Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Life Sci Alliance ; 6(9)2023 09.
Article in English | MEDLINE | ID: mdl-37385752

ABSTRACT

The Hippo signalling pathway is a master regulator of cell growth, proliferation, and cancer. The transcriptional coregulators of the Hippo pathway, YAP and TAZ, are central in various cancers. However, how YAP and TAZ get activated in most types of cancers is not well understood. Here, we show that androgens activate YAP/TAZ via the androgen receptor (AR) in prostate cancer (PCa), and that this activation is differential. AR regulates YAP translation while inducing transcription of the TAZ encoding gene, WWTR1 Furthermore, we show that AR-mediated YAP/TAZ activation is regulated by the RhoA GTPases transcriptional mediator, serum response factor (SRF). Importantly, in prostate cancer patients, SRF expression positively correlates with TAZ and the YAP/TAZ target genes CYR61 and CTGF We demonstrate that YAP/TAZ are not essential for sustaining AR activity, however, targeting YAP/TAZ or SRF sensitize PCa cells to AR inhibition in anchorage-independent growth conditions. Our findings dissect the cellular roles of YAP, TAZ, and SRF in prostate cancer cells. Our data emphasize the interplay between these transcriptional regulators and their roles in prostate tumorigenesis and highlight how these insights might be exploited therapeutically.


Subject(s)
Prostatic Neoplasms , Receptors, Androgen , Humans , Male , Androgens , Carcinogenesis , Prostate , Prostatic Neoplasms/genetics , Receptors, Androgen/genetics , Transcriptional Coactivator with PDZ-Binding Motif Proteins/metabolism
2.
J Microsc ; 2023 May 08.
Article in English | MEDLINE | ID: mdl-37156549

ABSTRACT

Standing wave (SW) microscopy is a method that uses an interference pattern to excite fluorescence from labelled cellular structures and produces high-resolution images of three-dimensional objects in a two-dimensional dataset. SW microscopy is performed with high-magnification, high-numerical aperture objective lenses, and while this results in high-resolution images, the field of view is very small. Here we report upscaling of this interference imaging method from the microscale to the mesoscale using the Mesolens, which has the unusual combination of a low-magnification and high-numerical aperture. With this method, we produce SW images within a field of view of 4.4 mm × 3.0 mm that can readily accommodate over 16,000 cells in a single dataset. We demonstrate the method using both single-wavelength excitation and the multi-wavelength SW method TartanSW. We show application of the method for imaging of fixed and living cells specimens, with the first application of SW imaging to study cells under flow conditions.

3.
Clin Transl Med ; 13(2): e1190, 2023 02.
Article in English | MEDLINE | ID: mdl-36740402

ABSTRACT

The Hippo signalling pathway is dysregulated across a wide range of cancer types and, although driver mutations that directly affect the core Hippo components are rare, a handful is found within pleural mesothelioma (PM). PM is a deadly disease of the lining of the lung caused by asbestos exposure. By pooling the largest-scale clinical datasets publicly available, we here interrogate associations between the most prevalent driver mutations within PM and Hippo pathway disruption in patients, while assessing correlations with a variety of clinical markers. This analysis reveals a consistent worse outcome in patients exhibiting transcriptional markers of YAP/TAZ activation, pointing to the potential of leveraging Hippo pathway transcriptional activation status as a metric by which patients may be meaningfully stratified. Preclinical models recapitulating disease are transformative in order to develop new therapeutic strategies. We here establish an isogenic cell-line model of PM, which represents the most frequently mutated genes and which faithfully recapitulates the molecular features of clinical PM. This preclinical model is developed to probe the molecular basis by which the Hippo pathway and key driver mutations affect cancer initiation and progression. Implementing this approach, we reveal the role of NF2 as a mechanosensory component of the Hippo pathway in mesothelial cells. Cellular NF2 loss upon physiological stiffnesses analogous to the tumour niche drive YAP/TAZ-dependent anchorage-independent growth. Consequently, the development and characterisation of this cellular model provide a unique resource to obtain molecular insights into the disease and progress new drug discovery programs together with future stratification of PM patients.


Subject(s)
Mesothelioma , Transcription Factors , Humans , Hippo Signaling Pathway , Mesothelioma/genetics , Mesothelioma/metabolism , Mesothelioma/pathology , Mutation/genetics , Protein Serine-Threonine Kinases/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Transcriptional Coactivator with PDZ-Binding Motif Proteins , YAP-Signaling Proteins
4.
Clin Sci (Lond) ; 136(3): 197-222, 2022 02 11.
Article in English | MEDLINE | ID: mdl-35119068

ABSTRACT

Tumorigenesis is a highly complex process, involving many interrelated and cross-acting signalling pathways. One such pathway that has garnered much attention in the field of cancer research over the last decade is the Hippo signalling pathway. Consisting of two antagonistic modules, the pathway plays an integral role in both tumour suppressive and oncogenic processes, generally via regulation of a diverse set of genes involved in a range of biological functions. This review discusses the history of the pathway within the context of cancer and explores some of the most recent discoveries as to how this critical transducer of cellular signalling can influence cancer progression. A special focus is on the various recent efforts to therapeutically target the key effectors of the pathway in both preclinical and clinical settings.


Subject(s)
Hippo Signaling Pathway , Neoplasms/drug therapy , Animals , Carcinogenesis , Humans , Neoplasms/metabolism , Trans-Activators , Transcription Factors , Transcriptional Coactivator with PDZ-Binding Motif Proteins , YAP-Signaling Proteins
5.
J Cell Sci ; 135(3)2022 02 01.
Article in English | MEDLINE | ID: mdl-35022745

ABSTRACT

Immunofluorescence microscopy is routinely used to visualise the spatial distribution of proteins that dictates their cellular function. However, unspecific antibody binding often results in high cytosolic background signals, decreasing the image contrast of a target structure. Recently, convolutional neural networks (CNNs) were successfully employed for image restoration in immunofluorescence microscopy, but current methods cannot correct for those background signals. We report a new method that trains a CNN to reduce unspecific signals in immunofluorescence images; we name this method label2label (L2L). In L2L, a CNN is trained with image pairs of two non-identical labels that target the same cellular structure. We show that after L2L training a network predicts images with significantly increased contrast of a target structure, which is further improved after implementing a multiscale structural similarity loss function. Here, our results suggest that sample differences in the training data decrease hallucination effects that are observed with other methods. We further assess the performance of a cycle generative adversarial network, and show that a CNN can be trained to separate structures in superposed immunofluorescence images of two targets.


Subject(s)
Image Processing, Computer-Assisted , Neural Networks, Computer , Cellular Structures , Image Processing, Computer-Assisted/methods , Microscopy, Fluorescence
6.
NPJ Regen Med ; 7(1): 9, 2022 Jan 27.
Article in English | MEDLINE | ID: mdl-35087046

ABSTRACT

The extent of tissue regeneration varies widely between species. Mammals have a limited regenerative capacity whilst lower vertebrates such as the zebrafish (Danio rerio), a freshwater teleost, can robustly regenerate a range of tissues, including the spinal cord, heart, and fin. The molecular and cellular basis of this altered response is one of intense investigation. In this review, we summarise the current understanding of the association between zebrafish regeneration and Hippo pathway function, a phosphorylation cascade that regulates cell proliferation, mechanotransduction, stem cell fate, and tumorigenesis, amongst others. We also compare this function to Hippo pathway activity in the regenerative response of other species. We find that the Hippo pathway effectors Yap/Taz facilitate zebrafish regeneration and that this appears to be latent in mammals, suggesting that therapeutically promoting precise and temporal YAP/TAZ signalling in humans may enhance regeneration and hence reduce morbidity.

7.
Bioinform Adv ; 2(1): vbac008, 2022.
Article in English | MEDLINE | ID: mdl-36699391

ABSTRACT

Motivation: Disrupted PERCC1 gene expression causes an intractable congenital diarrhoea in infants. However, this gene's molecular mechanism is unknown and no homologous proteins have been reported. Results: Our detailed evolutionary analysis of PERCC1 sequence reveals it to be a previously unappreciated member of the YAP/TAZ/FAM181 family of homologous transcriptional regulators. Like YAP and TAZ, PERCC1 likely interacts with DNA via binding to TEA/ATTS domain transcription factors (TEADs) using its conserved interface-2 and -3 sequences. We compare the expression patterns of PERCC1 with those of YAP, TAZ, TEADs. Our report provides the identification and first in-depth bioinformatic analysis of a YAP/TAZ homologue, and a likely new regulator of the YAP/TAZ-TEAD transcriptional complex. Availability and implementation: The data underlying this article are available in UniProt Database. Supplementary information: Supplementary data are available at Bioinformatics Advances online.

8.
Biochem Soc Trans ; 49(4): 1515-1527, 2021 08 27.
Article in English | MEDLINE | ID: mdl-34374419

ABSTRACT

The Hippo pathway is a dynamic cellular signalling nexus that regulates differentiation and controls cell proliferation and death. If the Hippo pathway is not precisely regulated, the functionality of the upstream kinase module is impaired, which increases nuclear localisation and activity of the central effectors, the transcriptional co-regulators YAP and TAZ. Pathological YAP and TAZ hyperactivity consequently cause cancer, fibrosis and developmental defects. The Hippo pathway controls an array of fundamental cellular processes, including adhesion, migration, mitosis, polarity and secretion of a range of biologically active components. Recent studies highlight that spatio-temporal regulation of Hippo pathway components are central to precisely controlling its context-dependent dynamic activity. Several levels of feedback are integrated into the Hippo pathway, which is further synergized with interactors outside of the pathway that directly regulate specific Hippo pathway components. Likewise, Hippo core kinases also 'moonlight' by phosphorylating multiple substrates beyond the Hippo pathway and thereby integrates further flexibility and robustness in the cellular decision-making process. This topic is still in its infancy but promises to reveal new fundamental insights into the cellular regulation of this therapeutically important pathway. We here highlight recent advances emphasising feedback dynamics and multilevel regulation of the Hippo pathway with a focus on mitosis and cell migration, as well as discuss potential productive future research avenues that might reveal novel insights into the overall dynamics of the pathway.


Subject(s)
Feedback , Hippo Signaling Pathway , Cell Movement , Humans , Male , Mitosis
9.
Cells ; 8(10)2019 09 30.
Article in English | MEDLINE | ID: mdl-31575050

ABSTRACT

The Hippo pathway is a cellular signalling network, which plays major roles in organ homeostasis and development[...].


Subject(s)
Protein Serine-Threonine Kinases/metabolism , Animals , Hippo Signaling Pathway , Homeostasis , Humans , Signal Transduction
10.
Cell Res ; 25(12): 1299-313, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26611634

ABSTRACT

YAP and TAZ are transcriptional co-activators and function as the major effectors of the Hippo tumor suppressor pathway, which controls cell growth, tissue homeostasis, and organ size. Here we show that YAP/TAZ play an essential role in amino acid-induced mTORC1 activation, particularly under nutrient-limiting conditions. Mechanistically, YAP/TAZ act via the TEAD transcription factors to induce expression of the high-affinity leucine transporter LAT1, which is a heterodimeric complex of SLC7A5 and SLC3A2. Deletion of YAP/TAZ abolishes expression of LAT1 and reduces leucine uptake. Re-expression of SLC7A5 in YAP/TAZ knockout cells restores leucine uptake and mTORC1 activation. Moreover, SLC7A5 knockout cells phenocopies YAP/TAZ knockout cells which exhibit defective mTORC1 activation in response to amino acids. We further demonstrate that YAP/TAZ act through SLC7A5 to provide cells with a competitive growth advantage. Our study provides molecular insight into the mechanism of YAP/TAZ target genes in cell growth regulation.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Amino Acids/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Multiprotein Complexes/metabolism , Phosphoproteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Adaptor Proteins, Signal Transducing/genetics , Cell Proliferation , Cells, Cultured , HEK293 Cells , Hippo Signaling Pathway , Humans , Intracellular Signaling Peptides and Proteins/genetics , Mechanistic Target of Rapamycin Complex 1 , Phosphoproteins/genetics , Trans-Activators , Transcription Factors , Transcriptional Coactivator with PDZ-Binding Motif Proteins , YAP-Signaling Proteins
11.
Trends Cell Biol ; 25(9): 499-513, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26045258

ABSTRACT

The Hippo pathway is a potent regulator of cellular proliferation, differentiation, and tissue homeostasis. Here we review the regulatory mechanisms of the Hippo pathway and discuss the function of Yes-associated protein (YAP)/transcriptional coactivator with a PDZ-binding domain (TAZ), the prime mediators of the Hippo pathway, in stem cell biology and tissue regeneration. We highlight their activities in both the nucleus and the cytoplasm and discuss their role as a signaling nexus and integrator of several other prominent signaling pathways such as the Wnt, G protein-coupled receptor (GPCR), epidermal growth factor (EGF), bone morphogenetic protein (BMP)/transforming growth factor beta (TGFß), and Notch pathways.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Phosphoproteins/physiology , Protein Serine-Threonine Kinases/physiology , Transcription Factors/physiology , Acyltransferases , Animals , Hippo Signaling Pathway , Humans , Intestinal Mucosa/metabolism , Signal Transduction , Stress, Physiological , Transcriptional Activation , YAP-Signaling Proteins
12.
Nat Commun ; 6: 6867, 2015 Apr 21.
Article in English | MEDLINE | ID: mdl-25897946

ABSTRACT

Caveolae have long been implicated in endocytosis. Recent data question this link, and in the absence of specific cargoes the potential cellular function of caveolar endocytosis remains unclear. Here we develop new tools, including doubly genome-edited cell lines, to assay the subcellular dynamics of caveolae using tagged proteins expressed at endogenous levels. We find that around 5% of the cellular pool of caveolae is present on dynamic endosomes, and is delivered to endosomes in a clathrin-independent manner. Furthermore, we show that caveolae are indeed likely to bud directly from the plasma membrane. Using a genetically encoded tag for electron microscopy and ratiometric light microscopy, we go on to show that bulk membrane proteins are depleted within caveolae. Although caveolae are likely to account for only a small proportion of total endocytosis, cells lacking caveolae show fundamentally altered patterns of membrane traffic when loaded with excess glycosphingolipid. Altogether, these observations support the hypothesis that caveolar endocytosis is specialized for transport of membrane lipid.


Subject(s)
Caveolin 1/metabolism , Glycosphingolipids/metabolism , Membrane Proteins/metabolism , Animals , Caveolin 1/genetics , Cell Membrane , Gene Expression Regulation/physiology , Genome , Green Fluorescent Proteins , HeLa Cells , Humans , Luminescent Proteins , Membrane Proteins/genetics , Mice , NIH 3T3 Cells , Photobleaching , Recombinant Proteins , Red Fluorescent Protein
13.
Nat Cell Biol ; 17(4): 500-10, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25751140

ABSTRACT

YAP (Yes-associated protein) is a transcription co-activator in the Hippo tumour suppressor pathway and controls cell growth, tissue homeostasis and organ size. YAP is inhibited by the kinase Lats, which phosphorylates YAP to induce its cytoplasmic localization and proteasomal degradation. YAP induces gene expression by binding to the TEAD family transcription factors. Dysregulation of the Hippo-YAP pathway is frequently observed in human cancers. Here we show that cellular energy stress induces YAP phosphorylation, in part due to AMPK-dependent Lats activation, thereby inhibiting YAP activity. Moreover, AMPK directly phosphorylates YAP Ser 94, a residue essential for the interaction with TEAD, thus disrupting the YAP-TEAD interaction. AMPK-induced YAP inhibition can suppress oncogenic transformation of Lats-null cells with high YAP activity. Our study establishes a molecular mechanism and functional significance of AMPK in linking cellular energy status to the Hippo-YAP pathway.


Subject(s)
AMP-Activated Protein Kinases/genetics , Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Phosphoproteins/antagonists & inhibitors , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , 3T3 Cells , Acyltransferases , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Line , Cell Transformation, Neoplastic/genetics , Cytoplasm , DNA-Binding Proteins/metabolism , Energy Metabolism , Enzyme Activation , HEK293 Cells , HeLa Cells , Hippo Signaling Pathway , Humans , Mice , Mice, Nude , Nuclear Proteins/metabolism , Phosphoproteins/genetics , Phosphoproteins/metabolism , Phosphorylation , RNA Interference , RNA, Small Interfering , TEA Domain Transcription Factors , Transcription Factors/genetics , Transcription Factors/metabolism , YAP-Signaling Proteins
14.
Nat Rev Cancer ; 15(2): 73-79, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25592648

ABSTRACT

Yes-associated protein (YAP) and transcriptional co-activator with PDZ-binding motif (TAZ) are the major downstream effectors of the Hippo pathway, which regulates tissue homeostasis, organ size, regeneration and tumorigenesis. In this Progress article, we summarize the current understanding of the biological functions of YAP and TAZ, and how the regulation of these two proteins can be disrupted in cancer. We also highlight recent findings on their expanding role in cancer progression and describe the potential of these targets for therapeutic intervention.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Cell Transformation, Neoplastic/genetics , Intracellular Signaling Peptides and Proteins/genetics , Neoplasms/genetics , Phosphoproteins/genetics , AMP-Activated Protein Kinase Kinases , Apoptosis , Cytoskeletal Proteins/genetics , GTP-Binding Protein alpha Subunits/genetics , GTP-Binding Protein alpha Subunits, Gq-G11 , Hippo Signaling Pathway , Humans , MicroRNAs/genetics , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins p21(ras) , Trans-Activators , Transcription Factors , Transcriptional Coactivator with PDZ-Binding Motif Proteins , Wnt Signaling Pathway/genetics , YAP-Signaling Proteins , ras Proteins/genetics
15.
Nat Commun ; 4: 1831, 2013.
Article in English | MEDLINE | ID: mdl-23652019

ABSTRACT

Caveolae are abundant in endothelial cells and are thought to have important roles in endothelial cell biology. The cavin proteins are key components of caveolae, and are expressed at varied amounts in different tissues. Here we use knockout mice to determine the roles of cavins 2 and 3 in caveolar morphogenesis in vivo. Deletion of cavin 2 causes loss of endothelial caveolae in lung and adipose tissue, but has no effect on the abundance of endothelial caveolae in heart and other tissues. Changes in the morphology of endothelium in cavin 2 null mice correlate with changes in caveolar abundance. Cavin 3 is not required for making caveolae in the tissues examined. Cavin 2 determines the size of cavin complexes, and acts to shape caveolae. Cavin 1, however, is essential for normal oligomerization of caveolin 1. Our data reveal that endothelial caveolae are heterogeneous, and identify cavin 2 as a determinant of this heterogeneity.


Subject(s)
Caveolae/metabolism , Endothelium/growth & development , Endothelium/metabolism , Gene Deletion , Membrane Proteins/genetics , Morphogenesis , Organ Specificity , Animals , Caveolin 1/chemistry , Caveolin 1/metabolism , Cell Shape , Endothelial Cells/cytology , Endothelial Cells/metabolism , Endothelial Cells/ultrastructure , Endothelium/ultrastructure , Lung/cytology , Lung/metabolism , Lung/ultrastructure , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Weight , Multiprotein Complexes/metabolism , Myocardium/metabolism , Myocardium/ultrastructure , Protein Structure, Quaternary , RNA-Binding Proteins
16.
J Cell Sci ; 124(Pt 16): 2777-85, 2011 Aug 15.
Article in English | MEDLINE | ID: mdl-21807942

ABSTRACT

The pacsin (also termed syndapin) protein family is well characterised structurally. They contain F-BAR domains associated with the generation or maintenance of membrane curvature. The cell biology of these proteins remains less understood. Here, we initially confirm that EHD2, a protein previously shown biochemically to be present in caveolar fractions and to bind to pacsins, is a caveolar protein. We go on to report that GFP-pacsin 2 can be recruited to caveolae, and that endogenous pacsin 2 partially colocalises with caveolin 1 at the plasma membrane. Analysis of the role of pacsin 2 in caveolar biogenesis using small interfering RNA (siRNA) reveals that loss of pacsin 2 function results in loss of morphologically defined caveolae and accumulation of caveolin proteins within the plasma membrane. Overexpression of the F-BAR domain of pacsin 2 (but not the related F-BAR domains of CIP4 and FBP17) disrupts caveolar morphogenesis or trafficking, implying that pacsin 2 interacts with components required for these processes. We propose that pacsin 2 has an important role in the formation of plasma membrane caveolae.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Carrier Proteins/metabolism , Caveolae/physiology , Caveolin 1/metabolism , Cell Membrane/metabolism , Fibroblasts/physiology , Proteins/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Caveolae/ultrastructure , Cell Membrane/ultrastructure , Cloning, Molecular , Cytoskeletal Proteins , Fibroblasts/ultrastructure , Humans , Mice , Microscopy, Electron , NIH 3T3 Cells , Protein Structure, Tertiary/genetics , Protein Transport/genetics , Proteins/genetics , RNA, Small Interfering/genetics , Transgenes/genetics
17.
Mol Pharmacol ; 69(4): 1242-50, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16434615

ABSTRACT

The serotonin transporter (SERT), which belongs to a family of sodium/chloride-dependent transporters, is the major pharmacological target in the treatment of several clinical disorders, including depression and anxiety. Interaction with a low-affinity allosteric site on SERT modulates the ligand affinity at the high-affinity binding site. Serotonin (5-hydroxytryptamine) and certain SERT inhibitors possess affinity for both sites. In the present study, we report the characterization of a severely attenuated allosteric mechanism at the recently cloned chicken serotonin transporter (gSERT). A cross-species chimera study was performed, followed by species scanning mutagenesis. Residues important for the allosteric mechanism were mapped to the C-terminal part of SERT containing the transmembrane domains 10 to 12. We identified nine residues located in four distinct amino acid segments. The contribution of each segment and individual residues was investigated. Consequently, a gSERT mutant with a restored allosteric mechanism, as well as a human SERT (hSERT) mutant with a severely attenuated allosteric mechanism, was generated. The nine residues confer a functional allosteric mechanism for different combinations of ligands, suggesting that they contribute to a general allosteric mechanism at SERT. The finding of an allosteric mechanism at SERT is likely to be of physiological importance, in that serotonin was also found to act as an allosteric effector at duloxetine, RTI-55 and (S)-citalopram. Furthermore, the allosteric potency of 5-HT was found to be conserved for both hSERT and gSERT.


Subject(s)
Serotonin Plasma Membrane Transport Proteins/metabolism , Allosteric Regulation , Animals , COS Cells , Chickens , Chimera , Chlorocebus aethiops , Citalopram/pharmacology , Mutagenesis, Site-Directed , Serotonin Plasma Membrane Transport Proteins/genetics , Species Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...