Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
1.
Nature ; 526(7571): 131-5, 2015 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-26416748

RESUMO

Despite major advances in understanding the molecular and genetic basis of cancer, metastasis remains the cause of >90% of cancer-related mortality. Understanding metastasis initiation and progression is critical to developing new therapeutic strategies to treat and prevent metastatic disease. Prevailing theories hypothesize that metastases are seeded by rare tumour cells with unique properties, which may function like stem cells in their ability to initiate and propagate metastatic tumours. However, the identity of metastasis-initiating cells in human breast cancer remains elusive, and whether metastases are hierarchically organized is unknown. Here we show at the single-cell level that early stage metastatic cells possess a distinct stem-like gene expression signature. To identify and isolate metastatic cells from patient-derived xenograft models of human breast cancer, we developed a highly sensitive fluorescence-activated cell sorting (FACS)-based assay, which allowed us to enumerate metastatic cells in mouse peripheral tissues. We compared gene signatures in metastatic cells from tissues with low versus high metastatic burden. Metastatic cells from low-burden tissues were distinct owing to their increased expression of stem cell, epithelial-to-mesenchymal transition, pro-survival, and dormancy-associated genes. By contrast, metastatic cells from high-burden tissues were similar to primary tumour cells, which were more heterogeneous and expressed higher levels of luminal differentiation genes. Transplantation of stem-like metastatic cells from low-burden tissues showed that they have considerable tumour-initiating capacity, and can differentiate to produce luminal-like cancer cells. Progression to high metastatic burden was associated with increased proliferation and MYC expression, which could be attenuated by treatment with cyclin-dependent kinase (CDK) inhibitors. These findings support a hierarchical model for metastasis, in which metastases are initiated by stem-like cells that proliferate and differentiate to produce advanced metastatic disease.


Assuntos
Neoplasias da Mama/patologia , Progressão da Doença , Metástase Neoplásica/patologia , Células-Tronco Neoplásicas/patologia , Análise de Célula Única , Animais , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/genética , Ciclo Celular/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Separação Celular , Transformação Celular Neoplásica/efeitos dos fármacos , Transformação Celular Neoplásica/patologia , Quinases Ciclina-Dependentes/antagonistas & inibidores , Modelos Animais de Doenças , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/patologia , Transição Epitelial-Mesenquimal/genética , Citometria de Fluxo , Perfilação da Expressão Gênica , Genes myc/genética , Humanos , Mesoderma/metabolismo , Mesoderma/patologia , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Metástase Neoplásica/tratamento farmacológico , Células-Tronco Neoplásicas/efeitos dos fármacos , Células-Tronco Neoplásicas/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
2.
Biochem Biophys Res Commun ; 526(1): 41-47, 2020 05 21.
Artigo em Inglês | MEDLINE | ID: mdl-32192771

RESUMO

Human breast tumors are not fully autonomous. They are dependent on nutrients and growth-promoting signals provided by the supporting stromal cells. Within the tumor microenvironment, one of the secreted macromolecules by tumor cells is activin A, where we show to downregulate CD36 in fibroblasts. Downregulation of CD36 in fibroblasts also increases the secretion of activin A by fibroblasts. We hypothesize that overexpression of CD36 in fibroblasts inhibits the formation of solid tumors in subtypes of breast cancer models. For the first time, we show that co-culturing organoid models of breast cancer cell lines of MDA-MB-231 (e.g., a triple-negative line) or MCF7 (e.g., a luminal-A line) with CD36+ fibroblasts inhibit the growth and normalizes basal and lateral polarities, respectively. In the long-term anchorage-independent growth assay, the rate of colony formation is also reduced for MDA-MB-231. These observations are consistent with the mechanism of tumor suppression involving the downregulation of pSMAD2/3 and YY1 expression levels. Our integrated analytical methods leverage and extend quantitative assays at cell- and colony-scales in both short- and long-term cultures using brightfield or immunofluorescent microscopy and robust image analysis. Conditioned media are profiled with the ELISA assay.


Assuntos
Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Antígenos CD36/metabolismo , Fibroblastos/metabolismo , Glândulas Mamárias Humanas/patologia , Ativinas/farmacologia , Linhagem Celular Tumoral , Polaridade Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Regulação para Baixo/efeitos dos fármacos , Feminino , Fibroblastos/efeitos dos fármacos , Fibroblastos/patologia , Humanos , Fosforilação/efeitos dos fármacos , Proteínas Smad/metabolismo , Ensaio Tumoral de Célula-Tronco , Fator de Transcrição YY1/metabolismo
3.
Breast Cancer Res Treat ; 176(1): 131-140, 2019 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-30993572

RESUMO

PURPOSE: Understanding how differentiation, microenvironment, and hormonal milieu influence human breast cell susceptibility to malignant transformation will require the use of physiologically relevant in vitro systems. We sought to develop a 3D culture model that enables the propagation of normal estrogen receptor alpha (ER) + cells. METHODS: We tested soluble factors and protocols for the ability to maintain progenitor and ER + cells in cultures established from primary cells. Optimized conditions were then used to profile estrogen-induced gene expression changes in cultures from three pathology-free individuals. RESULTS: Long-term representation of ER + cells was optimal in medium that included three different TGFß/activin receptor-like kinase inhibitors. We found that omitting the BMP signaling antagonist, Noggin, enhanced the responsiveness of the PGR gene to estradiol exposure without altering the proportions of ER + cells in the cultures. Profiling of estradiol-exposed cultures showed that while all the cultures showed immediate and robust induction of PGR, LRP2, and IGFB4, other responses varied qualitatively and quantitatively across specimens. CONCLUSIONS: We successfully identified conditions for the maintenance and propagation of functional ER + cells from normal human breast tissues. We propose that these 3D cultures will overcome limitations of conventional 2D cultures of partially or fully transformed cell lines by sustaining normal endocrine function and growth regulation of the cell populations that comprise intact breasts.


Assuntos
Neoplasias da Mama/metabolismo , Receptores de Estrogênio/metabolismo , Biomarcadores Tumorais , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Feminino , Imunofluorescência , Regulação Neoplásica da Expressão Gênica , Humanos , Células-Tronco Neoplásicas/metabolismo , Receptores de Estrogênio/genética , Esferoides Celulares , Ativação Transcricional , Células Tumorais Cultivadas
4.
Genes Dev ; 24(24): 2800-11, 2010 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-21159820

RESUMO

Organization into polarized three-dimensional structures defines whether epithelial cells are normal or malignant. In a model of morphogenesis, we show that inhibiting key signaling pathways in human breast cancer cells leads to "phenotypic reversion" of the malignant cells. Using architecture as an endpoint, we report that, in all cases, signaling through Raf/MEK/ERK disrupted tissue polarity via matrix metalloproteinase9 (MMP9) activity. Induction of Raf or activation of an engineered, functionally inducible MMP9 in nonmalignant cells led to loss of tissue polarity, and reinitiated proliferation. Conversely, inhibition of Raf or MMP9 with small molecule inhibitors or shRNAs restored the ability of cancer cells to form polarized quiescent structures. Silencing MMP9 expression also reduced tumor growth dramatically in a murine xenograft model. LC-MS/MS analysis comparing conditioned medium from nonmalignant cells with or without active MMP9 revealed laminin 111 (LM1) as an important target of MMP9. LM1 has been implicated in acinar morphogenesis; thus, its degradation by MMP9 provides a mechanism for loss of tissue polarity and reinitiation of growth associated with MMP9 activity. These findings underscore the importance of the dynamic reciprocity between the extracellular matrix integrity, tissue polarity, and Raf/MEK/ERK and MMP9 activities, providing an axis for either tissue homeostasis or malignant progression.


Assuntos
Neoplasias da Mama/patologia , Mama/citologia , Metaloproteinase 9 da Matriz/metabolismo , Morfogênese , Quinases raf/fisiologia , Animais , Técnicas de Cultura de Células , Polaridade Celular , Proliferação de Células , Humanos , Laminina/metabolismo , Camundongos , Neoplasias Experimentais , Transdução de Sinais , Transplante Heterólogo
5.
Semin Cancer Biol ; 35 Suppl: S104-S128, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25869441

RESUMO

One of the hallmarks of malignant cell populations is the ability to undergo continuous proliferation. This property allows clonal lineages to acquire sequential aberrations that can fuel increasingly autonomous growth, invasiveness, and therapeutic resistance. Innate cellular mechanisms have evolved to regulate replicative potential as a hedge against malignant progression. When activated in the absence of normal terminal differentiation cues, these mechanisms can result in a state of persistent cytostasis. This state, termed "senescence," can be triggered by intrinsic cellular processes such as telomere dysfunction and oncogene expression, and by exogenous factors such as DNA damaging agents or oxidative environments. Despite differences in upstream signaling, senescence often involves convergent interdependent activation of tumor suppressors p53 and p16/pRB, but can be induced, albeit with reduced sensitivity, when these suppressors are compromised. Doses of conventional genotoxic drugs required to achieve cancer cell senescence are often much lower than doses required to achieve outright cell death. Additional therapies, such as those targeting cyclin dependent kinases or components of the PI3K signaling pathway, may induce senescence specifically in cancer cells by circumventing defects in tumor suppressor pathways or exploiting cancer cells' heightened requirements for telomerase. Such treatments sufficient to induce cancer cell senescence could provide increased patient survival with fewer and less severe side effects than conventional cytotoxic regimens. This positive aspect is countered by important caveats regarding senescence reversibility, genomic instability, and paracrine effects that may increase heterogeneity and adaptive resistance of surviving cancer cells. Nevertheless, agents that effectively disrupt replicative immortality will likely be valuable components of new combinatorial approaches to cancer therapy.


Assuntos
Proliferação de Células/genética , Senescência Celular/genética , Terapia de Alvo Molecular , Neoplasias/tratamento farmacológico , Neoplasias/genética , Antineoplásicos/uso terapêutico , Proliferação de Células/efeitos dos fármacos , Transformação Celular Neoplásica/genética , Instabilidade Genômica/efeitos dos fármacos , Humanos , Neoplasias/patologia , Fosfatidilinositol 3-Quinases/genética , Transdução de Sinais/genética , Telomerase/efeitos dos fármacos , Telomerase/genética , Proteína Supressora de Tumor p53/genética
6.
Proc Natl Acad Sci U S A ; 109(39): E2595-604, 2012 Sep 25.
Artigo em Inglês | MEDLINE | ID: mdl-22923691

RESUMO

Breast cancer progression involves genetic changes and changes in the extracellular matrix (ECM). To test the importance of the ECM in tumor cell dissemination, we cultured epithelium from primary human breast carcinomas in different ECM gels. We used basement membrane gels to model the normal microenvironment and collagen I to model the stromal ECM. In basement membrane gels, malignant epithelium either was indolent or grew collectively, without protrusions. In collagen I, epithelium from the same tumor invaded with protrusions and disseminated cells. Importantly, collagen I induced a similar initial response of protrusions and dissemination in both normal and malignant mammary epithelium. However, dissemination of normal cells into collagen I was transient and ceased as laminin 111 localized to the basal surface, whereas dissemination of carcinoma cells was sustained throughout culture, and laminin 111 was not detected. Despite the large impact of ECM on migration strategy, transcriptome analysis of our 3D cultures revealed few ECM-dependent changes in RNA expression. However, we observed many differences between normal and malignant epithelium, including reduced expression of cell-adhesion genes in tumors. Therefore, we tested whether deletion of an adhesion gene could induce sustained dissemination of nontransformed cells into collagen I. We found that deletion of P-cadherin was sufficient for sustained dissemination, but exclusively into collagen I. Our data reveal that metastatic tumors preferentially disseminate in specific ECM microenvironments. Furthermore, these data suggest that breaks in the basement membrane could induce invasion and dissemination via the resulting direct contact between cancer cells and collagen I.


Assuntos
Neoplasias da Mama , Movimento Celular , Regulação Neoplásica da Expressão Gênica , Neoplasias Mamárias Animais , Microambiente Tumoral , Animais , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Feminino , Humanos , Neoplasias Mamárias Animais/genética , Neoplasias Mamárias Animais/metabolismo , Neoplasias Mamárias Animais/patologia , Camundongos , Invasividade Neoplásica
7.
Nat Genet ; 36(9): 984-8, 2004 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15300252

RESUMO

Transition through telomere crisis is thought to be a crucial event in the development of most breast carcinomas. Our goal in this study was to determine where this occurs in the context of histologically defined breast cancer progression. To this end, we assessed genome instability (using fluorescence in situ hybridization) and other features associated with telomere crisis in normal ductal epithelium, usual ductal hyperplasia, ductal carcinoma in situ and invasive cancer. We modeled this process in vitro by measuring these same features in human mammary epithelial cell cultures during ZNF217-mediated transition through telomere crisis and immortalization. Taken together, the data suggest that transition through telomere crisis and immortalization in breast cancer occurs during progression from usual ductal hyperplasia to ductal carcinoma in situ.


Assuntos
Neoplasias da Mama/genética , Instabilidade Cromossômica , Telômero/diagnóstico por imagem , Neoplasias da Mama/patologia , Carcinoma Ductal de Mama/genética , Carcinoma Ductal de Mama/patologia , Carcinoma Intraductal não Infiltrante/genética , Carcinoma Intraductal não Infiltrante/patologia , Progressão da Doença , Humanos , Hiperplasia/genética , Hiperplasia/patologia , Hibridização In Situ , Células Tumorais Cultivadas , Ultrassonografia
8.
Breast Cancer Res ; 13(3): 308, 2011 May 25.
Artigo em Inglês | MEDLINE | ID: mdl-21635707

RESUMO

Two recent reports identify ZNF703 as an oncogene driving selection of frequent chromosome 8p12 amplifications in luminal B breast tumors. The estrogen-responsive ZNF703 gene encodes a transcriptional cofactor that, when overexpressed, induces cell proliferation and interferes with transforming growth factor beta signaling. In MCF7 cells, increased ZNF703 expression results in activation of genes involved in stem cell self-renewal - while in primary human mammary epithelial cells, ZNF703 increases the ratio of luminal to basal progenitors. Expression of the murine homolog of ZNF703 reduces cell adhesion and promotes metastasis. ZNF703 overexpression thus alters regulation of proliferation and differentiation in luminal B tumors.


Assuntos
Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Animais , Adesão Celular/genética , Diferenciação Celular , Linhagem Celular Tumoral , Proliferação de Células , Cromossomos Humanos Par 8/genética , Feminino , Amplificação de Genes , Humanos , Camundongos , Metástase Neoplásica , Células-Tronco Neoplásicas/metabolismo , Transdução de Sinais/genética , Fatores de Transcrição/biossíntese
9.
Inorg Chem ; 50(1): 271-84, 2011 Jan 03.
Artigo em Inglês | MEDLINE | ID: mdl-21121684

RESUMO

The reactions of the breast cancer drug metabolite derivatives of tamoxifen, cis and trans-hydroxytamoxifen, cis-1 and trans-2, with [Cp*Rh(L)(3)](2+) complexes (L = H(2)O or MeOH), in CH(2)Cl(2) and CH(3)OH solvents, initially provided the kinetic η(1)-N complexes, cis-4 (OTf(-), CH(3)OH) and trans-5 (OTf(-), CH(3)OH), which underwent a novel, regioselective, intramolecular N-π rearrangement to give the cis and trans-η(6)-phenol substituted complexes, cis-6 and trans-7, via η(2)-N,O, η(1)-O, and ether aromatic ring η(6) intermediates. Recent density functional theory (DFT) calculations showed a preferred ground state for η(1)-N; η(2)-N,O; η(1)-O; and the η(6) complexes, including the prominent roles of the triflate anion (OTf(-)), and solvent molecules (CH(2)Cl(2) and CH(3)OH), and provided further steric, electronic, and thermodynamic data on the mechanism of the N-π rearrangement. The η(6) complex, cis-6, was shown to be an antagonist for ERα estrogen receptor binding, in a competition experiment with the female hormone, estradiol; therefore, computer docking studies of this biologically active complex at the estrogen receptors, ERα and ERß, also provided information on the binding modes and thermodynamic parameters, while bioassay results provided growth inhibition data on both hormone dependent and independent breast cancer cell lines.


Assuntos
Antineoplásicos Hormonais , Receptor alfa de Estrogênio , Receptor beta de Estrogênio , Compostos Organometálicos , Tamoxifeno/análogos & derivados , Antineoplásicos Hormonais/síntese química , Antineoplásicos Hormonais/farmacologia , Neoplasias da Mama/tratamento farmacológico , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Receptor alfa de Estrogênio/antagonistas & inibidores , Receptor alfa de Estrogênio/metabolismo , Receptor beta de Estrogênio/antagonistas & inibidores , Receptor beta de Estrogênio/metabolismo , Estrogênios/metabolismo , Feminino , Humanos , Modelos Moleculares , Compostos Organometálicos/síntese química , Compostos Organometálicos/farmacologia , Ligação Proteica , Ródio/química , Estereoisomerismo , Tamoxifeno/química , Tamoxifeno/farmacologia , Termodinâmica
10.
Cancers (Basel) ; 13(18)2021 Sep 08.
Artigo em Inglês | MEDLINE | ID: mdl-34572749

RESUMO

Reprogramming the tumor stroma is an emerging approach to circumventing the challenges of conventional cancer therapies. This strategy, however, is hampered by the lack of a specific molecular target. We previously reported that stromal fibroblasts (FBs) with high expression of CD36 could be utilized for this purpose. These studies are now expanded to identify the secreted factors responsible for tumor suppression. Methodologies included 3D colonies, fluorescent microscopy coupled with quantitative techniques, proteomics profiling, and bioinformatics analysis. The results indicated that the conditioned medium (CM) of the CD36+ FBs caused growth suppression via apoptosis in the triple-negative cell lines of MDA-MB-231, BT549, and Hs578T, but not in the ERBB2+ SKBR3. Following the proteomics and bioinformatic analysis of the CM of CD36+ versus CD36- FBs, we determined KLF10 as one of the transcription factors responsible for growth suppression. We also identified FBLN1, SLIT3, and PENK as active ligands, where their minimum effective concentrations were determined. Finally, in MDA-MB-231, we showed that a mixture of FBLN1, SLIT3, and PENK could induce an amount of growth suppression similar to the CM of CD36+ FBs. In conclusion, our findings suggest that these ligands, secreted by CD36+ FBs, can be targeted for breast cancer treatment.

11.
Breast Cancer Res ; 12(1): R11, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20146798

RESUMO

INTRODUCTION: Most human mammary epithelial cells (HMEC) cultured from histologically normal breast tissues enter a senescent state termed stasis after 5 to 20 population doublings. These senescent cells display increased size, contain senescence associated beta-galactosidase activity, and express cyclin-dependent kinase inhibitor, p16INK4A (CDKN2A; p16). However, HMEC grown in a serum-free medium, spontaneously yield, at low frequency, variant (v) HMEC that are capable of long-term growth and are susceptible to genomic instability. We investigated whether ionizing radiation, which increases breast cancer risk in women, affects the rate of vHMEC outgrowth. METHODS: Pre-stasis HMEC cultures were exposed to 5 to 200 cGy of sparsely (X- or gamma-rays) or densely (1 GeV/amu 56Fe) ionizing radiation. Proliferation (bromodeoxyuridine incorporation), senescence (senescence-associated beta-galactosidase activity), and p16 expression were assayed in subcultured irradiated or unirradiated populations four to six weeks following radiation exposure, when patches of vHMEC became apparent. Long-term growth potential and p16 promoter methylation in subsequent passages were also monitored. Agent-based modeling, incorporating a simple set of rules and underlying assumptions, was used to simulate vHMEC outgrowth and evaluate mechanistic hypotheses. RESULTS: Cultures derived from irradiated cells contained significantly more vHMEC, lacking senescence associated beta-galactosidase or p16 expression, than cultures derived from unirradiated cells. As expected, post-stasis vHMEC cultures derived from both unirradiated and irradiated cells exhibited more extensive methylation of the p16 gene than pre-stasis HMEC cultures. However, the extent of methylation of individual CpG sites in vHMEC samples did not correlate with passage number or treatment. Exposure to sparsely or densely ionizing radiation elicited similar increases in the numbers of vHMEC compared to unirradiated controls. Agent-based modeling indicated that radiation-induced premature senescence of normal HMEC most likely accelerated vHMEC outgrowth through alleviation of spatial constraints. Subsequent experiments using defined co-cultures of vHMEC and senescent cells supported this mechanism. CONCLUSIONS: Our studies indicate that ionizing radiation can promote the outgrowth of epigenetically altered cells with pre-malignant potential.


Assuntos
Mama/efeitos da radiação , Adolescente , Mama/patologia , Proliferação de Células/efeitos da radiação , Células Cultivadas , Relação Dose-Resposta à Radiação , Células Epiteliais/patologia , Células Epiteliais/efeitos da radiação , Feminino , Inativação Gênica , Genes p16 , Humanos , Pessoa de Meia-Idade , Modelos Biológicos
12.
Mol Cell Biol ; 26(21): 8159-72, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16940172

RESUMO

Numerous transcription factors recruit C-terminal binding protein (CtBP) corepressors. We show that the large zinc finger protein ZNF217 contacts CtBP. ZNF217 is encoded by an oncogene frequently amplified in tumors. ZNF217 contains a typical Pro-X-Asp-Leu-Ser (PXDLS) motif that binds in CtBP's PXDLS-binding cleft. However, ZNF217 also contains a second motif, Arg-Arg-Thr (RRT), that binds a separate surface on CtBP. The crystal structure of CtBP bound to an RRTGAPPAL peptide shows that it contacts a surface crevice distinct from the PXDLS binding cleft. Interestingly, both PXDLS and RRT motifs are also found in other zinc finger proteins, such as RIZ. Finally, we show that ZNF217 represses several promoters, including one from a known CtBP target gene, and mutations preventing ZNF217's contact with CtBP reduce repression. These results identify a new CtBP interaction motif and establish ZNF217 as a transcriptional repressor protein that functions, at least in part, by associating with CtBP.


Assuntos
Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/metabolismo , Fosfoproteínas/química , Fosfoproteínas/metabolismo , Conformação Proteica , Proteínas Repressoras/química , Proteínas Repressoras/metabolismo , Transativadores/metabolismo , Dedos de Zinco , Oxirredutases do Álcool , Sequência de Aminoácidos , Animais , Sítios de Ligação , Linhagem Celular , Proteínas Correpressoras , Cristalografia por Raios X , Proteínas de Ligação a DNA/genética , Regulação da Expressão Gênica , Humanos , Camundongos , Modelos Moleculares , Dados de Sequência Molecular , Mutação , Proteínas de Neoplasias/química , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Fosfoproteínas/genética , Regiões Promotoras Genéticas , Ligação Proteica , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Repressoras/genética , Alinhamento de Sequência , Transativadores/genética , Técnicas do Sistema de Duplo-Híbrido
13.
Biochim Biophys Acta ; 1775(2): 333-40, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17572303

RESUMO

Chromosome 20q13 is highly amplified in human cancers, including 20-30% of early stage human breast cancers. The amplification correlates with poor prognosis. Over-expression of the zinc-finger protein 217 (ZNF217), a candidate oncogene on 20q13.2, in cultured human mammary and ovarian epithelial cells can lead to their immortalization, indicating that selection for ZNF217 expression may drive 20q13 amplification during critical early stages of cancer progression. ZNF217 can also attenuate apoptotic signals resulting from exposure to doxorubicin, suggesting that ZNF217 expression may also be involved in resistance to chemotherapy. Recent findings indicate that ZNF217 binds specific DNA sequences, recruits the co-repressor C-terminal binding protein (CtBP), and represses the transcription of a variety of genes. Inappropriate expression of ZNF217 may lead to aberrant down-regulation of genes involved in limiting the proliferation, survival, and/or invasiveness of cancer cells. Better understanding of ZNF217 and its associated pathways may provide new targets for therapeutic intervention in human cancers.


Assuntos
Amplificação de Genes , Neoplasias/genética , Transativadores/genética , Progressão da Doença , Humanos , Proteínas de Neoplasias/química , Proteínas de Neoplasias/genética , Neoplasias/patologia , Neoplasias/fisiopatologia , Oncogenes
14.
Cancer Res ; 66(14): 7095-102, 2006 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-16849555

RESUMO

Nonmalignant human mammary epithelial cells (HMEC) seeded in laminin-rich extracellular matrix (lrECM) form polarized acini and, in doing so, transit from a disorganized proliferating state to an organized growth-arrested state. We hypothesized that the gene expression pattern of organized and growth-arrested HMECs would share similarities with breast tumors with good prognoses. Using Affymetrix HG-U133A microarrays, we analyzed the expression of 22,283 gene transcripts in 184 (finite life span) and HMT3522 S1 (immortal nonmalignant) HMECs on successive days after seeding in a lrECM assay. Both HMECs underwent growth arrest in G0-G1 and differentiated into polarized acini between days 5 and 7. We identified gene expression changes with the same temporal pattern in both lines and examined the expression of these genes in a previously published panel of microarray data for 295 breast cancer samples. We show that genes that are significantly lower in the organized, growth-arrested HMEC than in their proliferating counterparts can be used to classify breast cancer patients into poor and good prognosis groups with high accuracy. This study represents a novel unsupervised approach to identifying breast cancer markers that may be of use clinically.


Assuntos
Neoplasias da Mama/genética , Glândulas Mamárias Humanas/fisiologia , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Processos de Crescimento Celular/fisiologia , Células Cultivadas , Perfilação da Expressão Gênica , Humanos , Glândulas Mamárias Humanas/citologia , Glândulas Mamárias Humanas/metabolismo , Prognóstico
15.
SLAS Discov ; 22(3): 298-308, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-27872202

RESUMO

Malignant melanomas often arise from nevi, which result from initial oncogene-induced hyperproliferation of melanocytes that are maintained in a CDKN2A/p16-mediated senescent state. Thus, genes that can bypass this senescence barrier are likely to contribute to melanoma development. We have performed a gain-of-function screen of 17,030 lentivirally expressed human open reading frames (ORFs) in a melanoma cell line containing an inducible p16 construct to identify such genes. Genes known to bypass p16-induced senescence arrest, including the human papilloma virus 18 E7 gene ( HPV18E7), and genes such as the p16-binding CDK6 with expected functions, as well as panel of novel genes, were identified, including high-mobility group box (HMGB) proteins. A number of these were further validated in two other models of p16-induced senescence. Tissue immunohistochemistry demonstrated higher levels of CDK6 in primary melanomas compared with normal skin and nevi. Reduction of CDK6 levels drove melanoma cells expressing functional p16 into senescence, demonstrating its contribution to bypass senescence.


Assuntos
Pontos de Checagem do Ciclo Celular , Quinase 6 Dependente de Ciclina/genética , Inibidor p16 de Quinase Dependente de Ciclina/genética , Regulação Neoplásica da Expressão Gênica , Ensaios de Triagem em Larga Escala , Melanócitos/metabolismo , Linhagem Celular Tumoral , Senescência Celular , Quinase 6 Dependente de Ciclina/metabolismo , Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Biblioteca Gênica , Vetores Genéticos/química , Vetores Genéticos/metabolismo , Genoma Humano , Células HEK293 , Proteínas HMGB/genética , Proteínas HMGB/metabolismo , Humanos , Lentivirus/genética , Lentivirus/metabolismo , Melanócitos/patologia , Melanoma/genética , Melanoma/metabolismo , Melanoma/patologia , Nevo/genética , Nevo/metabolismo , Nevo/patologia , Proteínas Oncogênicas Virais/genética , Proteínas Oncogênicas Virais/metabolismo , Fases de Leitura Aberta , Neoplasias Cutâneas/genética , Neoplasias Cutâneas/metabolismo , Neoplasias Cutâneas/patologia
16.
Oncogene ; 24(20): 3369-76, 2005 May 05.
Artigo em Inglês | MEDLINE | ID: mdl-15735711

RESUMO

We have used cultured human mammary epithelial cells (HMEC) and breast tumor-derived lines to gain information on defects that occur during breast cancer progression. HMEC immortalized by a variety of agents (the chemical carcinogen benzo(a)pyrene, oncogenes c-myc and ZNF217, and/or dominant negative p53 genetic suppressor element GSE22) displayed marked upregulation (10-15 fold) of the telomere-binding protein, TRF2. Upregulation of TRF2 protein was apparently due to differences in post-transcriptional regulation, as mRNA levels remained comparable in finite lifespan and immortal HMEC. TRF2 protein was not upregulated by the oncogenic agents alone in the absence of immortalization, nor by expression of exogenously introduced hTERT genes. We found TRF2 levels to be at least twofold higher than in control cells in 11/15 breast tumor cell lines, suggesting that elevated TRF2 levels are a frequent occurrence during the transformation of breast tumor cells in vivo. The dispersed distribution of TRF2 throughout the nuclei in some immortalized and tumor-derived cells indicated that not all the TRF2 was associated with telomeres in these cells. The process responsible for accumulation of TRF2 in immortalized HMEC and breast tumor-derived cell lines may promote tumorigenesis by contributing to the cells' ability to maintain an indefinite lifespan.


Assuntos
Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Proteína 2 de Ligação a Repetições Teloméricas/biossíntese , Mama/patologia , Linhagem Celular Transformada , Linhagem Celular Tumoral , Proliferação de Células , Senescência Celular , Proteínas de Ligação a DNA , Progressão da Doença , Genes Dominantes , Humanos , Imuno-Histoquímica , RNA Mensageiro/metabolismo , Telomerase/metabolismo , Telômero/metabolismo , Telômero/ultraestrutura , Proteína 2 de Ligação a Repetições Teloméricas/metabolismo , Fatores de Tempo , Proteína Supressora de Tumor p53 , Regulação para Cima
17.
Environ Health Perspect ; 124(5): 563-9, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-26502914

RESUMO

BACKGROUND: Xenoestrogens are synthetic compounds that mimic endogenous estrogens by binding to and activating estrogen receptors. Exposure to estrogens and to some xenoestrogens has been associated with cell proliferation and an increased risk of breast cancer. Despite evidence of estrogenicity, parabens are among the most widely used xenoestrogens in cosmetics and personal-care products and are generally considered safe. However, previous cell-based studies with parabens do not take into account the signaling cross-talk between estrogen receptor α (ERα) and the human epidermal growth factor receptor (HER) family. OBJECTIVES: We investigated the hypothesis that the potency of parabens can be increased with HER ligands, such as heregulin (HRG). METHODS: The effects of HER ligands on paraben activation of c-Myc expression and cell proliferation were determined by real-time polymerase chain reaction, Western blots, flow cytometry, and chromatin immunoprecipitation assays in ERα- and HER2-positive human BT-474 breast cancer cells. RESULTS: Butylparaben (BP) and HRG produced a synergistic increase in c-Myc mRNA and protein levels in BT-474 cells. Estrogen receptor antagonists blocked the synergistic increase in c-Myc protein levels. The combination of BP and HRG also stimulated proliferation of BT-474 cells compared with the effects of BP alone. HRG decreased the dose required for BP-mediated stimulation of c-Myc mRNA expression and cell proliferation. HRG caused the phosphorylation of serine 167 in ERα. BP and HRG produced a synergistic increase in ERα recruitment to the c-Myc gene. CONCLUSION: Our results show that HER ligands enhanced the potency of BP to stimulate oncogene expression and breast cancer cell proliferation in vitro via ERα, suggesting that parabens might be active at exposure levels not previously considered toxicologically relevant from studies testing their effects in isolation. CITATION: Pan S, Yuan C, Tagmount A, Rudel RA, Ackerman JM, Yaswen P, Vulpe CD, Leitman DC. 2016. Parabens and human epidermal growth factor receptor ligand cross-talk in breast cancer cells. Environ Health Perspect 124:563-569; http://dx.doi.org/10.1289/ehp.1409200.


Assuntos
Estrogênios/toxicidade , Parabenos/toxicidade , Linhagem Celular Tumoral , Receptores ErbB/metabolismo , Receptor alfa de Estrogênio/metabolismo , Genes myc , Humanos , Neuregulina-1/metabolismo , Proteínas Proto-Oncogênicas c-myc/genética , Proteínas Proto-Oncogênicas c-myc/metabolismo
18.
Nat Med ; 22(11): 1321-1329, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27775705

RESUMO

Triple-negative breast cancer (TNBC), in which cells lack expression of the estrogen receptor (ER), the progesterone receptor (PR) and the ERBB2 (also known as HER2) receptor, is the breast cancer subtype with the poorest outcome. No targeted therapy is available against this subtype of cancer owing to a lack of validated molecular targets. We previously reported that signaling involving MYC-an essential, pleiotropic transcription factor that regulates the expression of hundreds of genes-is disproportionally higher in triple-negative (TN) tumors than in receptor-positive (RP) tumors. Direct inhibition of the oncogenic transcriptional activity of MYC has been challenging to achieve. Here, by conducting a shRNA screen targeting the kinome, we identified PIM1, a non-essential serine-threonine kinase, in a synthetic lethal interaction with MYC. PIM1 expression was higher in TN tumors than in RP tumors and was associated with poor prognosis in patients with hormone- and HER2-negative tumors. Small-molecule PIM kinase inhibitors halted the growth of human TN tumors with elevated MYC expression in patient-derived tumor xenograft (PDX) and MYC-driven transgenic mouse models of breast cancer by inhibiting the oncogenic transcriptional activity of MYC and restoring the function of the endogenous cell cycle inhibitor, p27. Our findings warrant clinical evaluation of PIM kinase inhibitors in patients with TN tumors that have elevated MYC expression.


Assuntos
Carcinoma Ductal de Mama/metabolismo , Neoplasias Mamárias Experimentais/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas c-myc/metabolismo , Proteínas Proto-Oncogênicas c-pim-1/antagonistas & inibidores , Neoplasias de Mama Triplo Negativas/metabolismo , Animais , Western Blotting , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Feminino , Humanos , Marcação In Situ das Extremidades Cortadas , Neoplasias Mamárias Experimentais/tratamento farmacológico , Neoplasias Mamárias Experimentais/genética , Camundongos Transgênicos , Microscopia de Fluorescência , Prognóstico , Proteínas Proto-Oncogênicas c-myc/genética , Proteínas Proto-Oncogênicas c-pim-1/metabolismo , RNA Interferente Pequeno , Reação em Cadeia da Polimerase em Tempo Real , Receptores de Estrogênio/metabolismo , Receptores de Progesterona/metabolismo , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Ensaios Antitumorais Modelo de Xenoenxerto
19.
Oncogene ; 21(41): 6328-39, 2002 Sep 12.
Artigo em Inglês | MEDLINE | ID: mdl-12214273

RESUMO

Using an estrogen-inducible retroviral system, we demonstrate that oncogenic Raf-1 induces growth arrest and morphological changes in finite lifespan human mammary epithelial cells (HMEC). This arrest does not rely on expression of the cyclin-dependent kinase inhibitor (CKI) p16(INK4a), nor on changes in expression of the CKIs p21(Cip1), p14(ARF), p27(Kip1) or p57(Kip2). The Raf-induced arrest is independent of viral oncogene mediated inactivation of p53 and pRB, or c-myc overexpression. Flow cytometric analysis demonstrates that cells arrest in both G1 and G2. The Raf-induced arrest is mitigated or eliminated in some immortally transformed HMEC. Immortal HMEC that have both overcome replicative senescence and undergone the recently described conversion process maintain growth in the presence of transduced oncogenic Raf-1; they also gain EGF-independent growth and a low frequency of anchorage-independent growth. However, HMEC that have overcome replicative senescence but have not undergone conversion and HMEC immortalized by transduction with the catalytic subunit of telomerase, hTERT, remain severely growth arrested. These results indicate that the molecular mechanisms responsible for the Raf-1-induced growth arrest may vary among different finite lifespan cell types, and that in HMEC, this mechanism is altered during the conversion process, rather than as a direct consequence of overcoming senescence or expressing hTERT.


Assuntos
Ciclo Celular/genética , Transformação Celular Neoplásica , Inibidor p16 de Quinase Dependente de Ciclina/genética , Células Epiteliais/patologia , Proteínas Proto-Oncogênicas c-raf/genética , Telomerase/genética , Mama/patologia , Transformação Celular Neoplásica/genética , Células Cultivadas , Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , Proteínas de Ligação a DNA , Células Epiteliais/metabolismo , Conversão Gênica
20.
Oncogene ; 22(34): 5238-51, 2003 Aug 14.
Artigo em Inglês | MEDLINE | ID: mdl-12917625

RESUMO

We describe novel effects of p53 loss on immortal transformation, based upon comparison of immortally transformed human mammary epithelial cell (HMEC) lines lacking functional p53 with closely related p53(+) lines. Our previous studies of p53(+) immortal HMEC lines indicated that overcoming the stringent replicative senescence step associated with critically short telomeres (agonescence), produced indefinite lifespan lines that maintained growth without immediately expressing telomerase activity. These telomerase(-) 'conditionally immortal' HMEC underwent an additional step, termed conversion, to become fully immortal telomerase(+) lines with uniform good growth. The very gradual conversion process was associated with slow heterogeneous growth and high expression of the cyclin-dependent kinase inhibitor p57(Kip2). We now show that p53 suppresses telomerase activity and is necessary for the p57 expression in early passage p53(+) conditionally immortal HMEC lines, and that p53(-/-) lines exhibit telomerase reactivation and attain full immortality much more rapidly. A p53-inhibiting genetic suppressor element introduced into early passages of a conditionally immortal telomerase(-) p53(+) HMEC line led to rapid induction of hTERT mRNA, expression of telomerase activity, loss of p57 expression, and quick attainment of uniform good growth. These studies indicate that derangements in p53 function may impact malignant progression through direct effects on the conversion process, a potentially rate-limiting step in HMEC acquisition of uniform unlimited growth potential. These studies also provide evidence that the function of p53 in suppression of telomerase activity is separable from its cell cycle checkpoint function.


Assuntos
Neoplasias da Mama/enzimologia , Mama/enzimologia , Transformação Celular Neoplásica/metabolismo , Telomerase/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Linhagem Celular Transformada , Transformação Celular Neoplásica/genética , Feminino , Deleção de Genes , Humanos , Proteína Supressora de Tumor p53/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA