear, treatment options are limited. Here, we showed an increased population of myeloid-derived immunosuppressor cells (MDSCs) in TNBC patients compared with non-TNBC patients. We found that high levels of the transcription factor ΔNp63 correlate with an increased number of MDSCs in basal TNBC patients, and that ΔNp63 promotes tumor growth, progression, and metastasis in human and mouse TNBC cells. Furthermore, we showed that MDSC recruitment to the primary tumor and metastatic sites occurs via direct ΔNp63-dependent activation of the chemokines CXCL2 and CCL22. CXCR2/CCR4 inhibitors reduced MDSC recruitment, angiogenesis, and metastasis, highlighting a novel treatment option for this subset of TNBC patients. Finally, we found that MDSCs secrete prometastatic factors such as MMP9 and chitinase 3-like 1 to promote TNBC cancer stem cell function, thereby identifying a nonimmunologic role for MDSCs in promoting TNBC progression. These findings identify a unique crosstalk between ΔNp63+ TNBC cells and MDSCs that promotes tumor progression and metastasis, which could be exploited in future combined immunotherapy/chemotherapy strategies for TNBC patients.


Asunto(s)
Células Supresoras de Origen Mieloide/inmunología , Células Madre Neoplásicas/inmunología , Factores de Transcripción/inmunología , Neoplasias de la Mama Triple Negativas/inmunología , Proteínas Supresoras de Tumor/inmunología , Animales , Quimiocina CCL22/genética , Quimiocina CCL22/inmunología , Quimiocina CXCL2/genética , Quimiocina CXCL2/inmunología , Proteína 1 Similar a Quitinasa-3/genética , Proteína 1 Similar a Quitinasa-3/inmunología , Femenino , Humanos , Células MCF-7 , Metaloproteinasa 9 de la Matriz/genética , Metaloproteinasa 9 de la Matriz/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Células Supresoras de Origen Mieloide/patología , Metástasis de la Neoplasia , Células Madre Neoplásicas/patología , Receptores CCR4/genética , Receptores CCR4/inmunología , Receptores CXCR4/genética , Receptores CXCR4/inmunología , Factores de Transcripción/genética , Neoplasias de la Mama Triple Negativas/genética , Neoplasias de la Mama Triple Negativas/patología , Neoplasias de la Mama Triple Negativas/terapia , Proteínas Supresoras de Tumor/genética
18.
Science ; 360(6396)2018 06 29.
Artículo en Inglés | MEDLINE | ID: mdl-29773667

RESUMEN

The stem cell niche is a specialized environment that dictates stem cell function during development and homeostasis. We show that Dll1, a Notch pathway ligand, is enriched in mammary gland stem cells (MaSCs) and mediates critical interactions with stromal macrophages in the surrounding niche in mouse models. Conditional deletion of Dll1 reduced the number of MaSCs and impaired ductal morphogenesis in the mammary gland. Moreover, MaSC-expressed Dll1 activates Notch signaling in stromal macrophages, increasing their expression of Wnt family ligands such as Wnt3, Wnt10A, and Wnt16, thereby initiating a feedback loop that promotes the function of Dll1-expressing MaSCs. Together, these findings reveal functionally important cross-talk between MaSCs and their macrophageal niche through Dll1-mediated Notch signaling.


Asunto(s)
Péptidos y Proteínas de Señalización Intercelular/metabolismo , Macrófagos/fisiología , Glándulas Mamarias Animales/citología , Glándulas Mamarias Animales/crecimiento & desarrollo , Receptores Notch/metabolismo , Nicho de Células Madre/fisiología , Células Madre/fisiología , Animales , Proteínas de Unión al Calcio , Recuento de Células , Femenino , Técnicas de Inactivación de Genes , Péptidos y Proteínas de Señalización Intercelular/genética , Ligandos , Macrófagos/citología , Glándulas Mamarias Animales/metabolismo , Ratones , Ratones Noqueados , Morfogénesis , Transducción de Señal , Células Madre/citología , Células del Estroma/citología , Células del Estroma/fisiología , Proteínas Wnt/metabolismo
19.
Biochim Biophys Acta Mol Basis Dis ; 1864(4 Pt A): 1060-1071, 2018 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-29309924

RESUMEN

Metastatic breast cancer is a leading cause of cancer-related deaths in women worldwide. Patients with triple negative breast cancer (TNBCs), a highly aggressive tumor subtype, have a particularly poor prognosis. Multiple reports demonstrate that altered content of the multicopy mitochondrial genome (mtDNA) in primary breast tumors correlates with poor prognosis. We earlier reported that mtDNA copy number reduction in breast cancer cell lines induces an epithelial-mesenchymal transition associated with metastasis. However, it is unknown whether the breast tumor subtypes (TNBC, Luminal and HER2+) differ in the nature and amount of mitochondrial defects and if mitochondrial defects can be used as a marker to identify tumors at risk for metastasis. By analyzing human primary tumors, cell lines and the TCGA dataset, we demonstrate a high degree of variability in mitochondrial defects among the tumor subtypes and TNBCs, in particular, exhibit higher frequency of mitochondrial defects, including reduced mtDNA content, mtDNA sequence imbalance (mtRNR1:ND4), impaired mitochondrial respiration and metabolic switch to glycolysis which is associated with tumorigenicity. We identified that genes involved in maintenance of mitochondrial structural and functional integrity are differentially expressed in TNBCs compared to non-TNBC tumors. Furthermore, we identified a subset of TNBC tumors that contain lower expression of epithelial splicing regulatory protein (ESRP)-1, typical of metastasizing cells. The overall impact of our findings reported here is that mitochondrial heterogeneity among TNBCs can be used to identify TNBC patients at risk of metastasis and the altered metabolism and metabolic genes can be targeted to improve chemotherapeutic response.


Asunto(s)
ADN Mitocondrial , Mitocondrias , Proteínas Mitocondriales , Proteínas de Neoplasias , Proteínas de Unión al ARN , Neoplasias de la Mama Triple Negativas , Línea Celular Tumoral , ADN Mitocondrial/genética , ADN Mitocondrial/metabolismo , Femenino , Humanos , Mitocondrias/genética , Mitocondrias/metabolismo , Mitocondrias/patología , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Neoplasias de la Mama Triple Negativas/genética , Neoplasias de la Mama Triple Negativas/metabolismo , Neoplasias de la Mama Triple Negativas/patología
20.
Nat Cell Biol ; 19(6): 711-723, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28530657

RESUMEN

Tumour-initiating cells, or cancer stem cells (CSCs), possess stem-cell-like properties observed in normal adult tissue stem cells. Normal and cancerous stem cells may therefore share regulatory mechanisms for maintaining self-renewing capacity and resisting differentiation elicited by cell-intrinsic or microenvironmental cues. Here, we show that miR-199a promotes stem cell properties in mammary stem cells and breast CSCs by directly repressing nuclear receptor corepressor LCOR, which primes interferon (IFN) responses. Elevated miR-199a expression in stem-cell-enriched populations protects normal and malignant stem-like cells from differentiation and senescence induced by IFNs that are produced by epithelial and immune cells in the mammary gland. Importantly, the miR-199a-LCOR-IFN axis is activated in poorly differentiated ER- breast tumours, functionally promotes tumour initiation and metastasis, and is associated with poor clinical outcome. Our study therefore reveals a common mechanism shared by normal and malignant stem cells to protect them from suppressive immune cytokine signalling.


Asunto(s)
Neoplasias de la Mama/metabolismo , Proliferación Celular , Transformación Celular Neoplásica/metabolismo , Interferones/metabolismo , Glándulas Mamarias Animales/metabolismo , Glándulas Mamarias Humanas/metabolismo , MicroARNs/metabolismo , Células Madre Neoplásicas/metabolismo , Proteínas Represoras/metabolismo , Factores de Transcripción/metabolismo , Animales , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Diferenciación Celular , Movimiento Celular , Autorrenovación de las Células , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/patología , Femenino , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Células HEK293 , Células HeLa , Humanos , Células MCF-7 , Glándulas Mamarias Animales/patología , Glándulas Mamarias Humanas/patología , Ratones Endogámicos C57BL , Ratones Transgénicos , MicroARNs/genética , Metástasis de la Neoplasia , Células Madre Neoplásicas/patología , Fenotipo , Proteínas Represoras/genética , Transducción de Señal , Factores de Transcripción/genética , Transfección , Microambiente Tumoral
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...