Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 135
Filter
2.
Nat Cell Biol ; 25(12): 1821-1832, 2023 Dec.
Article in English | MEDLINE | ID: mdl-38049604

ABSTRACT

Lineage transitions are a central feature of prostate development, tumourigenesis and treatment resistance. While epigenetic changes are well known to drive prostate lineage transitions, it remains unclear how upstream metabolic signalling contributes to the regulation of prostate epithelial identity. To fill this gap, we developed an approach to perform metabolomics on primary prostate epithelial cells. Using this approach, we discovered that the basal and luminal cells of the prostate exhibit distinct metabolomes and nutrient utilization patterns. Furthermore, basal-to-luminal differentiation is accompanied by increased pyruvate oxidation. We establish the mitochondrial pyruvate carrier and subsequent lactate accumulation as regulators of prostate luminal identity. Inhibition of the mitochondrial pyruvate carrier or supplementation with exogenous lactate results in large-scale chromatin remodelling, influencing both lineage-specific transcription factors and response to antiandrogen treatment. These results establish reciprocal regulation of metabolism and prostate epithelial lineage identity.


Subject(s)
Monocarboxylic Acid Transporters , Prostate , Male , Humans , Prostate/metabolism , Monocarboxylic Acid Transporters/metabolism , Cell Differentiation/physiology , Epithelial Cells/metabolism , Androgen Antagonists/pharmacology , Androgen Antagonists/metabolism , Lactates/metabolism
3.
4.
Nature ; 623(7988): 828-835, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37968399

ABSTRACT

The skin epidermis is constantly renewed throughout life1,2. Disruption of the balance between renewal and differentiation can lead to uncontrolled growth and tumour initiation3. However, the ways in which oncogenic mutations affect the balance between renewal and differentiation and lead to clonal expansion, cell competition, tissue colonization and tumour development are unknown. Here, through multidisciplinary approaches that combine in vivo clonal analysis using intravital microscopy, single-cell analysis and functional analysis, we show how SmoM2-a constitutively active oncogenic mutant version of Smoothened (SMO) that induces the development of basal cell carcinoma-affects clonal competition and tumour initiation in real time. We found that expressing SmoM2 in the ear epidermis of mice induced clonal expansion together with tumour initiation and invasion. By contrast, expressing SmoM2 in the back-skin epidermis led to a clonal expansion that induced lateral cell competition without dermal invasion and tumour formation. Single-cell analysis showed that oncogene expression was associated with a cellular reprogramming of adult interfollicular cells into an embryonic hair follicle progenitor (EHFP) state in the ear but not in the back skin. Comparisons between the ear and the back skin revealed that the dermis has a very different composition in these two skin types, with increased stiffness and a denser collagen I network in the back skin. Decreasing the expression of collagen I in the back skin through treatment with collagenase, chronic UV exposure or natural ageing overcame the natural resistance of back-skin basal cells to undergoing EHFP reprogramming and tumour initiation after SmoM2 expression. Altogether, our study shows that the composition of the extracellular matrix regulates how susceptible different regions of the body are to tumour initiation and invasion.


Subject(s)
Cell Transformation, Neoplastic , Extracellular Matrix , Skin Neoplasms , Tumor Microenvironment , Animals , Mice , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Collagen/metabolism , Epidermis/pathology , Extracellular Matrix/metabolism , Extracellular Matrix/pathology , Skin Neoplasms/pathology , Carcinoma, Basal Cell/pathology , Ear/pathology , Collagenases/metabolism , Aging , Ultraviolet Rays , Mutant Proteins/genetics , Mutant Proteins/metabolism
5.
Nature ; 620(7973): 402-408, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37532929

ABSTRACT

Epithelial-to-mesenchymal transition (EMT) regulates tumour initiation, progression, metastasis and resistance to anti-cancer therapy1-7. Although great progress has been made in understanding the role of EMT and its regulatory mechanisms in cancer, no therapeutic strategy to pharmacologically target EMT has been identified. Here we found that netrin-1 is upregulated in a primary mouse model of skin squamous cell carcinoma (SCC) exhibiting spontaneous EMT. Pharmacological inhibition of netrin-1 by administration of NP137, a netrin-1-blocking monoclonal antibody currently used in clinical trials in human cancer (ClinicalTrials.gov identifier NCT02977195 ), decreased the proportion of EMT tumour cells in skin SCC, decreased the number of metastases and increased the sensitivity of tumour cells to chemotherapy. Single-cell RNA sequencing revealed the presence of different EMT states, including epithelial, early and late hybrid EMT, and full EMT states, in control SCC. By contrast, administration of NP137 prevented the progression of cancer cells towards a late EMT state and sustained tumour epithelial states. Short hairpin RNA knockdown of netrin-1 and its receptor UNC5B in EPCAM+ tumour cells inhibited EMT in vitro in the absence of stromal cells and regulated a common gene signature that promotes tumour epithelial state and restricts EMT. To assess the relevance of these findings to human cancers, we treated mice transplanted with the A549 human cancer cell line-which undergoes EMT following TGFß1 administration8,9-with NP137. Netrin-1 inhibition decreased EMT in these transplanted A549 cells. Together, our results identify a pharmacological strategy for targeting EMT in cancer, opening up novel therapeutic interventions for anti-cancer therapy.


Subject(s)
Antibodies, Monoclonal , Carcinoma, Squamous Cell , Epithelial-Mesenchymal Transition , Netrin-1 , Skin Neoplasms , Animals , Humans , Mice , A549 Cells , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Epithelial-Mesenchymal Transition/drug effects , Netrin Receptors/antagonists & inhibitors , Netrin Receptors/deficiency , Netrin Receptors/genetics , Netrin-1/antagonists & inhibitors , Netrin-1/deficiency , Netrin-1/genetics , RNA, Small Interfering/genetics , RNA, Small Interfering/pharmacology , Disease Models, Animal , Skin Neoplasms/drug therapy , Skin Neoplasms/pathology , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Neoplasm Metastasis/drug therapy , Single-Cell Gene Expression Analysis , RNA-Seq , Epithelial Cell Adhesion Molecule/metabolism , Xenograft Model Antitumor Assays , Transforming Growth Factor beta1/pharmacology
6.
Nature ; 620(7973): 409-416, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37532934

ABSTRACT

Netrin-1 is upregulated in cancers as a protumoural mechanism1. Here we describe netrin-1 upregulation in a majority of human endometrial carcinomas (ECs) and demonstrate that netrin-1 blockade, using an anti-netrin-1 antibody (NP137), is effective in reduction of tumour progression in an EC mouse model. We next examined the efficacy of NP137, as a first-in-class single agent, in a Phase I trial comprising 14 patients with advanced EC. As best response we observed 8 stable disease (8 out of 14, 57.1%) and 1 objective response as RECIST v.1.1 (partial response, 1 out of 14 (7.1%), 51.16% reduction in target lesions at 6 weeks and up to 54.65% reduction during the following 6 months). To evaluate the NP137 mechanism of action, mouse tumour gene profiling was performed, and we observed, in addition to cell death induction, that NP137 inhibited epithelial-to-mesenchymal transition (EMT). By performing bulk RNA sequencing (RNA-seq), spatial transcriptomics and single-cell RNA-seq on paired pre- and on-treatment biopsies from patients with EC from the NP137 trial, we noted a net reduction in tumour EMT. This was associated with changes in immune infiltrate and increased interactions between cancer cells and the tumour microenvironment. Given the importance of EMT in resistance to current standards of care2, we show in the EC mouse model that a combination of NP137 with carboplatin-paclitaxel outperformed carboplatin-paclitaxel alone. Our results identify netrin-1 blockade as a clinical strategy triggering both tumour debulking and EMT inhibition, thus potentially alleviating resistance to standard treatments.


Subject(s)
Endometrial Neoplasms , Epithelial-Mesenchymal Transition , Netrin-1 , Animals , Female , Humans , Mice , Biopsy , Carboplatin/administration & dosage , Carboplatin/pharmacology , Carboplatin/therapeutic use , Disease Models, Animal , Drug Resistance, Neoplasm/drug effects , Endometrial Neoplasms/drug therapy , Endometrial Neoplasms/genetics , Endometrial Neoplasms/immunology , Endometrial Neoplasms/pathology , Epithelial-Mesenchymal Transition/drug effects , Gene Expression Profiling , Netrin-1/antagonists & inhibitors , Paclitaxel/administration & dosage , Paclitaxel/pharmacology , Paclitaxel/therapeutic use , RNA-Seq , Single-Cell Gene Expression Analysis , Tumor Microenvironment/drug effects
7.
Nat Cancer ; 4(8): 1063-1082, 2023 08.
Article in English | MEDLINE | ID: mdl-37537300

ABSTRACT

Cell plasticity represents the ability of cells to be reprogrammed and to change their fate and identity, enabling homeostasis restoration and tissue regeneration following damage. Cell plasticity also contributes to pathological conditions, such as cancer, enabling cells to acquire new phenotypic and functional features by transiting across distinct cell states that contribute to tumor initiation, progression, metastasis and resistance to therapy. Here, we review the intrinsic and extrinsic mechanisms driving cell plasticity that promote tumor growth and proliferation as well as metastasis and drug tolerance. Finally, we discuss how cell plasticity could be exploited for anti-cancer therapy.


Subject(s)
Cell Plasticity , Neoplasms , Humans , Cell Plasticity/physiology , Neoplasms/drug therapy , Cell Transformation, Neoplastic , Homeostasis
8.
Heliyon ; 9(7): e17842, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37456014

ABSTRACT

The mammary gland (MG) is composed of three main epithelial lineages, the basal cells (BC), the estrogen receptor (ER) positive luminal cells (ER+ LC), and the ER negative LC (ER- LC). Defining the cell identity of each lineage and how it is modulated throughout the different stages of life is important to understand how these cells function and communicate throughout life. Here, we used transgenic mice specifically labelling ER+ LC combined to cell surface markers to isolate with high purity the 3 distinct cell lineages of the mammary gland and defined their expression profiles and chromatin landscapes by performing bulk RNAseq and ATACseq of these isolated populations in puberty, adulthood and mid-pregnancy. Our analysis identified conserved genes, ligands and transcription factor (TF) associated with a specific lineage throughout life as well as genes, ligands and TFs specific for a particular stage of the MG. In summary, our study identified genes and TF network associated with the identity, function and cell-cell communication of the different epithelial lineages of the MG at different stages of life.

9.
Nature ; 620(7972): 192-199, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37495690

ABSTRACT

Sympathetic activation during cold exposure increases adipocyte thermogenesis via the expression of mitochondrial protein uncoupling protein 1 (UCP1)1. The propensity of adipocytes to express UCP1 is under a critical influence of the adipose microenvironment and varies between sexes and among various fat depots2-7. Here we report that mammary gland ductal epithelial cells in the adipose niche regulate cold-induced adipocyte UCP1 expression in female mouse subcutaneous white adipose tissue (scWAT). Single-cell RNA sequencing shows that glandular luminal epithelium subtypes express transcripts that encode secretory factors controlling adipocyte UCP1 expression under cold conditions. We term these luminal epithelium secretory factors 'mammokines'. Using 3D visualization of whole-tissue immunofluorescence, we reveal sympathetic nerve-ductal contact points. We show that mammary ducts activated by sympathetic nerves limit adipocyte UCP1 expression via the mammokine lipocalin 2. In vivo and ex vivo ablation of mammary duct epithelium enhance the cold-induced adipocyte thermogenic gene programme in scWAT. Since the mammary duct network extends throughout most of the scWAT in female mice, females show markedly less scWAT UCP1 expression, fat oxidation, energy expenditure and subcutaneous fat mass loss compared with male mice, implicating sex-specific roles of mammokines in adipose thermogenesis. These results reveal a role of sympathetic nerve-activated glandular epithelium in adipocyte UCP1 expression and suggest that mammary duct luminal epithelium has an important role in controlling glandular adiposity.


Subject(s)
Adipocytes , Adipose Tissue, White , Epithelium , Mammary Glands, Animal , Thermogenesis , Animals , Female , Male , Mice , Adipocytes/metabolism , Adipose Tissue, White/cytology , Adipose Tissue, White/metabolism , Epithelium/innervation , Epithelium/metabolism , Uncoupling Protein 1/genetics , Uncoupling Protein 1/metabolism , Mammary Glands, Animal/cytology , Mammary Glands, Animal/innervation , Mammary Glands, Animal/physiology , Cold Temperature , Sympathetic Nervous System/physiology , Energy Metabolism , Oxidation-Reduction , Sex Characteristics
10.
Nature ; 616(7955): 168-175, 2023 04.
Article in English | MEDLINE | ID: mdl-36949199

ABSTRACT

The resistance of cancer cells to therapy is responsible for the death of most patients with cancer1. Epithelial-to-mesenchymal transition (EMT) has been associated with resistance to therapy in different cancer cells2,3. However, the mechanisms by which EMT mediates resistance to therapy remain poorly understood. Here, using a mouse model of skin squamous cell carcinoma undergoing spontaneous EMT during tumorigenesis, we found that EMT tumour cells are highly resistant to a wide range of anti-cancer therapies both in vivo and in vitro. Using gain and loss of function studies in vitro and in vivo, we found that RHOJ-a small GTPase that is preferentially expressed in EMT cancer cells-controls resistance to therapy. Using genome-wide transcriptomic and proteomic profiling, we found that RHOJ regulates EMT-associated resistance to chemotherapy by enhancing the response to replicative stress and activating the DNA-damage response, enabling tumour cells to rapidly repair DNA lesions induced by chemotherapy. RHOJ interacts with proteins that regulate nuclear actin, and inhibition of actin polymerization sensitizes EMT tumour cells to chemotherapy-induced cell death in a RHOJ-dependent manner. Together, our study uncovers the role and the mechanisms through which RHOJ acts as a key regulator of EMT-associated resistance to chemotherapy.


Subject(s)
Carcinoma, Squamous Cell , Drug Resistance, Neoplasm , Epithelial-Mesenchymal Transition , Skin Neoplasms , rho GTP-Binding Proteins , Actins/drug effects , Actins/metabolism , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Epithelial-Mesenchymal Transition/drug effects , Proteomics , rho GTP-Binding Proteins/genetics , rho GTP-Binding Proteins/metabolism , Animals , Mice , Skin Neoplasms/drug therapy , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Gene Expression Profiling , Genome
12.
Nature ; 610(7930): 190-198, 2022 10.
Article in English | MEDLINE | ID: mdl-36131018

ABSTRACT

Although melanoma is notorious for its high degree of heterogeneity and plasticity1,2, the origin and magnitude of cell-state diversity remains poorly understood. Equally, it is unclear whether growth and metastatic dissemination are supported by overlapping or distinct melanoma subpopulations. Here, by combining mouse genetics, single-cell and spatial transcriptomics, lineage tracing and quantitative modelling, we provide evidence of a hierarchical model of tumour growth that mirrors the cellular and molecular logic underlying the cell-fate specification and differentiation of the embryonic neural crest. We show that tumorigenic competence is associated with a spatially localized perivascular niche, a phenotype acquired through an intercellular communication pathway established by endothelial cells. Consistent with a model in which only a fraction of cells are fated to fuel growth, temporal single-cell tracing of a population of melanoma cells with a mesenchymal-like state revealed that these cells do not contribute to primary tumour growth but, instead, constitute a pool of metastatic initiating cells that switch cell identity while disseminating to secondary organs. Our data provide a spatially and temporally resolved map of the diversity and trajectories of melanoma cell states and suggest that the ability to support growth and metastasis are limited to distinct pools of cells. The observation that these phenotypic competencies can be dynamically acquired after exposure to specific niche signals warrant the development of therapeutic strategies that interfere with the cancer cell reprogramming activity of such microenvironmental cues.


Subject(s)
Cell Proliferation , Melanoma , Neoplasm Metastasis , Animals , Cell Communication , Cell Differentiation , Cell Lineage , Cell Tracking , Cellular Reprogramming , Endothelial Cells , Melanoma/genetics , Melanoma/pathology , Mesoderm/pathology , Mice , Neoplasm Metastasis/pathology , Neural Crest/embryology , Phenotype , Single-Cell Analysis , Transcriptome , Tumor Microenvironment
13.
Nat Cell Biol ; 24(7): 1114-1128, 2022 07.
Article in English | MEDLINE | ID: mdl-35817961

ABSTRACT

The mammalian heart arises from various populations of Mesp1-expressing cardiovascular progenitors (CPs) that are specified during the early stages of gastrulation. Mesp1 is a transcription factor that acts as a master regulator of CP specification and differentiation. However, how Mesp1 regulates the chromatin landscape of nascent mesodermal cells to define the temporal and spatial patterning of the distinct populations of CPs remains unknown. Here, by combining ChIP-seq, RNA-seq and ATAC-seq during mouse pluripotent stem cell differentiation, we defined the dynamic remodelling of the chromatin landscape mediated by Mesp1. We identified different enhancers that are temporally regulated to erase the pluripotent state and specify the pools of CPs that mediate heart development. We identified Zic2 and Zic3 as essential cofactors that act with Mesp1 to regulate its transcription-factor activity at key mesodermal enhancers, thereby regulating the chromatin remodelling and gene expression associated with the specification of the different populations of CPs in vivo. Our study identifies the dynamics of the chromatin landscape and enhancer remodelling associated with temporal patterning of early mesodermal cells into the distinct populations of CPs that mediate heart development.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors , Chromatin , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Differentiation/genetics , Chromatin/genetics , Chromatin/metabolism , Enhancer Elements, Genetic/genetics , Gene Expression Regulation, Developmental , Heart , Homeodomain Proteins/metabolism , Mammals/metabolism , Mesoderm , Mice , Transcription Factors/genetics , Transcription Factors/metabolism
14.
Nat Rev Urol ; 19(4): 201-218, 2022 04.
Article in English | MEDLINE | ID: mdl-35079142

ABSTRACT

Stem and progenitor cells of the adult prostate epithelium have historically been believed to reside mainly or exclusively within the basal cell compartment and to possess basal-like phenotypic characteristics. Within the past decade, evidence of the existence of luminal epithelial cells exhibiting stem/progenitor properties has been obtained by lineage tracing and by functional characterization of sorted luminal-like cells. In 2020, the boom of single-cell transcriptomics led to increasingly exhaustive profiling of putative mouse luminal progenitor cells and, importantly, to the identification of cognate cells in the human prostate. The enrichment of luminal progenitor cells in genetically modified mouse models of prostate inflammation, benign prostate hypertrophy and prostate cancer, and the intrinsic castration tolerance of these cells, suggest their potential role in prostate pathogenesis and in resistance to androgen deprivation therapy. This Review bridges different approaches that have been used in the field to characterize luminal progenitor cells, including the unification of multiple identifiers employed to define these cells (names and markers). It also provides an overview of the intrinsic functional properties of luminal progenitor cells, and addresses their relevance in mouse and human prostate pathophysiology.


Subject(s)
Prostatic Hyperplasia , Prostatic Neoplasms , Androgen Antagonists , Animals , Epithelial Cells , Humans , Male , Mice , Prostate/pathology , Prostatic Hyperplasia/pathology , Prostatic Neoplasms/pathology , Stem Cells
15.
EMBO J ; 41(2): e109221, 2022 12 17.
Article in English | MEDLINE | ID: mdl-34918370

ABSTRACT

Within a tumor, cancer cells exist in different states that are associated with distinct tumor functions, including proliferation, differentiation, invasion, metastasis, and resistance to anti-cancer therapy. The identification of the gene regulatory networks underpinning each state is essential for better understanding functional tumor heterogeneity and revealing tumor vulnerabilities. Here, we review the different studies identifying tumor states by single-cell sequencing approaches and the mechanisms that promote and sustain these functional states and regulate their transitions. We also describe how different tumor states are spatially distributed and interact with the specific stromal cells that compose the tumor microenvironment. Finally, we discuss how the understanding of tumor plasticity and transition states can be used to develop new strategies to improve cancer therapy.


Subject(s)
Neoplasms/metabolism , Single-Cell Analysis/methods , Animals , Humans , Neoplasms/genetics , Neoplasms/pathology , RNA-Seq/methods
17.
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
18.
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
19.
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
20.
EMBO Mol Med ; 12(11): e13347, 2020 11 06.
Article in English | MEDLINE | ID: mdl-33063436

ABSTRACT

Merkel cell carcinoma (MCC) is a rare but very aggressive neuroendocrine cancer of the skin, with very limited therapeutic options. Although immunotherapy is effective in some cases, there is an unmet need for new therapeutic approaches in MCCs. In this issue of EMBO Molecular Medicine, Leiendecker et al identify a selective vulnerability of MCC for inhibitors of the lysine-specific histone demethylase 1A (LSD1). LSD1 inhibitors promote differentiation of tumor cells toward normal Merkel cell fate, impairing tumor cell growth in vivo, and opening new avenues for the treatment of patients with MCC.


Subject(s)
Carcinoma, Merkel Cell , Skin Neoplasms , Carcinoma, Merkel Cell/drug therapy , Carcinoma, Merkel Cell/genetics , Epigenesis, Genetic , Humans , Immunotherapy , Skin , Skin Neoplasms/drug therapy , Skin Neoplasms/genetics
SELECTION OF CITATIONS
SEARCH DETAIL