Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Carcinogenesis ; 44(6): 485-496, 2023 08 18.
Artículo en Inglés | MEDLINE | ID: mdl-37463322

RESUMEN

The chromobox-containing protein CBX4 is an important regulator of epithelial cell proliferation and differentiation, and has been implicated in several cancer types. The cancer stem cell (CSC) population is a key driver of metastasis and recurrence. The undifferentiated, plastic state characteristic of CSCs relies on cues from the microenvironment. Cancer-associated fibroblasts (CAFs) are a major component of the microenvironment that can influence the CSC population through the secretion of extracellular matrix and a variety of growth factors. Here we show CBX4 is a critical regulator of the CSC phenotype in squamous cell carcinomas of the skin and hypopharynx. Moreover, CAFs can promote the expression of CBX4 in the CSC population through the secretion of interleukin-6 (IL-6). IL-6 activates JAK/STAT3 signaling to increase ∆Np63α-a key transcription factor that is essential for epithelial stem cell function and the maintenance of proliferative potential that is capable of regulating CBX4. Targeting the JAK/STAT3 axis or CBX4 directly suppresses the aggressive phenotype of CSCs and represents a novel opportunity for therapeutic intervention.


Asunto(s)
Fibroblastos Asociados al Cáncer , Carcinoma de Células Escamosas , Humanos , Fibroblastos Asociados al Cáncer/metabolismo , Interleucina-6/metabolismo , Línea Celular Tumoral , Carcinoma de Células Escamosas/patología , Proliferación Celular/genética , Cromatina/metabolismo , Células Madre Neoplásicas/patología , Fibroblastos/metabolismo , Microambiente Tumoral/genética , Ligasas/genética , Ligasas/metabolismo , Proteínas del Grupo Polycomb/genética , Proteínas del Grupo Polycomb/metabolismo
2.
Trends Cell Biol ; 33(4): 280-292, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36115734

RESUMEN

Our understanding of cancer and the key pathways that drive cancer survival has expanded rapidly over the past several decades. However, there are still important challenges that continue to impair patient survival, including our inability to target cancer stem cells (CSCs), metastasis, and drug resistance. The transcription factor p63 is a p53 family member with multiple isoforms that carry out a wide array of functions. Here, we discuss the critical importance of the ΔNp63α isoform in cancer and potential therapeutic strategies to target ΔNp63α expression to impair the CSC population, as well as to prevent metastasis and drug resistance to improve patient survival.


Asunto(s)
Neoplasias , Proteínas Supresoras de Tumor , Humanos , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo , Neoplasias/genética , Neoplasias/terapia , Factores de Transcripción/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Regulación Neoplásica de la Expresión Génica , Línea Celular Tumoral
3.
Oncogene ; 41(35): 4130-4144, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35864175

RESUMEN

Enhancer of zeste homolog 2 (EZH2) and SET domain bifurcated 1 (SETDB1, also known as ESET) are oncogenic methyltransferases implicated in a number of human cancers. These enzymes typically function as epigenetic repressors of target genes by methylating histone H3 K27 and H3-K9 residues, respectively. Here, we show that EZH2 and SETDB1 are essential to proliferation in 3 SCC cell lines, HSC-5, FaDu, and Cal33. Additionally, we find both of these proteins highly expressed in an aggressive stem-like SCC sub-population. Depletion of either EZH2 or SETDB1 disrupts these stem-like cells and their associated phenotypes of spheroid formation, invasion, and tumor growth. We show that SETDB1 regulates this SCC stem cell phenotype through cooperation with ΔNp63α, an oncogenic isoform of the p53-related transcription factor p63. Furthermore, EZH2 is upstream of both SETDB1 and ΔNp63α, activating these targets via repression of the tumor suppressor RUNX3. We show that targeting this pathway with inhibitors of EZH2 results in activation of RUNX3 and repression of both SETDB1 and ΔNp63α, antagonizing the SCC cancer stem cell phenotype. This work highlights a novel pathway that drives an aggressive cancer stem cell phenotype and demonstrates a means of pharmacological intervention.


Asunto(s)
Carcinoma de Células Escamosas , Proteína Potenciadora del Homólogo Zeste 2 , Línea Celular Tumoral , Subunidad alfa 3 del Factor de Unión al Sitio Principal , N-Metiltransferasa de Histona-Lisina , Humanos , Células Madre Neoplásicas , Fenotipo , Regiones Promotoras Genéticas , Factores de Transcripción
4.
Cancer Res ; 81(24): 6246-6258, 2021 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-34697072

RESUMEN

Bromodomain containing protein 4 (BRD4) plays a critical role in controlling the expression of genes involved in development and cancer. Inactivation of BRD4 inhibits cancer growth, making it a promising anticancer drug target. The cancer stem cell (CSC) population is a key driver of recurrence and metastasis in patients with cancer. Here we show that cancer stem-like cells can be enriched from squamous cell carcinomas (SCC), and that these cells display an aggressive phenotype with enhanced stem cell marker expression, migration, invasion, and tumor growth. BRD4 is highly elevated in this aggressive subpopulation of cells, and its function is critical for these CSC-like properties. Moreover, BRD4 regulates ΔNp63α, a key transcription factor that is essential for epithelial stem cell function that is often overexpressed in cancers. BRD4 regulates an EZH2/STAT3 complex that leads to increased ΔNp63α-mediated transcription. Targeting BRD4 in human SCC reduces ΔNp63α, leading to inhibition of spheroid formation, migration, invasion, and tumor growth. These studies identify a novel BRD4-regulated signaling network in a subpopulation of cancer stem-like cells, elucidating a possible avenue for effective therapeutic intervention. SIGNIFICANCE: This study identifies a signaling cascade driven by BRD4 that upregulates ΔNp63α to promote cancer stem-like properties, which has potential therapeutic implications for the treatment of squamous cell carcinomas.


Asunto(s)
Carcinoma de Células Escamosas/patología , Proteínas de Ciclo Celular/metabolismo , Proteína Potenciadora del Homólogo Zeste 2/metabolismo , Regulación Neoplásica de la Expresión Génica , Células Madre Neoplásicas/patología , Factor de Transcripción STAT3/metabolismo , Factores de Transcripción/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Animales , Apoptosis , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/metabolismo , Proteínas de Ciclo Celular/genética , Proliferación Celular , Proteína Potenciadora del Homólogo Zeste 2/genética , Humanos , Ratones , Ratones Desnudos , Células Madre Neoplásicas/metabolismo , Factor de Transcripción STAT3/genética , Factores de Transcripción/genética , Células Tumorales Cultivadas , Proteínas Supresoras de Tumor/genética , Ensayos Antitumor por Modelo de Xenoinjerto
5.
J Cell Sci ; 133(17)2020 09 11.
Artículo en Inglés | MEDLINE | ID: mdl-32917730

RESUMEN

p63 (also known as TP63) is a transcription factor of the p53 family, along with p73. Multiple isoforms of p63 have been discovered and these have diverse functions encompassing a wide array of cell biology. p63 isoforms are implicated in lineage specification, proliferative potential, differentiation, cell death and survival, DNA damage response and metabolism. Furthermore, p63 is linked to human disease states including cancer. p63 is critical to many aspects of cell signaling, and in this Cell science at a glance article and the accompanying poster, we focus on the signaling cascades regulating TAp63 and ΔNp63 isoforms and those that are regulated by TAp63 and ΔNp63, as well the role of p63 in disease.


Asunto(s)
Neoplasias , Factores de Transcripción , Humanos , Neoplasias/genética , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Transducción de Señal , Factores de Transcripción/genética
6.
Mol Cancer Res ; 17(8): 1665-1674, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31123173

RESUMEN

Melanoma is a lethal skin cancer prone to progression and metastasis, and resistant to therapy. Metastasis and therapy resistance of melanoma and other cancers are driven by tumor cell plasticity, largely via acquisition/loss of stem-like characteristics and transitions between epithelial and mesenchymal phenotypes (EMT/MET). NME1 is a metastasis suppressor gene that inhibits metastatic potential when its expression is enforced in melanoma and other cancers. Herein, we have unmasked a novel role for NME1 as a driver of melanoma growth distinct from its canonical function as a metastasis suppressor. NME1 promotes expansion of stem-like melanoma cells that exhibit elevated expression of stem cell markers (e.g., Sox2, Sox10, Oct-4, KLF4, and Ccnb-1), enhanced growth as melanoma spheres in culture, and enhanced tumor growth and lung colonizing activities in vivo. In contrast, NME1 expression did not affect the proliferation of melanoma cell lines in monolayer culture conditions. Silencing of NME1 expression resulted in a dramatic reduction in melanoma sphere size, and impaired tumor growth and metastatic activities of melanoma sphere cells when xenografted in immunocompromised mice. Individual cells within melanoma sphere cultures displayed a wide range of NME1 expression across multiple melanoma cell lines. Cell subpopulations with elevated NME1 expression were fast cycling and displayed enhanced expression of stem cell markers. IMPLICATIONS: Our findings suggest the current model of NME1 as a metastasis-suppressing factor requires refinement, bringing into consideration its heterogeneous expression within melanoma sphere cultures and its novel role in promoting the expansion and tumorigenicity of stem-like cells.


Asunto(s)
Proliferación Celular , Regulación Neoplásica de la Expresión Génica , Neoplasias Pulmonares/secundario , Melanoma/patología , Nucleósido Difosfato Quinasas NM23/metabolismo , Células Madre Neoplásicas/patología , Animales , Apoptosis , Humanos , Factor 4 Similar a Kruppel , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Melanoma/genética , Melanoma/metabolismo , Ratones , Ratones Endogámicos NOD , Ratones SCID , Nucleósido Difosfato Quinasas NM23/genética , Células Madre Neoplásicas/metabolismo , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
7.
Oncotarget ; 8(43): 73407-73418, 2017 Sep 26.
Artículo en Inglés | MEDLINE | ID: mdl-29088716

RESUMEN

Epidermal squamous cell carcinoma (SCC) is among the most common cancers. SCC can be treated by surgical excision, but recurrence of therapy-resistant disease is a major problem. We recently showed that YAP1, the Hippo signaling transcription adaptor protein, and ∆Np63α, a key epidermal stem cell survival protein, form a complex to drive epidermal cancer stem cell survival. In the present study, we demonstrate that YAP1 and ∆Np63α are important sulforaphane cancer prevention targets. We show that sulforaphane treatment increases YAP1 phosphorylation and proteolytic degradation. The loss of YAP1 is associated with a reduction in ∆Np63α level and a reduction in ECS cell survival, spheroid formation, invasion and migration. Loss of YAP1 and ∆Np63α is mediated by the proteasome and can be inhibited by lactacystin treatment. YAP1 or ∆Np63α knockdown replicates the responses to sulforaphane, and restoration of YAP1 or ∆Np63α antagonizes sulforaphane action. Sulforaphane suppresses ECS cell tumor formation and this is associated with reduced levels of YAP1 and ∆Np63α. These studies suggest that YAP1 and ∆Np63α may be important sulforaphane cancer preventive targets in epidermal squamous cell carcinoma.

8.
Carcinogenesis ; 38(8): 827-836, 2017 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-28854561

RESUMEN

Protein arginine methyltransferase 5 (PRMT5) cooperates with methylosome protein 50 (MEP50) to arginine methylate histone H3 and H4 to silence gene expression, and increased PRMT5 activity is associated with enhanced cancer cell survival. We have studied the role of PRMT5 and MEP50 in epidermal squamous cell carcinoma. We show that knockdown of PRMT5 or MEP50 results in reduced H4R3me2s formation, and reduced cell proliferation, invasion, migration and tumor formation. We further show that treatment with sulforaphane (SFN), a cancer preventive agent derived from cruciferous vegetables, reduces PRMT5 and MEP50 level and H4R3me2s formation, and this is associated with reduced cell proliferation, invasion and migration. The SFN-dependent reduction in PRMT5 and MEP50 level requires proteasome activity. Moreover, SFN-mediated responses are partially reversed by forced PRMT5 or MEP50 expression. SFN treatment of tumors results in reduced MEP50 level and H4R3me2s formation, confirming that that SFN impacts this complex in vivo. These studies suggest that the PRMT5/MEP50 is required for tumor growth and that reduced expression of this complex is a part of the mechanism of SFN suppression of tumor formation.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Carcinoma de Células Escamosas/tratamiento farmacológico , Isotiocianatos/administración & dosificación , Proteína-Arginina N-Metiltransferasas/genética , Neoplasias Cutáneas/tratamiento farmacológico , Proteínas Adaptadoras Transductoras de Señales/biosíntesis , Animales , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/patología , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Metilación de ADN/efectos de los fármacos , Epidermis/efectos de los fármacos , Epidermis/patología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Humanos , Ratones , Invasividad Neoplásica/genética , Proteína-Arginina N-Metiltransferasas/biosíntesis , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/patología , Sulfóxidos , Ensayos Antitumor por Modelo de Xenoinjerto
10.
Oncotarget ; 8(66): 110257-110272, 2017 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-29299145

RESUMEN

Treating BRAF inhibitor-resistant melanoma is an important therapeutic goal. Thus, it is important to identify and target mechanisms of resistance to improve therapy. The YAP1 and TAZ proteins of the Hippo signaling pathway are important drivers of cancer cell survival, and are BRAF inhibitor resistant factors in melanoma. We examine the role of YAP1/TAZ in melanoma cancer stem cells (MCS cells). We demonstrate that YAP1, TAZ and TEAD (TEA domain transcription factor) levels are elevated in BRAF inhibitor resistant MCS cells and enhance cell survival, spheroid formation, matrigel invasion and tumor formation. Moreover, increased YAP1, TAZ and TEAD are associated with sustained ERK1/2 activity that is not suppressed by BRAF inhibitor. Xenograft studies show that treating BRAF inhibitor-resistant tumors with verteporfin, an agent that interferes with YAP1 function, reduces YAP1/TAZ level, restores BRAF inhibitor suppression of ERK1/2 signaling and reduces tumor growth. Verteporfin is highly effective as concentrations of verteporfin that do not impact tumor formation restore BRAF inhibitor suppression of tumor formation, suggesting that co-treatment with agents that inhibit YAP1 and BRAF(V600E) may be a viable therapy for cancer stem cell-derived BRAF inhibitor-resistant melanoma.

11.
Cancer Res ; 76(24): 7265-7276, 2016 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-27780825

RESUMEN

Transglutaminase 2 (TG2) expression is required for epidermal squamous cell carcinoma cancer stem cell survival. However, the molecular signaling mechanisms triggered by TG2 that mediate this survival action are not well understood. Here we show that TG2 is constitutively expressed in ECS cells, where it interacts with α6/ß4 integrin to stimulate FAK and Src signaling, leading to PI3K activation of phosphoinositide-dependent kinase 1 (PDK1). PDK1 inhibits Hippo signaling, leading to enhanced nuclear accumulation of YAP1, which interacted with and stabilized ΔNp63α to enhance epidermal squamous cell carcinoma spheroid formation, invasion, and migration. Overall, these findings suggest that constitutive TG2 expression results in stabilization of ΔNp63α, leading to maintenance of cancer stem cell properties and enhanced tumor formation. Cancer Res; 76(24); 7265-76. ©2016 AACR.


Asunto(s)
Carcinoma de Células Escamosas/patología , Proteínas de Unión al GTP/metabolismo , Células Madre Neoplásicas/patología , Neoplasias Cutáneas/patología , Factores de Transcripción/metabolismo , Transglutaminasas/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Carcinoma de Células Escamosas/metabolismo , Línea Celular Tumoral , Supervivencia Celular , Regulación Neoplásica de la Expresión Génica/fisiología , Xenoinjertos , Humanos , Integrina alfa6beta4/metabolismo , Ratones , Ratones Endogámicos NOD , Ratones Noqueados , Ratones SCID , Células Madre Neoplásicas/metabolismo , Fosfoproteínas/metabolismo , Proteína Glutamina Gamma Glutamiltransferasa 2 , Transducción de Señal/fisiología , Neoplasias Cutáneas/metabolismo , Proteínas Señalizadoras YAP
12.
Mol Carcinog ; 55(12): 2024-2036, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-26693692

RESUMEN

Melanoma is a metastatic cancer associated with poor survival. Here, we study a subpopulation of melanoma cancer cells displaying melanoma cancer stem cell (MCS cells) properties including elevated expression of stem cell markers, increased ability to survive as spheroids, and enhanced cell migration and invasion. We show that the Ezh2 stem cell survival protein is enriched in MCS cells and that Ezh2 knockdown or treatment with small molecule Ezh2 inhibitors, GSK126 or EPZ-6438, reduces Ezh2 activity. This reduction is associated with a reduced MCS cell spheroid formation, migration, and invasion. Moreover, the diet-derived cancer prevention agent, sulforaphane (SFN), suppresses MCS cell survival and this is associated with loss of Ezh2. Forced expression of Ezh2 partially reverses SFN suppression of MCS cell spheroid formation, migration, and invasion. A375 melanoma cell-derived MCS cells form rapidly growing tumors in immune-compromised mice and SFN treatment of these tumors reduces tumor growth and this is associated with reduced Ezh2 level and H3K27me3 formation, reduced matrix metalloproteinase expression, increased TIMP3 expression and increased apoptosis. These studies identify Ezh2 as a MCS cell marker and cancer stem cell prevention target, and suggest that SFN acts to reduce melanoma tumor formation via a mechanism that includes suppression of Ezh2 function. © 2015 Wiley Periodicals, Inc.


Asunto(s)
Anticarcinógenos/uso terapéutico , Proteína Potenciadora del Homólogo Zeste 2/metabolismo , Isotiocianatos/uso terapéutico , Melanoma/patología , Melanoma/prevención & control , Células Madre Neoplásicas/patología , Animales , Movimiento Celular/efectos de los fármacos , Proteína Potenciadora del Homólogo Zeste 2/genética , Técnicas de Silenciamiento del Gen , Melanoma/genética , Melanoma/metabolismo , Ratones Endogámicos NOD , Ratones Noqueados , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/metabolismo , Sulfóxidos , Regulación hacia Arriba
13.
Mol Carcinog ; 54(10): 947-58, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26258961

RESUMEN

Recent studies indicate that cancer cells express elevated levels of type II transglutaminase (TG2), and that expression is further highly enriched in cancer stem cells derived from these cancers. Moreover, elevated TG2 expression is associated with enhanced cancer stem cell marker expression, survival signaling, proliferation, migration, invasion, integrin-mediated adhesion, epithelial-mesenchymal transition, and drug resistance. TG2 expression is also associated with formation of aggressive and metastatic tumors that are resistant to conventional therapeutic intervention. This review summarizes the role of TG2 as a cancer cell survival factor in a range of tumor types, and as a target for preventive and therapeutic intervention. The literature supports the idea that TG2, in the closed/GTP-binding/signaling conformation, drives cancer cell and cancer stem cell survival, and that TG2, in the open/crosslinking conformation, is associated with cell death.


Asunto(s)
Supervivencia Celular/fisiología , Proteínas de Unión al GTP/metabolismo , Neoplasias/metabolismo , Células Madre Neoplásicas/metabolismo , Factor de Células Madre/metabolismo , Transglutaminasas/metabolismo , Animales , Humanos , Proteína Glutamina Gamma Glutamiltransferasa 2
14.
Oncotarget ; 6(24): 20525-39, 2015 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-25971211

RESUMEN

Type II transglutaminase (TG2) is a multifunctional protein that has recently been implicated as having a role in ECS cell survival. In the present study we investigate the role of TG2 in regulating epithelial mesenchymal transition (EMT) in ECS cells. Our studies show that TG2 knockdown or treatment with TG2 inhibitor, results in a reduced EMT marker expression, and reduced cell migration and invasion. TG2 has several activities, but the most prominent are its transamidase and GTP binding activity. Analysis of a series of TG2 mutants reveals that TG2 GTP binding activity, but not the transamidase activity, is required for expression of EMT markers (Twist, Snail, Slug, vimentin, fibronectin, N-cadherin and HIF-1α), and increased ECS cell invasion and migration. This coupled with reduced expression of E-cadherin. Additional studies indicate that NFÏ°B signaling, which has been implicated as mediating TG2 impact on EMT in breast cancer cells, is not involved in TG2 regulation of EMT in skin cancer. These studies suggest that TG2 is required for maintenance of ECS cell EMT, invasion and migration, and suggests that inhibiting TG2 GTP binding/G-protein related activity may reduce skin cancer tumor survival.


Asunto(s)
Carcinoma de Células Escamosas/enzimología , Proteínas de Unión al GTP/metabolismo , Células Madre Neoplásicas/enzimología , Células Madre Neoplásicas/patología , Transglutaminasas/metabolismo , Carcinoma de Células Escamosas/patología , Células Epiteliales/enzimología , Células Epiteliales/metabolismo , Células Epiteliales/patología , Transición Epitelial-Mesenquimal , Células HEK293 , Humanos , Células Madre Neoplásicas/metabolismo , Proteína Glutamina Gamma Glutamiltransferasa 2 , Transducción de Señal
15.
Mol Cancer Res ; 13(7): 1083-94, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25934691

RESUMEN

UNLABELLED: Cancer stem cells are thought to be responsible for rapid tumor growth, metastasis, and enhanced tumor survival following drug treatment. For this reason, there is a major emphasis on identifying proteins that can be targeted to kill cancer stem cells or control their growth, and transglutaminase type II (TGM2/TG2) is such a target in epidermal squamous cell carcinoma. TG2 was originally described as a transamidase in the extracellular matrix that crosslinks proteins by catalyzing ε-(γ-glutamyl)lysine bonds. However, subsequent studies have shown that TG2 is a GTP-binding protein that plays an important role in cell signaling and survival. In the present study, TG2 shows promise as a target for anticancer stem cell therapy in human squamous cell carcinoma. TG2 was determined to be highly elevated in epidermal cancer stem cells (ECS cells), and TG2 knockdown or suppression of TG2 function with inhibitors reduced ECS cell survival, spheroid formation, Matrigel invasion, and migration. The reduction in survival is associated with activation of apoptosis. Mechanistic studies, using TG2 mutants, revealed that the GTP-binding activity is required for maintenance of ECS cell growth and survival, and that the action of TG2 in ECS cells is not mediated by NF-κB signaling. IMPLICATIONS: This study suggests that TG2 has an important role in maintaining cancer stem cell survival, invasive, and metastatic behavior and is an important therapeutic target to reduce survival of cancer stem cells in epidermal squamous cell carcinoma.


Asunto(s)
Carcinoma de Células Escamosas/metabolismo , Células Epiteliales/metabolismo , Proteínas de Unión al GTP/genética , Proteínas de Unión al GTP/metabolismo , Células Madre Neoplásicas/metabolismo , Transglutaminasas/genética , Transglutaminasas/metabolismo , Apoptosis/efectos de los fármacos , Línea Celular , Supervivencia Celular/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Células Epiteliales/efectos de los fármacos , Proteínas de Unión al GTP/antagonistas & inhibidores , Técnicas de Silenciamiento del Gen , Vectores Genéticos , Guanosina Trifosfato/metabolismo , Humanos , FN-kappa B/metabolismo , Invasividad Neoplásica , Unión Proteica , Proteína Glutamina Gamma Glutamiltransferasa 2 , Transducción de Señal/efectos de los fármacos , Transglutaminasas/antagonistas & inhibidores
16.
PLoS One ; 8(12): e84324, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24376802

RESUMEN

Epidermal squamous cell carcinoma is among the most common cancers in humans. These tumors are comprised of phenotypically diverse populations of cells that display varying potential for proliferation and differentiation. An important goal is identifying cells from this population that drive tumor formation. To enrich for tumor-forming cells, cancer cells were grown as spheroids in non-attached conditions. We show that spheroid-selected cells form faster growing and larger tumors in immune-compromised mice as compared to non-selected cells. Moreover, spheroid-selected cells gave rise to tumors following injection of as few as one hundred cells, suggesting these cells have enhanced tumor-forming potential. Cells isolated from spheroid-selected tumors retain an enhanced ability to grow as spheroids when grown in non-attached culture conditions. Thus, these tumor-forming cells retain their phenotype following in vivo passage as tumors. Detailed analysis reveals that spheroid-selected cultures are highly enriched for expression of epidermal stem cell and embryonic stem cell markers, including aldehyde dehydrogenase 1, keratin 15, CD200, keratin 19, Oct4, Bmi-1, Ezh2 and trimethylated histone H3. These studies indicate that a subpopulation of cells that possess stem cell-like properties and express stem cell markers can be derived from human epidermal cancer cells and that these cells display enhanced ability to drive tumor formation.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Carcinoma de Células Escamosas/fisiopatología , Transformación Celular Neoplásica/metabolismo , Células Epidérmicas , Células Madre Neoplásicas/fisiología , Esferoides Celulares/citología , Animales , Técnicas de Cultivo de Célula/métodos , Humanos , Immunoblotting , Ratones , Esferoides Celulares/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...