Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 89
Filter
Add more filters

Publication year range
1.
Immunity ; 52(4): 668-682.e7, 2020 04 14.
Article in English | MEDLINE | ID: mdl-32294407

ABSTRACT

The primary mechanisms supporting immunoregulatory polarization of myeloid cells upon infiltration into tumors remain largely unexplored. Elucidation of these signals could enable better strategies to restore protective anti-tumor immunity. Here, we investigated the role of the intrinsic activation of the PKR-like endoplasmic reticulum (ER) kinase (PERK) in the immunoinhibitory actions of tumor-associated myeloid-derived suppressor cells (tumor-MDSCs). PERK signaling increased in tumor-MDSCs, and its deletion transformed MDSCs into myeloid cells that activated CD8+ T cell-mediated immunity against cancer. Tumor-MDSCs lacking PERK exhibited disrupted NRF2-driven antioxidant capacity and impaired mitochondrial respiratory homeostasis. Moreover, reduced NRF2 signaling in PERK-deficient MDSCs elicited cytosolic mitochondrial DNA elevation and, consequently, STING-dependent expression of anti-tumor type I interferon. Reactivation of NRF2 signaling, conditional deletion of STING, or blockade of type I interferon receptor I restored the immunoinhibitory potential of PERK-ablated MDSCs. Our findings demonstrate the pivotal role of PERK in tumor-MDSC functionality and unveil strategies to reprogram immunosuppressive myelopoiesis in tumors to boost cancer immunotherapy.


Subject(s)
Carcinoma, Lewis Lung/immunology , Carcinoma, Ovarian Epithelial/immunology , Gene Expression Regulation, Neoplastic , Melanoma, Experimental/immunology , Membrane Proteins/immunology , Skin Neoplasms/immunology , eIF-2 Kinase/immunology , Animals , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Carcinoma, Lewis Lung/genetics , Carcinoma, Lewis Lung/metabolism , Carcinoma, Lewis Lung/pathology , Carcinoma, Ovarian Epithelial/genetics , Carcinoma, Ovarian Epithelial/metabolism , Carcinoma, Ovarian Epithelial/pathology , Female , Humans , Immunosuppression Therapy , Interferon-alpha/genetics , Interferon-alpha/immunology , Interferon-beta/genetics , Interferon-beta/immunology , Male , Melanoma, Experimental/genetics , Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/immunology , Mitochondria/metabolism , Myeloid-Derived Suppressor Cells/immunology , Myeloid-Derived Suppressor Cells/pathology , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/immunology , Receptors, Interferon/genetics , Receptors, Interferon/immunology , Signal Transduction , Skin Neoplasms/genetics , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Unfolded Protein Response/immunology , eIF-2 Kinase/deficiency , eIF-2 Kinase/genetics
2.
Mol Cell ; 70(5): 825-841.e6, 2018 06 07.
Article in English | MEDLINE | ID: mdl-29861161

ABSTRACT

Super-enhancers are large clusters of enhancers that activate gene expression. Broad trimethyl histone H3 lysine 4 (H3K4me3) often defines active tumor suppressor genes. However, how these epigenomic signatures are regulated for tumor suppression is little understood. Here we show that brain-specific knockout of the H3K4 methyltransferase MLL4 (a COMPASS-like enzyme, also known as KMT2D) in mice spontaneously induces medulloblastoma. Mll4 loss upregulates oncogenic Ras and Notch pathways while downregulating neuronal gene expression programs. MLL4 enhances DNMT3A-catalyzed DNA methylation and SIRT1/BCL6-mediated H4K16 deacetylation, which antagonize expression of Ras activators and Notch pathway components, respectively. Notably, Mll4 loss downregulates tumor suppressor genes (e.g., Dnmt3a and Bcl6) by diminishing broad H3K4me3 and super-enhancers and also causes widespread impairment of these epigenomic signatures during medulloblastoma genesis. These findings suggest an anti-tumor role for super-enhancers and provide a unique tumor-suppressive mechanism in which MLL4 is necessary to maintain broad H3K4me3 and super-enhancers at tumor suppressor genes.


Subject(s)
Cerebellar Neoplasms/genetics , DNA Methylation , Genes, Tumor Suppressor , Histone-Lysine N-Methyltransferase/genetics , Medulloblastoma/genetics , Oncogenes , Protein Processing, Post-Translational , Acetylation , Animals , Cell Proliferation , Cerebellar Neoplasms/metabolism , Cerebellar Neoplasms/pathology , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methyltransferase 3A , Gene Expression Regulation, Neoplastic , Genes, ras , Histone-Lysine N-Methyltransferase/deficiency , Lysine , Medulloblastoma/metabolism , Medulloblastoma/pathology , Mice, Knockout , Proto-Oncogene Proteins c-bcl-6/genetics , Proto-Oncogene Proteins c-bcl-6/metabolism , Receptors, Notch/genetics , Receptors, Notch/metabolism , Signal Transduction , Sirtuin 1/genetics , Sirtuin 1/metabolism
5.
Genes Dev ; 31(17): 1715-1716, 2017 09 01.
Article in English | MEDLINE | ID: mdl-28982757

ABSTRACT

Medulloblastomas are among the most common malignant brain cancers in the pediatric population and consist of at least four distinct subgroups with unique molecular and genetic features and clinical outcomes. In this issue of Genes & Development, Niklison-Chirou and colleagues (pp. 1738-1753) identify the p53 family member and p73 isoform TAp73 as a crucial factor causing glutamine addiction in aggressive medulloblastomas. Their findings pave the way for the use of glutamine restriction as an adjuvant treatment for TAp73-expressing medulloblastomas.


Subject(s)
Cerebellar Neoplasms , Medulloblastoma , Child , DNA-Binding Proteins , Diet , Glutamine , Humans , Nuclear Proteins , Tumor Protein p73 , Tumor Suppressor Proteins
6.
Genes Dev ; 30(11): 1253-4, 2016 06 01.
Article in English | MEDLINE | ID: mdl-27298333

ABSTRACT

Multiciliogenesis is essential for the function of different epithelia, and its failure results in brain defects, respiratory diseases, and infertility. In this issue of Genes & Development, Nemajerova and colleagues (pp. 1300-1312) reveal the p53 family member and p73 isoform TAp73 as a transcription factor dictating the differentiation of multiciliated cells. Their findings provide the long-awaited unifying explanation for the diverse phenotypes of the p73 knockout mice.


Subject(s)
Nuclear Proteins/genetics , Tumor Protein p73 , Animals , Cell Differentiation , DNA-Binding Proteins/genetics , Phenotype , Transcription Factors/genetics , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Proteins/genetics
7.
Nucleic Acids Res ; 49(5): 2740-2758, 2021 03 18.
Article in English | MEDLINE | ID: mdl-33619536

ABSTRACT

The major clinical problem in human cancer is metastasis. Metastases are the cause of 90% of human cancer deaths. TAp63 is a critical suppressor of tumorigenesis and metastasis. ΔNp63 acts as a dominant-negative inhibitor to block the function of p53 and TAp63. Although several ubiquitin E3 ligases have been reported to regulate p63 stability, the mechanism of p63 regulation remains partially understood. Herein, we show that CHIP, an E3 ligase with a U-box domain, physically interacts with p63 and promotes p63 degradation. Notably, Hsp70 depletion by siRNA stabilizes TAp63 in H1299 cells and destabilizes ΔNp63 in SCC9 cells. Loss of Hsp70 results in a reduction in the TAp63-CHIP interaction in H1299 cells and an increase in the interaction between ΔNp63 and CHIP in SCC9 cells. Our results reveal that Hsp70 acts as a molecular switch to control CHIP-mediated ubiquitination and degradation of p63 isoforms. Furthermore, regulation of p63 by the Hsp70-CHIP axis contributes to the migration and invasion of tumor cells. Hence, our findings demonstrate that Hsp70 is a crucial regulator of CHIP-mediated ubiquitination and degradation of p63 isoforms and identify a new pathway for maintaining TAp63 or ΔNp63 stability in cancers.


Subject(s)
HSP70 Heat-Shock Proteins/metabolism , Transcription Factors/metabolism , Tumor Suppressor Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitination , Animals , Apoptosis , Cell Line, Tumor , Cell Movement , Cells, Cultured , HSP70 Heat-Shock Proteins/physiology , Humans , Mice , Neoplasm Invasiveness , Neoplasms/metabolism , Neoplasms/mortality , Trans-Activators/metabolism , Transcriptional Activation
8.
PLoS Genet ; 16(4): e1008642, 2020 04.
Article in English | MEDLINE | ID: mdl-32310940

ABSTRACT

Undifferentiated embryonal sarcoma of the liver (UESL) is a rare and aggressive malignancy. Though the molecular underpinnings of this cancer have been largely unexplored, recurrent chromosomal breakpoints affecting a noncoding region on chr19q13, which includes the chromosome 19 microRNA cluster (C19MC), have been reported in several cases. We performed comprehensive molecular profiling on samples from 14 patients diagnosed with UESL. Congruent with prior reports, we identified structural variants in chr19q13 in 10 of 13 evaluable tumors. From whole transcriptome sequencing, we observed striking expressional activity of the entire C19MC region. Concordantly, in 7 of 7 samples undergoing miRNAseq, we observed hyperexpression of the miRNAs within this cluster to levels >100 fold compared to matched normal tissue or a non-C19MC amplified cancer cell line. Concurrent TP53 mutation or copy number loss was identified in all evaluable tumors with evidence of C19MC overexpression. We find that C19MC miRNAs exhibit significant negative correlation to TP53 regulatory miRNAs and K-Ras regulatory miRNAs. Using RNA-seq we identified that pathways relevant to cellular differentiation as well as mRNA translation machinery are transcriptionally enriched in UESL. In summary, utilizing a combination of next-generation sequencing and high-density arrays we identify the combination of C19MC hyperexpression via chromosomal structural event with TP53 mutation or loss as highly recurrent genomic features of UESL.


Subject(s)
Chromosome Breakpoints , Chromosomes, Human, Pair 19/genetics , Gene Expression Regulation, Neoplastic , Liver Neoplasms/genetics , MicroRNAs/genetics , Mutation , Neoplasms, Germ Cell and Embryonal/genetics , Sarcoma/genetics , Tumor Suppressor Protein p53/genetics , Aneuploidy , Child , Child, Preschool , Female , Genes, ras/genetics , Genomic Instability/genetics , Humans , Infant , Male , Transcription Initiation Site , Tumor Suppressor Protein p53/deficiency , Up-Regulation
9.
Bioinformatics ; 37(20): 3681-3683, 2021 Oct 25.
Article in English | MEDLINE | ID: mdl-33901274

ABSTRACT

SUMMARY: The heterogeneous cell types of the tumor-immune microenvironment (TIME) play key roles in determining cancer progression, metastasis and response to treatment. We report the development of TIMEx, a novel TIME deconvolution method emphasizing on estimating infiltrating immune cells for bulk transcriptomics using pan-cancer single-cell RNA-seq signatures. We also implemented a comprehensive, user-friendly web-portal for users to evaluate TIMEx and other deconvolution methods with bulk transcriptomic profiles. AVAILABILITY AND IMPLEMENTATION: TIMEx web-portal is freely accessible at http://timex.moffitt.org. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.

10.
Cancer ; 127(23): 4413-4420, 2021 12 01.
Article in English | MEDLINE | ID: mdl-34358340

ABSTRACT

BACKGROUND: The survival benefit of elective neck dissection (END) for patients with cutaneous squamous cell carcinoma (cSCC) of the head and neck and no evidence of regional metastasis (cN0) has never been reported. The aim of this study was to determine the effect of END on patient survival. METHODS: The authors included patients with head and neck cSCC who had undergone primary surgery from 1995 to 2017. The primary end point was survival, and the secondary end points were the incidence of occult regional disease and regional disease control. To assess the impact of END on survival, the authors used multivariable Cox proportional hazards models with propensity score and matching techniques for internal validation. RESULTS: A total of 1111 patients presented with no evidence of nodal disease; 173 had END, and 938 were observed. Adjuvant radiotherapy to the neck was administered to 101 patients (9%). END resulted in a 5-year overall survival rate of 52%, whereas the rate was 63% in the observation group (P = .003 [log-rank]). The 5-year disease-free survival rate for patients undergoing END was similar to that for the observation group (73% vs 75%; P = .429). A multivariate regression model showed that the performance of END was not associated with improved rates of overall, disease-specific, or disease-free survival; similarly, among patients with advanced disease (T3-4), those who underwent END did not have improved survival rates. CONCLUSIONS: Among patients with cSCC of the head and neck, observation of the neck nodes resulted in noninferior survival rates in comparison with END at the time of primary surgery. Further studies are required to elucidate the role of END in patients with advanced disease.


Subject(s)
Carcinoma, Squamous Cell , Head and Neck Neoplasms , Skin Neoplasms , Carcinoma, Squamous Cell/pathology , Head and Neck Neoplasms/pathology , Head and Neck Neoplasms/surgery , Humans , Lymphatic Metastasis , Neck Dissection/methods , Neoplasm Staging , Retrospective Studies , Skin Neoplasms/pathology , Skin Neoplasms/surgery
12.
Nature ; 517(7536): 626-30, 2015 Jan 29.
Article in English | MEDLINE | ID: mdl-25409149

ABSTRACT

TP53 is commonly altered in human cancer, and Tp53 reactivation suppresses tumours in vivo in mice (TP53 and Tp53 are also known as p53). This strategy has proven difficult to implement therapeutically, and here we examine an alternative strategy by manipulating the p53 family members, Tp63 and Tp73 (also known as p63 and p73, respectively). The acidic transactivation-domain-bearing (TA) isoforms of p63 and p73 structurally and functionally resemble p53, whereas the ΔN isoforms (lacking the acidic transactivation domain) of p63 and p73 are frequently overexpressed in cancer and act primarily in a dominant-negative fashion against p53, TAp63 and TAp73 to inhibit their tumour-suppressive functions. The p53 family interacts extensively in cellular processes that promote tumour suppression, such as apoptosis and autophagy, thus a clear understanding of this interplay in cancer is needed to treat tumours with alterations in the p53 pathway. Here we show that deletion of the ΔN isoforms of p63 or p73 leads to metabolic reprogramming and regression of p53-deficient tumours through upregulation of IAPP, the gene that encodes amylin, a 37-amino-acid peptide co-secreted with insulin by the ß cells of the pancreas. We found that IAPP is causally involved in this tumour regression and that amylin functions through the calcitonin receptor (CalcR) and receptor activity modifying protein 3 (RAMP3) to inhibit glycolysis and induce reactive oxygen species and apoptosis. Pramlintide, a synthetic analogue of amylin that is currently used to treat type 1 and type 2 diabetes, caused rapid tumour regression in p53-deficient thymic lymphomas, representing a novel strategy to target p53-deficient cancers.


Subject(s)
Islet Amyloid Polypeptide/metabolism , Lymphoma/metabolism , Lymphoma/pathology , Tumor Suppressor Protein p53/deficiency , Animals , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Female , Genes, Tumor Suppressor , Humans , Islet Amyloid Polypeptide/pharmacology , Islet Amyloid Polypeptide/therapeutic use , Lymphoma/drug therapy , Lymphoma/genetics , Male , Mice , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Phosphoproteins/genetics , Phosphoproteins/metabolism , Receptor Activity-Modifying Protein 3/metabolism , Receptors, Calcitonin/metabolism , Thymus Gland/metabolism , Thymus Gland/pathology , Trans-Activators/genetics , Trans-Activators/metabolism , Tumor Protein p73 , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
13.
Genes Dev ; 27(9): 971-4, 2013 May 01.
Article in English | MEDLINE | ID: mdl-23651851

ABSTRACT

Autophagy is a biological process that is crucial to maintain cellular homeostasis and is regulated by several metabolic pathways, including the p53 tumor suppressor pathway. In this issue of Genes & Development, Kenzelmann Broz and colleagues (pp. 1016-1031) show how the p53 family as a whole, including p63 and p73, collaborate in controlling autophagy to support tumor suppression.


Subject(s)
Autophagy/genetics , DNA Damage/genetics , Fibroblasts/cytology , Fibroblasts/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Up-Regulation , Animals
14.
RNA Biol ; 17(11): 1636-1647, 2020 11.
Article in English | MEDLINE | ID: mdl-31910062

ABSTRACT

The tumour suppressor p53 and its paralogues, p63 and p73, are essential to maintain cellular homoeostasis and the integrity of the cell's genetic material, thus meriting the title of 'guardians of the genome'. The p53 family members are transcription factors and fulfill their activities by controlling the expression of protein-coding and non-coding genes. Here, we review how the latter group transcended from the 'dark matter' of the transcriptome, providing unexpected and intriguing anti-cancer therapeutic strategies.


Subject(s)
Biomarkers, Tumor , Gene Expression Regulation, Neoplastic , Multigene Family , Neoplasms/genetics , Tumor Suppressor Protein p53/genetics , Cell Nucleus/genetics , Cell Nucleus/metabolism , Gene Regulatory Networks , Humans , MicroRNAs/genetics , Neoplasms/metabolism , Neoplasms/pathology , RNA Stability , RNA, Circular/genetics , RNA, Long Noncoding/genetics , RNA, Transfer/genetics , Ribosomes/metabolism , Signal Transduction , Stress, Physiological , Tumor Suppressor Protein p53/metabolism
15.
Genes Dev ; 26(18): 1997-2000, 2012 Sep 15.
Article in English | MEDLINE | ID: mdl-22987633

ABSTRACT

p73 and p63 are evolving members of the p53 tumor suppressor family. TAp73 is a p73 isoform with a potent transcriptional activation domain, and loss of TAp73 predisposes mice to tumor development. In this issue of Genes & Development, Rufini and colleagues (pp. 2009-2014) discuss how TAp73-null mice display an aging phenotype that is due to mitochondrial dysfunction. Specifically, decreased levels of cytochrome C oxidase subunit 4 isoform 1 (Cox4i1) impair cytochrome C oxidase (COX) function, the multimeric enzyme that executes the last step in aerobic respiration. An emerging theme is that defects in metabolism account for both cancer and aging.


Subject(s)
Aging/genetics , Aging/metabolism , Mitochondria/metabolism , Oxygen Consumption , Transcription Factors/genetics , Transcription Factors/metabolism , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism , Animals , Humans
16.
Cancer ; 125(14): 2409-2422, 2019 07 15.
Article in English | MEDLINE | ID: mdl-31012964

ABSTRACT

BACKGROUND: Over 96% of high-grade ovarian carcinomas and 50% of all cancers are characterized by alterations in the p53 gene. Therapeutic strategies to restore and/or reactivate the p53 pathway have been challenging. By contrast, p63, which shares many of the downstream targets and functions of p53, is rarely mutated in cancer. METHODS: A novel strategy is presented for circumventing alterations in p53 by inducing the tumor-suppressor isoform TAp63 (transactivation domain of tumor protein p63) through its direct downstream target, microRNA-130b (miR-130b), which is epigenetically silenced and/or downregulated in chemoresistant ovarian cancer. RESULTS: Treatment with miR-130b resulted in: 1) decreased migration/invasion in HEYA8 cells (p53 wild-type) and disruption of multicellular spheroids in OVCAR8 cells (p53-mutant) in vitro, 2) sensitization of HEYA8 and OVCAR8 cells to cisplatin (CDDP) in vitro and in vivo, and 3) transcriptional activation of TAp63 and the B-cell lymphoma (Bcl)-inhibitor B-cell lymphoma 2-like protein 11 (BIM). Overexpression of TAp63 was sufficient to decrease cell viability, suggesting that it is a critical downstream effector of miR-130b. In vivo, combined miR-130b plus CDDP exhibited greater therapeutic efficacy than miR-130b or CDDP alone. Mice that carried OVCAR8 xenograft tumors and were injected with miR-130b in 1,2-dioleoyl-sn-glycero-3-phosphatidylcholine (DOPC) liposomes had a significant decrease in tumor burden at rates similar to those observed in CDDP-treated mice, and 20% of DOPC-miR-130b plus CDDP-treated mice were living tumor free. Systemic injections of scL-miR-130b plus CDDP in a clinically tested, tumor-targeted nanocomplex (scL) improved survival in 60% and complete remissions in 40% of mice that carried HEYA8 xenografts. CONCLUSIONS: The miR-130b/TAp63 axis is proposed as a new druggable pathway that has the potential to uncover broad-spectrum therapeutic options for the majority of p53-altered cancers.


Subject(s)
MicroRNAs/therapeutic use , Mutation, Missense , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/genetics , Transcription Factors/genetics , Transcriptional Activation/genetics , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Proteins/genetics , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Binding Sites , Cell Line, Tumor , Cell Movement/drug effects , Cisplatin/pharmacology , Cisplatin/therapeutic use , Drug Resistance, Neoplasm/drug effects , Female , Humans , Liposomes , Mice , Mice, Nude , MicroRNAs/administration & dosage , MicroRNAs/genetics , MicroRNAs/metabolism , Neoplasm Invasiveness/prevention & control , Protein Isoforms/genetics , Signal Transduction/drug effects , Transcription Factors/metabolism , Transfection , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins/metabolism , Xenograft Model Antitumor Assays
17.
Stem Cells ; 36(12): 1875-1889, 2018 12.
Article in English | MEDLINE | ID: mdl-30267595

ABSTRACT

Mammary gland development is fueled by stem cell self-renewal and differentiation. External cues from the microenvironment coupled with internal cues such as post-transcriptional regulation exerted by microRNAs regulate stem cell behavior and fate. Here, we have identified a miR-205 regulatory network required for mammary gland ductal development and stem cell regeneration following transplantation into the cleared mammary fat pad. In the postnatal mammary gland, miR-205 is predominantly expressed in the basal/stem cell enriched population. Conditional deletion of miR-205 in mammary epithelial cells impairs stem cell self-renewal and mammary regenerative potential in the in vitro mammosphere formation assay and in vivo mammary reconstitution. miR-205 null transplants display significant changes in basal cells, basement membrane, and stroma. NKD1 and PTPA, which inhibit the Wnt signaling pathway, and AMOT, which causes YAP cytoplasmic retention and inactivation were identified as miR-205 downstream mediators. These studies also confirmed that miR-205 is a direct ΔNp63 target gene that is critical for the regulation of basal cell identity. Stem Cells 2018;36:1875-15.


Subject(s)
Cell Self Renewal/physiology , Mammary Glands, Animal/growth & development , MicroRNAs/metabolism , Animals , Disease Models, Animal , Female , Mammary Glands, Animal/cytology , Mammary Glands, Animal/metabolism , Mice , Mice, Knockout , Transfection
18.
Br J Cancer ; 116(2): 149-155, 2017 Jan 17.
Article in English | MEDLINE | ID: mdl-27884017

ABSTRACT

The p53 family of transcription factors is essential to counteract tumour formation and progression. Although previously this was exclusively associated with the ability of the p53 family to induce cell cycle arrest and apoptosis, an increasing number of reports have now indisputably demonstrated that the tumour suppressive functions of the p53 family members also rely on their ability to control and regulate cellular metabolism and maintain cellular oxidative homeostasis. Here, we review how each p53 family member, including p63 and p73, controls metabolic pathways in physiological conditions, and how these mechanisms could be exploited to provide anticancer therapeutic opportunities.


Subject(s)
Metabolism/genetics , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/therapy , Tumor Suppressor Protein p53/physiology , Animals , Gene Expression Regulation, Neoplastic , Genes, Tumor Suppressor , Humans
19.
Proc Natl Acad Sci U S A ; 111(5): E572-81, 2014 Feb 04.
Article in English | MEDLINE | ID: mdl-24449888

ABSTRACT

The roles of microRNAs (miRNAs) and the miRNA processing machinery in the regulation of stem cell biology are not well understood. Here, we show that the p53 family member and p63 isoform, ΔNp63, is a transcriptional activator of a cofactor critical for miRNA processing (DGCR8). This regulation gives rise to a unique miRNA signature resulting in reprogramming cells to multipotency. Strikingly, ΔNp63(-/-) epidermal cells display profound defects in terminal differentiation and express a subset of markers and miRNAs present in embryonic stem cells and fibroblasts induced to pluripotency using Yamanaka factors. Moreover, ΔNp63(-/-) epidermal cells transduced with an inducible DGCR8 plasmid can differentiate into multiple cell fates in vitro and in vivo. We found that human primary keratinocytes depleted of ΔNp63 or DGCR8 can be reprogrammed in 6 d and express a unique miRNA and gene expression signature that is similar but not identical to human induced pluripotent stem cells. Our data reveal a role for ΔNp63 in the transcriptional regulation of DGCR8 to reprogram adult somatic cells into multipotent stem cells.


Subject(s)
Down-Regulation/genetics , Keratinocytes/metabolism , Multipotent Stem Cells/cytology , Phosphoproteins/genetics , Proteins/genetics , RNA-Binding Proteins/genetics , Trans-Activators/genetics , Transcription Factors/genetics , Tumor Suppressor Proteins/genetics , Adult , Animals , Cell Differentiation , Cell Line , Cell Lineage , Cell Proliferation , Chimera , Embryo, Mammalian/cytology , Epidermal Cells , Gene Expression Profiling , Homeodomain Proteins/metabolism , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Keratinocytes/cytology , Mice , MicroRNAs/genetics , MicroRNAs/metabolism , Multipotent Stem Cells/metabolism , Nanog Homeobox Protein , Octamer Transcription Factor-3/metabolism , Phosphoproteins/deficiency , Phosphoproteins/metabolism , Proteins/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA-Binding Proteins/metabolism , SOXB1 Transcription Factors/metabolism , Trans-Activators/deficiency , Trans-Activators/metabolism , Transcription Factors/deficiency , Transcription Factors/metabolism , Transcription, Genetic , Tumor Suppressor Proteins/deficiency , Tumor Suppressor Proteins/metabolism
20.
Nature ; 467(7318): 986-90, 2010 Oct 21.
Article in English | MEDLINE | ID: mdl-20962848

ABSTRACT

Aberrant expression of microRNAs (miRNAs) and the enzymes that control their processing have been reported in multiple biological processes including primary and metastatic tumours, but the mechanisms governing this are not clearly understood. Here we show that TAp63, a p53 family member, suppresses tumorigenesis and metastasis, and coordinately regulates Dicer and miR-130b to suppress metastasis. Metastatic mouse and human tumours deficient in TAp63 express Dicer at very low levels, and we found that modulation of expression of Dicer and miR-130b markedly affected the metastatic potential of cells lacking TAp63. TAp63 binds to and transactivates the Dicer promoter, demonstrating direct transcriptional regulation of Dicer by TAp63. These data provide a novel understanding of the roles of TAp63 in tumour and metastasis suppression through the coordinate transcriptional regulation of Dicer and miR-130b and may have implications for the many processes regulated by miRNAs.


Subject(s)
DEAD-box RNA Helicases/metabolism , Endoribonucleases/metabolism , Gene Expression Regulation, Neoplastic , MicroRNAs/biosynthesis , Neoplasm Metastasis/genetics , Phosphoproteins/metabolism , Ribonuclease III/metabolism , Trans-Activators/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Cell Line , Cell Line, Tumor , Cellular Senescence , DEAD-box RNA Helicases/biosynthesis , DEAD-box RNA Helicases/deficiency , DEAD-box RNA Helicases/genetics , Endoribonucleases/genetics , Female , Genes, Tumor Suppressor/physiology , Genomic Instability , Humans , Male , Mice , Mice, Inbred C57BL , MicroRNAs/genetics , MicroRNAs/metabolism , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Phosphoproteins/deficiency , Phosphoproteins/genetics , Promoter Regions, Genetic/genetics , Ribonuclease III/biosynthesis , Ribonuclease III/deficiency , Ribonuclease III/genetics , Trans-Activators/deficiency , Trans-Activators/genetics , Transcription Factors , Transcriptional Activation , Tumor Suppressor Proteins/deficiency , Tumor Suppressor Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL