Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 13 de 13
Filtrer
1.
Cell Death Differ ; 31(6): 779-791, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38654072

RÉSUMÉ

Cell plasticity has been found to play a critical role in tumor progression and therapy resistance. However, our understanding of the characteristics and markers of plastic cellular states during cancer cell lineage transition remains limited. In this study, multi-omics analyses show that prostate cancer cells undergo an intermediate state marked by Zeb1 expression with epithelial-mesenchymal transition (EMT), stemness, and neuroendocrine features during the development of neuroendocrine prostate cancer (NEPC). Organoid-formation assays and in vivo lineage tracing experiments demonstrate that Zeb1+ epithelioid cells are putative cells of origin for NEPC. Mechanistically, Zeb1 transcriptionally regulates the expression of several key glycolytic enzymes, thereby predisposing tumor cells to utilize glycolysis for energy metabolism. During this process, lactate accumulation-mediated histone lactylation enhances chromatin accessibility and cellular plasticity including induction of neuro-gene expression, which promotes NEPC development. Collectively, Zeb1-driven metabolic rewiring enables the epigenetic reprogramming of prostate cancer cells to license the adeno-to-neuroendocrine lineage transition.


Sujet(s)
Tumeurs de la prostate , Facteur de transcription Zeb1 , Mâle , Facteur de transcription Zeb1/métabolisme , Facteur de transcription Zeb1/génétique , Tumeurs de la prostate/métabolisme , Tumeurs de la prostate/anatomopathologie , Tumeurs de la prostate/génétique , Humains , Animaux , Chromatine/métabolisme , Transition épithélio-mésenchymateuse , Lignée cellulaire tumorale , Souris , Régulation de l'expression des gènes tumoraux , Tumeurs neuroendocrines/métabolisme , Tumeurs neuroendocrines/anatomopathologie , Tumeurs neuroendocrines/génétique , Plasticité cellulaire , Glycolyse , Assemblage et désassemblage de la chromatine
2.
J Clin Invest ; 133(24)2023 12 15.
Article de Anglais | MEDLINE | ID: mdl-38099497

RÉSUMÉ

Cell lineage plasticity is one of the major causes for the failure of targeted therapies in various cancers. However, the driver and actionable drug targets in promoting cancer cell lineage plasticity are scarcely identified. Here, we found that a G protein-coupled receptor, ADORA2A, is specifically upregulated during neuroendocrine differentiation, a common form of lineage plasticity in prostate cancer and lung cancer following targeted therapies. Activation of the ADORA2A signaling rewires the proline metabolism via an ERK/MYC/PYCR cascade. Increased proline synthesis promotes deacetylases SIRT6/7-mediated deacetylation of histone H3 at lysine 27 (H3K27), and thereby biases a global transcriptional output toward a neuroendocrine lineage profile. Ablation of Adora2a in genetically engineered mouse models inhibits the development and progression of neuroendocrine prostate and lung cancers, and, intriguingly, prevents the adenocarcinoma-to-neuroendocrine phenotypic transition. Importantly, pharmacological blockade of ADORA2A profoundly represses neuroendocrine prostate and lung cancer growth in vivo. Therefore, we believe that ADORA2A can be used as a promising therapeutic target to govern the epigenetic reprogramming in neuroendocrine malignancies.


Sujet(s)
Tumeurs du poumon , Tumeurs de la prostate , Sirtuines , Animaux , Humains , Mâle , Souris , Lignée cellulaire tumorale , Épigenèse génétique , Tumeurs du poumon/génétique , Tumeurs du poumon/anatomopathologie , Proline/métabolisme , Proline/usage thérapeutique , Prostate/métabolisme , Prostate/anatomopathologie , Tumeurs de la prostate/anatomopathologie , Sirtuines/métabolisme
3.
Adv Sci (Weinh) ; 10(17): e2206889, 2023 06.
Article de Anglais | MEDLINE | ID: mdl-37092583

RÉSUMÉ

Great attention is paid to the role of androgen receptor (AR) as a central transcriptional factor in driving the growth of prostate cancer (PCa) epithelial cells. However, the understanding of the role of androgen in PCa-infiltrated immune cells and the impact of androgen deprivation therapy (ADT), the first-line treatment for advanced PCa, on the PCa immune microenvironment remains limited. On the other hand, immune checkpoint blockade has revolutionized the treatment of certain cancer types, but fails to achieve any benefit in advanced PCa, due to an immune suppressive environment. In this study, it is reported that AR signaling pathway is evidently activated in tumor-associated macrophages (TAMs) of PCa both in mice and humans. AR acts as a transcriptional repressor for IL1B in TAMs. ADT releases the restraint of AR on IL1B and therefore leads to an excessive expression and secretion of IL-1ß in TAMs. IL-1ß induces myeloid-derived suppressor cells (MDSCs) accumulation that inhibits the activation of cytotoxic T cells, leading to the immune suppressive microenvironment. Critically, anti-IL-1ß antibody coupled with ADT and the immune checkpoint inhibitor anti-PD-1 antibody exerts a stronger anticancer effect on PCa following castration. Together, IL-1ß is an important androgen-responsive immunotherapeutic target for advanced PCa.


Sujet(s)
Tumeurs de la prostate , Animaux , Humains , Mâle , Souris , Antagonistes des androgènes , Androgènes , Immunothérapie , Macrophages/métabolisme , Tumeurs de la prostate/immunologie , Tumeurs de la prostate/thérapie , Microenvironnement tumoral
4.
Cell Rep ; 42(2): 112033, 2023 02 28.
Article de Anglais | MEDLINE | ID: mdl-36724072

RÉSUMÉ

Cell plasticity and neuroendocrine differentiation in prostate and lung adenocarcinomas are one of the major reasons for therapeutic resistance to targeted therapy. Whether and how metabolic changes contribute to this adenocarcinoma-to-neuroendocrine cell fate transition remains largely unclear. Here we show that neuroendocrine prostate or lung cancer cells possess mostly fragmented mitochondria with low membrane potential and rely on glycolysis for energy metabolism. We further show an important role of the cell fate determinant Numb in mitochondrial quality control via binding to Parkin and facilitating Parkin-mediated mitophagy. Deficiency in the Numb/Parkin pathway in prostate or lung adenocarcinomas causes a metabolic reprogramming featured with a significant increase in production of lactate acid, which subsequently leads to an upregulation of histone lactylation and transcription of neuroendocrine-associated genes. Collectively, the Numb/Parkin-directed mitochondrial fitness is a key metabolic switch and a promising therapeutic target on cancer cell plasticity through the regulation of histone lactylation.


Sujet(s)
Adénocarcinome pulmonaire , Tumeurs du poumon , Mâle , Humains , Histone/métabolisme , Mitochondries/métabolisme , Différenciation cellulaire , Tumeurs du poumon/métabolisme , Ubiquitin-protein ligases/métabolisme , Adénocarcinome pulmonaire/métabolisme , Protéines membranaires/métabolisme , Protéines de tissu nerveux/métabolisme
5.
Nat Cancer ; 3(5): 565-580, 2022 05.
Article de Anglais | MEDLINE | ID: mdl-35624341

RÉSUMÉ

Among the greatest hurdles in clinical management of prostate cancer (PCa) are the progression to lethal castration-resistant prostate cancer (CRPC) and the lack of suitable targeted therapies for advanced disease. Here we identify Gremlin1 as a ligand for fibroblast growth factor receptor 1 (FGFR1), which promotes lineage plasticity and drives castration resistance. Importantly, we generate a specific anti-Gremlin1 therapeutic antibody and demonstrate synergistic effect with androgen deprivation therapy (ADT) in CRPC. GREM1 transcription is suppressed by androgen receptor (AR) and released following ADT. We show that Gremlin1 binds to FGFR1 and activates downstream MAPK signaling. Gremlin1 interacts with FGFR1 differently to its canonical ligand FGF1, as revealed through protein structure docking and mutagenesis experiments. Altogether, our data indicate Gremlin1 as a promising candidate therapeutic target for CRPC.


Sujet(s)
Tumeurs prostatiques résistantes à la castration , Antagonistes des androgènes/pharmacologie , Castration , Humains , Protéines et peptides de signalisation intercellulaire/métabolisme , Ligands , Mâle , Tumeurs prostatiques résistantes à la castration/traitement médicamenteux , Tumeurs prostatiques résistantes à la castration/génétique , Tumeurs prostatiques résistantes à la castration/métabolisme , Récepteur FGFR1/génétique , Transduction du signal
6.
Cell Prolif ; 54(7): e13056, 2021 Jul.
Article de Anglais | MEDLINE | ID: mdl-34021647

RÉSUMÉ

OBJECTIVES: In contrast to extensive studies on bone metastasis in advanced prostate cancer (PCa), liver metastasis has been under-researched so far. In order to decipher molecular and cellular mechanisms underpinning liver metastasis of advanced PCa, we develop a rapid and immune sufficient mouse model for liver metastasis of PCa via orthotopic injection of organoids from PbCre+ ; rb1f/f ;p53f/f mice. MATERIALS AND METHODS: PbCre+ ;rb1f/f ;p53f/f and PbCre+ ;ptenf/f ;p53f/f mice were used to generate PCa organoid cultures in vitro. Immune sufficient liver metastasis models were established via orthotopic transplantation of organoids into the prostate of C57BL/6 mice. Immunofluorescent and immunohistochemical staining were performed to characterize the lineage profile in primary tumour and organoid-derived tumour (ODT). The growth of niche-labelling reporter infected ODT can be visualized by bioluminescent imaging system. Immune cells that communicated with tumour cells in the liver metastatic niche were determined by flow cytometry. RESULTS: A PCa liver metastasis model with full penetrance is established in immune-intact mouse. This model reconstitutes the histological and lineage features of original tumours and reveals dynamic tumour-immune cell communication in liver metastatic foci. Our results suggest that a lack of CD8+ T cell and an enrichment of CD163+ M2-like macrophage as well as PD1+ CD4+ T cell contribute to an immuno-suppressive microenvironment of PCa liver metastasis. CONCLUSIONS: Our model can be served as a reliable tool for analysis of the molecular pathogenesis and tumour-immune cell crosstalk in liver metastasis of PCa, and might be used as a valuable in vivo model for therapy development.


Sujet(s)
Tumeurs du foie/anatomopathologie , Tumeurs de la prostate/anatomopathologie , Animaux , Lymphocytes T CD4+/cytologie , Lymphocytes T CD4+/immunologie , Lymphocytes T CD4+/métabolisme , Lymphocytes T CD8+/cytologie , Lymphocytes T CD8+/immunologie , Lymphocytes T CD8+/métabolisme , Communication cellulaire , Modèles animaux de maladie humaine , Tumeurs du foie/secondaire , Macrophages/cytologie , Macrophages/immunologie , Macrophages/métabolisme , Mâle , Souris , Souris de lignée C57BL , Souris knockout , Phosphohydrolase PTEN/déficit , Phosphohydrolase PTEN/génétique , Tumeurs de la prostate/métabolisme , Microenvironnement tumoral , Protéine p53 suppresseur de tumeur/déficit , Protéine p53 suppresseur de tumeur/génétique
7.
Nat Commun ; 11(1): 706, 2020 02 05.
Article de Anglais | MEDLINE | ID: mdl-32024836

RÉSUMÉ

The basal cell compartment in many epithelial tissues is generally believed to serve as an important pool of stem cells. However, basal cells are heterogenous and the stem cell subpopulation within basal cells is not well elucidated. Here we uncover that the core epithelial-to-mesenchymal transition (EMT) inducer Zeb1 is expressed in a prostate basal cell subpopulation. The Zeb1+ prostate epithelial cells are multipotent prostate basal stem cells (PBSCs) that can self-renew and generate functional prostatic glandular structures at the single-cell level. Genetic ablation studies reveal an indispensable role for Zeb1 in prostate basal cell development. Utilizing unbiased single-cell transcriptomic analysis of over 9000 mouse prostate basal cells, we confirm the existence of the Zeb1+ basal cell subset. Moreover, Zeb1+ epithelial cells can be detected in mouse and human prostate tumors. Identification of the PBSC and its transcriptome profile is crucial to advance our understanding of prostate development and tumorigenesis.


Sujet(s)
Prostate/cytologie , Cellules souches/physiologie , Facteur de transcription Zeb1/métabolisme , Animaux , Cellules épithéliales/métabolisme , Transition épithélio-mésenchymateuse , Femelle , Humains , Mâle , Souris de lignée C57BL , Souris nude , Souris transgéniques , Techniques de culture d'organes , Grossesse , Prostate/métabolisme , Prostate/anatomopathologie , Tumeurs de la prostate/anatomopathologie , Rat Sprague-Dawley , Analyse sur cellule unique , Voie de signalisation Wnt , Facteur de transcription Zeb1/génétique
8.
Nat Commun ; 10(1): 3353, 2019 07 26.
Article de Anglais | MEDLINE | ID: mdl-31350389

RÉSUMÉ

The diverse repertoire of T cell receptors (TCR) and immunoglobulins is generated through the somatic rearrangement of respective V, D and J gene segments, termed V(D)J recombination, during early T or B cell development. However, epigenetic regulation of V(D)J recombination is still not fully understood. Here we show that the deficiency of Setd2, a histone methyltransferase that catalyzes lysine 36 trimethylation on histone 3 (H3K36me3) in mice, causes a severe developmental block of thymocytes at the CD4-CD8- DN3 stage. While H3K36me3 is normally enriched at the TCRß locus, Setd2 deficiency reduces TCRß H3K36me3 and suppresses TCRß V(D)J rearrangement by impairing RAG1 binding to TCRß loci and the DNA double-strand break repair. Similarly, Setd2 ablation also impairs immunoglobulin V(D)J rearrangement to induce B cell development block at the pro-B stage. Lastly, SETD2 is frequently mutated in patients with primary immunodeficiency. Our study thus demonstrates that Setd2 is required for optimal V(D)J recombination and normal lymphocyte development.


Sujet(s)
Histone-lysine N-methyltransferase/métabolisme , Recombinaison V(D)J , Motifs d'acides aminés , Animaux , Lymphocytes B/cytologie , Lymphocytes B/enzymologie , Différenciation cellulaire , Enfant d'âge préscolaire , Épigenèse génétique , Histone-lysine N-methyltransferase/génétique , Histone/composition chimique , Histone/métabolisme , Humains , Nourrisson , Lysine/génétique , Lysine/métabolisme , Mâle , Méthylation , Souris , Souris de lignée C57BL , Souris knockout , Mutation , Maladies d'immunodéficience primaire/enzymologie , Maladies d'immunodéficience primaire/génétique , Récepteur lymphocytaire T antigène, alpha-bêta/génétique , Récepteur lymphocytaire T antigène, alpha-bêta/métabolisme , Lymphocytes T/cytologie
9.
Cell Prolif ; 52(4): e12611, 2019 Jul.
Article de Anglais | MEDLINE | ID: mdl-31054182

RÉSUMÉ

OBJECTIVES: Epigenetic modifiers were important players in the development of haematological malignancies and sensitivity to therapy. Mutations of SET domain-containing 2 (SETD2), a methyltransferase that catalyses the trimethylation of histone 3 on lysine 36 (H3K36me3), were found in various myeloid malignancies. However, the detailed mechanisms through which SETD2 confers chronic myeloid leukaemia progression and resistance to therapy targeting on BCR-ABL remain unclear. MATERIALS AND METHODS: The level of SETD2 in imatinib-sensitive and imatinib-resistant chronic myeloid leukaemia (CML) cells was examined by immunoblotting and quantitative real-time PCR. We analysed CD34+ CD38- leukaemic stem cells by flow cytometry and colony formation assays upon SETD2 knockdown or overexpression. The impact of SETD2 expression alterations or small-molecule inhibitor JIB-04 targeting H3K36me3 loss on imatinib sensitivity was assessed by IC50, cell apoptosis and proliferation assays. Finally, RNA sequencing and ChIP-quantitative PCR were performed to verify putative downstream targets. RESULTS: SETD2 was found to act as a tumour suppressor in CML. The novel oncogenic targets MYCN and ERG were shown to be the direct downstream targets of SETD2, where their overexpression induced by SETD2 knockdown caused imatinib insensitivity and leukaemic stem cell enrichment in CML cell lines. Treatment with JIB-04, an inhibitor that restores H3K36me3 levels through blockade of its demethylation, successfully improved the cell imatinib sensitivity and enhanced the chemotherapeutic effect. CONCLUSIONS: Our study not only emphasizes the regulatory mechanism of SETD2 in CML, but also provides promising therapeutic strategies for overcoming the imatinib resistance in patients with CML.


Sujet(s)
Régulation négative/génétique , Résistance aux médicaments antinéoplasiques/génétique , Histone méthyltransférases/génétique , Histone-lysine N-methyltransferase/génétique , Mésilate d'imatinib/pharmacologie , Leucémie myéloïde chronique BCR-ABL positive/génétique , Aminopyridines/pharmacologie , Antinéoplasiques/pharmacologie , Apoptose/effets des médicaments et des substances chimiques , Apoptose/génétique , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Prolifération cellulaire/génétique , Régulation négative/effets des médicaments et des substances chimiques , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Protéines de fusion bcr-abl/génétique , Humains , Hydrazones/pharmacologie , Leucémie myéloïde chronique BCR-ABL positive/traitement médicamenteux , Inhibiteurs de protéines kinases/pharmacologie
10.
Theranostics ; 8(17): 4633-4648, 2018.
Article de Anglais | MEDLINE | ID: mdl-30279728

RÉSUMÉ

Tumor metastasis is the major cause of death for prostate cancer (PCa) patients. However, the treatment options for metastatic PCa are very limited. Epithelial-mesenchymal transition (EMT) has been reported to be an indispensable step for tumor metastasis and is suggested to associate with acquisition of cancer stem cell (CSC) attributes. We propose that small-molecule compounds that can reverse EMT or induce mesenchymal-epithelial transition (MET) of PCa cells may serve as drug candidates for anti-metastasis therapy. Methods: The promoters of CDH1 and VIM genes were sub-cloned to drive the expression of firefly and renilla luciferase reporter in a lentiviral vector. Mesenchymal-like PCa cells were infected with the luciferase reporter lentivirus and subjected to drug screening from a 1274 approved small-molecule drug library for the identification of agents to reverse EMT. The dosage-dependent effect of candidate compounds was confirmed by luciferase reporter assay and immunoblotting. Wound-healing assay, sphere formation, transwell migration assay, and in vivo intracardiac and orthotopic tumor xenograft experiments were used to evaluate the mobility, metastasis and tumor initiating capacity of PCa cells upon treatment. Possible downstream signaling pathways affected by the candidate compound treatment were analyzed by RNA sequencing and immunoblotting. Results: Drug screening identified Amlexanox, a drug used for recurrent aphthous ulcers, as a strong agent to reverse EMT. Amlexanox induced significant suppression of cell mobility, invasion, serial sphere formation and in vivo metastasis and tumor initiating capacity of PCa cells. Amlexanox treatment led to downregulation of the IKK-ɛ/ TBK1/ NF-κB signaling pathway. The effect of Amlexanox on EMT reversion and cell mobility inhibition can be mimicked by other IKK-ɛ/TBK1 inhibitors and rescued by reconstitution of dominant active NF-κB. Conclusions: Amlexanox can sufficiently suppress PCa metastasis by reversing EMT through downregulating the IKK-ɛ/TBK1/NF-κB signaling axis.


Sujet(s)
Aminopyridines/pharmacologie , Antinéoplasiques/pharmacologie , Transition épithélio-mésenchymateuse/effets des médicaments et des substances chimiques , Métastase tumorale/prévention et contrôle , Tumeurs de la prostate/secondaire , Transduction du signal/effets des médicaments et des substances chimiques , Aminopyridines/administration et posologie , Aminopyridines/isolement et purification , Animaux , Antinéoplasiques/administration et posologie , Antinéoplasiques/isolement et purification , Lignée cellulaire tumorale , Mouvement cellulaire/effets des médicaments et des substances chimiques , Modèles animaux de maladie humaine , Évaluation préclinique de médicament/méthodes , Humains , I-kappa B Kinase/métabolisme , Mâle , Souris , Modèles théoriques , Facteur de transcription NF-kappa B/métabolisme , Protein-Serine-Threonine Kinases/métabolisme , Résultat thérapeutique
11.
PLoS Genet ; 14(8): e1007609, 2018 08.
Article de Anglais | MEDLINE | ID: mdl-30118484

RÉSUMÉ

Cell polarity and correct mitotic spindle positioning are essential for the maintenance of a proper prostate epithelial architecture, and disruption of the two biological features occurs at early stages in prostate tumorigenesis. However, whether and how these two epithelial attributes are connected in vivo is largely unknown. We herein report that conditional genetic deletion of E-cadherin, a key component of adherens junctions, in a mouse model results in loss of prostate luminal cell polarity and randomization of spindle orientations. Critically, E-cadherin ablation causes prostatic hyperplasia which progresses to invasive adenocarcinoma. Mechanistically, E-cadherin and the spindle positioning determinant LGN interacts with the PDZ domain of cell polarity protein SCRIB and form a ternary protein complex to bridge cell polarity and cell division orientation. These findings provide a novel mechanism by which E-cadherin acts an anchor to maintain prostate epithelial integrity and to prevent carcinogenesis in vivo.


Sujet(s)
Cadhérines/physiologie , Polarité de la cellule , Prostate/cytologie , Appareil du fuseau/physiologie , Animaux , Cadhérines/génétique , Carcinogenèse , Division cellulaire , Lignée cellulaire , Prolifération cellulaire , Modèles animaux de maladie humaine , Épithélium , Délétion de gène , Régulation de l'expression des gènes , Humains , Mâle , Souris knockout , Tumeurs de la prostate/anatomopathologie
12.
Clin Cancer Res ; 23(21): 6744-6756, 2017 Nov 01.
Article de Anglais | MEDLINE | ID: mdl-28751447

RÉSUMÉ

Purpose: To elucidate the role and molecular mechanism of Numb in prostate cancer and the functional contribution of Numb-/low prostate cancer cells in castration resistance.Experimental Design: The expression of Numb was assessed using multiple Oncomine datasets and prostate cancer tissues from both humans and mice. The biological effects of the overexpression and knockdown of Numb in human prostate cancer cell lines were investigated in vitro and in vivo In addition, we developed a reliable approach to distinguish between prostate cancer cell populations with a high or low endogenous expression of Numb protein using a Numb promoter-based lentiviral reporter system. The difference between Numb-/low and Numbhigh prostate cancer cells in the response to androgen-deprivation therapy (ADT) was then tested. The likely downstream factors of Numb were analyzed using luciferase reporter assays, immunoblotting, and quantitative real-time PCR.Results: We show here that Numb was downregulated and negatively correlated with prostate cancer advancement. Functionally, Numb played an inhibitory role in xenograft prostate tumor growth and castration-resistant prostate cancer development by suppressing Notch and Hedgehog signaling. Using a Numb promoter-based lentiviral reporter system, we were able to distinguish Numb-/low prostate cancer cells from Numbhigh cells. Numb-/low prostate cancer cells were smaller and quiescent, preferentially expressed Notch and Hedgehog downstream and stem-cell-associated genes, and associated with a greater resistance to ADT. The inhibition of the Notch and Hedgehog signaling pathways significantly increased apoptosis in Numb-/low cells in response to ADT.Conclusions: Numb-/low enriches a castration-resistant prostate cancer cell subpopulation that is associated with unregulated Notch and Hedgehog signaling. Clin Cancer Res; 23(21); 6744-56. ©2017 AACR.


Sujet(s)
Androgènes/usage thérapeutique , Protéines membranaires/génétique , Protéines de tissu nerveux/génétique , Tumeurs prostatiques résistantes à la castration/traitement médicamenteux , Tumeurs prostatiques résistantes à la castration/génétique , Androgènes/métabolisme , Animaux , Lignage cellulaire/effets des médicaments et des substances chimiques , Régulation de l'expression des gènes tumoraux , Protéines Hedgehog/génétique , Humains , Mâle , Souris , Tumeurs prostatiques résistantes à la castration/anatomopathologie , Récepteurs Notch/génétique , Transduction du signal/génétique , Tests d'activité antitumorale sur modèle de xénogreffe
13.
Cancer Res ; 77(9): 2534-2547, 2017 05 01.
Article de Anglais | MEDLINE | ID: mdl-28209613

RÉSUMÉ

Cancer stem-like cells (CSC) drive cancer progression and recurrence. Self-renewal expansion of CSC is achieved through symmetric cell division, yet how external stimuli affect intracellular regulatory programs of CSC division modes and stemness remains obscure. Here, we report that the hTERThigh prostate cancer cells exhibit CSC properties, including a stem cell-associated gene expression signature, long-term tumor-propagating capacity and epithelial-to-mesenchymal transition. In promoting the self-renewal symmetric division of hTERThigh prostate cancer cells, WNT3a dramatically decreased the ratio of hTERThigh prostate cancer cells undergoing asymmetric division. Increased WNT/ß-catenin signal activation was also detected in hTERThigh prostate cancer cells. hTERT-mediated CSC properties were at least partially dependent on ß-catenin. These findings provide novel cellular and molecular mechanisms for the self-renewal of CSC orchestrated by tumor microenvironmental stimuli and intracellular signals. Cancer Res; 77(9); 2534-47. ©2017 AACR.


Sujet(s)
Cellules souches tumorales/métabolisme , Tumeurs de la prostate/génétique , Telomerase/génétique , Protéine Wnt3A/génétique , bêta-Caténine/génétique , Division cellulaire/effets des médicaments et des substances chimiques , Auto-renouvellement cellulaire/génétique , Transition épithélio-mésenchymateuse/génétique , Régulation de l'expression des gènes tumoraux/génétique , Humains , Mâle , Cellules souches tumorales/anatomopathologie , Tumeurs de la prostate/anatomopathologie , Voie de signalisation Wnt/génétique
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE