Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
2.
Nat Commun ; 10(1): 5494, 2019 12 02.
Artículo en Inglés | MEDLINE | ID: mdl-31792211

RESUMEN

LRIG1 has been reported to be a tumor suppressor in gastrointestinal tract and epidermis. However, little is known about the expression, regulation and biological functions of LRIG1 in prostate cancer (PCa). We find that LRIG1 is overexpressed in PCa, but its expression correlates with better patient survival. Functional studies reveal strong tumor-suppressive functions of LRIG1 in both AR+ and AR- xenograft models, and transgenic expression of LRIG1 inhibits tumor development in Hi-Myc and TRAMP models. LRIG1 also inhibits castration-resistant PCa and exhibits therapeutic efficacy in pre-established tumors. We further show that 1) AR directly transactivates LRIG1 through binding to several AR-binding sites in LRIG1 locus, and 2) LRIG1 dampens ERBB expression in a cell type-dependent manner and inhibits ERBB2-driven tumor growth. Collectively, our study indicates that LRIG1 represents a pleiotropic AR-regulated feedback tumor suppressor that functions to restrict oncogenic signaling from AR, Myc, ERBBs, and, likely, other oncogenic drivers.


Asunto(s)
Glicoproteínas de Membrana/metabolismo , Neoplasias de la Próstata/metabolismo , Receptores Androgénicos/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Animales , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Glicoproteínas de Membrana/genética , Ratones Endogámicos NOD , Ratones SCID , Proteína Oncogénica p55(v-myc)/genética , Proteína Oncogénica p55(v-myc)/metabolismo , Neoplasias de la Próstata/genética , Unión Proteica , Receptor ErbB-2/genética , Receptor ErbB-2/metabolismo , Receptores Androgénicos/genética , Transducción de Señal , Proteínas Supresoras de Tumor/genética
3.
J Sch Health ; 89(12): 969-976, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-31612505

RESUMEN

BACKGROUND: Evidence suggests a rise in childhood disability rates across the United States with males and those with lower socioeconomic status bearing greater burden. We investigated childhood disability rates in the Mississippi Delta (MDR) and Appalachian regions (AR) in comparison to other parts of the country. METHODS: Using data from the US Census Bureau, we calculated childhood disability rates by type and sex at national, regional, and subregional levels. We used risk ratios (RR) to compare childhood disability rates by sex, type, and region. We generated choropleth maps to represent the geographic distribution of disability. RESULTS: Childhood disability was more prevalent, at the national level, among boys (6.64%) than girls (4.08%). Children in the MDR (boys = 8.60%; girls = 5.08%) and AR (boys = 7.81%; girls = 4.83%) had greater risk of disability than those elsewhere in the country (boys = 6.47%; girls = 3.98%), with rates generally higher in rural compared to urban areas in said regions. CONCLUSIONS: Childhood disability affects rural areas of the country more extensively, with the MDR and AR affected to an even greater extent. School-based health centers, in particular, which are disproportionately located in urban areas, could benefit disabled children living in the MDR and AR.


Asunto(s)
Niños con Discapacidad , Adolescente , Región de los Apalaches/epidemiología , Niño , Preescolar , Bases de Datos Factuales , Niños con Discapacidad/estadística & datos numéricos , Estudios Epidemiológicos , Femenino , Disparidades en el Estado de Salud , Encuestas Epidemiológicas , Humanos , Masculino , Mississippi , Prevalencia , Distribución por Sexo , Estados Unidos
4.
AIDS Behav ; 23(11): 2936-2945, 2019 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-31321638

RESUMEN

This project established a faith-based, university-community partnership with the African Methodist Episcopal (AME) church in Alabama to develop a statewide training model to address HIV knowledge and stigma, promote discussion and generate action plans to address HIV in the Deep South. A community-engaged research team consisting of church leadership and university researchers developed and implemented the model, "Love with No Exceptions." Mixed methods were used to evaluate the model delivered in 3-h sessions in five state regions (N = 146 clergy and laity). The majority of participants reported feeling better prepared to serve those living with or affected by HIV and would implement education and awareness activities in their churches. Participants' HIV knowledge increased from pre- to post-training. Stigma-related attitudes showed minor changes from baseline. These results reflect that partnerships between academic institutions and churches can deliver promising steps towards impactful HIV education in the Deep South.


Asunto(s)
Negro o Afroamericano/psicología , Clero , Infecciones por VIH/prevención & control , Infecciones por VIH/psicología , Educación en Salud/métodos , Conocimientos, Actitudes y Práctica en Salud , Religión , Estigma Social , Adulto , Anciano , Alabama , Creación de Capacidad , Investigación Participativa Basada en la Comunidad , Discriminación en Psicología , Femenino , Infecciones por VIH/etnología , Promoción de la Salud/métodos , Humanos , Liderazgo , Amor , Masculino , Persona de Mediana Edad , Estados Unidos , Universidades
5.
Nat Commun ; 9(1): 3600, 2018 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-30190514

RESUMEN

Expression of androgen receptor (AR) in prostate cancer (PCa) is heterogeneous but the functional significance of AR heterogeneity remains unclear. Screening ~200 castration-resistant PCa (CRPC) cores and whole-mount sections (from 89 patients) reveals 3 AR expression patterns: nuclear (nuc-AR), mixed nuclear/cytoplasmic (nuc/cyto-AR), and low/no expression (AR-/lo). Xenograft modeling demonstrates that AR+ CRPC is enzalutamide-sensitive but AR-/lo CRPC is resistant. Genome editing-derived AR+ and AR-knockout LNCaP cell clones exhibit distinct biological and tumorigenic properties and contrasting responses to enzalutamide. RNA-Seq and biochemical analyses, coupled with experimental combinatorial therapy, identify BCL-2 as a critical therapeutic target and provide proof-of-concept therapeutic regimens for both AR+/hi and AR-/lo CRPC. Our study links AR expression heterogeneity to distinct castration/enzalutamide responses and has important implications in understanding the cellular basis of prostate tumor responses to AR-targeting therapies and in facilitating development of novel therapeutics to target AR-/lo PCa cells/clones.


Asunto(s)
Feniltiohidantoína/análogos & derivados , Neoplasias de la Próstata Resistentes a la Castración/tratamiento farmacológico , Neoplasias de la Próstata Resistentes a la Castración/genética , Receptores Androgénicos/genética , Animales , Antineoplásicos/farmacología , Benzamidas , Línea Celular Tumoral , Resistencia a Antineoplásicos/efectos de los fármacos , Resistencia a Antineoplásicos/genética , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Ratones Endogámicos NOD , Ratones Noqueados , Terapia Molecular Dirigida , Nitrilos , Feniltiohidantoína/farmacología , Neoplasias de la Próstata Resistentes a la Castración/patología , Proteínas Proto-Oncogénicas c-bcl-2/genética , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Receptores Androgénicos/metabolismo , Transducción de Señal , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Stem Cells Transl Med ; 6(3): 748-760, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-28297567

RESUMEN

Elucidating the cell of origin of cancer has great significance in stratifying patients into appropriate treatment groups and for developing novel targeted therapies. Early studies demonstrate that only stem-like basal cells in the normal human prostate (NHP) can function as the cell of origin for prostate cancer (PCa). Here, we show that the organoids derived from bulk NHP luminal cells can also be tumorigenically transformed. We further show that the WIT medium, which is used to culture human mammary epithelial progenitor cells, when combined with the ROCK inhibitor, can readily propagate a population of progenitor-like cells from the primary NHP luminal cell isolates. Such functionally defined luminal progenitors can be transformed by distinct sets of genetic perturbations (i.e., AR+AKT/ERG or c-MYC+PTEN knockout) to form tumor glands. Genome-wide RNA-Seq analysis of freshly purified unperturbed human benign prostatic basal and luminal cells and culture-expanded lineage-specific stem/progenitor populations reveals that the luminal progenitors possess a distinct gene expression profile that is greatly enriched in advanced, castration-resistant, and metastatic PCa, and it associates with poor patient survival. The ability of the simple two-dimensional culture system reported herein to greatly enrich NHP progenitor-like cells should facilitate biological and biochemical studies as well as high-throughput screening in these cells and in progenitor-like PCa cells. Stem Cells Translational Medicine 2017;6:748-760.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Próstata/patología , Neoplasias de la Próstata/patología , Células Madre/patología , Castración , Linaje de la Célula , Transformación Celular Neoplásica/patología , Células Cultivadas , Medios de Cultivo , Células Epiteliales/metabolismo , Células Epiteliales/patología , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Invasividad Neoplásica , Metástasis de la Neoplasia , Organoides/patología , Próstata/metabolismo , Neoplasias de la Próstata/genética , Regeneración , Células Madre/metabolismo , Análisis de Supervivencia , Transcriptoma/genética
7.
Cell Discov ; 2: 16041, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27867534

RESUMEN

The pluripotency transcription factor NANOG has been implicated in tumor development, and NANOG-expressing cancer cells manifest stem cell properties that sustain tumor homeostasis, mediate therapy resistance and fuel tumor progression. However, how NANOG converges on somatic circuitry to trigger oncogenic reprogramming remains obscure. We previously reported that inducible NANOG expression propels the emergence of aggressive castration-resistant prostate cancer phenotypes. Here we first show that endogenous NANOG is required for the growth of castration-resistant prostate cancer xenografts. Genome-wide chromatin immunoprecipitation sequencing coupled with biochemical assays unexpectedly reveals that NANOG co-occupies a distinctive proportion of androgen receptor/Forkhead box A1 genomic loci and physically interacts with androgen receptor and Forkhead box A1. Integrative analysis of chromatin immunoprecipitation sequencing and time-resolved RNA sequencing demonstrates that NANOG dynamically alters androgen receptor/Forkhead box A1 signaling leading to both repression of androgen receptor-regulated pro-differentiation genes and induction of genes associated with cell cycle, stem cells, cell motility and castration resistance. Our studies reveal global molecular mechanisms whereby NANOG reprograms prostate cancer cells to a clinically relevant castration-resistant stem cell-like state driven by distinct NANOG-regulated gene clusters that correlate with patient survival. Thus, reprogramming factors such as NANOG may converge on and alter lineage-specific master transcription factors broadly in somatic cancers, thereby facilitating malignant disease progression and providing a novel route for therapeutic resistance.

8.
Oncotarget ; 7(35): 56628-56642, 2016 Aug 30.
Artículo en Inglés | MEDLINE | ID: mdl-27447749

RESUMEN

Human cancers exhibit significant cellular heterogeneity featuring tumorigenic cancer stem cells (CSCs) in addition to more differentiated progeny with limited tumor-initiating capabilities. Recent studies suggest that microRNAs (miRNAs) regulate CSCs and tumor development. A previous library screening for differential miRNA expression in CD44+ (and other) prostate CSC vs. non-CSC populations identified miR-199a-3p to be among the most highly under-expressed miRNAs in CSCs. In this study, we characterized the biological functions of miR-199a-3p in CD44+ prostate cancer (PCa) cells and in tumor regeneration. Overexpression of miR-199a-3p in purified CD44+ or bulk PCa cells, including primary PCa, inhibited proliferation and clonal expansion without inducing apoptosis. miR-199a-3p overexpression also diminished tumor-initiating capacities of CD44+ PCa cells as well as tumor regeneration from bulk PCa cells. Importantly, inducible miR-199a-3p expression in pre-established prostate tumors in NOD/SCID mice inhibited tumor growth. Using target prediction program and luciferase assays, we show mechanistically that CD44 is a direct functional target of miR-199a-3p in PCa cells. Moreover, miR-199a-3p also directly or indirectly targeted several additional mitogenic molecules, including c-MYC, cyclin D1 (CCND1) and EGFR. Taken together, our results demonstrate how the aberrant loss of a miRNA-mediated mechanism can lead to the expansion and tumorigenic activity of prostate CSCs, further supporting the development and implementation of miRNA mimics for cancer treatment.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , MicroARNs/metabolismo , Células Madre Neoplásicas/metabolismo , Neoplasias de la Próstata/metabolismo , Animales , Apoptosis/genética , Carcinogénesis/genética , Línea Celular Tumoral , Proliferación Celular , Ciclina D1/metabolismo , Receptores ErbB/metabolismo , Humanos , Receptores de Hialuranos/metabolismo , Masculino , Ratones , Ratones Endogámicos NOD , Ratones SCID , Trasplante de Neoplasias , Neoplasias de la Próstata/genética , Transducción de Señal
9.
Mol Cell ; 62(4): 558-71, 2016 05 19.
Artículo en Inglés | MEDLINE | ID: mdl-27132940

RESUMEN

Histone H2B monoubiquitination (H2Bub1) is centrally involved in gene regulation. The deubiquitination module (DUBm) of the SAGA complex is a major regulator of global H2Bub1 levels, and components of this DUBm are linked to both neurodegenerative diseases and cancer. Unexpectedly, we find that ablation of USP22, the enzymatic center of the DUBm, leads to a reduction, rather than an increase, in global H2bub1 levels. In contrast, depletion of non-enzymatic components, ATXN7L3 or ENY2, results in increased H2Bub1. These observations led us to discover two H2Bub1 DUBs, USP27X and USP51, which function independently of SAGA and compete with USP22 for ATXN7L3 and ENY2 for activity. Like USP22, USP51 and USP27X are required for normal cell proliferation, and their depletion suppresses tumor growth. Our results reveal that ATXN7L3 and ENY2 orchestrate activities of multiple deubiquitinating enzymes and that imbalances in these activities likely potentiate human diseases including cancer.


Asunto(s)
Neoplasias de la Mama/enzimología , Proliferación Celular , Enzimas Desubicuitinizantes/metabolismo , Histonas/metabolismo , Factores de Transcripción/metabolismo , Carga Tumoral , Animales , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Enzimas Desubicuitinizantes/genética , Endopeptidasas/genética , Endopeptidasas/metabolismo , Femenino , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Genotipo , Células HEK293 , Humanos , Células MCF-7 , Ratones Endogámicos NOD , Ratones Noqueados , Ratones SCID , Fenotipo , Interferencia de ARN , Transducción de Señal , Tioléster Hidrolasas/genética , Tioléster Hidrolasas/metabolismo , Factores de Tiempo , Factores de Transcripción/genética , Transfección , Ubiquitina Tiolesterasa , Proteasas Ubiquitina-Específicas/genética , Proteasas Ubiquitina-Específicas/metabolismo , Ubiquitinación
10.
Clin Cancer Res ; 22(17): 4505-16, 2016 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-27060154

RESUMEN

PURPOSE: We have shown that the phenotypically undifferentiated (PSA(-/lo)) prostate cancer cell population harbors long-term self-renewing cancer stem cells (CSC) that resist castration, and a subset of the cells within the PSA(-/lo) population bearing the ALDH(hi)CD44(+)α2ß1(+) phenotype (Triple Marker(+)/TM(+)) is capable of robustly initiating xenograft tumors in castrated mice. The goal of the current project is to further characterize the biologic properties of TM(+) prostate cancer cell population, particularly in the context of initiating and propagating castration-resistant prostate cancer (CRPC). EXPERIMENTAL DESIGN: The in vivo CSC activities were measured by limiting-dilution serial tumor transplantation assays in both androgen-dependent and androgen-independent prostate cancer xenograft models. In vitro clonal, clonogenic, and sphere-formation assays were conducted in cells purified from xenograft and patient tumors. qPCR, Western blot, lentiviral-mediated gene knockdown, and human microRNA arrays were performed for mechanistic studies. RESULTS: By focusing on the LAPC9 model, we show that the TM(+) cells are CSCs with both tumor-initiating and tumor-propagating abilities for CRPC. Moreover, primary patient samples have TM(+) cells, which possess CSC activities in "castrated" culture conditions. Mechanistically, we find that (i) the phenotypic markers are causally involved in CRPC development; (ii) the TM(+) cells preferentially express castration resistance and stem cell-associated molecules that regulate their CSC characteristics; and (iii) the TM(+) cells possess distinct microRNA expression profiles and miR-499-5p functions as an oncomir. CONCLUSIONS: Our results define the TM(+) prostate cancer cells as a population of preexistent stem-like cancer cells that can both mediate and propagate CRPC and highlight the TM(+) cell population as a therapeutic target. Clin Cancer Res; 22(17); 4505-16. ©2016 AACR.


Asunto(s)
Células Madre Neoplásicas/metabolismo , Neoplasias de la Próstata Resistentes a la Castración/metabolismo , Andrógenos/metabolismo , Animales , Biomarcadores de Tumor , Castración , Línea Celular Tumoral , Modelos Animales de Enfermedad , Perfilación de la Expresión Génica , Xenoinjertos , Humanos , Inmunofenotipificación , Masculino , Ratones , Ratones Noqueados , MicroARNs/genética , Células Madre Neoplásicas/patología , Fenotipo , Neoplasias de la Próstata Resistentes a la Castración/genética , Neoplasias de la Próstata Resistentes a la Castración/patología , Factor de Transcripción SOX9/genética , Factor de Transcripción SOX9/metabolismo , Células Tumorales Cultivadas
11.
Nat Commun ; 7: 10798, 2016 Feb 29.
Artículo en Inglés | MEDLINE | ID: mdl-26924072

RESUMEN

The prostate gland mainly contains basal and luminal cells constructed as a pseudostratified epithelium. Annotation of prostate epithelial transcriptomes provides a foundation for discoveries that can impact disease understanding and treatment. Here we describe a genome-wide transcriptome analysis of human benign prostatic basal and luminal epithelial populations using deep RNA sequencing. Through molecular and biological characterizations, we show that the differential gene-expression profiles account for their distinct functional properties. Strikingly, basal cells preferentially express gene categories associated with stem cells, neurogenesis and ribosomal RNA (rRNA) biogenesis. Consistent with this profile, basal cells functionally exhibit intrinsic stem-like and neurogenic properties with enhanced rRNA transcription activity. Of clinical relevance, the basal cell gene-expression profile is enriched in advanced, anaplastic, castration-resistant and metastatic prostate cancers. Therefore, we link the cell-type-specific gene signatures to aggressive subtypes of prostate cancer and identify gene signatures associated with adverse clinical features.


Asunto(s)
Regulación de la Expresión Génica/fisiología , Próstata/citología , Neoplasias de la Próstata/metabolismo , Células Madre/fisiología , Transcriptoma , Adenocarcinoma/metabolismo , Biomarcadores , Linaje de la Célula , Técnicas de Silenciamiento del Gen , Humanos , Masculino , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas Proto-Oncogénicas c-myc/metabolismo , ARN Ribosómico/genética , ARN Ribosómico/metabolismo , Ribosomas/metabolismo
12.
Oncotarget ; 6(27): 23959-86, 2015 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-26246472

RESUMEN

Human cancers are heterogeneous containing stem-like cancer cells operationally defined as cancer stem cells (CSCs) that possess great tumor-initiating and long-term tumor-propagating properties. In this study, we systematically dissect the phenotypic, functional and tumorigenic heterogeneity in human prostate cancer (PCa) using xenograft models and >70 patient tumor samples. In the first part, we further investigate the PSA-/lo PCa cell population, which we have recently shown to harbor self-renewing long-term tumor-propagating cells and present several novel findings. We show that discordant AR and PSA expression in both untreated and castration-resistant PCa (CRPC) results in AR+PSA+, AR+PSA-, AR-PSA-, and AR-PSA+ subtypes of PCa cells that manifest differential sensitivities to therapeutics. We further demonstrate that castration leads to a great enrichment of PSA-/lo PCa cells in both xenograft tumors and CRPC samples and systemic androgen levels dynamically regulate the relative abundance of PSA+ versus PSA-/lo PCa cells that impacts the kinetics of tumor growth. We also present evidence that the PSA-/lo PCa cells possess distinct epigenetic profiles. As the PSA-/lo PCa cell population is heterogeneous, in the second part, we employ two PSA- (Du145 and PC3) and two PSA+ (LAPC9 and LAPC4) PCa models as well as patient tumor cells to further dissect the clonogenic and tumorigenic subsets. We report that different PCa models possess distinct tumorigenic subpopulations that both commonly and uniquely express important signaling pathways that could represent therapeutic targets. Our results have important implications in understanding PCa cell heterogeneity, response to clinical therapeutics, and cellular mechanisms underlying CRPC.


Asunto(s)
Neoplasias de la Próstata/metabolismo , Animales , Antineoplásicos/química , Apoptosis , Línea Celular Tumoral , Epigénesis Genética , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Ratones , Trasplante de Neoplasias , Células Madre Neoplásicas/citología , Fenotipo , Regiones Promotoras Genéticas , Antígeno Prostático Específico/sangre , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/patología , ARN Mensajero/metabolismo , Receptores Androgénicos/metabolismo , Transducción de Señal
13.
PLoS One ; 8(2): e56903, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23451107

RESUMEN

Reconstitution of tumor development in immunodeficient mice from disaggregated primary human tumor cells is always challenging. The main goal of the present study is to establish a reliable assay system that would allow us to reproducibly reconstitute human prostate tumor regeneration in mice using patient tumor-derived single cells. Using many of the 114 untreated primary human prostate cancer (HPCa) samples we have worked on, here we show that: 1) the subcutaneum represents the most sensitive site that allows the grafting of the implanted HPCa pieces; 2) primary HPCa cells by themselves fail to regenerate tumors in immunodeficient hosts; 3) when coinjected in Matrigel with rUGM (rat urogenital sinus mesenchyme), CAF (carcinoma-associated fibroblasts), or Hs5 (immortalized bone marrow derived stromal) cells, primary HPCa cells fail to initiate serially transplantable tumors in NOD/SCID mice; and 4) however, HPCa cells coinjected with the Hs5 cells into more immunodeficient NOD/SCID-IL2Rγ(-/-) (NSG) mice readily regenerate serially transplantable tumors. The HPCa/Hs5 reconstituted 'prostate' tumors present an overall epithelial morphology, are of the human origin, and contain cells positive for AR, CK8, and racemase. Cytogenetic analysis provides further evidence for the presence of karyotypically abnormal HPCa cells in the HPCa/Hs5 tumors. Of importance, HPCa/Hs5 xenograft tumors contain EpCAM(+) cells that are both clonogenic and tumorigenic. Surprisingly, all HPCa/Hs5 reconstituted tumors are undifferentiated, even for HPCa cells derived from Gleason 7 tumors. Our results indicate that primary HPCa cells coinjected with the immortalized Hs5 stromal cells generate undifferentiated tumors in NSG mice and we provide evidence that undifferentiated HPCa cells might be the cells that possessed tumorigenic potential and regenerated HPCa/Hs5 xenograft tumors.


Asunto(s)
Células Madre Mesenquimatosas/patología , Trasplante de Neoplasias/métodos , Neoplasias de la Próstata/patología , Animales , Línea Celular Tumoral , Modelos Animales de Enfermedad , Humanos , Masculino , Ratones , Ratones Endogámicos NOD , Ratones SCID , Trasplante Heterólogo
14.
Cell Stem Cell ; 10(5): 556-69, 2012 May 04.
Artículo en Inglés | MEDLINE | ID: mdl-22560078

RESUMEN

Prostate cancer (PCa) is heterogeneous and contains both differentiated and undifferentiated tumor cells, but the relative functional contribution of these two cell populations remains unclear. Here we report distinct molecular, cellular, and tumor-propagating properties of PCa cells that express high (PSA(+)) and low (PSA(-/lo)) levels of the differentiation marker PSA. PSA(-/lo) PCa cells are quiescent and refractory to stresses including androgen deprivation, exhibit high clonogenic potential, and possess long-term tumor-propagating capacity. They preferentially express stem cell genes and can undergo asymmetric cell division to generate PSA(+) cells. Importantly, PSA(-/lo) PCa cells can initiate robust tumor development and resist androgen ablation in castrated hosts, and they harbor highly tumorigenic castration-resistant PCa cells that can be prospectively enriched using ALDH(+)CD44(+)α2ß1(+) phenotype. In contrast, PSA(+) PCa cells possess more limited tumor-propagating capacity, undergo symmetric division, and are sensitive to castration. Altogether, our study suggests that PSA(-/lo) cells may represent a critical source of castration-resistant PCa cells.


Asunto(s)
Adenocarcinoma/patología , Antígenos de Diferenciación/metabolismo , Células Madre Neoplásicas/metabolismo , Antígeno Prostático Específico/metabolismo , Neoplasias de la Próstata/patología , Adenocarcinoma/diagnóstico , Adenocarcinoma/cirugía , Animales , División Celular Asimétrica , Castración , Diferenciación Celular , Línea Celular Tumoral , Supervivencia Celular , Transformación Celular Neoplásica , Humanos , Masculino , Ratones , Ratones Desnudos , Ratones SCID , Trasplante de Neoplasias , Células Madre Neoplásicas/clasificación , Células Madre Neoplásicas/patología , Neoplasias de la Próstata/diagnóstico , Neoplasias de la Próstata/cirugía
15.
PLoS One ; 6(9): e24397, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21935404

RESUMEN

Cancer stem cells (CSCs) possess high tumor-initiating capacity and have been reported to be resistant to therapeutics. Vice versa, therapy-resistant cancer cells seem to manifest CSC phenotypes and properties. It has been generally assumed that drug-resistant cancer cells may all be CSCs although the generality of this assumption is unknown. Here, we chronically treated Du145 prostate cancer cells with etoposide, paclitaxel and some experimental drugs (i.e., staurosporine and 2 paclitaxel analogs), which led to populations of drug-tolerant cells (DTCs). Surprisingly, these DTCs, when implanted either subcutaneously or orthotopically into NOD/SCID mice, exhibited much reduced tumorigenicity or were even non-tumorigenic. Drug-tolerant DLD1 colon cancer cells selected by a similar chronic selection protocol also displayed reduced tumorigenicity whereas drug-tolerant UC14 bladder cancer cells demonstrated either increased or decreased tumor-regenerating capacity. Drug-tolerant Du145 cells demonstrated low proliferative and clonogenic potential and were virtually devoid of CD44(+) cells. Prospective knockdown of CD44 in Du145 cells inhibited cell proliferation and tumor regeneration, whereas restoration of CD44 expression in drug-tolerant Du145 cells increased cell proliferation and partially increased tumorigenicity. Interestingly, drug-tolerant Du145 cells showed both increases and decreases in many "stemness" genes. Finally, evidence was provided that chronic drug exposure generated DTCs via epigenetic mechanisms involving molecules such as CD44 and KDM5A. Our results thus reveal that 1) not all DTCs are necessarily CSCs; 2) conventional chemotherapeutic drugs such as taxol and etoposide may directly target CD44(+) tumor-initiating cells; and 3) DTCs generated via chronic drug selection involve epigenetic mechanisms.


Asunto(s)
Antineoplásicos/uso terapéutico , Epigénesis Genética/genética , Receptores de Hialuranos/metabolismo , Células Madre Neoplásicas/citología , Células Madre Neoplásicas/metabolismo , Animales , Antineoplásicos/farmacología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Etopósido/farmacología , Etopósido/uso terapéutico , Técnica del Anticuerpo Fluorescente , Humanos , Receptores de Hialuranos/genética , Immunoblotting , Masculino , Ratones , Ratones SCID , Paclitaxel/farmacología , Paclitaxel/uso terapéutico , Proteína 2 de Unión a Retinoblastoma/genética , Proteína 2 de Unión a Retinoblastoma/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Estaurosporina/farmacología , Estaurosporina/uso terapéutico
16.
Nat Med ; 17(2): 211-5, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21240262

RESUMEN

Cancer stem cells (CSCs), or tumor-initiating cells, are involved in tumor progression and metastasis. MicroRNAs (miRNAs) regulate both normal stem cells and CSCs, and dysregulation of miRNAs has been implicated in tumorigenesis. CSCs in many tumors--including cancers of the breast, pancreas, head and neck, colon, small intestine, liver, stomach, bladder and ovary--have been identified using the adhesion molecule CD44, either individually or in combination with other marker(s). Prostate CSCs with enhanced clonogenic and tumor-initiating and metastatic capacities are enriched in the CD44(+) cell population, but whether miRNAs regulate CD44(+) prostate cancer cells and prostate cancer metastasis remains unclear. Here we show, through expression analysis, that miR-34a, a p53 target, was underexpressed in CD44(+) prostate cancer cells purified from xenograft and primary tumors. Enforced expression of miR-34a in bulk or purified CD44(+) prostate cancer cells inhibited clonogenic expansion, tumor regeneration, and metastasis. In contrast, expression of miR-34a antagomirs in CD44(-) prostate cancer cells promoted tumor development and metastasis. Systemically delivered miR-34a inhibited prostate cancer metastasis and extended survival of tumor-bearing mice. We identified and validated CD44 as a direct and functional target of miR-34a and found that CD44 knockdown phenocopied miR-34a overexpression in inhibiting prostate cancer regeneration and metastasis. Our study shows that miR-34a is a key negative regulator of CD44(+) prostate cancer cells and establishes a strong rationale for developing miR-34a as a novel therapeutic agent against prostate CSCs.


Asunto(s)
Receptores de Hialuranos/efectos de los fármacos , MicroARNs/uso terapéutico , Células Madre Neoplásicas/efectos de los fármacos , Neoplasias de la Próstata/tratamiento farmacológico , Animales , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Masculino , Ratones , Metástasis de la Neoplasia , Trasplante de Neoplasias , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
17.
Methods Mol Biol ; 568: 85-138, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19582423

RESUMEN

The cancer stem cell (CSC) theory posits that only a small population of tumor cells within the tumor has the ability to reinitiate tumor development and is responsible for tumor homeostasis and progression. Tumor initiation is a defining property of putative CSCs, which have been reported in both blood malignancies and solid tumors. In order to test whether any given human tumor cell population has CSC properties, the relatively enriched single cells have to be put into a foreign microenvironment in a recipient animal to test their tumorigenic potential. Furthermore, various in vitro assays need be performed to demonstrate that the presumed CSCs have certain biological properties normally associated with the stem cells (SCs). Herein, we present a comprehensive review of the experimental methodologies that our lab has been using in assaying putative prostate cancer (PCa) SCs in culture, xenograft tumors, and primary tumor samples.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Células Madre Neoplásicas/patología , Neoplasias de la Próstata/patología , Animales , Bioensayo , Biomarcadores de Tumor/metabolismo , Separación Celular , Humanos , Masculino , Ratones , Ratones SCID , Metástasis de la Neoplasia/patología , Trasplante de Neoplasias
18.
Stem Cells ; 27(5): 993-1005, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19415763

RESUMEN

Tumor development has long been known to resemble abnormal embryogenesis. The embryonic stem cell (ESC) self-renewal gene NANOG is purportedly expressed by some epithelial cancer cells but a causal role in tumor development has remained unclear. Here, we provide compelling evidence that cultured cancer cells, as well as xenograft- and human primary prostate cancer cells express a functional variant of NANOG. NANOG mRNA in cancer cells is derived predominantly from a retrogene locus termed NANOGP8. NANOG protein is detectable in the nucleus of cancer cells and is expressed higher in patient prostate tumors than matched benign tissues. NANOGP8 mRNA and/or NANOG protein levels are enriched in putative cancer stem/progenitor cell populations. Importantly, extensive loss-of-function analysis reveals that RNA interference-mediated NANOG knockdown inhibits tumor development, establishing a functional significance for NANOG expression in cancer cells. Nanog short hairpin RNA transduced cancer cells exhibit decreased long-term clonal and clonogenic growth, reduced proliferation and, in some cases, altered differentiation. Thus, our results demonstrate that NANOG, a cell-fate regulatory molecule known to be important for ESC self-renewal, also plays a novel role in tumor development.


Asunto(s)
Proteínas de Homeodominio/genética , Neoplasias/genética , Neoplasias/patología , Animales , Diferenciación Celular , Línea Celular Tumoral , Proliferación Celular , Células Clonales , Regulación Neoplásica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Proteínas de Homeodominio/metabolismo , Humanos , Receptores de Hialuranos/metabolismo , Masculino , Proteína Homeótica Nanog , Seudogenes , Interferencia de ARN , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Interferente Pequeño/metabolismo , Ratas , Transcripción Genética , Transducción Genética
19.
J Biol Chem ; 283(41): 27957-27972, 2008 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-18662989

RESUMEN

Normal human prostate (NHP) epithelial cells undergo senescence in vitro and in vivo, but the underlying molecular mechanisms remain obscure. Here we show that the senescence of primary NHP cells, which are immunophenotyped as intermediate basal-like cells expressing progenitor cell markers CD44, alpha2beta1, p63, hTERT, and CK5/CK18, involves loss of telomerase expression, up-regulation of p16, and activation of p53. Using genetically defined manipulations of these three signaling pathways, we show that p16 is the primary determinant of the NHP cell proliferative capacity and that hTERT is required for unlimited proliferative life span. Hence, suppression of p16 significantly extends NHP cell life span, but both p16 inhibition and hTERT are required to immortalize NHP cells. Importantly, immortalized NHP cells retain expression of most progenitor markers, demonstrate gene expression profiles characteristic of proliferating progenitor cells, and possess multilineage differentiation potential generating functional prostatic glands. Our studies shed important light on the molecular mechanisms regulating the proliferative life span of NHP progenitor cells.


Asunto(s)
Proliferación Celular , Senescencia Celular/fisiología , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Células Epiteliales/metabolismo , Próstata/metabolismo , Células Madre/metabolismo , Telomerasa/metabolismo , Antígenos de Diferenciación/biosíntesis , Línea Celular , Células Epiteliales/citología , Humanos , Masculino , Próstata/citología , Transducción de Señal/fisiología , Células Madre/citología , Proteína p53 Supresora de Tumor/biosíntesis , Regulación hacia Arriba/fisiología
20.
Cancer Res ; 68(6): 1820-5, 2008 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-18339862

RESUMEN

Primary keratinocytes exhibit three typical clonal morphologies represented by holoclones, meroclones, and paraclones, with holoclones containing self-renewing stem cells, and meroclones and paraclones containing more mature and differentiated cells. Interestingly, long-term-cultured human epithelial cancer cells in clonal cultures also form holoclones, meroclones, and paraclones, and tumor cell holoclones have been hypothesized to harbor stem-like cells or cancer stem cells. However, the key question of whether tumor cell holoclones genuinely contain tumor-initiating cells has not been directly addressed. Here, using PC3 human prostate carcinoma cells as a model, we provide direct experimental evidence that tumor cell holoclones contain stem-like cells that can initiate serially transplantable tumors. Importantly, holoclones derived from either cultured PC3 cells or holoclone-initiated tumors can be serially passaged and regenerate all three types of clones. In contrast, meroclones and paraclones cannot be continuously propagated and fail to initiate tumor development. Phenotypic characterizations reveal high levels of CD44, alpha(2)beta(1) integrin, and beta-catenin expression in holoclones, whereas meroclones and paraclones show markedly reduced expression of these stem cell markers. The present results have important implications in understanding morphologic heterogeneities and tumorigenic hierarchies in human epithelial cancer cells.


Asunto(s)
Neoplasias de la Próstata/patología , Animales , Procesos de Crecimiento Celular/fisiología , Línea Celular Tumoral , Células Clonales , Humanos , Receptores de Hialuranos/biosíntesis , Integrina alfa2beta1/biosíntesis , Masculino , Ratones , Ratones Endogámicos NOD , Ratones SCID , Trasplante de Neoplasias , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Neoplasias de la Próstata/metabolismo , Receptores Androgénicos/metabolismo , Trasplante Heterólogo , beta Catenina/biosíntesis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...