Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Más filtros

País/Región como asunto
Tipo del documento
Intervalo de año de publicación
2.
Nature ; 540(7634): 588-592, 2016 Dec 22.
Artículo en Inglés | MEDLINE | ID: mdl-27974798

RESUMEN

Metastasis is the leading cause of cancer-related deaths; metastatic lesions develop from disseminated cancer cells (DCCs) that can remain dormant. Metastasis-initiating cells are thought to originate from a subpopulation present in progressed, invasive tumours. However, DCCs detected in patients before the manifestation of breast-cancer metastasis contain fewer genetic abnormalities than primary tumours or than DCCs from patients with metastases. These findings, and those in pancreatic cancer and melanoma models, indicate that dissemination might occur during the early stages of tumour evolution. However, the mechanisms that might allow early disseminated cancer cells (eDCCs) to complete all steps of metastasis are unknown. Here we show that, in early lesions in mice and before any apparent primary tumour masses are detected, there is a sub-population of Her2+p-p38lop-Atf2loTwist1hiE-cadlo early cancer cells that is invasive and can spread to target organs. Intra-vital imaging and organoid studies of early lesions showed that Her2+ eDCC precursors invaded locally, intravasated and lodged in target organs. Her2+ eDCCs activated a Wnt-dependent epithelial-mesenchymal transition (EMT)-like dissemination program but without complete loss of the epithelial phenotype, which was reversed by Her2 or Wnt inhibition. Notably, although the majority of eDCCs were Twist1hiE-cadlo and dormant, they eventually initiated metastasis. Our work identifies a mechanism for early dissemination in which Her2 aberrantly activates a program similar to mammary ductal branching that generates eDCCs that are capable of forming metastasis after a dormancy phase.

3.
Proc Natl Acad Sci U S A ; 107(26): 11811-6, 2010 Jun 29.
Artículo en Inglés | MEDLINE | ID: mdl-20547842

RESUMEN

Sin3A/B is a master transcriptional scaffold and corepressor that plays an essential role in the regulation of gene transcription and maintenance of chromatin structure, and its inappropriate recruitment has been associated with aberrant gene silencing in cancer. Sin3A/B are highly related, large, multidomian proteins that interact with a wide variety of transcription factors and corepressor components, and we examined whether disruption of the function of a specific domain could lead to epigenetic reprogramming and derepression of specific subsets of genes. To this end, we selected the Sin3A/B-paired amphipathic alpha-helices (PAH2) domain based on its established role in mediating the effects of a relatively small number of transcription factors containing a PAH2-binding motif known as the Sin3 interaction domain (SID). Here, we show that in both human and mouse breast cancer cells, the targeted disruption of Sin3 function by introduction of a SID decoy that interferes with PAH2 binding to SID-containing partner proteins reverted the silencing of genes involved in cell growth and differentiation. In particular, the SID decoy led to epigenetic reprogramming and reexpression of the important breast cancer-associated silenced genes encoding E-cadherin, estrogen receptor alpha, and retinoic acid receptor beta and impaired tumor growth in vivo. Interestingly, the SID decoy was effective in the triple-negative M.D. Anderson-Metastatic Breast-231 (MDA-MB-231) breast cancer cell line, restoring sensitivity to 17beta-estradiol, tamoxifen, and retinoids. Therefore, the development of small molecules that can block interactions between PAH2 and SID-containing proteins offers a targeted epigenetic approach for treating this type of breast cancer that may also have wider therapeutic implications.


Asunto(s)
Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Neoplasias de la Mama/patología , Neoplasias de la Mama/terapia , Diferenciación Celular/genética , Línea Celular Tumoral , Ensamble y Desensamble de Cromatina , Cartilla de ADN/genética , Resistencia a Antineoplásicos/genética , Epigénesis Genética , Receptor alfa de Estrógeno/genética , Femenino , Marcación de Gen , Terapia Genética , Humanos , Neoplasias Mamarias Experimentales/genética , Neoplasias Mamarias Experimentales/metabolismo , Neoplasias Mamarias Experimentales/patología , Ratones , Unión Proteica , Estructura Terciaria de Proteína , Receptores de Ácido Retinoico/genética , Proteínas Represoras/antagonistas & inhibidores , Proteínas Represoras/química , Complejo Correpresor Histona Desacetilasa y Sin3
4.
Cell Rep ; 42(6): 112560, 2023 06 27.
Artículo en Inglés | MEDLINE | ID: mdl-37267946

RESUMEN

Disseminated cancer cells (DCCs) in secondary organs can remain dormant for years to decades before reactivating into overt metastasis. Microenvironmental signals leading to cancer cell chromatin remodeling and transcriptional reprogramming appear to control onset and escape from dormancy. Here, we reveal that the therapeutic combination of the DNA methylation inhibitor 5-azacytidine (AZA) and the retinoic acid receptor ligands all-trans retinoic acid (atRA) or AM80, an RARα-specific agonist, promotes stable dormancy in cancer cells. Treatment of head and neck squamous cell carcinoma (HNSCC) or breast cancer cells with AZA+atRA induces a SMAD2/3/4-dependent transcriptional program that restores transforming growth factor ß (TGF-ß)-signaling and anti-proliferative function. Significantly, either combination, AZA+atRA or AZA+AM80, strongly suppresses HNSCC lung metastasis formation by inducing and maintaining solitary DCCs in a SMAD4+/NR2F1+ non-proliferative state. Notably, SMAD4 knockdown is sufficient to drive resistance to AZA+atRA-induced dormancy. We conclude that therapeutic doses of AZA and RAR agonists may induce and/or maintain dormancy and significantly limit metastasis development.


Asunto(s)
Neoplasias de la Mama , Transducción de Señal , Proteína Smad4 , Carcinoma de Células Escamosas de Cabeza y Cuello , Tretinoina , Humanos , Azacitidina/farmacología , Neoplasias de Cabeza y Cuello/tratamiento farmacológico , Carcinoma de Células Escamosas de Cabeza y Cuello/tratamiento farmacológico , Factor de Crecimiento Transformador beta/metabolismo , Tretinoina/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Línea Celular Tumoral/efectos de los fármacos , Transducción de Señal/efectos de los fármacos
5.
Clin Cancer Res ; 29(24): 5155-5172, 2023 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-37982738

RESUMEN

PURPOSE: The integrated stress response (ISR) kinase PERK serves as a survival factor for both proliferative and dormant cancer cells. We aim to validate PERK inhibition as a new strategy to specifically eliminate solitary disseminated cancer cells (DCC) in secondary sites that eventually reawake and originate metastasis. EXPERIMENTAL DESIGN: A novel clinical-grade PERK inhibitor (HC4) was tested in mouse syngeneic and PDX models that present quiescent/dormant DCCs or growth-arrested cancer cells in micro-metastatic lesions that upregulate ISR. RESULTS: HC4 significantly blocks metastasis, by killing quiescent/slow-cycling ISRhigh, but not proliferative ISRlow DCCs. HC4 blocked expansion of established micro-metastasis that contained ISRhigh slow-cycling cells. Single-cell gene expression profiling and imaging revealed that a significant proportion of solitary DCCs in lungs were indeed dormant and displayed an unresolved ER stress as revealed by high expression of a PERK-regulated signature. In human breast cancer metastasis biopsies, GADD34 expression (PERK-regulated gene) and quiescence were positively correlated. HC4 effectively eradicated dormant bone marrow DCCs, which usually persist after rounds of therapies. Importantly, treatment with CDK4/6 inhibitors (to force a quiescent state) followed by HC4 further reduced metastatic burden. In HNSCC and HER2+ cancers HC4 caused cell death in dormant DCCs. In HER2+ tumors, PERK inhibition caused killing by reducing HER2 activity because of sub-optimal HER2 trafficking and phosphorylation in response to EGF. CONCLUSIONS: Our data identify PERK as a unique vulnerability in quiescent or slow-cycling ISRhigh DCCs. The use of PERK inhibitors may allow targeting of pre-existing or therapy-induced growth arrested "persister" cells that escape anti-proliferative therapies.


Asunto(s)
Neoplasias de la Mama , Humanos , Animales , Ratones , Femenino , Línea Celular Tumoral , Ciclo Celular , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Proliferación Celular , Muerte Celular , eIF-2 Quinasa/genética
6.
Dev Biol ; 349(2): 125-36, 2011 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-20974122

RESUMEN

We generated a transgenic (Tg)-mouse model expressing a dominant negative-(DN)-RARα, (RARαG303E) under adipocytes-specific promoter to explore the paracrine role of adipocyte retinoic acid receptors (RARs) in mammary morphogenesis. Transgenic adipocytes had reduced level of RARα, ß and γ, which coincided with a severely underdeveloped pubertal and mature ductal tree with profoundly decreased epithelial cell proliferation. Transplantation experiments of mammary epithelium and of whole mammary glands implicated a fat-pad dependent paracrine mechanism in the stunted phenotype of the epithelial ductal tree. Co-cultures of primary adipocytes, or in vitro differentiated adipocyte cell line, with mammary epithelium showed that when activated, adipocyte-RARs contribute to generation of secreted proliferative and pro-migratory factors. Gene expression microarrays revealed a large number of genes regulated by adipocyte-RARs. Among them, pleiotrophin (PTN) was identified as the paracrine effectors of epithelial cell migration. Its expression was found to be strongly inhibited by DN-RARα, an inhibition relieved by pharmacological doses of all-trans retinoic acid (atRA) in culture and in vivo. Moreover, adipocyte-PTHR, another atRA responsive gene, was found to be an up-stream regulator of PTN. Overall, these results support the existence of a novel paracrine loop controlled by adipocyte-RAR that regulates the mammary ductal tree morphogenesis.


Asunto(s)
Adipocitos/metabolismo , Regulación del Desarrollo de la Expresión Génica/fisiología , Glándulas Mamarias Animales/embriología , Morfogénesis/fisiología , Comunicación Paracrina/fisiología , Receptores de Ácido Retinoico/metabolismo , Células 3T3-L1 , Animales , Proteínas Portadoras/metabolismo , Medios de Cultivo Condicionados/química , Citocinas/metabolismo , Cartilla de ADN/genética , Femenino , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Immunoblotting , Inmunohistoquímica , Glándulas Mamarias Animales/trasplante , Ratones , Ratones Transgénicos , Receptor de Hormona Paratiroídea Tipo 1/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Tretinoina/farmacología
7.
Breast Cancer Res ; 14(4): R121, 2012 Aug 24.
Artículo en Inglés | MEDLINE | ID: mdl-22920668

RESUMEN

INTRODUCTION: Retinoic acid signaling plays key roles in embryonic development and in maintaining the differentiated status of adult tissues. Recently, the nuclear retinoic acid receptor (RAR) isotypes α, ß and γ were found to play specific functions in the expansion and differentiation of the stem compartments of various tissues. For instance, RARγ appears to be involved in stem cell compartment expansion, while RARα and RARß are implicated in the subsequent cell differentiation. We found that over-expressing c-Myc in normal mouse mammary epithelium and in a c-Myc-driven transgenic model of mammary cancer, disrupts the balance between RARγ and RARα/ß in favor of RARγ. METHODS: The effects of c-Myc on RAR isotype expression were evaluated in normal mouse mammary epithelium, mammary tumor cells obtained from the MMTV-Myc transgenic mouse model as well as human normal immortalized breast epithelial and breast cancer cell lines. The in vivo effect of the RARα-selective agonist 4-[(5,6,7,8-tetrahydro-5,5,8,8-tetramethyl-2-naphthyl)carboxamido]benzoic acid (Am580) was examined in the MMTV-Myc mouse model of mammary tumorigenesis. RESULTS: Modulation of the RARα/ß to RARγ expression in mammary glands of normal mice, oncomice, and human mammary cell lines through the alteration of RAR-target gene expression affected cell proliferation, survival and tumor growth. Treatment of MMTV-Myc mice with the RARα-selective agonist Am580 led to significant inhibition of mammary tumor growth (~90%, P<0.001), lung metastasis (P<0.01) and extended tumor latency in 63% of mice. Immunocytochemical analysis showed that in these mice, RARα responsive genes such as Cyp26A1, E-cadherin, cellular retinol-binding protein 1 (CRBP1) and p27, were up-regulated. In contrast, the mammary gland tumors of mice that responded poorly to Am580 treatment (37%) expressed significantly higher levels of RARγ. In vitro experiments indicated that the rise in RARγ was functionally linked to promotion of tumor growth and inhibition of differentiation. Thus, activation of the RARα pathway is linked to tumor growth inhibition, differentiation and cell death. CONCLUSIONS: The functional consequence of the interplay between c-Myc oncogene expression and the RARγ to RARα/ß balance suggests that prevalence of RARγ over-RARα/ß expression levels in breast cancer accompanied by c-Myc amplification or over-expression in breast cancer should be predictive of response to treatment with RARα-isotype-specific agonists and warrant monitoring during clinical trials.


Asunto(s)
Benzoatos/farmacología , Neoplasias de la Mama/genética , Transformación Celular Neoplásica/efectos de los fármacos , Transformación Celular Neoplásica/genética , Genes myc , Receptores de Ácido Retinoico/agonistas , Receptores de Ácido Retinoico/genética , Tetrahidronaftalenos/farmacología , Animales , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Proliferación Celular , Supervivencia Celular/genética , Modelos Animales de Enfermedad , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Xenoinjertos , Humanos , Neoplasias Pulmonares/secundario , Ratones , ARN Interferente Pequeño/genética , Receptores de Ácido Retinoico/metabolismo , Receptor alfa de Ácido Retinoico , Proteínas de Unión al Retinol/genética , Transcripción Genética , Receptor de Ácido Retinoico gamma
8.
Transl Oncol ; 16: 101320, 2022 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-34968869

RESUMEN

SIN3A, a scaffold protein has regulatory functions in tumor biology. Through its Paired amphipathic helix (PAH2) domain, SIN3A interacts with PHF12 (PF1), a protein with SIN3 interaction domain (SID) that forms a complex with MRG15 and KDM5A/B. These components are often overexpressed in cancer. In the present study, we evaluated the role of SIN3A and its interacting partner PF1 in mediating inhibition of tumor growth and invasion in triple negative breast cancer (TNBC). We found profound inhibition of invasion, migration, and induction of cellular senescence by specific disruption of the PF1/SIN3A PAH2 domain interaction in TNBC cells expressing PF1-SID transcript or peptide treatment. Genome-wide transcriptomic analysis by RNA-seq revealed that PF1-SID downregulates several gene sets and pathways linked to invasion and migration. Integrin α6 (ITGA6) and integrin ß1 (ITGB1) and their downstream target proteins were downregulated in PF1-SID cells. We further determined increased presence of SIN3A and transcriptional repressor, KLF9, on promoters of ITGA6 and ITGB1 in PF1-SID cells. Knockdown of KLF9 leads to re-expression of ITGA6 and ITGB1 and restoration of the invasive phenotype, functionally linking KLF9 to this process. Overall, these data demonstrate that specific disruption of PF1/SIN3A, inhibits tumor growth, migration, and invasion. Also, PF1-SID not only inhibits tumor growth by senescence induction and reduced proliferation, but it also targets cancer stem cell gene expression and blocks mammosphere formation. Overall, these data demonstrate a mechanism whereby invasion and metastasis of TNBC can be suppressed by inhibiting SIN3A-PF1 interaction and enhancing KLF9 mediated suppression of ITGA6 and ITGB1.

9.
Nat Cancer ; 3(1): 90-107, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-35121989

RESUMEN

Cancer cells disseminate and seed in distant organs, where they can remain dormant for many years before forming clinically detectable metastases. Here we studied how disseminated tumor cells sense and remodel the extracellular matrix (ECM) to sustain dormancy. ECM proteomics revealed that dormant cancer cells assemble a type III collagen-enriched ECM niche. Tumor-derived type III collagen is required to sustain tumor dormancy, as its disruption restores tumor cell proliferation through DDR1-mediated STAT1 signaling. Second-harmonic generation two-photon microscopy further revealed that the dormancy-to-reactivation transition is accompanied by changes in type III collagen architecture and abundance. Analysis of clinical samples revealed that type III collagen levels were increased in tumors from patients with lymph node-negative head and neck squamous cell carcinoma compared to patients who were positive for lymph node colonization. Our data support the idea that the manipulation of these mechanisms could serve as a barrier to metastasis through disseminated tumor cell dormancy induction.


Asunto(s)
Colágeno Tipo III , Neoplasias de Cabeza y Cuello , Proliferación Celular , Matriz Extracelular , Humanos , Carcinoma de Células Escamosas de Cabeza y Cuello
10.
Kidney Int ; 79(6): 624-634, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21150871

RESUMEN

All-trans retinoic acid protects against the development of HIV-associated nephropathy (HIVAN) in HIV-1 transgenic mice (Tg26). In vitro, all-trans retinoic acid inhibits HIV-induced podocyte proliferation and restores podocyte differentiation markers by activating its receptor-α (RARα). Here, we report that Am580, a water-soluble RARα-specific agonist, attenuated proteinuria, glomerosclerosis, and podocyte proliferation, and restored podocyte differentiation markers in kidneys of Tg26 mice. Furthermore, RARα-/- Tg26 mice developed more severe kidney and podocyte injury than did RARα+/- Tg26 mice. Am580 failed to ameliorate kidney injury in RARα-/- Tg26 mice, confirming our hypothesis that Am580 acts through RARα. Although the expression of RARα-target genes was suppressed in the kidneys of Tg26 mice and of patients with HIVAN, the expression of RARα in the kidney was not different between patients with HIVAN and minimal change disease. However, the tissue levels of retinoic acid were reduced in the kidney cortex and isolated glomeruli of Tg26 mice. Consistent with this, the expression of two key enzymes in the retinoic acid synthetic pathway, retinol dehydrogenase type 1 and 9, and the overall enzymatic activity for retinoic acid synthesis were significantly reduced in the glomeruli of Tg26 mice. Thus, a defect in the endogenous synthesis of retinoic acid contributes to loss of the protection by retinoic acid in HIVAN. Hence, RARα agonists may be potential agents for the treatment of HIVAN.


Asunto(s)
Nefropatía Asociada a SIDA/metabolismo , VIH-1/genética , Podocitos/metabolismo , Receptores de Ácido Retinoico/metabolismo , Transducción de Señal , Nefropatía Asociada a SIDA/genética , Nefropatía Asociada a SIDA/patología , Nefropatía Asociada a SIDA/prevención & control , Nefropatía Asociada a SIDA/virología , Oxidorreductasas de Alcohol/metabolismo , Animales , Benzoatos/farmacología , Diferenciación Celular , Proliferación Celular , Modelos Animales de Enfermedad , Femenino , Glomerulonefritis/metabolismo , Glomerulonefritis/prevención & control , Glomerulonefritis/virología , Humanos , Hidroxiesteroide Deshidrogenasas/metabolismo , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos , Podocitos/efectos de los fármacos , Podocitos/patología , Podocitos/virología , Proteinuria/metabolismo , Proteinuria/prevención & control , Proteinuria/virología , Receptores de Ácido Retinoico/agonistas , Receptores de Ácido Retinoico/deficiencia , Receptores de Ácido Retinoico/genética , Receptor alfa de Ácido Retinoico , Retinoides/metabolismo , Índice de Severidad de la Enfermedad , Transducción de Señal/efectos de los fármacos , Tetrahidronaftalenos/farmacología , Factores de Tiempo
11.
Breast Cancer Res ; 12(5): R79, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20923554

RESUMEN

INTRODUCTION: Retinoic acid signaling pathways are disabled in human breast cancer suggesting a controlling role in normal mammary growth that might be lost in tumorigenesis. We tested a single receptor isotype, RARα1, for its role in mouse mammary gland morphogenesis and MMTV-wnt1-induced oncogenesis. METHODS: The role of RARα1 in mammary morphogenesis was tested in RARα1-knockout (KO) mice and in mammary tumorigenesis in bi-genic (RARα1/KO crossed with MMTV-wnt1) mice. We used whole mounts analysis, stem cells/progenitor quantification, mammary gland repopulation, Q-PCR, test of tumor-free survival, tumor fragments and cell transplantation. RESULTS: In 2 genetic backgrounds (129/Bl-6 and FVB) the neo-natal RARα1/KO-mammary epithelial tree was 2-fold larger and the pubertal tree had 2-fold more branch points and 5-fold more mature end buds, a phenotype that was predominantly epithelial cell autonomous. The stem/progenitor compartment of the RARα1/KO mammary, defined as CD24(low)/ALDH(high activity) was increased by a median 1.7 fold, but the mammary stem cell (MaSC)-containing compartment, (CD24(low)/CD29(high)), was larger (~1.5 fold) in the wt-glands, and the mammary repopulating ability of the wt-gland epithelium was ~2-fold greater. In MMTV-wnt1 transgenic glands the progenitor (CD24(low)/ALDH(high activity)) content was 2.6-fold greater than in the wt and was further increased in the RARα1/KO-wnt1 glands. The tumor-free survival of RARα1/KO-wnt1 mice was significantly (p=0.0002, Kaplan Meier) longer, the in vivo growth of RARα1/KO-wnt1 transplanted tumor fragments was significantly (p=0.01) slower and RARα1/KO-wnt1 tumors cell suspension produced tumors after much longer latency. CONCLUSIONS: In vitamin A-replete mice, RARα1 is required to maintain normal mammary morphogenesis, but paradoxically, also efficient tumorigenesis. While its loss increases the density of the mammary epithelial tree and the content of luminal mammary progenitors, it appears to reduce the size of the MaSC-containing compartment, the mammary repopulating activity, and to delay significantly the MMTV-wnt1-mammary tumorigenesis. Whether the delay in tumorigenesis is solely due to a reduction in wnt1 target cells or due to additional mechanisms remains to be determined. These results reveal the intricate nature of the retinoid signaling pathways in mammary development and carcinogenesis and suggest that a better understanding will be needed before retinoids can join the armament of effective anti- breast cancer therapies.


Asunto(s)
Transformación Celular Neoplásica , Glándulas Mamarias Animales/crecimiento & desarrollo , Neoplasias Mamarias Animales/genética , Neoplasias Mamarias Animales/metabolismo , Receptores de Ácido Retinoico/genética , Receptores de Ácido Retinoico/metabolismo , Proteína Wnt1/metabolismo , Familia de Aldehído Deshidrogenasa 1 , Animales , Antígeno CD24/análisis , Femenino , Integrina beta1/análisis , Isoenzimas/metabolismo , Glándulas Mamarias Animales/citología , Glándulas Mamarias Animales/metabolismo , Glándulas Mamarias Animales/trasplante , Virus del Tumor Mamario del Ratón/genética , Virus del Tumor Mamario del Ratón/fisiología , Ratones , Ratones Noqueados , Morfogénesis , Retinal-Deshidrogenasa/metabolismo , Receptor alfa de Ácido Retinoico , Transducción de Señal , Células Madre/citología , Proteína Wnt1/genética
12.
BMC Cell Biol ; 10: 64, 2009 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-19754954

RESUMEN

BACKGROUND: The ErbB2/Her2/Neu receptor tyrosine kinase is amplified in approximately 30% of human breast cancers. Phosphorylation of the translation initiation factor, eIF2alpha inhibits global protein synthesis and activates a stress signaling and growth suppressive program. We have shown that forced phosphorylation of eIF2alpha can suppress head and neck, colorectal carcinoma and multiple myeloma tumor growth and/or survival. Here we explore whether ErbB2 modulates eIF2alpha phosphorylation and whether forced phosphorylation of the latter can antagonize ErbB2 deregulation of mammary acinar morphogenesis. RESULTS: We tested whether ErbB2 signaling influenced eIF2alpha signaling and whether enhanced phosphorylation of the latter affected ErbB2-deregulated mammary acinar development. We obtained stable MCF10A cells overexpressing wild-type (Wt) Neu/ErbB2 or a constitutively active (CA) variant via retroviral delivery or mammary tumor cells from MMTV-Neu tumors. Western blotting, RT-PCR and confocal microscopy were used to analyze the effects of ErbB2 activation on eIF2alpha signaling and the effect of the GADD34-PP1C inhibitor salubrinal. Wt- and MMTV-Neu cells formed aberrant acini structures resembling DCIS, while CA-ErbB2 overexpression induced invasive lesions. In these structures we found that CA-ErbB2 but not the Wt variant significantly down-regulated the pro-apoptotic gene CHOP. This occurred without apparent modulation of basal phosphorylation of PERK and eIF2alpha or induction of its downstream target ATF4. However, inhibition of eIF2alpha dephosphorylation with salubrinal was sufficient to inhibit Wt- and CA-ErbB2- as well as MMTV-Neu-induced deregulation of acinar growth. This was linked to enhanced CHOP expression, inhibition of proliferation, induction of apoptosis and luminal clearing in Wt-ErbB2 and to inhibition of cyclin D1 levels and subsequent proliferation in CA-ErbB2 cells. CONCLUSION: Depending on the strength of ErbB2 signaling there is a differential regulation of CHOP and eIF2alpha phosphorylation. ErbB2 uncouples in basal conditions eIF2alpha phosphorylation from CHOP induction. However, this signal was restored by salubrinal treatment in Wt-ErbB2 expressing MCF10A cells as these DCIS-like structures underwent luminal clearing. In CA-ErbB2 structures apoptosis is not induced by salubrinal and instead a state of quiescence with reduced proliferation was achieved. Treatments that stabilize P-eIF2alpha levels may be effective in treating ErbB2 positive cancers without severely disrupting normal tissue function and structure.


Asunto(s)
Factor 2 Eucariótico de Iniciación/metabolismo , Glándulas Mamarias Animales/metabolismo , Morfogénesis , Receptor ErbB-2/metabolismo , Transducción de Señal , Factor de Transcripción Activador 4/metabolismo , Animales , Apoptosis , Células Cultivadas , Ciclina D1/genética , Factor 2 Eucariótico de Iniciación/genética , Femenino , Humanos , Glándulas Mamarias Animales/citología , Glándulas Mamarias Animales/crecimiento & desarrollo , Ratones , Fosforilación , Receptor ErbB-2/genética , Factor de Transcripción CHOP/metabolismo
13.
Oncotarget ; 8(51): 88421-88436, 2017 Oct 24.
Artículo en Inglés | MEDLINE | ID: mdl-29179446

RESUMEN

Cancer cell invasion is an obligatory step for metastatic dissemination that contributes to rapid relapse and a poorer survival in triple negative breast cancer (TNBC) patients. Development of novel therapeutic strategies to block tumor invasion is an unmet need in the treatment of cancer. We reported that the selective inhibition of the PAH2 domain of SIN3A protein function markedly suppressed metastatic dissemination to the lungs in TNBC xenograft bearing mice. Here, we show that TNBC cell lines treated with Sin3 interaction domain (SID) decoy peptides that bind to PAH2 display a strong in vitro inhibition of transwell invasion. This is accompanied by actin cytoskeleton reorganization with increased cortical actin deposition and downregulation of known Wnt target genes that are associated with epithelial to mesenchymal transition (EMT) and cancer cell invasion. Wnt pathway inhibition by SID decoy peptide was confirmed by decreased Wnt reporter activity and altered cytoplasmic localization of nuclear ß-catenin. TGIF1, a transcription factor that modulates Wnt signaling and known to interact with the PAH2 domain of SIN3A, can be dissociated from the SIN3A complex by SID decoys. TGIF1 knockdown inhibits WNT target genes and in vitro cell invasion suggesting that TGIF1 might be a key target of the SID decoys to block tumor invasion. Taken together, targeting SIN3 function using SID decoys is a novel strategy to reverse invasion and the EMT program in TNBC translating into the inhibition of metastasis dissemination and eradication of residual disease.

14.
Oncogene ; 24(9): 1598-606, 2005 Feb 24.
Artículo en Inglés | MEDLINE | ID: mdl-15608670

RESUMEN

Downregulation of the cellular retinol-binding protein-I (CRBP-I) occurs in breast and other human cancers, but its significance is not well understood. Recently, we showed that restoration of CRBP-I expression in transformed MTSV1-7 breast epithelial cells increased retinoic receptor activity, inhibited anoikis, promoted acinar differentiation and inhibited tumorigenicity, suggesting that CRBP-I suppresses tumor progression. However, the mechanism underlying these effects of CRBP-I was not elucidated. Here we demonstrate, using genetic and pharmacological approaches, that CRBP-I inhibits, in a retinoic acid receptor-dependent manner, the PI3K/Akt survival pathway. Inhibition of PI3K/Akt was necessary and sufficient to explain the antitumor effects of CRBP-I and was mediated by decreased p85 regulatory and p110 catalytic subunit heterodimerization. We present evidence consistent with the idea that this effect is due to CRBP-I inhibition of p85 phosphorylation at Y688. To our knowledge, this is the first demonstration of PI3K regulation at the level of p85-p110 heterodimerization. These findings lead us to hypothesize that CRBP-I downregulation in cancer promotes tumor progression through inhibition of retinoic acid receptor activity and derepression of PI3K/Akt signaling via a novel mechanism.


Asunto(s)
Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Receptores de Ácido Retinoico/fisiología , Proteínas de Unión al Retinol/fisiología , Transducción de Señal/fisiología , Mama , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Diferenciación Celular , Línea Celular , Línea Celular Transformada , Dimerización , Progresión de la Enfermedad , Femenino , Humanos , Proteínas Proto-Oncogénicas c-akt , Proteínas Recombinantes/metabolismo , Proteínas Celulares de Unión al Retinol , Transfección
15.
Mol Cancer ; 5: 12, 2006 Mar 24.
Artículo en Inglés | MEDLINE | ID: mdl-16563162

RESUMEN

BACKGROUND: Retinoic acid suppresses cell growth and promotes cell differentiation, and pharmacological retinoic acid receptor (RAR) activation is anti-tumorigenic. This begs the question of whether chronic physiological RAR activation by endogenous retinoids is likewise anti-tumorigenic. RESULTS: To address this question, we generated transgenic mice in which expression of a ligand binding defective dominant negative RARalpha (RARalphaG303E) was under the control of the mouse mammary tumor virus (MMTV) promoter. The transgene was expressed in the lymphoid compartment and in the mammary epithelium. Observation of aging mice revealed that transgenic mice, unlike their wild type littermates, developed B cell lymphomas at high penetrance, with a median latency of 40 weeks. MMTV-RARalphaG303E lymphomas were high grade Pax-5+, surface H+L Ig negative, CD69+ and BCL6- and cytologically and phenotypically resembled human adult high grade (Burkitt's or lymphoblastic) lymphomas. We postulated that mammary tumors might arise after a long latency period as seen in other transgenic models of breast cancer. We tested this idea by transplanting transgenic epithelium into the cleared fat pads of wild type hosts, thus bypassing lymphomagenesis. At 17 months post-transplantation, a metastatic mammary adenocarcinoma developed in one of four transplanted glands whereas no tumors developed in sixteen of sixteen endogenous glands with wild type epithelium. CONCLUSION: These findings suggest that physiological RAR activity may normally suppress B lymphocyte and mammary epithelial cell growth and that global RAR inactivation is sufficient to initiate a stochastic process of tumor development requiring multiple transforming events. Our work makes available to the research community a new animal resource that should prove useful as an experimental model of aggressive sporadic lymphoma in immunologically uncompromised hosts. We anticipate that it may also prove useful as a model of breast cancer.


Asunto(s)
Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Transformación Celular Neoplásica/genética , Genes Dominantes/genética , Receptores de Ácido Retinoico/genética , Receptores de Ácido Retinoico/metabolismo , Animales , Neoplasias de la Mama/irrigación sanguínea , Neoplasias de la Mama/genética , Transformación Celular Neoplásica/patología , Modelos Animales de Enfermedad , Células Epiteliales/metabolismo , Células Epiteliales/trasplante , Glicina/genética , Glicina/metabolismo , Tejido Linfoide/citología , Tejido Linfoide/metabolismo , Glándulas Mamarias Animales/metabolismo , Glándulas Mamarias Animales/patología , Ratones , Ratones Transgénicos , Trasplante de Neoplasias , Receptor alfa de Ácido Retinoico , Tasa de Supervivencia
16.
Int J Cardiol ; 108(2): 181-8, 2006 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-15922464

RESUMEN

BACKGROUND: Tissue Doppler imaging (TDI) is useful in the evaluation of systolic and diastolic function. It allows assessment of ventricular dynamics in its longitudinal axis. We sought to investigate the difference in systolic and diastolic longitudinal function in patients with chronic heart failure (CHF) with normal and reduced ejection fraction. METHODS AND RESULTS: One hundred ten outpatients with CHF and 68 controls were included. Ejection fraction (EF) was obtained and longitudinal systolic (S) and diastolic (E' and A') wall velocities were recorded from basal septum. Group A (controls) were normal and CHF patients were classified by EF in Group B1: > 45% and B2: < or = 45%. In A, B1 and B2 the mean S peak was 7.74; 5.45 and 4.89 cm/s (p<0.001); the mean E' peak was 8.56; 5.72 and 6.1 cm/s (p<0.001); and the mean A' peak was 10.2; 7.3 and 5.3 cm/s (p<0.001). Also, isovolumic contraction and relaxation time were different among control and CHF groups, (both p<0.001). The most useful parameters for identifying diastolic CHF were IVRT and S peak, with area under ROC curves of 0.93 and 0.89. The cut-off of 115 ms for IVRT and 5.8 cm/s for S peak showed a sensitivity of 94 and 97%, with a specificity of 82 and 73%, respectively. CONCLUSION: These findings suggest that impairment of left ventricular systolic function is present even in those with diastolic heart failure, and that abnormalities may have an important role to identifying the condition.


Asunto(s)
Ecocardiografía Doppler , Insuficiencia Cardíaca/diagnóstico por imagen , Insuficiencia Cardíaca/fisiopatología , Volumen Sistólico , Anciano , Diástole , Femenino , Humanos , Masculino , Persona de Mediana Edad , Estudios Prospectivos , Curva ROC , Sístole , Disfunción Ventricular Izquierda/diagnóstico por imagen , Disfunción Ventricular Izquierda/fisiopatología
17.
Cell Rep ; 16(2): 472-486, 2016 07 12.
Artículo en Inglés | MEDLINE | ID: mdl-27346354

RESUMEN

Chromatin-mediated processes influence the development and progression of breast cancer. Using murine mammary carcinoma-derived tumorspheres as a functional readout for an aggressive breast cancer phenotype, we performed a loss-of-function screen targeting 60 epigenetic regulators. We identified the Polycomb protein Cbx8 as a key regulator of mammary carcinoma both in vitro and in vivo. Accordingly, Cbx8 is overexpressed in human breast cancer and correlates with poor survival. Our genomic analyses revealed that Cbx8 positively regulates Notch signaling by maintaining H3K4me3 levels on Notch-network gene promoters. Ectopic expression of Notch1 partially rescues tumorsphere formation in Cbx8-depleted cells. We find that Cbx8 associates with non-PRC1 complexes containing the H3K4 methyltransferase complex component WDR5, which together regulate Notch gene expression. Thus, our study implicates a key non-canonical role for Cbx8 in promoting breast tumorigenesis.


Asunto(s)
Neoplasias Mamarias Animales/metabolismo , Proteínas de Transporte de Membrana Mitocondrial/fisiología , Proteínas del Grupo Polycomb/fisiología , Proteínas/fisiología , Animales , Carcinogénesis/metabolismo , Línea Celular Tumoral , Epigénesis Genética , Células Epiteliales/metabolismo , Femenino , Expresión Génica , Regulación Neoplásica de la Expresión Génica , Sitios Genéticos , Histonas/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intracelular , Neoplasias Mamarias Animales/genética , Neoplasias Mamarias Animales/patología , Ratones Transgénicos , Células Madre Neoplásicas/metabolismo , Complejo Represivo Polycomb 1 , Procesamiento Proteico-Postraduccional , Receptores Notch/genética , Receptores Notch/metabolismo , Transducción de Señal , Esferoides Celulares/metabolismo , Carga Tumoral
18.
Am Heart J ; 149(3): 451-7, 2005 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15864233

RESUMEN

BACKGROUND: C-reactive protein (CRP) levels are associated with cardiovascular risk. We assessed the hypothesis that atorvastatin might have anti-inflammatory effects in acute coronary syndromes (ACS) as shown by CRP reduction. METHODS: This study was a prospective, randomized, double-blind, placebo-controlled study of 90 consecutive patients admitted within 48 hours of onset of ACS with CRP levels > or =1.4 mg/dL. Patients were assigned to atorvastatin 40 mg daily or placebo over 30 days. C-reactive protein levels, lipid profiles, serum fibrinogen, white cell count, and erythrocyte sedimentation rate were measured at entry, hospital discharge, and 1 month later. RESULTS: Baseline clinical characteristics did not differ between atorvastatin and placebo groups (mean age 59.3 +/- 13.4 vs 61.1 +/- 11.5, P = ns); myocardial infarction 52.3% versus 67.4% ( P = ns). In both groups, median baseline CRP levels were comparable (5.97 +/- 6.2 vs 4.64 +/- 4.2 mg/dL, P = ns). C-reactive protein levels were lower in the atorvastatin group versus control group at discharge (1.68 +/- 1.65 vs 4.12 +/- 4.18 mg/dL) and at 30 days (0.50 +/- 0.71 vs 2.91 +/- 2.68 mg/dL, both P < .0001). C-reactive protein levels significantly decreased from baseline to discharge and 1 month later in placebo and atorvastatin groups (both P < .0001); however, the reduction was greater in the atorvastatin group (62% vs 11% at discharge [P < .0001]; 84% vs 30% at 1 month [P < .0001]). In addition, atorvastatin was associated with a reduction in total and low-density lipoprotein cholesterol and erythrocyte sedimentation rate at discharge and at 30 days (P < .0001 for all comparisons). No correlation was found between changes in CRP and cholesterol levels. CONCLUSIONS: C-reactive protein levels in ACS were rapidly reduced with atorvastatin. These data provide evidence that statins have fast and early anti-inflammatory effects in addition to lipid-lowering effects in ACS.


Asunto(s)
Antiinflamatorios/administración & dosificación , Proteína C-Reactiva/metabolismo , Enfermedad Coronaria/tratamiento farmacológico , Enfermedad Coronaria/metabolismo , Ácidos Heptanoicos/administración & dosificación , Inhibidores de Hidroximetilglutaril-CoA Reductasas/administración & dosificación , Pirroles/administración & dosificación , Enfermedad Aguda , Proteínas de Fase Aguda/efectos de los fármacos , Proteínas de Fase Aguda/metabolismo , Atorvastatina , Biomarcadores/metabolismo , Proteína C-Reactiva/efectos de los fármacos , LDL-Colesterol/efectos de los fármacos , LDL-Colesterol/metabolismo , Enfermedad Coronaria/complicaciones , Complicaciones de la Diabetes , Método Doble Ciego , Esquema de Medicación , Femenino , Guías como Asunto , Humanos , Hiperlipidemias/complicaciones , Lípidos/sangre , Masculino , Persona de Mediana Edad , Estudios Prospectivos , Prevención Secundaria , Síndrome
19.
Int J Cardiol ; 99(2): 253-61, 2005 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-15749184

RESUMEN

BACKGROUND: The progression of chronic heart failure (CHF) is characterized by frequent exacerbation requiring hospitalization and high mortality. Clinical deterioration is triggered by many factors that could promote ongoing myocytes injury. We sought to determine whether a specific marker of cardiac injury, troponin T (cTnT), is associated with prognosis in acute decompensated heart failure (ADHF). METHODS: One hundred and eighty-four consecutive patients with ADHF were enrolled in the absence of an acute coronary syndrome. A cTnT value> or =0.1 ng/ml in samples drawn at 6, 12 or 24 h after hospital admission was considered abnormal. RESULTS: Increased levels of cTnT were found in 58 patients (31.5%, group 1). There were no significant differences between group 1 and patients with cTnT<0.1 ng/ml (group 2) in terms of demographic and clinical characteristics, although ischemic etiology was more prevalent in group 1 (51.7% vs. 31.7%, p=0.009). During follow-up, the mortality in groups 1 and 2 was 31% and 17.5% (p=0.038, OR=2.13, 95% CI: 1.03-4.69), respectively. The 3-year free-CHF readmission survival in group 1 and 2 was 25% and 53% (log rank test p=0.015). In a Cox proportional hazard model, poor tissue perfusion (HR=2.46, 95% CI=1.31-4.6), previous infarction (HR=1.99, 95% CI=1.02-3.9) and cTnT> or =0.1 ng/ml (HR=1.74, 95% CI=1.05-2.9) emerged as the independent predictors of long-term outcome. CONCLUSIONS: One third of patients with decompensated CHF had elevated levels of cTnT. Troponin T was an independent long-term prognostic marker of morbidity and mortality and it suggests a role of biochemical risk stratification in this setting.


Asunto(s)
Insuficiencia Cardíaca/sangre , Isquemia Miocárdica/sangre , Troponina T/sangre , Enfermedad Aguda , Anciano , Progresión de la Enfermedad , Ecocardiografía , Electrocardiografía , Femenino , Insuficiencia Cardíaca/complicaciones , Insuficiencia Cardíaca/mortalidad , Ventrículos Cardíacos/diagnóstico por imagen , Ventrículos Cardíacos/fisiopatología , Mortalidad Hospitalaria , Humanos , Masculino , Persona de Mediana Edad , Isquemia Miocárdica/etiología , Isquemia Miocárdica/fisiopatología , Valor Predictivo de las Pruebas , Pronóstico , Estudios Prospectivos , Función Ventricular Izquierda/fisiología
20.
Mol Cell Biol ; 35(9): 1543-56, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25713103

RESUMEN

Xbp1, a key mediator of the unfolded protein response (UPR), is activated by IRE1α-mediated splicing, which results in a frameshift to encode a protein with transcriptional activity. However, the direct function of Xbp1 in epithelial cells during mammary gland development is unknown. Here we report that the loss of Xbp1 in the mammary epithelium through targeted deletion leads to poor branching morphogenesis, impaired terminal end bud formation, and spontaneous stromal fibrosis during the adult virgin period. Additionally, epithelial Xbp1 deletion induces endoplasmic reticulum (ER) stress in the epithelium and dramatically inhibits epithelial proliferation and differentiation during lactation. The synthesis of milk and its major components, α/ß-casein and whey acidic protein (WAP), is significantly reduced due to decreased prolactin receptor (Prlr) and ErbB4 expression in Xbp1-deficient mammary epithelium. Reduction of Prlr and ErbB4 expression and their diminished availability at the cell surface lead to reduced phosphorylated Stat5, an essential regulator of cell proliferation and differentiation during lactation. As a result, lactating mammary glands in these mice produce less milk protein, leading to poor pup growth and postnatal death. These findings suggest that the loss of Xbp1 induces a terminal UPR which blocks proliferation and differentiation during mammary gland development.


Asunto(s)
Proliferación Celular , Proteínas de Unión al ADN/metabolismo , Lactancia , Glándulas Mamarias Animales/citología , Glándulas Mamarias Animales/crecimiento & desarrollo , Factores de Transcripción/metabolismo , Animales , Diferenciación Celular , Células Cultivadas , Proteínas de Unión al ADN/análisis , Proteínas de Unión al ADN/genética , Células Epiteliales/citología , Células Epiteliales/metabolismo , Femenino , Eliminación de Gen , Glándulas Mamarias Animales/ultraestructura , Ratones , Ratones Endogámicos C57BL , Prolactina/análisis , Prolactina/metabolismo , Receptor ErbB-4/análisis , Receptor ErbB-4/metabolismo , Factores de Transcripción del Factor Regulador X , Factor de Transcripción STAT5/análisis , Factor de Transcripción STAT5/metabolismo , Factores de Transcripción/análisis , Factores de Transcripción/genética , Respuesta de Proteína Desplegada , Proteína 1 de Unión a la X-Box
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA