Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Front Cell Dev Biol ; 11: 1221784, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37440925

RESUMO

Breast cancer has emerged as the most commonly diagnosed cancer and primary cause of cancer-related deaths among women worldwide. Although significant progress has been made in targeting the primary tumor, the effectiveness of systemic treatments to prevent metastasis remains limited. Metastatic disease continues to be the predominant factor leading to fatality in the majority of breast cancer patients. The existence of a prolonged latency period between initial treatment and eventual recurrence in certain patients indicates that tumors can both adapt to and interact with the systemic environment of the host, facilitating and sustaining the progression of the disease. In order to identify potential therapeutic interventions for metastasis, it will be crucial to gain a comprehensive framework surrounding the mechanisms driving the growth, survival, and spread of tumor cells, as well as their interaction with supporting cells of the microenvironment. This review aims to consolidate recent discoveries concerning critical aspects of breast cancer metastasis, encompassing the intricate network of cells, molecules, and physical factors that contribute to metastasis, as well as the molecular mechanisms governing cancer dormancy.

2.
Dev Cell ; 58(13): 1123-1125, 2023 07 10.
Artigo em Inglês | MEDLINE | ID: mdl-37433279

RESUMO

In this issue of Developmental Cell, Pfannenstein and Macara leverage the experimental versatility of the mammary gland to show that intercalation within the terminal end bud is regulated by the tight junction organizing protein ZO-1 and dynamic actin cytoskeletal rearrangements to propel ductal elongation.


Assuntos
Actinas , Citoesqueleto
3.
Cell Stem Cell ; 30(6): 743-744, 2023 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-37267909

RESUMO

In this issue of Cell Stem Cell, Beziaud et al.1 show that immunotherapy induces stem-like properties in models of breast cancer. Strikingly, T-cell-derived IFNγ promotes cancer stem cell (CSC) phenotypes, therapy resistance, and metastasis. Targeting BCAT1 downstream offers promise for enhancing immunotherapy outcomes.


Assuntos
Inibidores de Checkpoint Imunológico , Neoplasias , Linfócitos T , Células-Tronco Neoplásicas , Neoplasias/terapia
4.
Mol Biol Cell ; 33(11): ar103, 2022 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-36001375

RESUMO

Cell-extracellular matrix (ECM) interactions represent fundamental exchanges during tumor progression, yet how particular signal-transduction factors prompt the conversion of tumor cells into migratory populations capable of systemic spread during metastasis remains elusive. We demonstrate that the noncanonical Wnt receptor, Ror2, regulates tumor cell-driven matrix remodeling and invasion in breast cancer. Ror2 loss-of-function (LOF) triggers the disruption of E-cadherin within tumor cells, accompanied by an increase in tumor cell invasion and collagen realignment in three-dimensional cultures. RNA sequencing of Ror2-deficient organoids further uncovered alterations in actin cytoskeleton, cell adhesion, and collagen cross-linking gene expression programs. Spatially, we pinpoint the up-regulation and redistribution of α5 and ß3 integrins together with the production of fibronectin in areas of invasion downstream of Ror2 loss. Wnt/ß-catenin-dependent and Wnt/Ror2 alternative Wnt signaling appear to regulate distinct functions for tumor cells regarding their ability to modify cell-ECM exchanges during invasion. Furthermore, blocking either integrin or focal adhesion kinase (FAK), a downstream mediator of integrin-mediated signal transduction, abrogates the enhanced migration observed upon Ror2 loss. These results reveal a critical function for the alternative Wnt receptor, Ror2, as a determinant of tumor cell-driven ECM exchanges during cancer invasion and metastasis.


Assuntos
Neoplasias da Mama , Receptores Órfãos Semelhantes a Receptor Tirosina Quinase/metabolismo , Adesão Celular , Linhagem Celular Tumoral , Junções Célula-Matriz , Feminino , Humanos , Integrinas , Via de Sinalização Wnt
5.
Nat Commun ; 12(1): 4262, 2021 07 12.
Artigo em Inglês | MEDLINE | ID: mdl-34253738

RESUMO

The epithelial-mesenchymal transition (EMT) has been implicated in conferring stem cell properties and therapeutic resistance to cancer cells. Therefore, identification of drugs that can reprogram EMT may provide new therapeutic strategies. Here, we report that cells derived from claudin-low mammary tumors, a mesenchymal subtype of triple-negative breast cancer, exhibit a distinctive organoid structure with extended "spikes" in 3D matrices. Upon a miR-200 induced mesenchymal-epithelial transition (MET), the organoids switch to a smoother round morphology. Based on these observations, we developed a morphological screening method with accompanying analytical pipelines that leverage deep neural networks and nearest neighborhood classification to screen for EMT-reversing drugs. Through screening of a targeted epigenetic drug library, we identified multiple class I HDAC inhibitors and Bromodomain inhibitors that reverse EMT. These data support the use of morphological screening of mesenchymal mammary tumor organoids as a platform to identify drugs that reverse EMT.


Assuntos
Antineoplásicos/farmacologia , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Neoplasias Mamárias Animais/patologia , Mesoderma/patologia , Organoides/patologia , Animais , Azacitidina/farmacologia , Benzamidas/farmacologia , Ensaios de Seleção de Medicamentos Antitumorais , Epigênese Genética , Feminino , Regulação Neoplásica da Expressão Gênica , Processamento de Imagem Assistida por Computador , Neoplasias Mamárias Animais/genética , Camundongos Endogâmicos BALB C , MicroRNAs/genética , MicroRNAs/metabolismo , Proteínas de Neoplasias/metabolismo , Organoides/efeitos dos fármacos , Pirimidinas/farmacologia , Reprodutibilidade dos Testes , Bibliotecas de Moléculas Pequenas/farmacologia
6.
Front Oncol ; 11: 645698, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33777805

RESUMO

While numerous therapies are highly efficacious in early-stage breast cancers and in particular subsets of breast cancers, therapeutic resistance and metastasis unfortunately arise in many patients. In many cases, tumors that are resistant to standard of care therapies, as well as tumors that have metastasized, are treatable but incurable with existing clinical strategies. Both therapy resistance and metastasis are multi-step processes during which tumor cells must overcome diverse environmental and selective hurdles. Mechanisms by which tumor cells achieve this are numerous and include acquisition of invasive and migratory capabilities, cell-intrinsic genetic and/or epigenetic adaptations, clonal selection, immune evasion, interactions with stromal cells, entering a state of dormancy or senescence, and maintaining self-renewal capacity. To overcome therapy resistance and metastasis in breast cancer, the ability to effectively model each of these mechanisms in the laboratory is essential. Herein we review historic and the current state-of-the-art laboratory model systems and experimental approaches used to investigate breast cancer metastasis and resistance to standard of care therapeutics. While each model system has inherent limitations, they have provided invaluable insights, many of which have translated into regimens undergoing clinical evaluation. We will discuss the limitations and advantages of a variety of model systems that have been used to investigate breast cancer metastasis and therapy resistance and outline potential strategies to improve experimental modeling to further our knowledge of these processes, which will be crucial for the continued development of effective breast cancer treatments.

7.
Cancer Res ; 78(15): 4229-4240, 2018 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-29844125

RESUMO

Cooperativity between WNT and FGF signaling is well documented in embryonic development and cancer progression, but the molecular mechanisms underlying this cross-talk remain elusive. In this study, we interrogated the dynamics of RNA levels, ribosome occupancy, and protein expression as a function of inducible FGF signaling in mouse mammary glands with constitutive WNT hyperactivation. Multiomics correlation analysis revealed a substantial discrepancy between RNA and ribosome occupancy levels versus protein levels. However, this discrepancy decreased as cells became premalignant and dynamically responded to FGF signaling, implicating the importance of stringent gene regulation in nontransformed cells. Analysis of individual genes demonstrated that acute FGF hyperactivation increased translation of many stem cell self-renewal regulators, including WNT signaling components, and decreased translation of genes regulating cellular senescence. WNT pathway components translationally upregulated by FGF signaling had long and structured 5' UTRs with a high frequency of polypurine sequences, several of which harbored (CGG)4 motifs that can fold into either stable G-quadruplexes or other stable secondary structures. The FGF-mediated increase in translation of WNT pathway components was compromised by silvestrol, an inhibitor of EIF4A that clamps EIF4A to polypurine sequences to block 43S scanning and inhibits its RNA-unwinding activity important for translation initiation. Moreover, silvestrol treatment significantly delayed FGF-WNT-driven tumorigenesis. Taken together, these results suggest that FGF signaling selectively enhances translation of structured mRNAs, particularly WNT signaling components, and highlight their vulnerability to inhibitors that target the RNA helicase EIF4A.Significance: The RNA helicase EIF4A may serve as a therapeutic target for breast cancers that require FGF and WNT signaling. Cancer Res; 78(15); 4229-40. ©2018 AACR.


Assuntos
Regiões 5' não Traduzidas/genética , Fator de Iniciação 4A em Eucariotos/genética , Biossíntese de Proteínas/genética , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/genética , Via de Sinalização Wnt/genética , Regiões 5' não Traduzidas/efeitos dos fármacos , Animais , Camundongos , Biossíntese de Proteínas/efeitos dos fármacos , RNA Helicases/genética , RNA Mensageiro/genética , Ribossomos/efeitos dos fármacos , Ribossomos/genética , Triterpenos/farmacologia , Via de Sinalização Wnt/efeitos dos fármacos
8.
Dev Cell ; 43(4): 436-448.e6, 2017 11 20.
Artigo em Inglês | MEDLINE | ID: mdl-29103953

RESUMO

Puberty is characterized by dynamic tissue remodeling in the mammary gland involving ductal elongation, resolution into the mature epithelial bilayer, and lumen formation. To decipher the cellular mechanisms underlying these processes, we studied the fate of putative stem cells, termed cap cells, present in terminal end buds of pubertal mice. Employing a p63CreERT2-based lineage-tracing strategy, we identified a unipotent fate for proliferative cap cells that only generated cells with basal features. Furthermore, we observed that dislocated "cap-in-body" cells underwent apoptosis, which aided lumen formation during ductal development. Basal lineage-specific profiling and genetic loss-of-function experiments revealed a critical role for FOXO transcription factors in mediating these proliferative versus apoptotic fates. Importantly, these studies revealed a mode of WNT signaling-mediated FOXO1 inhibition, potentially mediated through AKT. Together, these data suggest that the WNT pathway confers proliferative and survival advantages on cap cells via regulation of FOXO1 localization.


Assuntos
Apoptose/fisiologia , Mama/metabolismo , Proteína Forkhead Box O1/metabolismo , Homeostase/fisiologia , Células-Tronco/citologia , Via de Sinalização Wnt/fisiologia , Animais , Mama/citologia , Linhagem da Célula/fisiologia , Proliferação de Células/fisiologia , Glândulas Mamárias Animais/citologia , Glândulas Mamárias Animais/crescimento & desenvolvimento , Glândulas Mamárias Animais/metabolismo , Camundongos Transgênicos
9.
Endocr Relat Cancer ; 22(6): T161-76, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26206777

RESUMO

The mammary gland is a dynamic organ that undergoes extensive morphogenesis during the different stages of embryonic development, puberty, estrus, pregnancy, lactation and involution. Systemic and local cues underlie this constant tissue remodeling and act by eliciting an intricate pattern of responses in the mammary epithelial and stromal cells. Decades of studies utilizing methods such as transplantation and lineage-tracing have identified a complex hierarchy of mammary stem cells, progenitors and differentiated epithelial cells that fuel mammary epithelial development. Importantly, these studies have extended our understanding of the molecular crosstalk between cell types and the signaling pathways maintaining normal homeostasis that often are deregulated during tumorigenesis. While several questions remain, this research has many implications for breast cancer. Fundamental among these are the identification of the cells of origin for the multiple subtypes of breast cancer and the understanding of tumor heterogeneity. A deeper understanding of these critical questions will unveil novel breast cancer drug targets and treatment paradigms. In this review, we provide a current overview of normal mammary development and tumorigenesis from a stem cell perspective.


Assuntos
Neoplasias da Mama/patologia , Mama/citologia , Células-Tronco Neoplásicas/citologia , Células-Tronco/citologia , Animais , Mama/embriologia , Mama/crescimento & desenvolvimento , Diferenciação Celular , Linhagem da Célula , Autorrenovação Celular , Células Epiteliais/classificação , Células Epiteliais/citologia , Estrogênios/fisiologia , Feminino , Xenoenxertos , Humanos , Lactação , Glândulas Mamárias Animais/citologia , Glândulas Mamárias Animais/embriologia , Glândulas Mamárias Animais/crescimento & desenvolvimento , Neoplasias Mamárias Experimentais/patologia , Camundongos , Morfogênese , Neoplasias Hormônio-Dependentes/patologia , Especificidade de Órgãos , Comunicação Parácrina , Gravidez , Progesterona/fisiologia , Puberdade , Receptores de Estrogênio/fisiologia , Receptores de Progesterona/fisiologia , Transdução de Sinais , Transplante de Células-Tronco
10.
J Cell Biol ; 208(3): 351-66, 2015 Feb 02.
Artigo em Inglês | MEDLINE | ID: mdl-25624393

RESUMO

Wnt signaling encompasses ß-catenin-dependent and -independent networks. How receptor context provides Wnt specificity in vivo to assimilate multiple concurrent Wnt inputs throughout development remains unclear. Here, we identified a refined expression pattern of Wnt/receptor combinations associated with the Wnt/ß-catenin-independent pathway in mammary epithelial subpopulations. Moreover, we elucidated the function of the alternative Wnt receptor Ror2 in mammary development and provided evidence for coordination of this pathway with Wnt/ß-catenin-dependent signaling in the mammary epithelium. Lentiviral short hairpin RNA (shRNA)-mediated depletion of Ror2 in vivo increased branching and altered the differentiation of the mammary epithelium. Microarray analyses identified distinct gene level alterations within the epithelial compartments in the absence of Ror2, with marked changes observed in genes associated with the actin cytoskeleton. Modeling of branching morphogenesis in vitro defined specific defects in cytoskeletal dynamics accompanied by Rho pathway alterations downstream of Ror2 loss. The current study presents a model of Wnt signaling coordination in vivo and assigns an important role for Ror2 in mammary development.


Assuntos
Citoesqueleto de Actina/metabolismo , Diferenciação Celular , Células Epiteliais/fisiologia , Glândulas Mamárias Animais/fisiologia , Receptores Órfãos Semelhantes a Receptor Tirosina Quinase/fisiologia , Animais , Forma Celular , Epitélio/fisiologia , Feminino , Glândulas Mamárias Animais/citologia , Camundongos , Via de Sinalização Wnt
11.
Stem Cells ; 32(10): 2571-82, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24891218

RESUMO

In breast cancer, a subset of tumor-initiating cells (TIC) or "cancer stem cells" are thought to be responsible for tumor maintenance, treatment resistance, and disease recurrence. While current breast cancer stem cell markers (e.g., CD44(high) /CD24(low/neg) , ALDH positive) have allowed enrichment for such cells, they are not universally expressed and may actually identify distinct TIC subpopulations in the same tumor. Thus, additional markers of functional stem cells are needed. The STAT3 pathway is a critical regulator of the function of normal stem cells, and evidence is accumulating for its important role in breast cancer stem cells. However, due to the lack of a method for separating live cells based on their level of STAT3 activity, it remains unknown whether STAT3 functions in the cancer stem cells themselves, or in surrounding niche cells, or in both. To approach this question, we constructed a series of lentiviral fluorescent (enhanced green fluorescent protein, EGFP) reporters that enabled flow cytometric enrichment of cells differing in STAT3-mediated transcriptional activity, as well as in vivo/in situ localization of STAT3 responsive cells. Using in vivo claudin-low cell line xenograft models of human breast cancer, we found that STAT3 signaling reporter activity (EGFP(+) ) is associated with a subpopulation of cancer cells enriched for mammosphere-forming efficiency, as well as TIC function in limiting dilution transplantation assays compared to negative or unsorted populations. Our results support STAT3 signaling activity as another functional marker for human breast cancer stem cells thus making it an attractive therapeutic target for stem-cell-directed therapy in some breast cancer subtypes.


Assuntos
Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Claudinas/metabolismo , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/patologia , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais , Animais , Carcinogênese/patologia , Linhagem Celular Tumoral , Feminino , Fluorescência , Genes Reporter , Proteínas de Fluorescência Verde/metabolismo , Humanos , Lentivirus/metabolismo , Camundongos SCID , Modelos Biológicos , Reprodutibilidade dos Testes , Esferoides Celulares/metabolismo , Esferoides Celulares/patologia , Ensaios Antitumorais Modelo de Xenoenxerto
12.
Breast Cancer Res ; 16(1): 202, 2014 Jan 29.
Artigo em Inglês | MEDLINE | ID: mdl-24476463

RESUMO

Paracrine signaling mechanisms play a critical role in both normal mammary gland development and breast cancer. Dissection of these mechanisms using genetically engineered mouse models has provided significant insight into our understanding of the mechanisms that guide intratumoral heterogeneity. In the following perspective, we briefly review some of the emerging concepts in this field and emphasize why elucidation of these pathways will be important for future progress in devising new and improved combinatorial therapeutic approaches for breast and other solid cancers.


Assuntos
Glândulas Mamárias Animais/embriologia , Glândulas Mamárias Animais/crescimento & desenvolvimento , Neoplasias Mamárias Animais/genética , Comunicação Parácrina/genética , Animais , Receptores ErbB/metabolismo , Feminino , Humanos , Neoplasias Mamárias Animais/patologia , Camundongos , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/metabolismo , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/metabolismo , Receptores de Estrogênio/metabolismo , Via de Sinalização Wnt/fisiologia
13.
Breast Cancer Res ; 15(6): 322, 2013 Dec 23.
Artigo em Inglês | MEDLINE | ID: mdl-24365076

RESUMO

The mammary gland requires the coordinated efforts of multiple epithelial cell lineages to build an organized bilayered ductal network. How epigenetic regulators direct decisions of cell-fate and lineage determination in cooperation with intrinsic transcriptional factors and extrinsic signaling factors remains a fundamental question in the field of developmental biology. Recent work sheds new light on the role of the histone methylation reader Pygopus 2 in coordinating the balance of self-renewal Wnt signals with luminal-specific Notch signals in mammary epithelial lineage determination.


Assuntos
Diferenciação Celular , Células Epiteliais/citologia , Peptídeos e Proteínas de Sinalização Intracelular/fisiologia , Glândulas Mamárias Animais/citologia , Receptores Notch/metabolismo , Células-Tronco/citologia , Proteínas Wnt/metabolismo , Animais , Feminino , Masculino
14.
Stem Cells ; 31(1): 178-89, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23097355

RESUMO

Fibroblast growth factor (FGF) signaling plays an important role in embryonic stem cells and adult tissue homeostasis, but the function of FGFs in mammary gland stem cells is less well defined. Both FGFR1 and FGFR2 are expressed in basal and luminal mammary epithelial cells (MECs), suggesting that together they might play a role in mammary gland development and stem cell dynamics. Previous studies have demonstrated that the deletion of FGFR2 resulted only in transient developmental defects in branching morphogenesis. Using a conditional deletion strategy, we investigated the consequences of FGFR1 deletion alone and then the simultaneous deletion of both FGFR1 and FGFR2 in the mammary epithelium. FGFR1 deletion using a keratin 14 promoter-driven Cre-recombinase resulted in an early, yet transient delay in development. However, no reduction in functional outgrowth potential was observed following limiting dilution transplantation analysis. In contrast, a significant reduction in outgrowth potential was observed upon the deletion of both FGFR1 and FGFR2 in MECs using adenovirus-Cre. Additionally, using a fluorescent reporter mouse model to monitor Cre-mediated recombination, we observed a competitive disadvantage following transplantation of both FGFR1/R2-null MECs, most prominently in the basal epithelial cells. This correlated with the complete loss of the mammary stem cell repopulating population in the FGFR1/R2-attenuated epithelium. FGFR1/R2-null MECs were partially rescued in chimeric outgrowths containing wild-type MECs, suggesting the potential importance of paracrine mechanisms involved in the maintenance of the basal epithelial stem cells. These studies document the requirement for functional FGFR signaling in mammary stem cells during development.


Assuntos
Glândulas Mamárias Animais/crescimento & desenvolvimento , Glândulas Mamárias Animais/metabolismo , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/metabolismo , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/metabolismo , Células-Tronco/fisiologia , Animais , Células Epiteliais/metabolismo , Feminino , Técnicas de Inativação de Genes , Glândulas Mamárias Animais/citologia , Glândulas Mamárias Animais/embriologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/genética , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/genética , Transdução de Sinais
15.
PLoS Genet ; 8(7): e1002840, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22911650

RESUMO

Pregnancy-induced noncoding RNA (PINC) and retinoblastoma-associated protein 46 (RbAp46) are upregulated in alveolar cells of the mammary gland during pregnancy and persist in alveolar cells that remain in the regressed lobules following involution. The cells that survive involution are thought to function as alveolar progenitor cells that rapidly differentiate into milk-producing cells in subsequent pregnancies, but it is unknown whether PINC and RbAp46 are involved in maintaining this progenitor population. Here, we show that, in the post-pubertal mouse mammary gland, mPINC is enriched in luminal and alveolar progenitors. mPINC levels increase throughout pregnancy and then decline in early lactation, when alveolar cells undergo terminal differentiation. Accordingly, mPINC expression is significantly decreased when HC11 mammary epithelial cells are induced to differentiate and produce milk proteins. This reduction in mPINC levels may be necessary for lactation, as overexpression of mPINC in HC11 cells blocks lactogenic differentiation, while knockdown of mPINC enhances differentiation. Finally, we demonstrate that mPINC interacts with RbAp46, as well as other members of the polycomb repressive complex 2 (PRC2), and identify potential targets of mPINC that are differentially expressed following modulation of mPINC expression levels. Taken together, our data suggest that mPINC inhibits terminal differentiation of alveolar cells during pregnancy to prevent abundant milk production and secretion until parturition. Additionally, a PRC2 complex that includes mPINC and RbAp46 may confer epigenetic modifications that maintain a population of mammary epithelial cells committed to the alveolar fate in the involuted gland.


Assuntos
Diferenciação Celular , Glândulas Mamárias Animais/metabolismo , Gravidez/metabolismo , RNA não Traduzido/metabolismo , Proteínas Repressoras/metabolismo , Proteína 7 de Ligação ao Retinoblastoma/metabolismo , Animais , Feminino , Técnicas de Silenciamento de Genes , Glândulas Mamárias Animais/citologia , Camundongos , Camundongos Endogâmicos BALB C , Proteínas do Grupo Polycomb , RNA não Traduzido/genética , Ratos
16.
Proc Natl Acad Sci U S A ; 108(9): 3665-70, 2011 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-21307310

RESUMO

The discovery of RNAi has revolutionized loss-of-function genetic studies in mammalian systems. However, significant challenges still remain to fully exploit RNAi for mammalian genetics. For instance, genetic screens and in vivo studies could be broadly improved by methods that allow inducible and uniform gene expression control. To achieve this, we built the lentiviral pINDUCER series of expression vehicles for inducible RNAi in vivo. Using a multicistronic design, pINDUCER vehicles enable tracking of viral transduction and shRNA or cDNA induction in a broad spectrum of mammalian cell types in vivo. They achieve this uniform temporal, dose-dependent, and reversible control of gene expression across heterogenous cell populations via fluorescence-based quantification of reverse tet-transactivator expression. This feature allows isolation of cell populations that exhibit a potent, inducible target knockdown in vitro and in vivo that can be used in human xenotransplantation models to examine cancer drug targets.


Assuntos
Técnicas Genéticas , Vetores Genéticos/genética , Lentivirus/genética , Interferência de RNA , Animais , Neoplasias da Mama/patologia , Linhagem Celular , DNA Complementar/genética , Diagnóstico por Imagem , Feminino , Expressão Gênica , Humanos , Luminescência , Glândulas Mamárias Animais/metabolismo , Glândulas Mamárias Animais/patologia , Camundongos , RNA Interferente Pequeno/metabolismo , Reprodutibilidade dos Testes , Ensaios Antitumorais Modelo de Xenoenxerto
17.
Curr Opin Pharmacol ; 10(6): 643-9, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20810315

RESUMO

The mammary stem cell and its local microenvironment are central for the maintenance of proper tissue homeostasis during normal development. Defining the hierarchical organization of the epithelial subtypes in the mammary gland and the molecular pathways guiding their development has begun to provide a framework for understanding how cancer stem cells sustain the progression and heterogeneity of breast cancers. The Wnt pathway plays a fundamental role in multiple adult stem cells, as well as in orchestrating proper mammary gland development and maintenance. These processes are intricately guided by the influence of systemic hormones and local factors. Alterations in Wnt signaling can skew the homeostatic balance of the mammary epithelium to drive malignant progression; however, complexities of Wnt pathway components present a challenge in understanding their physiological function.


Assuntos
Mama/metabolismo , Glândulas Mamárias Animais/metabolismo , Glândulas Mamárias Humanas/fisiopatologia , Células-Tronco Neoplásicas/metabolismo , Transdução de Sinais , Células-Tronco/metabolismo , Proteínas Wnt/metabolismo , beta Catenina/metabolismo , Animais , Mama/citologia , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Regulação para Baixo , Epitélio/metabolismo , Receptor alfa de Estrogênio/metabolismo , Feminino , Hormônios/metabolismo , Hormônios/fisiologia , Humanos , Proteínas Relacionadas a Receptor de LDL/metabolismo , Proteína-5 Relacionada a Receptor de Lipoproteína de Baixa Densidade , Proteína-6 Relacionada a Receptor de Lipoproteína de Baixa Densidade , Glândulas Mamárias Animais/citologia , Glândulas Mamárias Humanas/metabolismo , Glândulas Mamárias Humanas/fisiologia , Células-Tronco Neoplásicas/patologia , Receptores de Progesterona/metabolismo , Células-Tronco/patologia
18.
J Immunol ; 183(1): 97-105, 2009 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-19535626

RESUMO

TGF-beta, together with IL-6 and IL-21, promotes Th17 cell development. IL-6 and IL-21 induce activation of STAT3, which is crucial for Th17 cell differentiation, as well as the expression of suppressor of cytokine signaling (SOCS)3, a major negative feedback regulator of STAT3-activating cytokines that negatively regulates Th17 cells. However, it is still largely unclear how TGF-beta regulates Th17 cell development and which TGF-beta signaling pathway is involved in Th17 cell development. In this report, we demonstrate that TGF-beta inhibits IL-6- and IL-21-induced SOCS3 expression, thus enhancing as well as prolonging STAT3 activation in naive CD4(+)CD25(-) T cells. TGF-beta inhibits IL-6-induced SOCS3 promoter activity in T cells. Also, SOCS3 small interfering RNA knockdown partially compensates for the action of TGF-beta on Th17 cell development. In mice with a dominant-negative form of TGF-beta receptor II and impaired TGF-beta signaling, IL-6-induced CD4(+) T cell expression of SOCS3 is higher whereas STAT3 activation is lower compared with wild-type B6 CD4(+) T cells. The addition of a TGF-beta receptor I kinase inhibitor that blocks Smad-dependent TGF-beta signaling greatly, but not completely, abrogates the effect of TGF-beta on Th17 cell differentiation. Our data indicate that inhibition of SOCS3 and, thus, enhancement of STAT3 activation is at least one of the mechanisms of TGF-beta promotion of Th17 cell development.


Assuntos
Diferenciação Celular/imunologia , Interleucina-17/biossíntese , Proteínas Supressoras da Sinalização de Citocina/antagonistas & inibidores , Linfócitos T Auxiliares-Indutores/imunologia , Linfócitos T Auxiliares-Indutores/metabolismo , Fator de Crescimento Transformador beta1/fisiologia , Animais , Diferenciação Celular/genética , Células Cultivadas , Regulação para Baixo/genética , Regulação para Baixo/imunologia , Interleucina-17/fisiologia , Interleucina-6/antagonistas & inibidores , Interleucina-6/fisiologia , Interleucinas/antagonistas & inibidores , Interleucinas/fisiologia , Mucosa Intestinal/citologia , Mucosa Intestinal/imunologia , Mucosa Intestinal/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Mucosa/citologia , Mucosa/imunologia , Mucosa/metabolismo , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/deficiência , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/fisiologia , Receptor do Fator de Crescimento Transformador beta Tipo I , Receptor do Fator de Crescimento Transformador beta Tipo II , Receptores de Fatores de Crescimento Transformadores beta/antagonistas & inibidores , Receptores de Fatores de Crescimento Transformadores beta/deficiência , Receptores de Fatores de Crescimento Transformadores beta/genética , Receptores de Fatores de Crescimento Transformadores beta/fisiologia , Fator de Transcrição STAT3/metabolismo , Fator de Transcrição STAT3/fisiologia , Transdução de Sinais/genética , Transdução de Sinais/imunologia , Proteína 3 Supressora da Sinalização de Citocinas , Proteínas Supressoras da Sinalização de Citocina/biossíntese , Proteínas Supressoras da Sinalização de Citocina/genética , Linfócitos T Auxiliares-Indutores/citologia , Fator de Crescimento Transformador beta1/antagonistas & inibidores , Fator de Crescimento Transformador beta1/genética , Regulação para Cima/genética , Regulação para Cima/imunologia
19.
Breast Cancer Res ; 11(2): R19, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19344510

RESUMO

INTRODUCTION: The tumour-suppressive effects of transforming growth factor-beta (TGF-beta) are well documented; however, the mechanistic basis of these effects is not fully understood. Previously, we showed that a non-canonical member of the Wingless-related protein family, Wnt5a, is required for TGF-beta-mediated effects on mammary development. Several lines of evidence support the hypothesis that Wnt5a acts as a tumour suppressor. In addition, it has been shown that Wnt5a can antagonise canonical Wnt/beta-catenin signalling in various cell types. Here we test the hypothesis that TGF-beta and Wnt5a can antagonise Wnt/beta-catenin signalling and redirect mammary tumour phenotype. The results provide a new mechanism for the tumour-suppressive effects of TGF-beta. METHODS: Wnt/beta-catenin signalling was measured in tumours with altered TGF-beta (dominant-negative TGF-beta type II receptor, DNIIR) or Wnt5a (Wnt5a-/-) signalling as the accumulation of nuclear beta-catenin using both confocal microscopy and cell fractionation. RT-PCR was used to measure the expression of Wnt/beta-catenin target genes. Sca1 expression was determined by western blot and keratin (K) 6- and K14-positive populations were determined by immunohistochemistry. RESULTS: Loss of TGF-beta or Wnt5a signalling resulted in stabilisation of nuclear beta-catenin and expression of Wnt/beta-catenin target genes suggesting that TGF-beta and Wnt5a act to inhibit Wnt/beta-catenin signalling in mammary epithelium. Increased expression of Sca-1 was observed in developing DNIIR and Wnt5a-/- mammary glands. DNIIR and Wnt5a-/- tumours demonstrated an expanded population of K6- and K14-expressing cells typically seen in Wnt/beta-catenin-induced tumours. CONCLUSIONS: The key findings here are that: TGF-beta and Wnt5a regulate Wnt/beta-catenin activity; and loss of TGF-beta and Wnt5a redirect the phenotype of tumours so that they resemble tumours induced by activation of Wnt/beta-catenin. The findings suggest a new mechanism for the tumour-suppressive effects of TGF-beta.


Assuntos
Glândulas Mamárias Animais/crescimento & desenvolvimento , Neoplasias Mamárias Experimentais/metabolismo , Neoplasias Mamárias Experimentais/patologia , Proteínas Serina-Treonina Quinases/metabolismo , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Proteínas Wnt/fisiologia , beta Catenina/metabolismo , Animais , Western Blotting , Proliferação de Células , Feminino , Genes Dominantes , Técnicas Imunoenzimáticas , Glândulas Mamárias Animais/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos ICR , Camundongos Knockout , Camundongos SCID , Fenótipo , Proteínas Serina-Treonina Quinases/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptor do Fator de Crescimento Transformador beta Tipo II , Receptores de Fatores de Crescimento Transformadores beta/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Frações Subcelulares , Fator de Crescimento Transformador beta/genética , Proteína Wnt-5a , beta Catenina/genética
20.
Clin Cancer Res ; 14(15): 4694-704, 2008 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-18676737

RESUMO

PURPOSE: STATs activate transcription in response to numerous cytokines, controlling proliferation, gene expression, and apoptosis. Aberrant activation of STAT proteins, particularly STAT-3, is implicated in the pathogenesis of many cancers, including GBM, by promoting cell cycle progression, stimulating angiogenesis, and impairing tumor immune surveillance. Little is known about the endogenous STAT inhibitors, the PIAS proteins, in human malignancies. The objective of this study was to examine the expression of STAT-3 and its negative regulator, PIAS3, in human tissue samples from control and GBM brains. EXPERIMENTAL DESIGN: Control and GBM human tissues were analyzed by immunoblotting and immunohistochemistry to determine the activation status of STAT-3 and expression of the PIAS3 protein. The functional consequence of PIAS3 inhibition by small interfering RNA or PIAS3 overexpression in GBM cells was determined by examining cell proliferation, STAT-3 transcriptional activity, and STAT-3 target gene expression. This was accomplished using [(3)H]TdR incorporation, STAT-3 dominant-negative constructs, reverse transcription-PCR, and immunoblotting. RESULTS AND CONCLUSIONS: STAT-3 activation, as assessed by tyrosine and serine phosphorylation, was elevated in GBM tissue compared with control tissue. Interestingly, we observed expression of PIAS3 in control tissue, whereas PIAS3 protein expression in GBM tissue was greatly reduced. Inhibition of PIAS3 resulted in enhanced glioblastoma cellular proliferation. Conversely, PIAS3 overexpression inhibited STAT-3 transcriptional activity, expression of STAT-3-regulated genes, and cell proliferation. We propose that the loss of PIAS3 in GBM contributes to enhanced STAT-3 transcriptional activity and subsequent cell proliferation.


Assuntos
Regulação Neoplásica da Expressão Gênica , Glioblastoma/tratamento farmacológico , Glioblastoma/metabolismo , Fator de Transcrição STAT3/metabolismo , Animais , Linhagem Celular Tumoral , Proliferação de Células , Humanos , Metaloproteinase 9 da Matriz/metabolismo , Fosforilação , RNA Interferente Pequeno/metabolismo , Serina/química , Proteína 3 Supressora da Sinalização de Citocinas , Proteínas Supressoras da Sinalização de Citocina/metabolismo , Transcrição Gênica , Ativação Transcricional , Tirosina/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA