Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 66
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Nature ; 495(7440): 241-5, 2013 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-23467088

RESUMEN

Epithelial ovarian cancer (EOC) is the fifth leading cause of cancer deaths among women in the United States, but its pathogenesis is poorly understood. Some epithelial cancers are known to occur in transitional zones between two types of epithelium, whereas others have been shown to originate in epithelial tissue stem cells. The stem cell niche of the ovarian surface epithelium (OSE), which is ruptured and regenerates during ovulation, has not yet been defined unequivocally. Here we identify the hilum region of the mouse ovary, the transitional (or junction) area between the OSE, mesothelium and tubal (oviductal) epithelium, as a previously unrecognized stem cell niche of the OSE. We find that cells of the hilum OSE are cycling slowly and express stem and/or progenitor cell markers ALDH1, LGR5, LEF1, CD133 and CK6B. These cells display long-term stem cell properties ex vivo and in vivo, as shown by our serial sphere generation and long-term lineage-tracing assays. Importantly, the hilum cells show increased transformation potential after inactivation of tumour suppressor genes Trp53 and Rb1, whose pathways are altered frequently in the most aggressive and common type of human EOC, high-grade serous adenocarcinoma. Our study supports experimentally the idea that susceptibility of transitional zones to malignant transformation may be explained by the presence of stem cell niches in those areas. Identification of a stem cell niche for the OSE may have important implications for understanding EOC pathogenesis.


Asunto(s)
Epitelio/patología , Neoplasias Glandulares y Epiteliales/patología , Neoplasias Ováricas/patología , Ovario/patología , Nicho de Células Madre , Animales , Biomarcadores/análisis , Biomarcadores/metabolismo , Carcinoma Epitelial de Ovario , Linaje de la Célula , Separación Celular , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Epitelio/metabolismo , Femenino , Masculino , Ratones , Neoplasias Glandulares y Epiteliales/metabolismo , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Neoplasias Ováricas/metabolismo , Ovario/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Proteína de Retinoblastoma/deficiencia , Proteína de Retinoblastoma/genética , Proteína de Retinoblastoma/metabolismo , Proteína p53 Supresora de Tumor/deficiencia , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
2.
Mod Pathol ; 30(9): 1241-1250, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28664938

RESUMEN

Recently it has been reported that serous tubal intraepithelial carcinoma (STIC), the likely precursor of ovarian/extra-uterine high-grade serous carcinoma, are frequently located in the vicinity of tubal-peritoneal junctions, consistent with the cancer-prone features of many epithelial transitional regions. To test if p53 (aka TP53)-signatures and secretory cell outgrowths (SCOUTs) also localize to tubal-peritoneal junctions, we examined these lesions in the fallopian tubes of patients undergoing salpingo-oophorectomy for sporadic high-grade serous carcinomas or as a prophylactic procedure for carriers of familial BRCA1 or 2 mutations. STICs were located closest to the tubal-peritoneal junctions with an average distance of 1.31 mm, while SCOUTs were not detected in the fimbriated end of the fallopian tube. As many epithelial transitional regions contain stem cells, we also determined the expression of stem cell markers in the normal fallopian tube, tubal intraepithelial lesions and high-grade serous carcinomas. Of those, LEF1 was consistently expressed in the tubal-peritoneal junctions and all lesions, independent of p53 status. All SCOUTs demonstrated strong nuclear expression of ß-catenin consistent with the LEF1 participation in the canonical WNT pathway. However, ß-catenin was preferentially located in the cytoplasm of cells comprising STICs and p53 signatures, suggesting WNT-independent function of LEF1 in those lesions. Both frequency of LEF1 expression and ß-catenin nuclear expression correlated with the worst 5-year patient survival, supporting important role of both proteins in high-grade serous carcinoma. Taken together, our findings suggest the existence of stem cell niche within the tubal-peritoneal junctions. Furthermore, they support the notion that the pathogenesis of SCOUTs is distinct from that of STICs and p53 signatures. The location and discrete patterns of LEF1 and ß-catenin expression may serve as highly sensitive and reliable ancillary markers for the detection and differential diagnosis of tubal intraepithelial lesions.


Asunto(s)
Biomarcadores de Tumor/análisis , Carcinoma in Situ/química , Neoplasias de las Trompas Uterinas/química , Factor de Unión 1 al Potenciador Linfoide/análisis , Células Madre Neoplásicas/química , Adulto , Anciano , Anciano de 80 o más Años , Proteína BRCA1/genética , Proteína BRCA2/genética , Carcinoma in Situ/genética , Carcinoma in Situ/patología , Carcinoma in Situ/cirugía , Estudios de Casos y Controles , Neoplasias de las Trompas Uterinas/genética , Neoplasias de las Trompas Uterinas/patología , Neoplasias de las Trompas Uterinas/cirugía , Femenino , Humanos , Inmunohistoquímica , Estimación de Kaplan-Meier , Persona de Mediana Edad , Mutación , Clasificación del Tumor , Células Madre Neoplásicas/patología , Valor Predictivo de las Pruebas , Modelos de Riesgos Proporcionales , Reproducibilidad de los Resultados , Salpingooforectomía , Nicho de Células Madre , Factores de Tiempo , Resultado del Tratamiento , Microambiente Tumoral , Proteína p53 Supresora de Tumor/análisis , beta Catenina/análisis
3.
Proc Natl Acad Sci U S A ; 109(33): 13380-5, 2012 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-22847442

RESUMEN

We examined the myogenic response to infarction in neonatal and adult mice to determine the role of c-kit(+) cardiovascular precursor cells (CPC) that are known to be present in early heart development. Infarction of postnatal day 1-3 c-kit(BAC)-EGFP mouse hearts induced the localized expansion of (c-kit)EGFP(+) cells within the infarct, expression of the c-kit and Nkx2.5 mRNA, myogenesis, and partial regeneration of the infarction, with (c-kit)EGFP(+) cells adopting myogenic and vascular fates. Conversely, infarction of adult mice resulted in a modest induction of (c-kit)EGFP(+) cells within the infarct, which did not express Nkx2.5 or undergo myogenic differentiation, but adopted a vascular fate within the infarction, indicating a lack of authentic CPC. Explantation of infarcted neonatal and adult heart tissue to scid mice, and adoptive transfer of labeled bone marrow, confirmed the cardiac source of myogenic (neonate) and angiogenic (neonate and adult) cells. FACS-purified (c-kit)EGFP(+)/(αMHC)mCherry(-) (noncardiac) cells from microdissected infarcts within 6 h of infarction underwent cardiac differentiation, forming spontaneously beating myocytes in vitro; cre/LoxP fate mapping identified a noncardiac population of (c-kit)EGFP(+) myocytes within infarctions, indicating that the induction of undifferentiated precursors contributes to localized myogenesis. Thus, adult postinfarct myogenic failure is likely not due to a context-dependent restriction of precursor differentiation, and c-kit induction following injury of the adult heart does not define precursor status.


Asunto(s)
Envejecimiento/patología , Desarrollo de Músculos , Infarto del Miocardio/patología , Proteínas Proto-Oncogénicas c-kit/metabolismo , Células Madre/citología , Envejecimiento/metabolismo , Animales , Animales Recién Nacidos , Linaje de la Célula , Células Endoteliales/metabolismo , Células Endoteliales/patología , Proteínas Fluorescentes Verdes/metabolismo , Ratones , Infarto del Miocardio/metabolismo , Regeneración , Células Madre/metabolismo
4.
Proc Natl Acad Sci U S A ; 109(25): 9786-91, 2012 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-22665775

RESUMEN

Multipotent adipose-derived stem cells (ASCs) are increasingly used for regenerative purposes such as soft tissue reconstruction following mastectomy; however, the ability of tumors to commandeer ASC functions to advance tumor progression is not well understood. Through the integration of physical sciences and oncology approaches we investigated the capability of tumor-derived chemical and mechanical cues to enhance ASC-mediated contributions to tumor stroma formation. Our results indicate that soluble factors from breast cancer cells inhibit adipogenic differentiation while increasing proliferation, proangiogenic factor secretion, and myofibroblastic differentiation of ASCs. This altered ASC phenotype led to varied extracellular matrix (ECM) deposition and contraction thereby enhancing tissue stiffness, a characteristic feature of breast tumors. Increased stiffness, in turn, facilitated changes in ASC behavior similar to those observed with tumor-derived chemical cues. Orthotopic mouse studies further confirmed the pathological relevance of ASCs in tumor progression and stiffness in vivo. In summary, altered ASC behavior can promote tumorigenesis and, thus, their implementation for regenerative therapy should be carefully considered in patients previously treated for cancer.


Asunto(s)
Tejido Adiposo/citología , Neoplasias de la Mama/terapia , Trasplante de Células Madre , Animales , Neoplasias de la Mama/patología , Diferenciación Celular , Línea Celular Tumoral , Progresión de la Enfermedad , Matriz Extracelular , Femenino , Humanos , Ratones , Trasplante de Neoplasias
5.
Proc Natl Acad Sci U S A ; 108(34): 14240-5, 2011 Aug 23.
Artículo en Inglés | MEDLINE | ID: mdl-21831840

RESUMEN

Recent observations suggest that p53 mutations are responsible not only for growth of primary tumors but also for their dissemination. However, mechanisms involved in p53-mediated control of cell motility and invasion remain poorly understood. By using the primary ovarian surface epithelium cell culture, we show that conditional inactivation of p53 or expression of its mutant forms results in overexpression of MET receptor tyrosine kinase, a crucial regulator of invasive growth. At the same time, cells acquire increased MET-dependent motility and invasion. Wild-type p53 negatively regulates MET expression by two mechanisms: (i) transactivation of MET-targeting miR-34, and (ii) inhibition of SP1 binding to MET promoter. Both mechanisms are not functional in p53 absence, but mutant p53 proteins retain partial MET promoter suppression. Accordingly, MET overexpression, cell motility, and invasion are particularly high in p53-null cells. These results identify MET as a critical effector of p53 and suggest that inhibition of MET may be an effective antimetastatic approach to treat cancers with p53 mutations. These results also show that the extent of advanced cancer traits, such as invasion, may be determined by alterations in individual components of p53/MET regulatory network.


Asunto(s)
Movimiento Celular , Regulación Neoplásica de la Expresión Génica , Proteínas Proto-Oncogénicas c-met/genética , Receptores de Factores de Crecimiento/genética , Proteína p53 Supresora de Tumor/metabolismo , Animales , Línea Celular Tumoral , Epitelio/metabolismo , Epitelio/patología , Femenino , Silenciador del Gen , Humanos , Ratones , MicroARNs/genética , MicroARNs/metabolismo , Proteínas Mutantes/metabolismo , Invasividad Neoplásica , Ovario/metabolismo , Ovario/patología , Regiones Promotoras Genéticas/genética , Unión Proteica , Proteínas Proto-Oncogénicas c-met/metabolismo , Receptores de Factores de Crecimiento/metabolismo , Factor de Transcripción Sp1/metabolismo
6.
bioRxiv ; 2024 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-38562813

RESUMEN

Serous endometrial carcinoma (SEC) constitutes about 10% of endometrial carcinomas and is one of the most aggressive and lethal types of uterine cancer. Due to the rapid progression of SEC, early detection of this disease is of utmost importance. However, molecular and cellular dynamics during the pre-dysplastic stage of this disease remain largely unknown. Here, we provide a comprehensive census of cell types and their states for normal, pre-dysplastic, and dysplastic endometrium in a mouse model of SEC. This model is associated with inactivation of tumor suppressor genes Trp53 and Rb1 , whose pathways are altered frequently in SEC. We report that pre-dysplastic changes are characterized by an expanded and increasingly diverse immature luminal epithelial cell populations. Consistent with transcriptome changes, cells expressing the luminal epithelial marker TROP2 begin to substitute FOXA2+ cells in the glandular epithelium. These changes are associated with a reduction in number and strength of predicted interactions between epithelial and stromal endometrial cells. By using a multi-level approach combining single-cell and spatial transcriptomics paired with screening for clinically relevant genes in human endometrial carcinoma, we identified a panel of 44 genes suitable for further testing of their validity as early diagnostic and prognostic markers. Among these genes are known markers of human SEC, such as C DKN2A, and novel markers, such as OAS2 and OASL, members of 2-5A synthetase family that is essential for the innate immune response. In summary, our results suggest an important role of the luminal epithelium in SEC pathogenesis, highlight aberrant cell-cell interactions in pre-dysplastic stages, and provide a new platform for comparative identification and characterization of novel, clinically relevant prognostic and diagnostic markers and potential therapeutic modalities.

7.
Proc Natl Acad Sci U S A ; 107(4): 1408-13, 2010 Jan 26.
Artículo en Inglés | MEDLINE | ID: mdl-20080707

RESUMEN

EGF receptor (EGFR) signaling in human cancers elicits changes in protein-expression patterns that are crucial for potentiating tumor growth. Identifying those proteins with expression regulated by the EGFR and determining how they contribute to malignancy is fundamental for the development of more effective strategies to treat cancer. Here, we show that tissue transglutaminase (tTG) is one such protein. EGF up-regulates tTG expression in human breast-cancer cells, and knock-downs of tTG or the treatment of breast cancer cells with a tTG inhibitor blocks their EGF-stimulated anchorage-independent growth. We further show that the combined actions of Ras and Cdc42, leading to the activation of PI 3-kinase and NFkappaB, provide a mechanism by which EGF can up-regulate tTG in breast-cancer cells. Moreover, overexpression of wild-type tTG, but not its transamidation-defective counterpart, fully mimics the growth advantages afforded by EGF to these cancer cells. Surprisingly, the tTG-promoted growth of breast-cancer cells is dependent on its ability to activate the Src tyrosine kinase as an outcome of a complex formed between tTG and the breast-cancer marker and intermediate filament protein keratin-19. These findings identify tTG as a key participant in an EGFR/Src-signaling pathway in breast-cancer cells and a potential target for inhibiting EGFR-promoted tumor progression.


Asunto(s)
Neoplasias de la Mama/metabolismo , Factor de Crecimiento Epidérmico/metabolismo , Transducción de Señal , Transglutaminasas/metabolismo , Familia-src Quinasas/metabolismo , Neoplasias de la Mama/genética , Línea Celular Tumoral , Proliferación Celular , Activación Enzimática , Proteínas de Unión al GTP , Regulación Enzimológica de la Expresión Génica , Humanos , Queratina-19/genética , Queratina-19/metabolismo , Proteína Oncogénica p21(ras)/metabolismo , Unión Proteica , Proteína Glutamina Gamma Glutamiltransferasa 2 , Transglutaminasas/genética , Proteína de Unión al GTP cdc42/metabolismo
8.
Cell Rep ; 42(4): 112396, 2023 04 25.
Artículo en Inglés | MEDLINE | ID: mdl-37061917

RESUMEN

Emerging evidence indicates that metabolic dysregulation drives prostate cancer (PCa) progression and metastasis. AMP-activated protein kinase (AMPK) is a master regulator of metabolism, although its role in PCa remains unclear. Here, we show that genetic and pharmacological activation of AMPK provides a protective effect on PCa progression in vivo. We show that AMPK activation induces PGC1α expression, leading to catabolic metabolic reprogramming of PCa cells. This catabolic state is characterized by increased mitochondrial gene expression, increased fatty acid oxidation, decreased lipogenic potential, decreased cell proliferation, and decreased cell invasiveness. Together, these changes inhibit PCa disease progression. Additionally, we identify a gene network involved in cell cycle regulation that is inhibited by AMPK activation. Strikingly, we show a correlation between this gene network and PGC1α gene expression in human PCa. Taken together, our findings support the use of AMPK activators for clinical treatment of PCa to improve patient outcome.


Asunto(s)
Proteínas Quinasas Activadas por AMP , Neoplasias de la Próstata , Masculino , Humanos , Proteínas Quinasas Activadas por AMP/metabolismo , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/metabolismo , Lipogénesis , Metabolismo de los Lípidos , Neoplasias de la Próstata/patología
9.
Genesis ; 50(1): 28-40, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21809434

RESUMEN

The role of hedgehog (HH) signaling in reproductive tract development was studied in mice in which a dominant active allele of the signal transducer smoothened (SmoM2) was conditionally expressed in the Müllerian duct and ovary. Mutant females are infertile, primarily because they fail to ovulate. Levels of mRNA for targets of HH signaling, Gli1, Ptch1, and Hhip, were elevated in reproductive tracts of 24-day-old mutant mice, confirming overactivation of HH signaling. The tracts of mutant mice developed abnormally. The uterine luminal epithelium had a simple columnar morphology in control mice, but in mutants contained stratified squamous cells typical of the cervix and vagina. In mutant mice, the number of uterine glands were reduced and the oviducts were not coiled. Expression of genes within the Hox and Wnt families that regulate patterning of the reproductive tract were altered. Hoxa13, which is normally expressed primarily in the vagina and cervix, was expressed at 12-fold higher levels in the uterus of mutant mice compared with controls. Wnt5a, which is required for development of the cervix and vagina and postnatal differentiation of the uterus, was expressed at higher levels in the oviduct and uterus of mutant mice compared with controls. Mating mutant females with fertile or vasectomized males induced a severe inflammatory response in the tract. In summary, overactivation of HH signaling causes aberrant development of the reproductive tract. The phenotype observed could be mediated by ectopic expression of Hoxa13 in the uterus and elevated levels of Wnt5a in the oviducts and uterus.


Asunto(s)
Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Reproducción/genética , Transducción de Señal/genética , Alelos , Animales , Diferenciación Celular , Epitelio/metabolismo , Femenino , Regulación del Desarrollo de la Expresión Génica , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Masculino , Ratones , Ratones Noqueados , Conductos Paramesonéfricos/metabolismo , Mutación , Ovario/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Útero/metabolismo , Proteínas Wnt/genética , Proteínas Wnt/metabolismo , Proteína Wnt-5a
10.
Am J Pathol ; 177(5): 2645-58, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20864684

RESUMEN

The cell of origin and pathogenesis of the majority of adult soft tissue sarcomas (STS) remains poorly understood. Because mutations in both the P53 and RB tumor suppressor genes are frequent in STS in humans, we inactivated these genes by Cre-loxP-mediated recombination in mice with floxed p53 and Rb. Ninety-three percent of mice developed spindle cell/pleomorphic sarcomas after a single subcutaneous injection of adenovirus carrying Cre-recombinase. Similar to human STS, these sarcomas overexpress Cxcr4, which contributes to their invasive properties. Using irradiation chimeras generated by transplanting bone marrow cells from mice carrying either the Rosa26StoploxPLacZ or the Z/EG reporter, as well as the floxed p53 and Rb genes, into irradiated p53loxP/loxPRbloxP/loxP mice, it was determined that sarcomas do not originate from bone marrow-derived cells, such as macrophages, but arise from the local resident cells. At the same time, dermal mesenchymal stem cells isolated by strict plastic adherence and low levels of Sca-1 expression (Sca-1low, CD31negCD45neg) have shown enhanced potential for malignant transformation according to soft agar, invasion, and tumorigenicity assays, after the conditional inactivation of both p53 and Rb. Sarcomas formed after transplantation of these cells have features typical for undifferentiated high-grade pleomorphic sarcomas. Taken together, our studies indicate that local Sca-1low dermal mesenchymal stem/progenitor cells are preferential targets for malignant transformation associated with deficiencies in both p53 and Rb.


Asunto(s)
Células Madre Mesenquimatosas/patología , Proteína de Retinoblastoma/metabolismo , Sarcoma/genética , Sarcoma/patología , Proteína p53 Supresora de Tumor/metabolismo , Adulto , Animales , Transformación Celular Neoplásica , Células Cultivadas , Humanos , Masculino , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones Transgénicos , Quimera por Radiación , Proteína de Retinoblastoma/genética , Proteína p53 Supresora de Tumor/genética
11.
Proc Natl Acad Sci U S A ; 105(21): 7546-51, 2008 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-18495924

RESUMEN

Retinoic acid (RA) displays potent anticarcinogenic activities that are mediated by the nuclear retinoic acid receptors (RARs). However, use of RA in oncology is limited by RA resistance acquired during carcinogenesis. Moreover, in some cancers, RA facilitates rather than inhibits growth. A clue to this paradoxical behavior was recently suggested by the findings that RA also activates PPARbeta/delta, a receptor involved in mitogenic and anti-apoptotic activities. The observations that partitioning of RA between its two receptors is regulated by two intracellular lipid-binding proteins-CRABP-II, which targets RA to RAR, and FABP5, which delivers it to PPARbeta/delta-further suggest that RA resistance may stem from the deregulation of the binding proteins, resulting in activation of PPARbeta/delta rather than RAR. Here, we show that, in the RA-resistant mouse model of breast cancer MMTV-neu, RA indeed activates the nonclassical RA receptor PPARbeta/delta. This behavior was traced to an aberrantly high intratumor FABP5/CRABP-II ratio. Decreasing this ratio in mammary tissue diverted RA from PPARbeta/delta to RAR and suppressed tumor growth. The data demonstrate the existence of a mechanism that underlies RA resistance in tumors, indicate that CRABP-II functions as a tumor suppressor, and suggest that the inhibition of FABP5 may comprise a therapeutic strategy for overcoming RA resistance in some tumors.


Asunto(s)
Antineoplásicos/farmacología , Carcinoma/metabolismo , Resistencia a Antineoplásicos , Proteínas de Unión a Ácidos Grasos/metabolismo , Neoplasias Mamarias Animales/metabolismo , Proteínas de Neoplasias/metabolismo , Receptores de Ácido Retinoico/agonistas , Tretinoina/farmacología , Animales , Línea Celular Tumoral , Modelos Animales de Enfermedad , Proteínas de Unión a Ácidos Grasos/antagonistas & inhibidores , Ratones , Ratones Transgénicos , Proteínas de Neoplasias/antagonistas & inhibidores , PPAR delta/agonistas , PPAR-beta/agonistas , Receptores de Ácido Retinoico/genética , Receptores de Ácido Retinoico/metabolismo , Transcripción Genética/efectos de los fármacos
12.
Toxicol Pathol ; 38(1): 62-71, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19920280

RESUMEN

The cancer stem cell concept assumes that cancers are mainly sustained by a small pool of neoplastic cells, known as cancer stem cells or tumor initiating cells, which are able to reproduce themselves and produce phenotypically heterogeneous cells with lesser tumorigenic potential. Cancer stem cells represent an appealing target for development of more selective and efficient therapies. However, direct testing of the cancer stem cell concept and assessment of its therapeutic implications in human cancers have been complicated by the use of immunocompromised mice. Genetically defined immunocompetent autochthonous mouse models of human cancer provide a valuable tool to address this problem. Furthermore, they allow for a better understanding of the relevance of mechanisms controlling normal stem cell compartment to carcinogenesis. Advantages and disadvantages of some of the existing mouse models are reviewed, and future challenges in cancer stem cell research are outlined.


Asunto(s)
Modelos Animales de Enfermedad , Células Madre Neoplásicas/patología , Animales , Neoplasias Encefálicas/patología , Neoplasias de la Mama/patología , Femenino , Humanos , Neoplasias Intestinales/patología , Masculino , Ratones , Trasplante de Neoplasias , Neoplasias Ováricas/patología , Neoplasias de la Próstata/patología , Trasplante Heterólogo
13.
Cell Rep ; 32(9): 108086, 2020 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-32877668

RESUMEN

High-grade serous ovarian carcinoma (HGSOC) is the fifth leading cause of cancer-related deaths of women in the United States. Disease-associated mutations have been identified by the Cancer Genome Atlas Research Network. However, aside from mutations in TP53 or the RB1 pathway that are common in HGSOC, the contributions of mutation combinations are unclear. Here, we report CRISPR mutagenesis of 20 putative HGSOC driver genes to identify combinatorial disruptions of genes that transform either ovarian surface epithelium stem cells (OSE-SCs) or non-stem cells (OSE-NSs). Our results support the OSE-SC theory of HGSOC initiation and suggest that most commonly mutated genes in HGSOC have no effect on OSE-SC transformation initiation. Our results indicate that disruption of TP53 and PTEN, combined with RB1 disruption, constitutes a core set of mutations driving efficient transformation in vitro. The combined data may contribute to more accurate modeling of HGSOC development.


Asunto(s)
Células Epiteliales/metabolismo , Neoplasias Ováricas/genética , Neoplasias Ováricas/metabolismo , Células Madre/metabolismo , Femenino , Humanos , Mutación , Clasificación del Tumor
14.
Dis Model Mech ; 13(10)2020 10 30.
Artículo en Inglés | MEDLINE | ID: mdl-32998907

RESUMEN

Humans and mice have cyclical regeneration of the endometrial epithelium. It is expected that such regeneration is ensured by tissue stem cells, but their location and hierarchy remain debatable. A number of recent studies have suggested the presence of stem cells in the mouse endometrial epithelium. At the same time, it has been reported that this tissue can be regenerated by stem cells of stromal/mesenchymal or bone marrow cell origin. Here, we describe a single-cell transcriptomic atlas of the main cell types of the mouse uterus and epithelial subset transcriptome and evaluate the contribution of epithelial cells expressing the transcription factor PAX8 to the homeostatic regeneration and malignant transformation of adult endometrial epithelium. According to lineage tracing, PAX8+ epithelial cells are responsible for long-term maintenance of both luminal and glandular epithelium. Furthermore, multicolor tracing shows that individual glands and contiguous areas of luminal epithelium are formed by clonal cell expansion. Inactivation of the tumor suppressor genes Trp53 and Rb1 in PAX8+ cells, but not in FOXJ1+ cells, leads to the formation of neoplasms with features of serous endometrial carcinoma, one of the most aggressive types of human endometrial malignancies. Taken together, our results show that the progeny of single PAX8+ cells represents the main source of regeneration of the adult endometrial epithelium. They also provide direct experimental genetic evidence for the key roles of the P53 and RB pathways in the pathogenesis of serous endometrial carcinoma and suggest that PAX8+ cells represent the cell of origin of this neoplasm.


Asunto(s)
Neoplasias Endometriales/patología , Endometrio/patología , Epitelio/patología , Homeostasis , Neoplasias Quísticas, Mucinosas y Serosas/patología , Factor de Transcripción PAX8/metabolismo , Regeneración , Envejecimiento , Animales , Proliferación Celular , Modelos Animales de Enfermedad , Neoplasias Endometriales/genética , Células Epiteliales/metabolismo , Células Epiteliales/patología , Epitelio/metabolismo , Femenino , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Perfilación de la Expresión Génica , Inmunofenotipificación , Integrasas/metabolismo , Ratones Transgénicos , Neoplasias Quísticas, Mucinosas y Serosas/genética , Factor de Transcripción PAX8/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Útero/metabolismo
15.
Oncogenesis ; 9(3): 38, 2020 Mar 23.
Artículo en Inglés | MEDLINE | ID: mdl-32205838

RESUMEN

Aberrant neuroendocrine signaling is frequent yet poorly understood feature of prostate cancers. Membrane metalloendopeptidase (MME) is responsible for the catalytic inactivation of neuropeptide substrates, and is downregulated in nearly 50% of prostate cancers. However its role in prostate carcinogenesis, including formation of castration-resistant prostate carcinomas, remains uncertain. Here we report that MME cooperates with PTEN in suppression of carcinogenesis by controlling activities of prostate stem/progenitor cells. Lack of MME and PTEN results in development of adenocarcinomas characterized by propensity for vascular invasion and formation of proliferative neuroendocrine clusters after castration. Effects of MME on prostate stem/progenitor cells depend on its catalytic activity and can be recapitulated by addition of the MME substrate, gastrin-releasing peptide (GRP). Knockdown or inhibition of GRP receptor (GRPR) abrogate effects of MME deficiency and delay growth of human prostate cancer xenografts by reducing the number of cancer-propagating cells. In sum, our study provides a definitive proof of tumor-suppressive role of MME, links GRP/GRPR signaling to the control of prostate stem/progenitor cells, and shows how dysregulation of such signaling may promote formation of castration-resistant prostate carcinomas. It also identifies GRPR as a valuable target for therapies aimed at eradication of cancer-propagating cells in prostate cancers with MME downregulation.

16.
Sci Rep ; 10(1): 9837, 2020 06 17.
Artículo en Inglés | MEDLINE | ID: mdl-32555344

RESUMEN

Many high-grade serous carcinomas (HGSCs) likely originate in the distal region of the Fallopian tube's epithelium (TE) before metastasizing to the ovary. Unfortunately, molecular mechanisms promoting malignancy in the distal TE are obfuscated, largely due to limited primary human TE gene expression data. Here we report an in depth bioinformatic characterization of 34 primary TE mRNA-seq samples. These samples were prepared from proximal and distal TE regions of 12 normal Fallopian tubes. Samples were segregated based on their aldehyde dehydrogenase (ALDH) activity. Distal cells form organoids with higher frequency and larger size during serial organoid formation assays when compared to proximal cells. Consistent with enrichment for stem/progenitor cells, ALDH+ cells have greater WNT signaling. Comparative evaluation of proximal and distal TE cell population's shows heightened inflammatory signaling in distal differentiated (ALDH-) TE. Furthermore, comparisons of proximal and distal TE cell populations finds that the distal ALDH+ TE cells exhibit pronounced expression of gene sets characteristic of HGSC sub-types. Overall, our study indicates increased organoid forming capacity, WNT/inflammatory signaling, and HGSC signatures underlie differences between distal and proximal regions of the human TE. These findings provide the basis for further mechanistic studies of distal TE susceptibility to the malignant transformation.


Asunto(s)
Biología Computacional , Células Epiteliales/citología , Trompas Uterinas/citología , Vía de Señalización Wnt , Aldehído Deshidrogenasa/metabolismo , Diferenciación Celular , Células Epiteliales/patología , Trompas Uterinas/patología , Femenino , Regulación de la Expresión Génica , Humanos , Inflamación/patología
17.
Nat Commun ; 11(1): 84, 2020 01 03.
Artículo en Inglés | MEDLINE | ID: mdl-31901081

RESUMEN

Areas of a junction between two types of epithelia are known to be cancer-prone in many organ systems. However, mechanisms for preferential malignant transformation at the junction areas remain insufficiently elucidated. Here we report that inactivation of tumor suppressor genes Trp53 and Rb1 in the gastric squamous-columnar junction (SCJ) epithelium results in preferential formation of metastatic poorly differentiated neoplasms, which are similar to human gastroesophageal carcinoma. Unlike transformation-resistant antral cells, SCJ cells contain a highly proliferative pool of immature Lgr5-CD44+ cells, which are prone to transformation in organoid assays, comprise early dysplastic lesions, and constitute up to 30% of all neoplastic cells. CD44 ligand osteopontin (OPN) is preferentially expressed in and promotes organoid formation ability and transformation of the SCJ glandular epithelium. OPN and CD44 overexpression correlate with the worst prognosis of human gastroesophageal carcinoma. Thus, detection and selective targeting of the active OPN-CD44 pathway may have direct clinical relevance.


Asunto(s)
Carcinoma de Células Escamosas/metabolismo , Unión Esofagogástrica/metabolismo , Receptores de Hialuranos/metabolismo , Osteopontina/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Neoplasias Gástricas/metabolismo , Anciano , Anciano de 80 o más Años , Animales , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/mortalidad , Carcinoma de Células Escamosas/patología , Transformación Celular Neoplásica , Estudios de Cohortes , Unión Esofagogástrica/patología , Femenino , Humanos , Receptores de Hialuranos/genética , Masculino , Ratones , Ratones Noqueados , Persona de Mediana Edad , Osteopontina/genética , Receptores Acoplados a Proteínas G/genética , Proteína de Retinoblastoma/genética , Proteína de Retinoblastoma/metabolismo , Neoplasias Gástricas/genética , Neoplasias Gástricas/mortalidad , Neoplasias Gástricas/patología , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
18.
Cancer Res ; 67(12): 5683-90, 2007 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-17553900

RESUMEN

Recently, we have shown that prostate epithelium-specific deficiency for p53 and Rb tumor suppressors leads to metastatic cancer, exhibiting features of both luminal and neuroendocrine differentiation. Using stage-by-stage evaluation of carcinogenesis in this model, we report that all malignant neoplasms arise from the proximal region of the prostatic ducts, the compartment highly enriched for prostatic stem/progenitor cells. In close similarity to reported properties of prostatic stem cells, the cells of the earliest neoplastic lesions express stem cell marker stem cell antigen 1 and are not sensitive to androgen withdrawal. Like a subset of normal cells located in the proximal region of prostatic ducts, the early neoplastic cells coexpress luminal epithelium markers cytokeratin 8, androgen receptor, and neuroendocrine markers synaptophysin and chromogranin A. Inactivation of p53 and Rb also takes place in the lineage-committed transit-amplifying and/or differentiated cells of the distal region of the prostatic ducts. However, the resulting prostatic intraepithelial neoplasms never progress to carcinoma by the time of mouse death. Interestingly, in an ectopic transplantation assay, early mutant cells derived from either region of the prostatic ducts are capable of forming neoplasms within 3 months. These findings indicate that p53 and Rb are critically important for the regulation of the prostatic stem cell compartment, the transformation in which may lead to particularly aggressive cancers in the context of microenvironment.


Asunto(s)
Transformación Celular Neoplásica , Neoplasias de la Próstata/patología , Proteína de Retinoblastoma/deficiencia , Células Madre/fisiología , Proteína p53 Supresora de Tumor/deficiencia , Animales , Inmunohistoquímica , Masculino , Ratones , Ratones Noqueados , Reacción en Cadena de la Polimerasa , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/metabolismo
19.
Cancer Res ; 67(18): 8433-8, 2007 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-17823410

RESUMEN

MicroRNAs (miRNA) are a recently discovered class of noncoding RNAs that negatively regulate gene expression. Recent evidence indicates that miRNAs may play an important role in cancer. However, the mechanism of their deregulation in neoplastic transformation has only begun to be understood. To elucidate the role of tumor suppressor p53 in regulation of miRNAs, we have analyzed changes in miRNA microarray expression profile immediately after conditional inactivation of p53 in primary mouse ovarian surface epithelium cells. Among the most significantly affected miRNAs were miR-34b and miR-34c, which were down-regulated 12-fold according to quantitative reverse transcription-PCR analysis. Computational promoter analysis of the mir-34b/mir-34c locus identified the presence of evolutionarily conserved p53 binding sites approximately 3 kb upstream of the miRNA coding sequence. Consistent with evolutionary conservation, mir-34b/mir-34c were also down-regulated in p53-null human ovarian carcinoma cells. Furthermore, as expected from p53 binding to the mir-34b/c promoter, doxorubicin treatment of wild-type, but not p53-deficient, cells resulted in an increase of mir-34b/mir-34c expression. Importantly, miR-34b and miR-34c cooperate in suppressing proliferation and soft-agar colony formation of neoplastic epithelial ovarian cells, in agreement with the partially overlapping spectrum of their predicted targets. Taken together, these results show the existence of a novel mechanism by which p53 suppresses such critical components of neoplastic growth as cell proliferation and adhesion-independent colony formation.


Asunto(s)
MicroARNs/genética , Neoplasias Ováricas/genética , Neoplasias Ováricas/patología , Animales , Secuencia de Bases , Adhesión Celular/genética , Procesos de Crecimiento Celular/genética , Línea Celular Tumoral , Femenino , Regulación Neoplásica de la Expresión Génica , Silenciador del Gen , Genes p53 , Humanos , Ratones , Análisis de Secuencia por Matrices de Oligonucleótidos , Ovario/citología , Ovario/metabolismo , Ovario/fisiología , Regiones Promotoras Genéticas , Transcripción Genética , Transfección , Proteína p53 Supresora de Tumor/biosíntesis , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
20.
Cancer Res ; 67(15): 7525-33, 2007 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-17671224

RESUMEN

The application of Cre/loxP technology has resulted in a new generation of conditional mouse models of prostate cancer. Here, we describe the improvement of the conditional Pten deletion model of prostate adenocarcinoma by combining it with either a conditional luciferase or enhanced green fluorescent protein reporter line. In these models, the recombination mechanism that inactivates the Pten alleles also activates the reporter gene. In the luciferase reporter model, the growth of the primary cancer can be followed noninvasively by bioluminescence imaging (BLI). Surgical castration of tumor-bearing animals leads to a reduced bioluminescence signal corresponding to tumor regression that is verified at necropsy. When castrated animals are maintained, the emergence of androgen depletion-independent cancer is detected using BLI at times varying from 7 to 28 weeks postcastration. The ability to monitor growth, regression, or relapse of the tumor with the use of BLI lead to the collection of tumors at different stages of development. By comparing the distribution of phenotypically distinct populations of epithelial cells in cancer tissues, we noted that the degree of hyperplasia of cells with neuroendocrine differentiation significantly increases in the recurrent cancer relative to the primary cancer, a characteristic which may parallel the appearance of a neuroendocrine phenotype in human androgen depletion-independent cancer. The enhanced green fluorescent protein model, at necropsy, can provide an opportunity to locate or assess tumor volume or to isolate enriched populations of cancer cells from tumor tissues via fluorescence-based technologies. These refined models should be useful in the elucidation of mechanisms of prostate cancer progression, and for the development of approaches to preclinical intervention.


Asunto(s)
Técnica del Anticuerpo Fluorescente , Interpretación de Imagen Asistida por Computador , Mediciones Luminiscentes , Neoplasias Pulmonares/diagnóstico , Recurrencia Local de Neoplasia/diagnóstico , Neoplasias de la Próstata/patología , Animales , Línea Celular Tumoral , Proteínas Fluorescentes Verdes , Humanos , Luciferasas , Neoplasias Pulmonares/secundario , Metástasis Linfática/diagnóstico , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Fosfohidrolasa PTEN/genética , Fosfohidrolasa PTEN/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA