Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
bioRxiv ; 2024 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-38559183

RESUMEN

Circulating Tumor Cells (CTCs), interrogated by sampling blood from patients with cancer, contain multiple analytes, including intact RNA, high molecular weight DNA, proteins, and metabolic markers. However, the clinical utility of tumor cell-based liquid biopsy has been limited since CTCs are very rare, and current technologies cannot process the blood volumes required to isolate a sufficient number of tumor cells for in-depth assays. We previously described a high-throughput microfluidic prototype utilizing high-flow channels and amplification of cell sorting forces through magnetic lenses. Here, we apply this technology to analyze patient-derived leukapheresis products, interrogating a mean blood volume of 5.83 liters from patients with metastatic cancer, with a median of 2,799 CTCs purified per patient. Isolation of many CTCs from individual patients enables characterization of their morphological and molecular heterogeneity, including cell and nuclear size and RNA expression. It also allows robust detection of gene copy number variation, a definitive cancer marker with potential diagnostic applications. High-volume microfluidic enrichment of CTCs constitutes a new dimension in liquid biopsies.

3.
JCO Precis Oncol ; 7: e2200532, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-37141550

RESUMEN

PURPOSE: For patients with hormone receptor-positive (HR+), human epidermal growth factor receptor 2-negative (HER2-) metastatic breast cancer (MBC), first-line treatment is endocrine therapy (ET) plus cyclin-dependent kinase 4/6 inhibition (CDK4/6i). After disease progression, which often comes with ESR1 resistance mutations (ESR1-MUT), which therapies to use next and for which patients are open questions. An active area of exploration is treatment with further CDK4/6i, particularly abemaciclib, which has distinct pharmacokinetic and pharmacodynamic properties compared with the other approved CDK4/6 inhibitors, palbociclib and ribociclib. We investigated a gene panel to prognosticate abemaciclib susceptibility in patients with ESR1-MUT MBC after palbociclib progression. METHODS: We examined a multicenter retrospective cohort of patients with ESR1-MUT MBC who received abemaciclib after disease progression on ET plus palbociclib. We generated a panel of CDK4/6i resistance genes and compared abemaciclib progression-free survival (PFS) in patients without versus with mutations in this panel (CDKi-R[-] v CDKi-R[+]). We studied how ESR1-MUT and CDKi-R mutations affect abemaciclib sensitivity of immortalized breast cancer cells and patient-derived circulating tumor cell lines in culture. RESULTS: In ESR1-MUT MBC with disease progression on ET plus palbociclib, the median PFS was 7.0 months for CDKi-R(-) (n = 17) versus 3.5 months for CDKi-R(+) (n = 11), with a hazard ratio of 2.8 (P = .03). In vitro, CDKi-R alterations but not ESR1-MUT induced abemaciclib resistance in immortalized breast cancer cells and were associated with resistance in circulating tumor cells. CONCLUSION: For ESR1-MUT MBC with resistance to ET and palbociclib, PFS on abemaciclib is longer for patients with CDKi-R(-) than CDKi-R(+). Although a small and retrospective data set, this is the first demonstration of a genomic panel associated with abemaciclib sensitivity in the postpalbociclib setting. Future directions include testing and improving this panel in additional data sets, to guide therapy selection for patients with HR+/HER2- MBC.


Asunto(s)
Neoplasias de la Mama , Humanos , Femenino , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/genética , Quinasa 4 Dependiente de la Ciclina/genética , Estudios Retrospectivos , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico , Progresión de la Enfermedad
4.
medRxiv ; 2023 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-36798390

RESUMEN

Innovative service delivery models are needed to increase access to genetics specialists. Electronic consultation (e-Consult) programs can connect clinicians with specialists. At Massachusetts General Hospital, an e-Consult service was created to address genomics-related questions. In its first year, the e-Consult service triaged 153 requests and completed 122 in an average of 3.2 days. Of the 95 e-Consults with actionable recommendations, there was documentation that most ordering clinicians followed through (82%). A variety of providers used the service, although the majority (77%) were generalists. E-Consult models should be considered as one way to increase access to genetics care.

5.
Proc Natl Acad Sci U S A ; 119(43): e2209563119, 2022 10 25.
Artículo en Inglés | MEDLINE | ID: mdl-36256815

RESUMEN

The successful application of antibody-based therapeutics in either primary or metastatic cancer depends upon the selection of rare cell surface epitopes that distinguish cancer cells from surrounding normal epithelial cells. By contrast, as circulating tumor cells (CTCs) transit through the bloodstream, they are surrounded by hematopoietic cells with dramatically distinct cell surface proteins, greatly expanding the number of targetable epitopes. Here, we show that an antibody (23C6) against cadherin proteins effectively suppresses blood-borne metastasis in mouse isogenic and xenograft models of triple negative breast and pancreatic cancers. The 23C6 antibody is remarkable in that it recognizes both the epithelial E-cadherin (CDH1) and mesenchymal OB-cadherin (CDH11), thus overcoming considerable heterogeneity across tumor cells. Despite its efficacy against single cells in circulation, the antibody does not suppress primary tumor formation, nor does it elicit detectable toxicity in normal epithelial organs, where cadherins may be engaged within intercellular junctions and hence inaccessible for antibody binding. Antibody-mediated suppression of metastasis is comparable in matched immunocompetent and immunodeficient mouse models. Together, these studies raise the possibility of antibody targeting CTCs within the vasculature, thereby suppressing blood-borne metastasis.


Asunto(s)
Neoplasias de la Mama , Células Neoplásicas Circulantes , Neoplasias Pancreáticas , Humanos , Animales , Ratones , Femenino , Transición Epitelial-Mesenquimal , Línea Celular Tumoral , Cadherinas/metabolismo , Células Neoplásicas Circulantes/patología , Procesos Neoplásicos , Neoplasias Pancreáticas/tratamiento farmacológico , Ratones Desnudos , Ratones SCID , Epítopos , Neoplasias de la Mama/tratamiento farmacológico , Metástasis de la Neoplasia , Neoplasias Pancreáticas
6.
Cancer Res ; 82(6): 1084-1097, 2022 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-35045985

RESUMEN

Cancer therapy often results in heterogeneous responses in different metastatic lesions in the same patient. Inter- and intratumor heterogeneity in signaling within various tumor compartments and its impact on therapy are not well characterized due to the limited sensitivity of single-cell proteomic approaches. To overcome this barrier, we applied single-cell mass cytometry with a customized 26-antibody panel to PTEN-deleted orthotopic prostate cancer xenograft models to measure the evolution of kinase activities in different tumor compartments during metastasis or drug treatment. Compared with primary tumors and circulating tumor cells (CTC), bone metastases, but not lung and liver metastases, exhibited elevated PI3K/mTOR signaling and overexpressed receptor tyrosine kinases (RTK) including c-MET protein. Suppression of c-MET impaired tumor growth in the bone. Intratumoral heterogeneity within tumor compartments also arose from highly proliferative EpCAM-high epithelial cells with increased PI3K and mTOR kinase activities coexisting with poorly proliferating EpCAM-low mesenchymal populations with reduced kinase activities; these findings were recapitulated in epithelial and mesenchymal CTC populations in patients with metastatic prostate and breast cancer. Increased kinase activity in EpCAM-high cells rendered them more sensitive to PI3K/mTOR inhibition, and drug-resistant EpCAM-low populations with reduced kinase activity emerged over time. Taken together, single-cell proteomics indicate that microenvironment- and cell state-dependent activation of kinase networks create heterogeneity and differential drug sensitivity among and within tumor populations across different sites, defining a new paradigm of drug responses to kinase inhibitors. SIGNIFICANCE: Single-cell mass cytometry analyses provide insights into the differences in kinase activities across tumor compartments and cell states, which contribute to heterogeneous responses to targeted therapies.


Asunto(s)
Neoplasias de la Próstata , Proteómica , Animales , Línea Celular Tumoral , Molécula de Adhesión Celular Epitelial , Humanos , Masculino , Fosfatidilinositol 3-Quinasas/metabolismo , Neoplasias de la Próstata/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/farmacología , Serina-Treonina Quinasas TOR/metabolismo , Microambiente Tumoral
7.
Cancer Res ; 81(3): 517-524, 2021 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-33479028

RESUMEN

Deregulation of the mRNA translational process has been observed during tumorigenesis. However, recent findings have shown that deregulation of translation also contributes specifically to cancer cell spread. During metastasis, cancer cells undergo changes in cellular state, permitting the acquisition of features necessary for cell survival, dissemination, and outgrowth. In addition, metastatic cells respond to external cues, allowing for their persistence under significant cellular and microenvironmental stresses. Recent work has revealed the importance of mRNA translation to these dynamic changes, including regulation of cell states through epithelial-to-mesenchymal transition and tumor dormancy and as a response to external stresses such as hypoxia and immune surveillance. In this review, we focus on examples of altered translation underlying these phenotypic changes and responses to external cues and explore how they contribute to metastatic progression. We also highlight the therapeutic opportunities presented by aberrant mRNA translation, suggesting novel ways to target metastatic tumor cells.


Asunto(s)
Transición Epitelial-Mesenquimal/fisiología , Metástasis de la Neoplasia/genética , Biosíntesis de Proteínas/fisiología , Carcinogénesis/metabolismo , Movimiento Celular , Supervivencia Celular/fisiología , Humanos , Metástasis de la Neoplasia/patología , Metástasis de la Neoplasia/terapia , Proteínas de Neoplasias/biosíntesis , Neoplasias/terapia , Neovascularización Patológica/etiología , Fenotipo , Serina-Treonina Quinasas TOR/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Escape del Tumor/fisiología , Hipoxia Tumoral/fisiología , Microambiente Tumoral/fisiología
8.
Nat Commun ; 11(1): 6311, 2020 12 09.
Artículo en Inglés | MEDLINE | ID: mdl-33298946

RESUMEN

Blood-borne metastasis to the brain is a major complication of breast cancer, but cellular pathways that enable cancer cells to selectively grow in the brain microenvironment are poorly understood. We find that cultured circulating tumor cells (CTCs), derived from blood samples of women with advanced breast cancer and directly inoculated into the mouse frontal lobe, exhibit striking differences in proliferative potential in the brain. Derivative cell lines generated by serial intracranial injections acquire selectively increased proliferative competency in the brain, with reduced orthotopic tumor growth. Increased Hypoxia Inducible Factor 1A (HIF1A)-associated signaling correlates with enhanced proliferation in the brain, and shRNA-mediated suppression of HIF1A or drug inhibition of HIF-associated glycolytic pathways selectively impairs brain tumor growth while minimally impacting mammary tumor growth. In clinical specimens, brain metastases have elevated HIF1A protein expression, compared with matched primary breast tumors, and in patients with brain metastases, hypoxic signaling within CTCs predicts decreased overall survival. The selective activation of hypoxic signaling by metastatic breast cancer in the brain may have therapeutic implications.


Asunto(s)
Neoplasias Encefálicas/secundario , Encéfalo/patología , Neoplasias de la Mama/patología , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Células Neoplásicas Circulantes/metabolismo , Animales , Neoplasias Encefálicas/sangre , Neoplasias Encefálicas/mortalidad , Neoplasias de la Mama/sangre , Neoplasias de la Mama/mortalidad , Hipoxia de la Célula , Proliferación Celular , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/antagonistas & inhibidores , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Glándulas Mamarias Animales/patología , Metabolómica , Ratones , ARN Interferente Pequeño/metabolismo , RNA-Seq , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Esferoides Celulares , Técnicas Estereotáxicas , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
9.
Science ; 367(6485): 1468-1473, 2020 03 27.
Artículo en Inglés | MEDLINE | ID: mdl-32029688

RESUMEN

Circulating tumor cells (CTCs) are shed into the bloodstream from primary tumors, but only a small subset of these cells generates metastases. We conducted an in vivo genome-wide CRISPR activation screen in CTCs from breast cancer patients to identify genes that promote distant metastasis in mice. Genes coding for ribosomal proteins and regulators of translation were enriched in this screen. Overexpression of RPL15, which encodes a component of the large ribosomal subunit, increased metastatic growth in multiple organs and selectively enhanced translation of other ribosomal proteins and cell cycle regulators. RNA sequencing of freshly isolated CTCs from breast cancer patients revealed a subset with strong ribosome and protein synthesis signatures; these CTCs expressed proliferation and epithelial markers and correlated with poor clinical outcome. Therapies targeting this aggressive subset of CTCs may merit exploration as potential suppressors of metastatic progression.


Asunto(s)
Neoplasias de la Mama/patología , Metástasis de la Neoplasia , Células Neoplásicas Circulantes/patología , Proteínas Ribosómicas/genética , Animales , Neoplasias de la Mama/genética , Sistemas CRISPR-Cas , Línea Celular Tumoral , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Ratones , Trasplante de Neoplasias , Análisis de Secuencia de ARN
11.
Nature ; 554(7692): 308-309, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-32094721
12.
Genes Dev ; 31(18): 1827-1840, 2017 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-29051388

RESUMEN

Advances in the enrichment and analysis of rare cells from the bloodstream have allowed for detection and characterization of circulating tumor cells (CTCs) from patients with cancer. The analysis of CTCs has provided significant insight into the metastatic process. Studies on the biology of CTCs have begun to elucidate the molecular mechanisms of CTC generation, intravasation, survival, interactions with components of the blood, extravasation, and colonization of distant organs. Additionally, the study of CTCs has exposed dramatic intrapatient and interpatient heterogeneity and their evolution over time. In this review, we focus on the current knowledge of CTC biology and the potential clinical implications.


Asunto(s)
Metástasis de la Neoplasia/patología , Células Neoplásicas Circulantes/patología , Animales , Plaquetas/fisiología , Comunicación Celular , Plasticidad de la Célula , Separación Celular , Heterogeneidad Genética , Humanos , Ratones , Mutación , Células Neoplásicas Circulantes/metabolismo , Especies Reactivas de Oxígeno/metabolismo
13.
Mol Oncol ; 11(7): 770-780, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28544498

RESUMEN

Metastasis of epithelial cancer cells to distant sites is a particularly critical stage of cancer progression that typically marks the incurability of the disease. It is governed by a complex series of events including invasion and intravasation of tumor cells into the stroma and blood, respectively. Epithelial-to-mesenchymal transition (EMT), a phenotypic change marked by the loss of epithelial characteristics and the acquisition of invasive mesenchymal properties, is implicated in the dissemination of tumor cells. Circulating tumor cells (CTCs), the precursors of metastasis, can be used to interrogate the contribution of EMT in metastasis and therapeutic responses. The analysis of these CTCs and in particular the presence of inter- and intrapatient heterogeneity for markers of EMT has provided new insights into the metastatic process. This review will focus on epithelial-mesenchymal plasticity in CTCs and its potential clinical implications.


Asunto(s)
Transición Epitelial-Mesenquimal , Células Neoplásicas Circulantes/metabolismo , Células Madre Neoplásicas/metabolismo , Animales , Humanos , Células Neoplásicas Circulantes/patología , Células Madre Neoplásicas/patología
14.
JAMA Neurol ; 72(9): 1061-5, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26213984

RESUMEN

A man in his 40s presented with 1 month of worsening confusion, fatigue, and headache. Results from laboratory analyses were notable for a complete white blood cell count of 17 000/µL (31% blast cells), a platelet count of 76 000/µL, and a hemoglobin level of 16.6 g/dL. Imaging studies revealed a large mixed-attenuation subdural collection in the right frontal region with prominent mass effect. The patient underwent an emergency neurosurgical procedure. The differential diagnosis, pathologic findings, and diagnosis are discussed.


Asunto(s)
Cefalea/etiología , Letargia/etiología , Leucemia/complicaciones , Leucemia/diagnóstico , Trastornos Mentales/etiología , Adulto , Antineoplásicos Hormonales/uso terapéutico , Craneotomía/métodos , ADN Nucleotidilexotransferasa/metabolismo , Dexametasona/uso terapéutico , Cefalea/complicaciones , Cefalea/terapia , Humanos , Letargia/complicaciones , Letargia/terapia , Leucemia/terapia , Leucocitos Mononucleares/patología , Levetiracetam , Imagen por Resonancia Magnética , Masculino , Trastornos Mentales/complicaciones , Trastornos Mentales/terapia , Nootrópicos/uso terapéutico , Piracetam/análogos & derivados , Piracetam/uso terapéutico , Tomografía Computarizada por Rayos X
15.
Breast Cancer Res ; 14(4): R100, 2012 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-22765220

RESUMEN

INTRODUCTION: Mammary-specific overexpression of Six1 in mice induces tumors that resemble human breast cancer, some having undergone epithelial to mesenchymal transition (EMT) and exhibiting stem/progenitor cell features. Six1 overexpression in human breast cancer cells promotes EMT and metastatic dissemination. We hypothesized that Six1 plays a role in the tumor initiating cell (TIC) population specifically in certain subtypes of breast cancer, and that by understanding its mechanism of action, we could potentially develop new means to target TICs. METHODS: We examined gene expression datasets to determine the breast cancer subtypes with Six1 overexpression, and then examined its expression in the CD24low/CD44+ putative TIC population in human luminal breast cancers xenografted through mice and in luminal breast cancer cell lines. Six1 overexpression, or knockdown, was performed in different systems to examine how Six1 levels affect TIC characteristics, using gene expression and flow cytometric analysis, tumorsphere assays, and in vivo TIC assays in immunocompromised and immune-competent mice. We examined the molecular pathways by which Six1 influences TICs using genetic/inhibitor approaches in vitro and in vivo. Finally, we examined the expression of Six1 and phosphorylated extracellular signal-regulated kinase (p-ERK) in human breast cancers. RESULTS: High levels of Six1 are associated with adverse outcomes in luminal breast cancers, particularly the luminal B subtype. Six1 levels are enriched in the CD24low/CD44+ TIC population in human luminal breast cancers xenografted through mice, and in tumorsphere cultures in MCF7 and T47D luminal breast cancer cells. When overexpressed in MCF7 cells, Six1expands the TIC population through activation of transforming growth factor-beta (TGF-ß) and mitogen activated protein kinase (MEK)/ERK signaling. Inhibition of ERK signaling in MCF7-Six1 cells with MEK1/2 inhibitors, U0126 and AZD6244, restores the TIC population of luminal breast cancer cells back to that observed in control cells. Administration of AZD6244 dramatically inhibits tumor formation efficiency and metastasis in cells that express high levels of Six1 ectopically or endogenously. Finally, we demonstrate that Six1 significantly correlates with phosphorylated ERK in human breast cancers. CONCLUSIONS: Six1 plays an important role in the TIC population in luminal breast cancers and induces a TIC phenotype by enhancing both TGF-ß and ERK signaling. MEK1/2 kinase inhibitors are potential candidates for targeting TICs in breast tumors.


Asunto(s)
Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Proteínas de Homeodominio/genética , Células Madre Neoplásicas/metabolismo , Transducción de Señal , Factor de Crecimiento Transformador beta/metabolismo , Animales , Neoplasias de la Mama/mortalidad , Neoplasias de la Mama/patología , Línea Celular Tumoral , Análisis por Conglomerados , Modelos Animales de Enfermedad , Femenino , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Xenoinjertos , Humanos , Ratones , Pronóstico
16.
J Clin Invest ; 122(5): 1895-906, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22466647

RESUMEN

An association between lymph node metastasis and poor prognosis in breast cancer was observed decades ago. However, the mechanisms by which tumor cells infiltrate the lymphatic system are not completely understood. Recently, it has been proposed that the lymphatic system has an active role in metastatic dissemination and that tumor-secreted growth factors stimulate lymphangiogenesis. We therefore investigated whether SIX1, a homeodomain-containing transcription factor previously associated in breast cancer with lymph node positivity, was involved in lymphangiogenesis and lymphatic metastasis. In a model in which human breast cancer cells were injected into immune-compromised mice, we found that SIX1 expression promoted peritumoral and intratumoral lymphangiogenesis, lymphatic invasion, and distant metastasis of breast cancer cells. SIX1 induced transcription of the prolymphangiogenic factor VEGF-C, and this was required for lymphangiogenesis and lymphatic metastasis. Using a mouse mammary carcinoma model, we found that VEGF-C was not sufficient to mediate all the metastatic effects of SIX1, indicating that SIX1 acts through additional, VEGF-C-independent pathways. Finally, we verified the clinical significance of this prometastatic SIX1/VEGF-C axis by demonstrating coexpression of SIX1 and VEGF-C in human breast cancer. These data define a critical role for SIX1 in lymphatic dissemination of breast cancer cells, providing a direct mechanistic explanation for how VEGF-C expression is upregulated in breast cancer, resulting in lymphangiogenesis and metastasis.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Proteínas de Homeodominio/fisiología , Neoplasias Pulmonares/secundario , Linfangiogénesis , Neoplasias Mamarias Experimentales/patología , Factor C de Crecimiento Endotelial Vascular/genética , Animales , Línea Celular Tumoral , Femenino , Perfilación de la Expresión Génica , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Metástasis Linfática , Neoplasias Mamarias Experimentales/metabolismo , Neoplasias Mamarias Experimentales/fisiopatología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos NOD , Ratones Desnudos , Ratones SCID , Trasplante de Neoplasias , Análisis de Secuencia por Matrices de Oligonucleótidos , Transcripción Genética , Carga Tumoral , Regulación hacia Arriba , Factor C de Crecimiento Endotelial Vascular/metabolismo
17.
Cancer Res ; 70(24): 10371-80, 2010 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-21056993

RESUMEN

The Six1 homeodomain protein is a developmental transcription factor that has been implicated in tumor onset and progression. Our recent work shows that Six1 overexpression in human breast cancer cell lines is sufficient to induce epithelial-to-mesenchymal transition (EMT) and metastasis. Importantly, Six1-induced EMT and metastasis are dependent on TGF-ß signaling. The TGF-ß pathway plays a dual role in cancer, acting as a tumor suppressor in early lesions but enhancing metastatic spread in more advanced tumors. Our previous work indicated that Six1 may be a critical mediator of the switch in TGF-ß signaling from tumor suppressive to tumor promotional. However, the mechanism by which Six1 impinges on the TGF-ß pathway was, until now, unclear. In this work, we identify the TGF-ß type I receptor (TßRI) as a target of Six1 and a critical effector of Six1-induced TGF-ß signaling and EMT. We show that Six1-induced upregulation of TßRI is both necessary and sufficient to activate TGF-ß signaling and induce properties of EMT. Interestingly, increased TßRI expression is not sufficient to induce experimental metastasis, providing in vivo evidence that Six1 overexpression is required to switch TGF-ß signaling to the prometastatic phenotype and showing that induction of EMT is not sufficient to induce experimental metastasis. Together, these results show a novel mechanism for the activation of TGF-ß signaling, identify TßRI as a new target of Six1, and implicate Six1 as a determinant of TGF-ß function in breast cancer.


Asunto(s)
Neoplasias de la Mama/metabolismo , Proteínas de Homeodominio/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Animales , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Línea Celular Tumoral , Transición Epitelial-Mesenquimal , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Ratones , Ratones Endogámicos NOD , Ratones Desnudos , Ratones SCID , Metástasis de la Neoplasia , Regiones Promotoras Genéticas , Proteínas Serina-Treonina Quinasas/biosíntesis , Proteínas Serina-Treonina Quinasas/genética , Receptor Tipo I de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta/biosíntesis , Receptores de Factores de Crecimiento Transformadores beta/genética , Transducción de Señal , Transcripción Genética , Regulación hacia Arriba
18.
J Mammary Gland Biol Neoplasia ; 15(2): 117-34, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20490631

RESUMEN

From the earliest stages of embryonic development, cells of epithelial and mesenchymal origin contribute to the structure and function of developing organs. However, these phenotypes are not always permanent, and instead, under the appropriate conditions, epithelial and mesenchymal cells convert between these two phenotypes. These processes, termed Epithelial-Mesenchymal Transition (EMT), or the reverse Mesenchymal-Epithelial Transition (MET), are required for complex body patterning and morphogenesis. In addition, epithelial plasticity and the acquisition of invasive properties without the full commitment to a mesenchymal phenotype are critical in development, particularly during branching morphogenesis in the mammary gland. Recent work in cancer has identified an analogous plasticity of cellular phenotypes whereby epithelial cancer cells acquire mesenchymal features that permit escape from the primary tumor. Because local invasion is thought to be a necessary first step in metastatic dissemination, EMT and epithelial plasticity are hypothesized to contribute to tumor progression. Similarities between developmental and oncogenic EMT have led to the identification of common contributing pathways, suggesting that the reactivation of developmental pathways in breast and other cancers contributes to tumor progression. For example, developmental EMT regulators including Snail/Slug, Twist, Six1, and Cripto, along with developmental signaling pathways including TGF-beta and Wnt/beta-catenin, are misexpressed in breast cancer and correlate with poor clinical outcomes. This review focuses on the parallels between epithelial plasticity/EMT in the mammary gland and other organs during development, and on a selection of developmental EMT regulators that are misexpressed specifically during breast cancer.


Asunto(s)
Neoplasias de la Mama/patología , Transdiferenciación Celular , Desarrollo Embrionario , Células Epiteliales/fisiología , Glándulas Mamarias Humanas/fisiología , Glándulas Mamarias Humanas/fisiopatología , Células Madre Mesenquimatosas/fisiología , Animales , Neoplasias de la Mama/fisiopatología , Desdiferenciación Celular , Diferenciación Celular , Progresión de la Enfermedad , Femenino , Humanos , Glándulas Mamarias Animales/crecimiento & desarrollo , Glándulas Mamarias Animales/fisiología , Glándulas Mamarias Animales/fisiopatología , Glándulas Mamarias Humanas/crecimiento & desarrollo , Neoplasias Mamarias Experimentales/patología , Neoplasias Mamarias Experimentales/fisiopatología , Neoplasias Mamarias Experimentales/secundario , Invasividad Neoplásica , Metástasis de la Neoplasia
19.
Future Oncol ; 5(8): 1129-43, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19852726

RESUMEN

The epithelial-mesenchymal transition (EMT) is a critical developmental process from the earliest events of embryogenesis to later morphogenesis and organ formation. EMT contributes to the complex architecture of the embryo by permitting the progression of embryogenesis from a simple single-cell layer epithelium to a complex three-dimensional organism composed of both epithelial and mesenchymal cells. However, in most tissues EMT is a developmentally restricted process and fully differentiated epithelia typically maintain their epithelial phenotype. Recently, elements of EMT, specifically the loss of epithelial markers and the gain of mesenchymal markers, have been observed in pathological states, including epithelial cancers. Analysis of the molecular mechanisms of this oncogenic epithelial plasticity have implicated the inappropriate expression and activation of developmental EMT programs, suggesting that cancer cells may reinstitute properties of developmental EMT including enhanced migration and invasion. Thus, in the context of cancer, an EMT-like process may permit dissemination of tumor cells from the primary tumor into the surrounding stroma, setting the stage for metastatic spread. Consistent with this hypothesis, activation of these developmental EMT programs in human cancer correlates with advanced disease and poor prognosis. This review will focus on the current knowledge regarding developmental EMT pathways that have been implicated in cancer progression.


Asunto(s)
Transformación Celular Neoplásica/patología , Epitelio/patología , Mesodermo/patología , Neoplasias/patología , Animales , Diferenciación Celular/fisiología , Transformación Celular Neoplásica/genética , Humanos , Metaplasia , Neoplasias/genética
20.
J Clin Invest ; 119(9): 2678-90, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19726885

RESUMEN

Inappropriate activation of developmental pathways is a well-recognized tumor-promoting mechanism. Here we show that overexpression of the homeoprotein Six1, normally a developmentally restricted transcriptional regulator, increases TGF-beta signaling in human breast cancer cells and induces an epithelial-mesenchymal transition (EMT) that is in part dependent on its ability to increase TGF-beta signaling. TGF-beta signaling and EMT have been implicated in metastatic dissemination of carcinoma. Accordingly, we used spontaneous and experimental metastasis mouse models to demonstrate that Six1 overexpression promotes breast cancer metastasis. In addition, we show that, like its induction of EMT, Six1-induced experimental metastasis is dependent on its ability to activate TGF-beta signaling. Importantly, in human breast cancers Six1 correlated with nuclear Smad3 and thus increased TGF-beta signaling. Further, breast cancer patients whose tumors overexpressed Six1 had a shortened time to relapse and metastasis and an overall decrease in survival. Finally, we show that the effects of Six1 on tumor progression likely extend beyond breast cancer, since its overexpression correlated with adverse outcomes in numerous other cancers including brain, cervical, prostate, colon, kidney, and liver. Our findings indicate that Six1, acting through TGF-beta signaling and EMT, is a powerful and global promoter of cancer metastasis.


Asunto(s)
Neoplasias de la Mama/patología , Neoplasias de la Mama/fisiopatología , Proteínas de Homeodominio/fisiología , Factor de Crecimiento Transformador beta/fisiología , Animales , Neoplasias de la Mama/genética , Línea Celular Tumoral , Epitelio/patología , Femenino , Proteínas de Homeodominio/genética , Humanos , Neoplasias Mamarias Experimentales/genética , Neoplasias Mamarias Experimentales/patología , Neoplasias Mamarias Experimentales/fisiopatología , Mesodermo/patología , Ratones , Ratones Endogámicos NOD , Ratones Desnudos , Ratones SCID , Metástasis de la Neoplasia/patología , Metástasis de la Neoplasia/fisiopatología , Trasplante de Neoplasias , Pronóstico , Transducción de Señal , Proteína smad3/metabolismo , Trasplante Heterólogo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...