Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 61
Filter
1.
Nat Commun ; 15(1): 2700, 2024 Mar 27.
Article in English | MEDLINE | ID: mdl-38538573

ABSTRACT

Hyperactivation of YAP/TAZ, the Hippo pathway downstream effectors, is common in human cancer. The requirement of YAP/TAZ for cancer cell survival in preclinical models, prompted the development of pharmacological inhibitors that suppress their transcriptional activity. However, systemic YAP/TAZ inhibition may sometimes have unpredictable patient outcomes, with limited or even adverse effects because YAP/TAZ action is not simply tumor promoting but also tumor suppressive in some cell types. Here, we review the role of the Hippo pathway in distinct tumor cell populations, discuss the impact of inhibiting Hippo output on tumor growth, and examine current developments in YAP/TAZ inhibitors.


Subject(s)
Neoplasms , Signal Transduction , Humans , Adaptor Proteins, Signal Transducing/metabolism , Transcription Factors/metabolism , Protein Serine-Threonine Kinases/metabolism , YAP-Signaling Proteins , Neoplasms/drug therapy , Neoplasms/genetics
2.
Nat Cell Biol ; 26(1): 153-167, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38182825

ABSTRACT

In the mammalian liver, hepatocytes exhibit diverse metabolic and functional profiles based on their location within the liver lobule. However, it is unclear whether this spatial variation, called zonation, is governed by a well-defined gene regulatory code. Here, using a combination of single-cell multiomics, spatial omics, massively parallel reporter assays and deep learning, we mapped enhancer-gene regulatory networks across mouse liver cell types. We found that zonation affects gene expression and chromatin accessibility in hepatocytes, among other cell types. These states are driven by the repressors TCF7L1 and TBX3, alongside other core hepatocyte transcription factors, such as HNF4A, CEBPA, FOXA1 and ONECUT1. To examine the architecture of the enhancers driving these cell states, we trained a hierarchical deep learning model called DeepLiver. Our study provides a multimodal understanding of the regulatory code underlying hepatocyte identity and their zonation state that can be used to engineer enhancers with specific activity levels and zonation patterns.


Subject(s)
Deep Learning , Multiomics , Mice , Animals , Gene Regulatory Networks , Liver/metabolism , Hepatocytes , Mammals
3.
Mol Cancer Ther ; 23(1): 3-13, 2024 Jan 03.
Article in English | MEDLINE | ID: mdl-37748190

ABSTRACT

The Hippo pathway and its downstream effectors, the YAP and TAZ transcriptional coactivators, are deregulated in multiple different types of human cancer and are required for cancer cell phenotypes in vitro and in vivo, while largely dispensable for tissue homeostasis in adult mice. YAP/TAZ and their main partner transcription factors, the TEAD1-4 factors, are therefore promising anticancer targets. Because of frequent YAP/TAZ hyperactivation caused by mutations in the Hippo pathway components NF2 and LATS2, mesothelioma is one of the prime cancer types predicted to be responsive to YAP/TAZ-TEAD inhibitor treatment. Mesothelioma is a devastating disease for which currently no effective treatment options exist. Here, we describe a novel covalent YAP/TAZ-TEAD inhibitor, SWTX-143, that binds to the palmitoylation pocket of all four TEAD isoforms. SWTX-143 caused irreversible and specific inhibition of the transcriptional activity of YAP/TAZ-TEAD in Hippo-mutant tumor cell lines. More importantly, YAP/TAZ-TEAD inhibitor treatment caused strong mesothelioma regression in subcutaneous xenograft models with human cells and in an orthotopic mesothelioma mouse model. Finally, SWTX-143 also selectively impaired the growth of NF2-mutant kidney cancer cell lines, suggesting that the sensitivity of mesothelioma models to these YAP/TAZ-TEAD inhibitors can be extended to other tumor types with aberrations in Hippo signaling. In brief, we describe a novel and specific YAP/TAZ-TEAD inhibitor that has potential to treat multiple Hippo-mutant solid tumor types.


Subject(s)
Mesothelioma, Malignant , Mesothelioma , Adult , Humans , Animals , Mice , Hippo Signaling Pathway , YAP-Signaling Proteins , Transcription Factors/genetics , Transcription Factors/metabolism , Mesothelioma/drug therapy , Mesothelioma/genetics , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Tumor Suppressor Proteins/metabolism
4.
Elife ; 122023 05 03.
Article in English | MEDLINE | ID: mdl-37133250

ABSTRACT

Wound response programs are often activated during neoplastic growth in tumors. In both wound repair and tumor growth, cells respond to acute stress and balance the activation of multiple programs, including apoptosis, proliferation, and cell migration. Central to those responses are the activation of the JNK/MAPK and JAK/STAT signaling pathways. Yet, to what extent these signaling cascades interact at the cis-regulatory level and how they orchestrate different regulatory and phenotypic responses is still unclear. Here, we aim to characterize the regulatory states that emerge and cooperate in the wound response, using the Drosophila melanogaster wing disc as a model system, and compare these with cancer cell states induced by rasV12scrib-/- in the eye disc. We used single-cell multiome profiling to derive enhancer gene regulatory networks (eGRNs) by integrating chromatin accessibility and gene expression signals. We identify a 'proliferative' eGRN, active in the majority of wounded cells and controlled by AP-1 and STAT. In a smaller, but distinct population of wound cells, a 'senescent' eGRN is activated and driven by C/EBP-like transcription factors (Irbp18, Xrp1, Slow border, and Vrille) and Scalloped. These two eGRN signatures are found to be active in tumor cells at both gene expression and chromatin accessibility levels. Our single-cell multiome and eGRNs resource offers an in-depth characterization of the senescence markers, together with a new perspective on the shared gene regulatory programs acting during wound response and oncogenesis.


Subject(s)
Drosophila Proteins , Neoplasms , Animals , Drosophila melanogaster/metabolism , Drosophila Proteins/metabolism , Gene Regulatory Networks , Transcription Factors/genetics , Transcription Factors/metabolism , Neoplasms/pathology , Chromatin/metabolism , DNA-Binding Proteins/metabolism
5.
Cell Death Dis ; 12(12): 1110, 2021 11 27.
Article in English | MEDLINE | ID: mdl-34839349

ABSTRACT

Activated hepatic stellate cells (aHSC) are the main source of extra cellular matrix in liver fibrosis. Activation is classically divided in two phases: initiation and perpetuation. Currently, HSC-based therapeutic candidates largely focus on targeting the aHSCs in the perpetuation phase. However, the importance of HSC initiation during chronic liver disease (CLD) remains unclear. Here, we identified transcriptional programs of initiating and activated HSCs by RNA sequencing, using in vitro and in vivo mouse models of fibrosis. Importantly, we show that both programs are active in HSCs during murine and human CLD. In human cirrhotic livers, scar associated mesenchymal cells employ both transcriptional programs at the single cell level. Our results indicate that the transcriptional programs that drive the initiation of HSCs are still active in humans suffering from CLD. We conclude that molecules involved in the initiation of HSC activation, or in the maintenance of aHSCs can be considered equally important in the search for druggable targets of chronic liver disease.


Subject(s)
Hepatic Stellate Cells/metabolism , Liver Diseases/physiopathology , Animals , Chronic Disease , Disease Models, Animal , Humans , Male , Mice
6.
Hepatology ; 74(3): 1445-1460, 2021 09.
Article in English | MEDLINE | ID: mdl-33768568

ABSTRACT

BACKGROUND AND AIMS: Earlier diagnosis and treatment of intrahepatic cholangiocarcinoma (iCCA) are necessary to improve therapy, yet limited information is available about initiation and evolution of iCCA precursor lesions. Therefore, there is a need to identify mechanisms driving formation of precancerous lesions and their progression toward invasive tumors using experimental models that faithfully recapitulate human tumorigenesis. APPROACH AND RESULTS: To this end, we generated a mouse model which combines cholangiocyte-specific expression of KrasG12D with 3,5-diethoxycarbonyl-1,4-dihydrocollidine (DDC) diet-induced inflammation to mimic iCCA development in patients with cholangitis. Histological and transcriptomic analyses of the mouse precursor lesions and iCCA were performed and compared with human analyses. The function of genes overexpressed during tumorigenesis was investigated in human cell lines. We found that mice expressing KrasG12D in cholangiocytes and fed a DDC diet developed cholangitis, ductular proliferations, intraductal papillary neoplasms of bile ducts (IPNBs), and, eventually, iCCAs. The histology of mouse and human IPNBs was similar, and mouse iCCAs displayed histological characteristics of human mucin-producing, large-duct-type iCCA. Signaling pathways activated in human iCCA were also activated in mice. The identification of transition zones between IPNB and iCCA on tissue sections, combined with RNA-sequencing analyses of the lesions supported that iCCAs derive from IPNBs. We further provide evidence that tensin-4 (TNS4), which is stimulated by KRASG12D and SRY-related HMG box transcription factor 17, promotes tumor progression. CONCLUSIONS: We developed a mouse model that faithfully recapitulates human iCCA tumorigenesis and identified a gene cascade which involves TNS4 and promotes tumor progression.


Subject(s)
Bile Duct Neoplasms/genetics , Carcinoma, Ductal/genetics , Cholangiocarcinoma/genetics , Disease Models, Animal , Liver Neoplasms, Experimental/genetics , Mice , Tensins/genetics , Animals , Bile Duct Neoplasms/chemically induced , Bile Duct Neoplasms/metabolism , Bile Duct Neoplasms/pathology , Carcinoma, Ductal/chemically induced , Carcinoma, Ductal/metabolism , Carcinoma, Ductal/pathology , Carcinoma, Papillary/chemically induced , Carcinoma, Papillary/genetics , Carcinoma, Papillary/metabolism , Carcinoma, Papillary/pathology , Cholangiocarcinoma/chemically induced , Cholangiocarcinoma/metabolism , Cholangiocarcinoma/pathology , Cholangitis/chemically induced , Cholangitis/complications , HMGB Proteins/genetics , HMGB Proteins/metabolism , Liver Neoplasms, Experimental/chemically induced , Liver Neoplasms, Experimental/metabolism , Liver Neoplasms, Experimental/pathology , Proto-Oncogene Proteins p21(ras)/genetics , Pyridines/toxicity , SOXF Transcription Factors/genetics , SOXF Transcription Factors/metabolism , Signal Transduction , Tensins/metabolism
7.
Gastroenterology ; 160(3): 847-862, 2021 02.
Article in English | MEDLINE | ID: mdl-33127392

ABSTRACT

BACKGROUND AND AIMS: The Hippo pathway and its downstream effectors YAP and TAZ (YAP/TAZ) are heralded as important regulators of organ growth and regeneration. However, different studies provided contradictory conclusions about their role during regeneration of different organs, ranging from promoting proliferation to inhibiting it. Here we resolve the function of YAP/TAZ during regeneration of the liver, where Hippo's role in growth control has been studied most intensely. METHODS: We evaluated liver regeneration after carbon tetrachloride toxic liver injury in mice with conditional deletion of Yap/Taz in hepatocytes and/or biliary epithelial cells, and measured the behavior of different cell types during regeneration by histology, RNA sequencing, and flow cytometry. RESULTS: We found that YAP/TAZ were activated in hepatocytes in response to carbon tetrachloride toxic injury. However, their targeted deletion in adult hepatocytes did not noticeably impair liver regeneration. In contrast, Yap/Taz deletion in adult bile ducts caused severe defects and delay in liver regeneration. Mechanistically, we showed that Yap/Taz mutant bile ducts degenerated, causing cholestasis, which stalled the recruitment of phagocytic macrophages and the removal of cellular corpses from injury sites. Elevated bile acids activated pregnane X receptor, which was sufficient to recapitulate the phenotype observed in mutant mice. CONCLUSIONS: Our data show that YAP/TAZ are practically dispensable in hepatocytes for liver development and regeneration. Rather, YAP/TAZ play an indirect role in liver regeneration by preserving bile duct integrity and securing immune cell recruitment and function.


Subject(s)
Adaptor Proteins, Signal Transducing/deficiency , Chemical and Drug Induced Liver Injury/pathology , Cholestasis/pathology , Liver Regeneration/genetics , Adaptor Proteins, Signal Transducing/genetics , Animals , Bile Ducts/pathology , Carbon Tetrachloride/administration & dosage , Carbon Tetrachloride/toxicity , Cell Proliferation/genetics , Chemical and Drug Induced Liver Injury/complications , Cholestasis/etiology , Disease Models, Animal , Hepatocytes/drug effects , Hepatocytes/pathology , Hippo Signaling Pathway , Humans , Liver/drug effects , Liver/pathology , Mice , Mice, Knockout , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , YAP-Signaling Proteins
8.
Science ; 366(6468): 1029-1034, 2019 11 22.
Article in English | MEDLINE | ID: mdl-31754005

ABSTRACT

The Hippo signaling pathway and its two downstream effectors, the YAP and TAZ transcriptional coactivators, are drivers of tumor growth in experimental models. Studying mouse models, we show that YAP and TAZ can also exert a tumor-suppressive function. We found that normal hepatocytes surrounding liver tumors displayed activation of YAP and TAZ and that deletion of Yap and Taz in these peritumoral hepatocytes accelerated tumor growth. Conversely, experimental hyperactivation of YAP in peritumoral hepatocytes triggered regression of primary liver tumors and melanoma-derived liver metastases. Furthermore, whereas tumor cells growing in wild-type livers required YAP and TAZ for their survival, those surrounded by Yap- and Taz-deficient hepatocytes were not dependent on YAP and TAZ. Tumor cell survival thus depends on the relative activity of YAP and TAZ in tumor cells and their surrounding tissue, suggesting that YAP and TAZ act through a mechanism of cell competition to eliminate tumor cells.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cell Cycle Proteins/metabolism , Cholangiocarcinoma/metabolism , Hepatocytes/metabolism , Liver Neoplasms, Experimental/metabolism , Trans-Activators/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Cycle Proteins/genetics , Cell Line, Tumor , Cell Survival , Cholangiocarcinoma/pathology , Hippo Signaling Pathway , Humans , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Liver Neoplasms/secondary , Liver Neoplasms, Experimental/pathology , Melanoma/metabolism , Melanoma/secondary , Mice, Inbred C57BL , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Trans-Activators/economics , Trans-Activators/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Transcriptional Coactivator with PDZ-Binding Motif Proteins , Tumor Burden , YAP-Signaling Proteins
9.
Cells ; 8(4)2019 04 25.
Article in English | MEDLINE | ID: mdl-31027317

ABSTRACT

Inducible cyclization recombinase (Cre) transgenic mouse strains are powerful tools for cell lineage tracing and tissue-specific knockout experiments. However, low efficiency or leaky expression can be important pitfalls. Here, we compared the efficiency and specificity of two commonly used cholangiocyte-specific Cre drivers, the Opn-iCreERT2 and Ck19-CreERT drivers, using a tdTomato reporter strain. We found that Opn-iCreERT2 triggered recombination of the tdTomato reporter in 99.9% of all cholangiocytes while Ck19-CreERT only had 32% recombination efficiency after tamoxifen injection. In the absence of tamoxifen, recombination was also induced in 2% of cholangiocytes for the Opn-iCreERT2 driver and in 13% for the Ck19-CreERT driver. For both drivers, Cre recombination was highly specific for cholangiocytes since recombination was rare in other liver cell types. Toxic liver injury ectopically activated Opn-iCreERT2 but not Ck19-CreERT expression in hepatocytes. However, ectopic recombination in hepatocytes could be avoided by applying a three-day long wash-out period between tamoxifen treatment and toxin injection. Therefore, the Opn-iCreERT2 driver is best suited for the generation of mutant bile ducts, while the Ck19-CreERT driver has near absolute specificity for bile duct cells and is therefore favorable for lineage tracing experiments.


Subject(s)
Genetic Engineering/methods , Keratin-19/metabolism , Osteopontin/metabolism , Animals , Animals, Genetically Modified/genetics , Animals, Genetically Modified/metabolism , Bile Ducts/metabolism , Cell Lineage/drug effects , Female , Gene Expression/genetics , Gene Expression/physiology , Integrases/biosynthesis , Integrases/genetics , Integrases/metabolism , Keratin-19/genetics , Keratin-19/physiology , Liver/metabolism , Male , Mice , Mice, Transgenic/genetics , Osteopontin/genetics , Osteopontin/physiology , Recombinant Proteins/metabolism , Tamoxifen/pharmacology
10.
Int J Mol Sci ; 20(3)2019 Feb 01.
Article in English | MEDLINE | ID: mdl-30717258

ABSTRACT

Primary liver cancer comprises a diverse group of liver tumors. The heterogeneity of these tumors is seen as one of the obstacles to finding an effective therapy. The Hippo pathway, with its downstream transcriptional co-activator Yes-associated protein (YAP) and transcriptional co-activator with PDZ-binding motif (TAZ), has a decisive role in the carcinogenesis of primary liver cancer. Therefore, we examined the expression pattern of YAP and TAZ in 141 patients with hepatocellular carcinoma keratin 19 positive (HCC K19⁺), hepatocellular carcinoma keratin 19 negative (HCC K19-), combined hepatocellular⁻cholangiocarcinoma carcinoma (cHCC-CCA), or cholangiocarcinoma (CCA). All cHCC-CCA and CCA patients showed high expression levels for YAP and TAZ, while only some patients of the HCC group were positive. Notably, we found that a histoscore of both markers is useful in the challenging diagnosis of cHCC-CCA. In addition, positivity for YAP and TAZ was observed in the hepatocellular and cholangiocellular components of cHCC-CCA, which suggests a single cell origin in cHCC-CCA. Within the K19- HCC group, our results demonstrate that the expression of YAP is a statistically significant predictor of poor prognosis when observed in the cytoplasm. Nuclear expression of TAZ is an even more specific and independent predictor of poor disease-free survival and overall survival of K19- HCC patients. Our results thus identify different levels of YAP/TAZ expression in various liver cancers that can be used for diagnostics.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Bile Duct Neoplasms/genetics , Biomarkers, Tumor/genetics , Carcinoma, Hepatocellular/genetics , Cholangiocarcinoma/genetics , Gene Expression Regulation, Neoplastic , Intracellular Signaling Peptides and Proteins/genetics , Liver Neoplasms/genetics , Phosphoproteins/genetics , Adaptor Proteins, Signal Transducing/metabolism , Aged , Bile Duct Neoplasms/diagnosis , Bile Duct Neoplasms/mortality , Bile Duct Neoplasms/pathology , Biomarkers, Tumor/metabolism , Carcinogenesis/genetics , Carcinogenesis/metabolism , Carcinogenesis/pathology , Carcinoma, Hepatocellular/diagnosis , Carcinoma, Hepatocellular/mortality , Carcinoma, Hepatocellular/pathology , Cell Nucleus/metabolism , Cell Nucleus/pathology , Cholangiocarcinoma/diagnosis , Cholangiocarcinoma/mortality , Cholangiocarcinoma/pathology , Cytosol/metabolism , Cytosol/pathology , Female , Genetic Heterogeneity , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Kaplan-Meier Estimate , Keratin-19/deficiency , Keratin-19/genetics , Ki-67 Antigen/genetics , Ki-67 Antigen/metabolism , Liver Neoplasms/diagnosis , Liver Neoplasms/mortality , Liver Neoplasms/pathology , Male , Middle Aged , Phosphoproteins/metabolism , Prognosis , Proportional Hazards Models , Retrospective Studies , Signal Transduction , Trans-Activators , Transcription Factors , Transcriptional Coactivator with PDZ-Binding Motif Proteins , YAP-Signaling Proteins
11.
Nat Rev Mol Cell Biol ; 20(4): 211-226, 2019 04.
Article in English | MEDLINE | ID: mdl-30546055

ABSTRACT

The Hippo pathway and its downstream effectors, the transcriptional co-activators Yes-associated protein (YAP) and transcriptional co-activator with PDZ-binding motif (TAZ), regulate organ growth and cell plasticity during animal development and regeneration. Remarkably, experimental activation of YAP/TAZ in the mouse can promote regeneration in organs with poor or compromised regenerative capacity, such as the adult heart and the liver and intestine of old or diseased mice. However, therapeutic YAP/TAZ activation may cause serious side effects. Most notably, YAP/TAZ are hyperactivated in human cancers, and prolonged activation of YAP/TAZ triggers cancer development in mice. Thus, can the power of YAP/TAZ to promote regeneration be harnessed in a safe way? Here, we review the role of Hippo signalling in animal regeneration, examine the promises and risks of YAP/TAZ activation for regenerative medicine and discuss strategies to activate YAP/TAZ for regenerative therapy while minimizing adverse side effects.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Protein Serine-Threonine Kinases/metabolism , Signal Transduction/physiology , Transcription Factors/metabolism , Animals , Humans , Regenerative Medicine/methods
12.
Proc Natl Acad Sci U S A ; 115(42): 10684-10689, 2018 10 16.
Article in English | MEDLINE | ID: mdl-30257938

ABSTRACT

The Hippo tumor-suppressor pathway regulates organ growth, cell proliferation, and stem cell biology. Defects in Hippo signaling and hyperactivation of its downstream effectors-Yorkie (Yki) in Drosophila and YAP/TAZ in mammals-result in progenitor cell expansion and overgrowth of multiple organs and contribute to cancer development. Deciphering the mechanisms that regulate the activity of the Hippo pathway is key to understanding its function and for therapeutic targeting. However, although the Hippo kinase cascade and several other upstream inputs have been identified, the mechanisms that regulate Yki/YAP/TAZ activity are still incompletely understood. To identify new regulators of Yki activity, we screened in Drosophila for suppressors of tissue overgrowth and Yki activation caused by overexpression of atypical protein kinase C (aPKC), a member of the apical cell polarity complex. In this screen, we identified mutations in the heterogeneous nuclear ribonucleoprotein Hrb27C that strongly suppressed the tissue defects induced by ectopic expression of aPKC. Hrb27C was required for aPKC-induced tissue growth and Yki target gene expression but did not affect general gene expression. Genetic and biochemical experiments showed that Hrb27C affects Yki phosphorylation. Other RNA-binding proteins known to interact with Hrb27C for mRNA transport in oocytes were also required for normal Yki activity, although they suppressed Yki output. Based on the known functions of Hrb27C, we conclude that Hrb27C-mediated control of mRNA splicing, localization, or translation is essential for coordinated activity of the Hippo pathway.


Subject(s)
Drosophila Proteins/metabolism , Drosophila melanogaster/growth & development , Gene Expression Regulation, Developmental , Nuclear Proteins/metabolism , RNA-Binding Proteins/metabolism , Trans-Activators/metabolism , Animals , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Heterogeneous-Nuclear Ribonucleoprotein Group F-H/genetics , Heterogeneous-Nuclear Ribonucleoprotein Group F-H/metabolism , Heterogeneous-Nuclear Ribonucleoproteins/genetics , Heterogeneous-Nuclear Ribonucleoproteins/metabolism , Nuclear Proteins/genetics , Poly(A)-Binding Protein II/genetics , Poly(A)-Binding Protein II/metabolism , RNA-Binding Proteins/genetics , Signal Transduction , Trans-Activators/genetics , YAP-Signaling Proteins
13.
J Hematol Oncol ; 11(1): 108, 2018 08 25.
Article in English | MEDLINE | ID: mdl-30144809

ABSTRACT

BACKGROUND: The CNOT3 protein is a subunit of the CCR4-NOT complex, which is involved in mRNA degradation. We recently identified CNOT3 loss-of-function mutations in patients with T-cell acute lymphoblastic leukemia (T-ALL). METHODS: Here, we use different Drosophila melanogaster eye cancer models to study the potential tumor suppressor function of Not3, the CNOT3 orthologue, and other members of the CCR4-NOT complex. RESULTS: Our data show that knockdown of Not3, the structural components Not1/Not2, and the deadenylases twin/Pop2 all result in increased tumor formation. In addition, overexpression of Not3 could reduce tumor formation. Not3 downregulation has a mild but broad effect on gene expression and leads to increased levels of genes involved in DNA replication and ribosome biogenesis. CycB upregulation also contributes to the Not3 tumor phenotype. Similar findings were obtained in human T-ALL cell lines, pointing out the conserved function of Not3. CONCLUSIONS: Together, our data establish a critical role for Not3 and the entire CCR4-NOT complex as tumor suppressor.


Subject(s)
Drosophila Proteins/metabolism , Drosophila melanogaster/pathogenicity , Eye Neoplasms/genetics , Genes, Tumor Suppressor/physiology , RNA-Binding Proteins/metabolism , Ribonucleases/metabolism , Animals , Disease Models, Animal , Eye Neoplasms/metabolism , Humans , Protein Binding
14.
Nat Genet ; 50(7): 1011-1020, 2018 07.
Article in English | MEDLINE | ID: mdl-29867222

ABSTRACT

Transcriptional enhancers function as docking platforms for combinations of transcription factors (TFs) to control gene expression. How enhancer sequences determine nucleosome occupancy, TF recruitment and transcriptional activation in vivo remains unclear. Using ATAC-seq across a panel of Drosophila inbred strains, we found that SNPs affecting binding sites of the TF Grainy head (Grh) causally determine the accessibility of epithelial enhancers. We show that deletion and ectopic expression of Grh cause loss and gain of DNA accessibility, respectively. However, although Grh binding is necessary for enhancer accessibility, it is insufficient to activate enhancers. Finally, we show that human Grh homologs-GRHL1, GRHL2 and GRHL3-function similarly. We conclude that Grh binding is necessary and sufficient for the opening of epithelial enhancers but not for their activation. Our data support a model positing that complex spatiotemporal expression patterns are controlled by regulatory hierarchies in which pioneer factors, such as Grh, establish tissue-specific accessible chromatin landscapes upon which other factors can act.


Subject(s)
DNA-Binding Proteins/genetics , Drosophila Proteins/genetics , Nucleosomes/genetics , Transcription Factors/genetics , Animals , Animals, Genetically Modified , Binding Sites , Cell Line, Tumor , Chromatin/genetics , Drosophila melanogaster/genetics , Enhancer Elements, Genetic , Epithelial Cells , Gene Expression Regulation, Developmental , Humans , MCF-7 Cells , Polymorphism, Single Nucleotide , Transcriptional Activation
15.
Article in English | MEDLINE | ID: mdl-28600393

ABSTRACT

The Hippo signal transduction pathway is an important regulator of organ growth and cell differentiation, and its deregulation contributes to the development of cancer. The activity of the Hippo pathway is strongly dependent on cell junctions, cellular architecture, and the mechanical properties of the microenvironment. In this review, we discuss recent advances in our understanding of how cell junctions transduce signals from the microenvironment and control the activity of the Hippo pathway. We also discuss how these mechanisms may control organ growth during development and regeneration, and how defects in them deregulate Hippo signaling in cancer cells.


Subject(s)
Intercellular Junctions , Models, Biological , Protein Serine-Threonine Kinases/physiology , Adherens Junctions/metabolism , Adherens Junctions/physiology , Animals , Cell Differentiation , Cell Proliferation , Cellular Microenvironment , Cytoskeleton/metabolism , Cytoskeleton/ultrastructure , Hippo Signaling Pathway , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Tight Junctions/metabolism , Tight Junctions/physiology
16.
Dev Cell ; 42(5): 462-478.e7, 2017 09 11.
Article in English | MEDLINE | ID: mdl-28867486

ABSTRACT

Vascular endothelial growth factor (VEGF) is a major driver of blood vessel formation. However, the signal transduction pathways culminating in the biological consequences of VEGF signaling are only partially understood. Here, we show that the Hippo pathway effectors YAP and TAZ work as crucial signal transducers to mediate VEGF-VEGFR2 signaling during angiogenesis. We demonstrate that YAP/TAZ are essential for vascular development as endothelium-specific deletion of YAP/TAZ leads to impaired vascularization and embryonic lethality. Mechanistically, we show that VEGF activates YAP/TAZ via its effects on actin cytoskeleton and that activated YAP/TAZ induce a transcriptional program to further control cytoskeleton dynamics and thus establish a feedforward loop that ensures a proper angiogenic response. Lack of YAP/TAZ also results in altered cellular distribution of VEGFR2 due to trafficking defects from the Golgi apparatus to the plasma membrane. Altogether, our study identifies YAP/TAZ as central mediators of VEGF signaling and therefore as important regulators of angiogenesis.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Neovascularization, Physiologic , Phosphoproteins/metabolism , Signal Transduction , Vascular Endothelial Growth Factor A/metabolism , Actin Cytoskeleton/genetics , Animals , Animals, Newborn , Brain/pathology , Cell Cycle Proteins , Cell Line, Tumor , Cell Membrane/metabolism , Cell Movement/genetics , Cell Nucleus/metabolism , Chromatin Immunoprecipitation , Embryonic Development/genetics , Endothelial Cells/metabolism , Gene Deletion , Gene Knockout Techniques , Gene Silencing , Golgi Apparatus/metabolism , Mice , Models, Biological , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Neovascularization, Physiologic/genetics , Signal Transduction/genetics , Trans-Activators , Transcription, Genetic , Vascular Endothelial Growth Factor Receptor-2/metabolism , YAP-Signaling Proteins
17.
Dev Cell ; 42(6): 667-680.e4, 2017 09 25.
Article in English | MEDLINE | ID: mdl-28950103

ABSTRACT

Hyperactivating mutations in Ras signaling are hallmarks of carcinomas. Ras signaling mediates cell fate decisions as well as proliferation during development. It is not known what dictates whether Ras signaling drives differentiation versus proliferation. Here we show that the Hippo pathway is critical for this decision. Loss of Hippo switches Ras activation from promoting cellular differentiation to aggressive cellular proliferation. Transcriptome analysis combined with genetic tests show that this excessive proliferation depends on the synergistic induction of Ras target genes. Using ChIP-nexus, we find that Hippo signaling keeps Ras targets in check by directly regulating the expression of two key downstream transcription factors of Ras signaling: the ETS-domain transcription factor Pointed and the repressor Capicua. Our results highlight how independent signaling pathways can impinge on each other at the level of transcription factors, thereby providing a safety mechanism to keep proliferation in check under normal developmental conditions.


Subject(s)
Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Signal Transduction , Transcription, Genetic , ras Proteins/metabolism , Animals , Cell Differentiation/genetics , Cell Proliferation/genetics , Drosophila melanogaster/cytology , Drosophila melanogaster/metabolism , ErbB Receptors/metabolism , Gene Expression Regulation, Developmental , Genes, Insect , Models, Biological , Mutation/genetics , Pupa/metabolism , Regulon/genetics , Sequence Analysis, RNA , Transcription Factors/metabolism
18.
Curr Opin Cell Biol ; 43: 62-68, 2016 12.
Article in English | MEDLINE | ID: mdl-27592171

ABSTRACT

We have a limited ability to stimulate cells in damaged tissues to regenerate properly patterned and functional organs. Excitingly, however, recent work shows that experimental modulation of the Hippo pathway can promote the regeneration of several organs in mice. The Hippo pathway plays pivotal and specific roles in organ growth, cellular plasticity, and stem cell biology, which are all important for regeneration. In this review we survey and compare the effects of experimental manipulation of Hippo signaling in mouse on the development, homeostasis, and regeneration of the heart, liver, intestine, and other organs. We also discuss the potential of targeting the Hippo pathway as a therapeutic approach for regenerative medicine.


Subject(s)
Cellular Reprogramming , Organ Specificity , Protein Serine-Threonine Kinases/metabolism , Regeneration , Signal Transduction , Animals , Humans , Neoplasms/pathology , Stem Cells/metabolism
19.
Curr Biol ; 26(16): 2101-13, 2016 08 22.
Article in English | MEDLINE | ID: mdl-27476594

ABSTRACT

Cancer cells have abnormal gene expression profiles; however, to what degree these are chaotic or driven by structured gene regulatory networks is often not known. Here we studied a model of Ras-driven invasive tumorigenesis in Drosophila epithelial tissues and combined in vivo genetics with next-generation sequencing and computational modeling to decipher the regulatory logic of tumor cells. Surprisingly, we discovered that the bulk of the tumor-specific gene expression is controlled by an ectopic network of a few transcription factors that are overexpressed and/or hyperactivated in tumor cells. These factors are Stat, AP-1, the bHLH proteins Myc and AP-4, the nuclear hormone receptor Ftz-f1, the nuclear receptor coactivator Taiman/SRC3, and Mef2. Notably, many of these transcription factors also are hyperactivated in human tumors. Bioinformatic analysis predicted that these factors directly regulate the majority of the tumor-specific gene expression, that they are interconnected by extensive cross-regulation, and that they show a high degree of co-regulation of target genes. Indeed, the factors of this network were required in multiple epithelia for tumor growth and invasiveness, and knockdown of several factors caused a reversion of the tumor-specific expression profile but had no observable effect on normal tissues. We further found that the Hippo pathway effector Yorkie was strongly activated in tumor cells and initiated cellular reprogramming by activating several transcription factors of this network. Thus, modeling regulatory networks identified an ectopic and ordered network of master regulators that control a large part of tumor cell-specific gene expression.


Subject(s)
Carcinogenesis , Drosophila Proteins/genetics , Drosophila melanogaster/physiology , Gene Expression Regulation, Neoplastic , Transcription Factors/genetics , Animals , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Gene Regulatory Networks , Signal Transduction , Transcription Factors/metabolism , Tumor Cells, Cultured
20.
Nat Commun ; 6: 8357, 2015 Oct 05.
Article in English | MEDLINE | ID: mdl-26437443

ABSTRACT

The Hippo pathway plays a central role in tissue homoeostasis, and its dysregulation contributes to tumorigenesis. Core components of the Hippo pathway include a kinase cascade of MST1/2 and LATS1/2 and the transcription co-activators YAP/TAZ. In response to stimulation, LATS1/2 phosphorylate and inhibit YAP/TAZ, the main effectors of the Hippo pathway. Accumulating evidence suggests that MST1/2 are not required for the regulation of YAP/TAZ. Here we show that deletion of LATS1/2 but not MST1/2 abolishes YAP/TAZ phosphorylation. We have identified MAP4K family members--Drosophila Happyhour homologues MAP4K1/2/3 and Misshapen homologues MAP4K4/6/7-as direct LATS1/2-activating kinases. Combined deletion of MAP4Ks and MST1/2, but neither alone, suppresses phosphorylation of LATS1/2 and YAP/TAZ in response to a wide range of signals. Our results demonstrate that MAP4Ks act in parallel to and are partially redundant with MST1/2 in the regulation of LATS1/2 and YAP/TAZ, and establish MAP4Ks as components of the expanded Hippo pathway.


Subject(s)
Nuclear Proteins/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Transcription Factors/metabolism , Tumor Suppressor Proteins/genetics , Acyltransferases , Animals , Blotting, Western , Carcinogenesis/genetics , Cell Cycle Proteins , Cell Line, Tumor , Drosophila , Drosophila Proteins , Fluorescent Antibody Technique , Germinal Center Kinases , HEK293 Cells , Hepatocyte Growth Factor/genetics , Hepatocyte Growth Factor/metabolism , Hippo Signaling Pathway , Homeostasis/genetics , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Phosphorylation , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Serine-Threonine Kinase 3 , Signal Transduction , Tumor Suppressor Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...