Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
2.
JCI Insight ; 6(5)2021 03 08.
Article in English | MEDLINE | ID: mdl-33497366

ABSTRACT

Cancer is caused primarily by genomic alterations resulting in deregulation of gene regulatory circuits in key growth, apoptosis, or DNA repair pathways. Multiple genes associated with the initiation and development of tumors are also regulated at the level of mRNA decay, through the recruitment of RNA-binding proteins to AU-rich elements (AREs) located in their 3'-untranslated regions. One of these ARE-binding proteins, tristetraprolin (TTP; encoded by Zfp36), is consistently dysregulated in many human malignancies. Herein, using regulated overexpression or conditional ablation in the context of cutaneous chemical carcinogenesis, we show that TTP represents a critical regulator of skin tumorigenesis. We provide evidence that TTP controlled both tumor-associated inflammation and key oncogenic pathways in neoplastic epidermal cells. We identify Areg as a direct target of TTP in keratinocytes and show that EGFR signaling potentially contributed to exacerbated tumor formation. Finally, single-cell RNA-Seq analysis indicated that ZFP36 was downregulated in human malignant keratinocytes. We conclude that TTP expression by epidermal cells played a major role in the control of skin tumorigenesis.


Subject(s)
Carcinogenesis/metabolism , Keratinocytes/metabolism , Skin Neoplasms/metabolism , Skin/metabolism , Tristetraprolin/metabolism , 3' Untranslated Regions , AU Rich Elements , Animals , Carcinogenesis/genetics , Disease Models, Animal , Down-Regulation , ErbB Receptors/metabolism , Gene Regulatory Networks , Humans , Inflammation/metabolism , Mice, Inbred C57BL , RNA Stability , RNA, Messenger/metabolism , RNA-Binding Proteins/metabolism , Signal Transduction , Skin Neoplasms/genetics
3.
Nature ; 589(7842): 448-455, 2021 01.
Article in English | MEDLINE | ID: mdl-33328637

ABSTRACT

FAT1, which encodes a protocadherin, is one of the most frequently mutated genes in human cancers1-5. However, the role and the molecular mechanisms by which FAT1 mutations control tumour initiation and progression are poorly understood. Here, using mouse models of skin squamous cell carcinoma and lung tumours, we found that deletion of Fat1 accelerates tumour initiation and malignant progression and promotes a hybrid epithelial-to-mesenchymal transition (EMT) phenotype. We also found this hybrid EMT state in FAT1-mutated human squamous cell carcinomas. Skin squamous cell carcinomas in which Fat1 was deleted presented increased tumour stemness and spontaneous metastasis. We performed transcriptional and chromatin profiling combined with proteomic analyses and mechanistic studies, which revealed that loss of function of FAT1 activates a CAMK2-CD44-SRC axis that promotes YAP1 nuclear translocation and ZEB1 expression that stimulates the mesenchymal state. This loss of function also inactivates EZH2, promoting SOX2 expression, which sustains the epithelial state. Our comprehensive analysis identified drug resistance and vulnerabilities in FAT1-deficient tumours, which have important implications for cancer therapy. Our studies reveal that, in mouse and human squamous cell carcinoma, loss of function of FAT1 promotes tumour initiation, progression, invasiveness, stemness and metastasis through the induction of a hybrid EMT state.


Subject(s)
Cadherins/deficiency , Epithelial-Mesenchymal Transition/genetics , Gene Deletion , Neoplasm Metastasis/genetics , Neoplasms/genetics , Neoplasms/pathology , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cadherins/genetics , Cadherins/metabolism , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Disease Progression , Enhancer of Zeste Homolog 2 Protein/metabolism , Epithelial Cells/metabolism , Epithelial Cells/pathology , Epithelial-Mesenchymal Transition/drug effects , Gene Expression Regulation, Neoplastic , Humans , Hyaluronan Receptors/metabolism , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Mesoderm/metabolism , Mesoderm/pathology , Mice , Neoplasm Metastasis/drug therapy , Neoplasms/drug therapy , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Phenotype , Phosphoproteins/analysis , Phosphoproteins/metabolism , Proteomics , SOXB1 Transcription Factors/metabolism , Signal Transduction , Skin Neoplasms/genetics , Skin Neoplasms/pathology , Transcription Factors/metabolism , YAP-Signaling Proteins , Zinc Finger E-box-Binding Homeobox 1/metabolism , src-Family Kinases/metabolism
4.
Nat Cancer ; 2(11): 1152-1169, 2021 11.
Article in English | MEDLINE | ID: mdl-35122061

ABSTRACT

The nongenetic mechanisms required to sustain malignant tumor state are poorly understood. During the transition from benign tumors to malignant carcinoma, tumor cells need to repress differentiation and acquire invasive features. Using transcriptional profiling of cancer stem cells from benign tumors and malignant skin squamous cell carcinoma (SCC), we identified the nuclear receptor NR2F2 as uniquely expressed in malignant SCC. Using genetic gain of function and loss of function in vivo, we show that NR2F2 is essential for promoting the malignant tumor state by controlling tumor stemness and maintenance in mouse and human SCC. We demonstrate that NR2F2 promotes tumor cell proliferation, epithelial-mesenchymal transition and invasive features, while repressing tumor differentiation and immune cell infiltration by regulating a common transcriptional program in mouse and human SCCs. Altogether, we identify NR2F2 as a key regulator of malignant cancer stem cell functions that promotes tumor renewal and restricts differentiation to sustain a malignant tumor state.


Subject(s)
Carcinoma, Squamous Cell , Skin Neoplasms , Animals , Carcinoma, Squamous Cell/genetics , Cell Differentiation/genetics , Epithelial-Mesenchymal Transition/genetics , Gene Expression Regulation, Neoplastic , Mice , Neoplastic Processes , Skin Neoplasms/genetics
5.
Nature ; 584(7820): 268-273, 2020 08.
Article in English | MEDLINE | ID: mdl-32728211

ABSTRACT

The ability of the skin to grow in response to stretching has been exploited in reconstructive surgery1. Although the response of epidermal cells to stretching has been studied in vitro2,3, it remains unclear how mechanical forces affect their behaviour in vivo. Here we develop a mouse model in which the consequences of stretching on skin epidermis can be studied at single-cell resolution. Using a multidisciplinary approach that combines clonal analysis with quantitative modelling and single-cell RNA sequencing, we show that stretching induces skin expansion by creating a transient bias in the renewal activity of epidermal stem cells, while a second subpopulation of basal progenitors remains committed to differentiation. Transcriptional and chromatin profiling identifies how cell states and gene-regulatory networks are modulated by stretching. Using pharmacological inhibitors and mouse mutants, we define the step-by-step mechanisms that control stretch-mediated tissue expansion at single-cell resolution in vivo.


Subject(s)
Mechanotransduction, Cellular/physiology , Single-Cell Analysis , Skin/cytology , Skin/growth & development , Adaptor Proteins, Signal Transducing/metabolism , Adherens Junctions/metabolism , Animals , Base Sequence , Cell Cycle Proteins/metabolism , Cell Differentiation/drug effects , Cell Self Renewal/drug effects , Chromatin/drug effects , Chromatin/genetics , Chromatin Assembly and Disassembly/drug effects , Clone Cells/cytology , Clone Cells/drug effects , Clone Cells/metabolism , Disease Models, Animal , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Regulatory Networks/drug effects , Hydrogels/administration & dosage , Hydrogels/pharmacology , Mechanotransduction, Cellular/drug effects , Mechanotransduction, Cellular/genetics , Mice , Mice, Transgenic , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinase Kinases/metabolism , Mutation , RNA, Messenger/genetics , RNA-Seq , Skin/drug effects , Stem Cells/cytology , Stem Cells/drug effects , Stem Cells/metabolism , Trans-Activators/antagonists & inhibitors , Trans-Activators/metabolism , Transcription Factor AP-1/metabolism , Transcription, Genetic/drug effects , YAP-Signaling Proteins
6.
Nature ; 562(7727): 434-438, 2018 10.
Article in English | MEDLINE | ID: mdl-30297799

ABSTRACT

Basal cell carcinoma (BCC) is the most frequent cancer in humans and results from constitutive activation of the Hedgehog pathway1. Several Smoothened inhibitors are used to treat Hedgehog-mediated malignancies, including BCC and medulloblastoma2. Vismodegib, a Smoothened inhibitor, leads to BCC shrinkage in the majority of patients with BCC3, but the mechanism by which it mediates BCC regression is unknown. Here we used two genetically engineered mouse models of BCC4 to investigate the mechanisms by which inhibition of Smoothened mediates tumour regression. We found that vismodegib mediates BCC regression by inhibiting a hair follicle-like fate and promoting the differentiation of tumour cells. However, a small population of tumour cells persists and is responsible for tumour relapse following treatment discontinuation, mimicking the situation found in humans5. In both mouse and human BCC, this persisting, slow-cycling tumour population expresses LGR5 and is characterized by active Wnt signalling. Combining Lgr5 lineage ablation or inhibition of Wnt signalling with vismodegib treatment leads to eradication of BCC. Our results show that vismodegib induces tumour regression by promoting tumour differentiation, and demonstrates that the synergy between Wnt and Smoothened inhibitors is a clinically relevant strategy for overcoming tumour relapse in BCC.


Subject(s)
Anilides/pharmacology , Anilides/therapeutic use , Carcinoma, Basal Cell/drug therapy , Carcinoma, Basal Cell/pathology , Neoplasm Recurrence, Local , Pyridines/pharmacology , Pyridines/therapeutic use , Receptors, G-Protein-Coupled/metabolism , Anilides/administration & dosage , Animals , Carcinoma, Basal Cell/genetics , Cell Differentiation/drug effects , Cell Lineage/drug effects , Disease Models, Animal , Female , Hair Follicle/cytology , Hair Follicle/drug effects , Hedgehog Proteins/antagonists & inhibitors , Hedgehog Proteins/metabolism , Humans , Male , Mice , Neoplasm Recurrence, Local/prevention & control , Patched-1 Receptor/deficiency , Pyridines/administration & dosage , Recurrence , Secondary Prevention , Skin Neoplasms/drug therapy , Skin Neoplasms/genetics , Skin Neoplasms/pathology , Smoothened Receptor/antagonists & inhibitors , Withholding Treatment , Wnt Proteins/antagonists & inhibitors , Wnt Proteins/metabolism , Wnt Signaling Pathway/drug effects
7.
Stem Cells ; 35(5): 1355-1364, 2017 05.
Article in English | MEDLINE | ID: mdl-28100039

ABSTRACT

The carcinogenic effect of ionizing radiation has been evaluated based on limited populations accidently exposed to high dose radiation. In contrast, insufficient data are available on the effect of low dose radiation (LDR), such as radiation deriving from medical investigations and interventions, as well as occupational exposure that concern a large fraction of western populations. Using mouse skin epidermis as a model, we showed that LDR results in DNA damage in sebaceous gland (SG) and bulge epidermal stem cells (SCs). While the first commit apoptosis upon low dose irradiation, the latter survive. Bulge SC survival coincides with higher HIF-1α expression and a metabolic switch upon LDR. Knocking down HIF-1α sensitizes bulge SCs to LDR-induced apoptosis, while upregulation of HIF-1α in the epidermis, including SG SCs, rescues cell death. Most importantly, we show that LDR results in cancer formation with full penetrance in the radiation-sensitive Patched1 heterozygous mice. Overall, our results demonstrate for the first time that LDR can be a potent carcinogen in individuals predisposed to cancer. Stem Cells 2017;35:1355-1364.


Subject(s)
Epidermis/radiation effects , Skin Neoplasms/etiology , Stem Cells/radiation effects , Animals , Carcinoma, Basal Cell/pathology , Cell Survival/radiation effects , DNA Damage , Dose-Response Relationship, Radiation , Heterozygote , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mice , Patched-1 Receptor/metabolism , Penetrance , Sebaceous Glands/pathology , Skin Neoplasms/pathology , Stem Cells/metabolism
8.
Nat Med ; 22(8): 861-8, 2016 08.
Article in English | MEDLINE | ID: mdl-27376578

ABSTRACT

In a search for mediators of the p53 tumor suppressor pathway, which induces pleiotropic and often antagonistic cellular responses, we identified the long noncoding RNA (lncRNA) NEAT1. NEAT1 is an essential architectural component of paraspeckle nuclear bodies, whose pathophysiological relevance remains unclear. Activation of p53, pharmacologically or by oncogene-induced replication stress, stimulated the formation of paraspeckles in mouse and human cells. Silencing Neat1 expression in mice, which prevents paraspeckle formation, sensitized preneoplastic cells to DNA-damage-induced cell death and impaired skin tumorigenesis. We provide mechanistic evidence that NEAT1 promotes ATR signaling in response to replication stress and is thereby engaged in a negative feedback loop that attenuates oncogene-dependent activation of p53. NEAT1 targeting in established human cancer cell lines induced synthetic lethality with genotoxic chemotherapeutics, including PARP inhibitors, and nongenotoxic activation of p53. This study establishes a key genetic link between NEAT1 paraspeckles, p53 biology and tumorigenesis and identifies NEAT1 as a promising target to enhance sensitivity of cancer cells to both chemotherapy and p53 reactivation therapy.


Subject(s)
Carcinogenesis/genetics , Gene Expression Regulation, Neoplastic/genetics , RNA, Long Noncoding/genetics , Tumor Suppressor Protein p53/genetics , Animals , Ataxia Telangiectasia Mutated Proteins , Cell Line, Tumor , DNA Damage , Flow Cytometry , Humans , Immunoblotting , Immunohistochemistry , In Situ Hybridization, Fluorescence , Kaplan-Meier Estimate , MCF-7 Cells , Mice , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Precancerous Conditions/genetics , Proportional Hazards Models , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Skin Neoplasms/genetics , Survival Analysis
9.
Cell Stem Cell ; 16(1): 67-79, 2015 Jan 08.
Article in English | MEDLINE | ID: mdl-25575080

ABSTRACT

Twist1 promotes epithelial-to-mesenchymal transition (EMT), invasion, metastasis, and cancer stem cell (CSC) properties. However, it remains unclear whether Twist1 is also required for tumor initiation and whether Twist1-induced cancer stemness and EMT are functionally linked. Using a conditional deletion of Twist1 at different stages of skin carcinogenesis, we show that Twist1 is required for skin tumor initiation and progression in a gene-dosage-dependent manner. Moreover, conditional ablation of Twist1 in benign tumors leads to increased apoptosis, reduced cell proliferation, and defective tumor maintenance and propagation independently of its EMT-inducing abilities. Concomitant deletion of Twist1 and p53 rescues the apoptotic response, but not the cell proliferation and propagation defects. These results reveal that Twist1 is required for tumor initiation and maintenance in a p53-dependent and -independent manner. Importantly, our findings also indicate that tumor stemness and EMT can be regulated by distinct mechanisms.


Subject(s)
Carcinogenesis/metabolism , Disease Progression , Neoplastic Stem Cells/metabolism , Nuclear Proteins/metabolism , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Twist-Related Protein 1/metabolism , Animals , Carcinogenesis/pathology , Epithelial-Mesenchymal Transition , Humans , Mice , Mice, Inbred NOD , Neoplastic Stem Cells/pathology , Protein Stability , Tumor Suppressor Protein p53/metabolism
10.
Nature ; 511(7508): 246-50, 2014 Jul 10.
Article in English | MEDLINE | ID: mdl-24909994

ABSTRACT

Cancer stem cells (CSCs) have been reported in various cancers, including in skin squamous-cell carcinoma (SCC). The molecular mechanisms regulating tumour initiation and stemness are still poorly characterized. Here we find that Sox2, a transcription factor expressed in various types of embryonic and adult stem cells, was the most upregulated transcription factor in the CSCs of squamous skin tumours in mice. SOX2 is absent in normal epidermis but begins to be expressed in the vast majority of mouse and human pre-neoplastic skin tumours, and continues to be expressed in a heterogeneous manner in invasive mouse and human SCCs. In contrast to other SCCs, in which SOX2 is frequently genetically amplified, the expression of SOX2 in mouse and human skin SCCs is transcriptionally regulated. Conditional deletion of Sox2 in the mouse epidermis markedly decreases skin tumour formation after chemical-induced carcinogenesis. Using green fluorescent protein (GFP) as a reporter of Sox2 transcriptional expression (SOX2-GFP knock-in mice), we showed that SOX2-expressing cells in invasive SCC are greatly enriched in tumour-propagating cells, which further increase upon serial transplantations. Lineage ablation of SOX2-expressing cells within primary benign and malignant SCCs leads to tumour regression, consistent with the critical role of SOX2-expressing cells in tumour maintenance. Conditional Sox2 deletion in pre-existing skin papilloma and SCC leads to tumour regression and decreases the ability of cancer cells to be propagated upon transplantation into immunodeficient mice, supporting the essential role of SOX2 in regulating CSC functions. Transcriptional profiling of SOX2-GFP-expressing CSCs and of tumour epithelial cells upon Sox2 deletion uncovered a gene network regulated by SOX2 in primary tumour cells in vivo. Chromatin immunoprecipitation identified several direct SOX2 target genes controlling tumour stemness, survival, proliferation, adhesion, invasion and paraneoplastic syndrome. We demonstrate that SOX2, by marking and regulating the functions of skin tumour-initiating cells and CSCs, establishes a continuum between tumour initiation and progression in primary skin tumours.


Subject(s)
Carcinoma, Squamous Cell , Cell Transformation, Neoplastic/genetics , Neoplastic Stem Cells/metabolism , SOXB1 Transcription Factors/metabolism , Skin Neoplasms , Animals , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Cell Adhesion/genetics , Cell Proliferation , Cell Transformation, Neoplastic/metabolism , Disease Models, Animal , Gene Deletion , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Gene Regulatory Networks/genetics , Mice , Mice, Inbred Strains , SOXB1 Transcription Factors/genetics , Skin Neoplasms/genetics , Skin Neoplasms/pathology
11.
Nat Cell Biol ; 14(12): 1282-94, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23178882

ABSTRACT

Basal cell carcinoma, the most frequent human skin cancer, arises from activating hedgehog (HH) pathway mutations; however, little is known about the temporal changes that occur in tumour-initiating cells from the first oncogenic hit to the development of invasive cancer. Using an inducible mouse model enabling the expression of a constitutively active Smoothened mutant (SmoM2) in the adult epidermis, we carried out transcriptional profiling of SmoM2-expressing cells at different times during cancer initiation. We found that tumour-initiating cells are massively reprogrammed into a fate resembling that of embryonic hair follicle progenitors (EHFPs). Wnt/ ß-catenin signalling was very rapidly activated following SmoM2 expression in adult epidermis and coincided with the expression of EHFP markers. Deletion of ß-catenin in adult SmoM2-expressing cells prevents EHFP reprogramming and tumour initiation. Finally, human basal cell carcinomas also express genes of the Wnt signalling and EHFP signatures.


Subject(s)
Carcinoma, Basal Cell/pathology , Hair Follicle/cytology , Neoplastic Stem Cells/cytology , Animals , Carcinoma, Basal Cell/metabolism , Flow Cytometry , Hair Follicle/metabolism , Immunohistochemistry , Keratinocytes/cytology , Keratinocytes/metabolism , Mice , Mice, Transgenic , Neoplastic Stem Cells/metabolism , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , beta Catenin/metabolism
12.
EMBO J ; 31(24): 4563-75, 2012 Dec 12.
Article in English | MEDLINE | ID: mdl-23188079

ABSTRACT

Cancer stem cells have been described in various cancers including squamous tumours of the skin by their ability to reform secondary tumours upon transplantation into immunodeficient mice. Here, we used transplantation of limiting dilution of different populations of FACS-isolated tumour cells from four distinct mouse models of squamous skin tumours to investigate the frequency of tumour propagating cells (TPCs) at different stages of tumour progression. We found that benign papillomas, despite growing rapidly in vivo and being clonogenic in vitro, reformed secondary tumours upon transplantation at very low frequency and only when tumour cells were co-transplanted together with tumour-associated fibroblasts or endothelial cells. In two models of skin squamous cell carcinoma (SCC), TPCs increased with tumour invasiveness. Interestingly, the frequency of TPCs increased in CD34(HI) but not in CD34(LO) SCC cells with serial transplantations, while the two populations initially gave rise to secondary tumours with the same frequency. Our results illustrate the progressive increase of squamous skin TPCs with tumour progression and invasiveness and reveal that serial transplantation may be required to define the long-term renewal potential of TPCs.


Subject(s)
Carcinoma, Squamous Cell/physiopathology , Models, Biological , Neoplasm Invasiveness/physiopathology , Neoplastic Stem Cells/physiology , Skin Neoplasms/physiopathology , Animals , Antigens, CD34/metabolism , Cell Proliferation , DNA Primers/genetics , Disease Progression , Flow Cytometry , Histological Techniques , Mice , Mice, SCID , Neoplastic Stem Cells/cytology , Papilloma/physiopathology , Real-Time Polymerase Chain Reaction , Stem Cell Transplantation , Tamoxifen
13.
Nature ; 478(7369): 399-403, 2011 Oct 19.
Article in English | MEDLINE | ID: mdl-22012397

ABSTRACT

Angiogenesis is critical during tumour initiation and malignant progression. Different strategies aimed at blocking vascular endothelial growth factor (VEGF) and its receptors have been developed to inhibit angiogenesis in cancer patients. It has become increasingly clear that in addition to its effect on angiogenesis, other mechanisms including a direct effect of VEGF on tumour cells may account for the efficiency of VEGF-blockade therapies. Cancer stem cells (CSCs) have been described in various cancers including squamous tumours of the skin. Here we use a mouse model of skin tumours to investigate the impact of the vascular niche and VEGF signalling on controlling the stemness (the ability to self renew and differentiate) of squamous skin tumours during the early stages of tumour progression. We show that CSCs of skin papillomas are localized in a perivascular niche, in the immediate vicinity of endothelial cells. Furthermore, blocking VEGFR2 caused tumour regression not only by decreasing the microvascular density, but also by reducing CSC pool size and impairing CSC renewal properties. Conditional deletion of Vegfa in tumour epithelial cells caused tumours to regress, whereas VEGF overexpression by tumour epithelial cells accelerated tumour growth. In addition to its well-known effect on angiogenesis, VEGF affected skin tumour growth by promoting cancer stemness and symmetric CSC division, leading to CSC expansion. Moreover, deletion of neuropilin-1 (Nrp1), a VEGF co-receptor expressed in cutaneous CSCs, blocked VEGF's ability to promote cancer stemness and renewal. Our results identify a dual role for tumour-cell-derived VEGF in promoting cancer stemness: by stimulating angiogenesis in a paracrine manner, VEGF creates a perivascular niche for CSCs, and by directly affecting CSCs through Nrp1 in an autocrine loop, VEGF stimulates cancer stemness and renewal. Finally, deletion of Nrp1 in normal epidermis prevents skin tumour initiation. These results may have important implications for the prevention and treatment of skin cancers.


Subject(s)
Carcinoma, Squamous Cell/blood supply , Carcinoma, Squamous Cell/pathology , Neuropilin-1/metabolism , Signal Transduction , Skin Neoplasms/blood supply , Skin Neoplasms/pathology , Vascular Endothelial Growth Factor A/metabolism , Animals , Cell Differentiation , Cell Proliferation , Cells, Cultured , Disease Models, Animal , Epithelial Cells/cytology , Gene Deletion , Gene Expression Regulation, Neoplastic , Mice , Neoplastic Stem Cells , Neuropilin-1/genetics , Vascular Endothelial Growth Factor A/genetics
14.
Proc Natl Acad Sci U S A ; 108(18): 7431-6, 2011 May 03.
Article in English | MEDLINE | ID: mdl-21502497

ABSTRACT

Squamous cell carcinoma (SCC) is the second most frequent skin cancer. The cellular origin of SCC remains controversial. Here, we used mouse genetics to determine the epidermal cell lineages at the origin of SCC. Using mice conditionally expressing a constitutively active KRas mutant (G12D) and an inducible CRE recombinase in different epidermal lineages, we activated Ras signaling in different cellular compartments of the skin epidermis and determined from which epidermal compartments Ras activation induces squamous tumor formation. Expression of mutant KRas in hair follicle bulge stem cells (SCs) and their immediate progeny (hair germ and outer root sheath), but not in their transient amplifying matrix cells, led to benign squamous skin tumor (papilloma). Expression of KRas(G12D) in interfollicular epidermis also led to papilloma formation, demonstrating that squamous tumor initiation is not restricted to the hair follicle lineages. Whereas no malignant tumor was observed after KRas(G12D) expression alone, expression of KRas(G12D) combined with the loss of p53 induced invasive SCC. Our studies demonstrate that different epidermal lineages including bulge SC are competent to initiate papilloma formation and that multiple genetic hits in the context of oncogenic KRas are required for the development of invasive SCC.


Subject(s)
Carcinoma, Squamous Cell/physiopathology , Cell Lineage/physiology , Epidermal Cells , Skin Neoplasms/physiopathology , Animals , Bromodeoxyuridine , Carcinoma, Squamous Cell/genetics , DNA Primers/genetics , Flow Cytometry , Hair Follicle/cytology , Integrases/metabolism , Mice , Mice, Transgenic , Proto-Oncogene Proteins p21(ras)/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Skin Neoplasms/genetics , Stem Cells/metabolism
16.
Nat Cell Biol ; 12(6): 572-82, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20473297

ABSTRACT

Adult stem cells (SCs) are at high risk of accumulating deleterious mutations because they reside and self-renew in adult tissues for extended periods. Little is known about how adult SCs sense and respond to DNA damage within their natural niche. Here, using mouse epidermis as a model, we define the functional consequences and the molecular mechanisms by which adult SCs respond to DNA damage. We show that multipotent hair-follicle-bulge SCs have two important mechanisms for increasing their resistance to DNA-damage-induced cell death: higher expression of the anti-apoptotic gene Bcl-2 and transient stabilization of p53 after DNA damage in bulge SCs. The attenuated p53 activation is the consequence of a faster DNA repair activity, mediated by a higher non-homologous end joining (NHEJ) activity, induced by the key protein DNA-PK. Because NHEJ is an error-prone mechanism, this novel characteristic of adult SCs may have important implications in cancer development and ageing.


Subject(s)
DNA Repair , Hair Follicle/cytology , Multipotent Stem Cells/cytology , Multipotent Stem Cells/physiology , Stem Cells/metabolism , Adult , Aging , Animals , Biochemical Phenomena , Cell Death , DNA/metabolism , DNA Damage , Epidermis/metabolism , Hair Follicle/metabolism , Hair Follicle/physiology , Humans , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred Strains , Mice, Knockout , Mice, SCID , Multipotent Stem Cells/metabolism , Tumor Suppressor Protein p53/metabolism
17.
Nat Cell Biol ; 12(3): 299-305, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20154679

ABSTRACT

For most types of cancers, the cell at the origin of tumour initiation is still unknown. Here, we used mouse genetics to identify cells at the origin of basal cell carcinoma (BCC), which is one of the most frequently occurring types of cancer in humans, and can result from the activation of the Hedgehog signalling pathway. Using mice conditionally expressing constitutively active Smoothened mutant (SmoM2), we activated Hedgehog signalling in different cellular compartments of the skin epidermis and determined in which compartments Hedgehog activation induces BCC formation. Activation of SmoM2 in hair follicle bulge stem cells and their transient amplifying progenies did not induce cancer formation, demonstrating that BCC does not originate from bulge stem cells, as previously thought. Using clonal analysis, we found that BCC arises from long-term resident progenitor cells of the interfollicular epidermis and the upper infundibulum. Our studies uncover the cells at the origin of BCC in mice and demonstrate that expression of differentiation markers in tumour cells is not necessarily predictive of the cancer initiating cells.


Subject(s)
Carcinoma, Basal Cell/pathology , Cell Lineage , Epidermis/pathology , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Cadherins/metabolism , Carcinoma, Basal Cell/metabolism , Cell Count , Cell Differentiation , Clone Cells/metabolism , Clone Cells/pathology , Ear, External/pathology , Epidermis/metabolism , Epithelial Cells/metabolism , Epithelial Cells/pathology , Genes, Reporter/genetics , Hair Follicle/metabolism , Hair Follicle/pathology , Hedgehog Proteins/genetics , Integrases/genetics , Integrin beta4/metabolism , Keratin-10/metabolism , Keratin-14/genetics , Keratin-15/genetics , Keratin-15/metabolism , Keratin-19/genetics , Kruppel-Like Transcription Factors/metabolism , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Mice , Mice, Inbred Strains , Mice, Transgenic , Models, Biological , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Patched Receptors , Proteins/genetics , Proteins/metabolism , RNA, Untranslated , Receptors, Cell Surface/metabolism , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Skin/metabolism , Skin/pathology , Smoothened Receptor , Tail/pathology
18.
Hum Mutat ; 31(1): 74-80, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19830810

ABSTRACT

Advanced prostate cancer (PCa) has emerged as a public health concern due to population aging. Although androgen deprivation has proven efficacy in this condition, most advanced PCa patients will have to face failure of androgen deprivation as a treatment. Mutations in the androgen receptor (AR) from tumor cells have been shown to induce androgen independency both in PCa cell lines and in the clinic. We have investigated the molecular events leading to androgen independency in the 22Rv1 cell line, a commonly used preclinical model of PCa. Besides AR mutants that have been described so far, including nonsense mutations, recent data have focused on AR pre-mRNA aberrant splicing as a new mechanism leading to constitutively active truncated AR variants. In this article, we describe two novel variants arising from aberrant splicing of AR pre-mRNA, characterized by long mRNA transcripts that encode truncated, constitutively active proteins. We also describe several new nonsense mutants that share ligand independency and transcriptional activity. Finally, we show that alongside these mutants, 22Rv1 cells also express a mutant AR lacking exon 3 tandem duplication, a major feature of this cell line. By describing unreported AR mutants in the 22Rv1 cell line, our data emphasize the complexity and heterogeneity of molecular events that occur in preclinical models, and supposedly in the clinic. Future work on the 22Rv1 cell line should take into account the concomitant expression of various AR mutants.


Subject(s)
Alternative Splicing , Mutation , Neoplasms, Hormone-Dependent/genetics , Prostatic Neoplasms/genetics , RNA Precursors , Receptors, Androgen/genetics , Androgens/metabolism , Cell Line, Tumor , Exons , Gene Expression Regulation, Neoplastic , Genetic Variation , Humans , Male , RNA Precursors/genetics , RNA Precursors/metabolism , Receptors, Androgen/metabolism
19.
Cell Stem Cell ; 3(1): 69-84, 2008 Jul 03.
Article in English | MEDLINE | ID: mdl-18593560

ABSTRACT

During embryonic development, multipotent cardiovascular progenitor cells are specified from early mesoderm. Using mouse ESCs in which gene expression can be temporally regulated, we have found that transient expression of Mesp1 dramatically accelerates and enhances multipotent cardiovascular progenitor specification through an intrinsic and cell autonomous mechanism. Genome-wide transcriptional analysis indicates that Mesp1 rapidly activates and represses a discrete set of genes, and chromatin immunoprecipitation shows that Mesp1 directly binds to regulatory DNA sequences located in the promoter of many key genes in the core cardiac transcriptional machinery, resulting in their rapid upregulation. Mesp1 also directly represses the expression of key genes regulating other early mesoderm and endoderm cell fates. Our results demonstrate that Mesp1 acts as a key regulatory switch during cardiovascular specification, residing at the top of the hierarchy of the gene network responsible for cardiovascular cell-fate determination.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/physiology , Cardiovascular System/cytology , Cell Differentiation/physiology , Pluripotent Stem Cells/cytology , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Embryonic Development , Gene Expression Regulation, Developmental , Heart/embryology , Heart/physiology , Helix-Loop-Helix Motifs/physiology , Homeostasis , Mice , Myocardium/cytology , Pluripotent Stem Cells/physiology
20.
Adv Exp Med Biol ; 617: 529-34, 2008.
Article in English | MEDLINE | ID: mdl-18497078

ABSTRACT

Mutations in the human androgen receptor (AR) gene that lead to C-terminus truncated AR variants are frequently detected in prostate cancer (PC). These AR variants lack both the ligand-binding domain (LBD) and the AF-2 region. The aim of this study was to delineate the alternative mechanisms that lead to the activation of such AR variants as they are unresponsive to hormone stimulation, and to outline consequences of the loss of the LBD/AF-2 region on their functional properties. By using an MMTV-luciferase reporter construct and LY294002, UO126, or ZD1839, inhibitor of PI3K, MEK1/2, and EGFR signaling pathway respectively, we demonstrated that phosphorylation was required for full transcriptional activities of one these AR variants, the Q640X mutant AR. Western-blot analyses confirmed that these inhibitors affect the phosphorylation status of this AR variant. Furthermore, studies of the intranuclear colocalization of the Q640X AR with cofactors, such as CBP, GRIP-1, and c-Jun, reveal that the transcriptional complex that forms around the mutant AR is different to that formed around the wild type AR. We demonstrated that CBP and c-Jun are highly recruited by the mutant AR, and this leads to an unexpected activation of AP-1, NFAT, and NFkappaB transcriptional activities. Similar enhanced activities of these transcription factors were not observed with the wild type AR. The importance of the LBD/AF-2 for the regulation of AR transcriptional activities, the impact of the presence of such AR variants on PC cells proliferation and survival, and on progression to androgen independence are discussed.


Subject(s)
Gene Expression Regulation, Neoplastic , Genetic Variation , Neoplasms, Hormone-Dependent/genetics , Prostatic Neoplasms/genetics , Receptors, Androgen/genetics , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/metabolism , Humans , MAP Kinase Kinase 1/antagonists & inhibitors , MAP Kinase Kinase 1/metabolism , MAP Kinase Kinase 2/antagonists & inhibitors , MAP Kinase Kinase 2/metabolism , Male , NF-kappa B/genetics , NF-kappa B/metabolism , NFATC Transcription Factors/genetics , NFATC Transcription Factors/metabolism , Neoplasms, Hormone-Dependent/metabolism , Neoplasms, Hormone-Dependent/pathology , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Receptors, Androgen/metabolism , Transcription Factor AP-1/genetics , Transcription Factor AP-1/metabolism , Transcription, Genetic , Transcriptional Activation , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...