Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
1.
Cell ; 186(1): 80-97.e26, 2023 01 05.
Article in English | MEDLINE | ID: mdl-36608661

ABSTRACT

Glucose is a universal bioenergy source; however, its role in controlling protein interactions is unappreciated, as are its actions during differentiation-associated intracellular glucose elevation. Azido-glucose click chemistry identified glucose binding to a variety of RNA binding proteins (RBPs), including the DDX21 RNA helicase, which was found to be essential for epidermal differentiation. Glucose bound the ATP-binding domain of DDX21, altering protein conformation, inhibiting helicase activity, and dissociating DDX21 dimers. Glucose elevation during differentiation was associated with DDX21 re-localization from the nucleolus to the nucleoplasm where DDX21 assembled into larger protein complexes containing RNA splicing factors. DDX21 localized to specific SCUGSDGC motif in mRNA introns in a glucose-dependent manner and promoted the splicing of key pro-differentiation genes, including GRHL3, KLF4, OVOL1, and RBPJ. These findings uncover a biochemical mechanism of action for glucose in modulating the dimerization and function of an RNA helicase essential for tissue differentiation.


Subject(s)
DEAD-box RNA Helicases , Glucose , Keratinocytes , Cell Nucleolus/metabolism , Cell Nucleus/metabolism , DEAD-box RNA Helicases/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Glucose/metabolism , Keratinocytes/cytology , Keratinocytes/metabolism , Humans
2.
Genes Dev ; 34(7-8): 511-525, 2020 04 01.
Article in English | MEDLINE | ID: mdl-32115406

ABSTRACT

The Hippo pathway is a master regulator of tissue homeostasis and organ size. NF2 is a well-established tumor suppressor, and loss of NF2 severely compromises Hippo pathway activity. However, the precise mechanism of how NF2 mediates upstream signals to regulate the Hippo pathway is not clear. Here we report that, in mammalian cells, NF2's lipid-binding ability is critical for its function in activating the Hippo pathway in response to osmotic stress. Mechanistically, osmotic stress induces PI(4,5)P2 plasma membrane enrichment by activating the PIP5K family, allowing for NF2 plasma membrane recruitment and subsequent downstream Hippo pathway activation. An NF2 mutant deficient in lipid binding is unable to activate the Hippo pathway in response to osmotic stress, as measured by LATS and YAP phosphorylation. Our findings identify the PIP5K family as novel regulators upstream of Hippo signaling, and uncover the importance of phosphoinositide dynamics, specifically PI(4,5)P2, in Hippo pathway regulation.


Subject(s)
Homeostasis/physiology , Neurofibromin 2/metabolism , Phosphatidylinositols/metabolism , Signal Transduction , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Cycle Proteins/metabolism , Cell Line , Hippo Signaling Pathway , Humans , Mice , Neurofibromin 2/genetics , Osmotic Pressure/physiology , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Signal Transduction/genetics , YAP-Signaling Proteins
3.
Nature ; 560(7720): 655-660, 2018 08.
Article in English | MEDLINE | ID: mdl-30135582

ABSTRACT

Mammalian cells are surrounded by neighbouring cells and extracellular matrix (ECM), which provide cells with structural support and mechanical cues that influence diverse biological processes1. The Hippo pathway effectors YAP (also known as YAP1) and TAZ (also known as WWTR1) are regulated by mechanical cues and mediate cellular responses to ECM stiffness2,3. Here we identified the Ras-related GTPase RAP2 as a key intracellular signal transducer that relays ECM rigidity signals to control mechanosensitive cellular activities through YAP and TAZ. RAP2 is activated by low ECM stiffness, and deletion of RAP2 blocks the regulation of YAP and TAZ by stiffness signals and promotes aberrant cell growth. Mechanistically, matrix stiffness acts through phospholipase Cγ1 (PLCγ1) to influence levels of phosphatidylinositol 4,5-bisphosphate and phosphatidic acid, which activates RAP2 through PDZGEF1 and PDZGEF2 (also known as RAPGEF2 and RAPGEF6). At low stiffness, active RAP2 binds to and stimulates MAP4K4, MAP4K6, MAP4K7 and ARHGAP29, resulting in activation of LATS1 and LATS2 and inhibition of YAP and TAZ. RAP2, YAP and TAZ have pivotal roles in mechanoregulated transcription, as deletion of YAP and TAZ abolishes the ECM stiffness-responsive transcriptome. Our findings show that RAP2 is a molecular switch in mechanotransduction, thereby defining a mechanosignalling pathway from ECM stiffness to the nucleus.


Subject(s)
Protein Serine-Threonine Kinases/metabolism , Signal Transduction , rap GTP-Binding Proteins/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Transformation, Neoplastic , Extracellular Matrix/chemistry , Extracellular Matrix/genetics , Extracellular Matrix/metabolism , Female , GTPase-Activating Proteins/metabolism , Germinal Center Kinases , Guanine Nucleotide Exchange Factors/metabolism , HEK293 Cells , Hippo Signaling Pathway , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Mice , Mice, Inbred NOD , Mice, Nude , Mice, SCID , Nerve Tissue Proteins/metabolism , Phospholipase C gamma/metabolism , Phosphoproteins/metabolism , Trans-Activators , Transcription Factors , Transcriptional Coactivator with PDZ-Binding Motif Proteins , Transcriptome , YAP-Signaling Proteins , rap GTP-Binding Proteins/genetics
4.
Mol Cell ; 64(5): 993-1008, 2016 12 01.
Article in English | MEDLINE | ID: mdl-27912098

ABSTRACT

The Hippo pathway is important for regulating tissue homeostasis, and its dysregulation has been implicated in human cancer. However, it is not well understood how the Hippo pathway becomes dysregulated because few mutations in core Hippo pathway components have been identified. Therefore, much work in the Hippo field has focused on identifying upstream regulators, and a complex Hippo interactome has been identified. Nevertheless, it is not always clear which components are the most physiologically relevant in regulating YAP/TAZ. To provide an overview of important Hippo pathway components, we created knockout cell lines for many of these components and compared their relative contributions to YAP/TAZ regulation in response to a wide range of physiological signals. By this approach, we provide an overview of the functional importance of many Hippo pathway components and demonstrate NF2 and RHOA as important regulators of YAP/TAZ and TAOK1/3 as direct kinases for LATS1/2.


Subject(s)
DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Gene Expression Regulation/physiology , Signal Transduction/genetics , Acyltransferases , Cell Cycle Proteins , Clustered Regularly Interspaced Short Palindromic Repeats , Gene Knockdown Techniques , HEK293 Cells , Hippo Signaling Pathway , Humans , Neurofibromin 2 , Nuclear Proteins , Phosphorylation , Protein Serine-Threonine Kinases , Transcription Factors , Tumor Suppressor Proteins , rhoA GTP-Binding Protein
5.
EMBO Rep ; 18(1): 72-86, 2017 01.
Article in English | MEDLINE | ID: mdl-27979971

ABSTRACT

YAP is the major downstream effector of the Hippo pathway, which controls cell growth, tissue homeostasis, and organ size. Aberrant YAP activation, resulting from dysregulation of the Hippo pathway, is frequently observed in human cancers. YAP is a transcription co-activator, and the key mechanism of YAP regulation is its nuclear and cytoplasmic translocation. The Hippo pathway component, LATS, inhibits YAP by phosphorylating YAP at Ser127, leading to 14-3-3 binding and cytoplasmic retention of YAP Here, we report that osmotic stress stimulates transient YAP nuclear localization and increases YAP activity even when YAP Ser127 is phosphorylated. Osmotic stress acts via the NLK kinase to induce YAP Ser128 phosphorylation. Phosphorylation of YAP at Ser128 interferes with its ability to bind to 14-3-3, resulting in YAP nuclear accumulation and induction of downstream target gene expression. This osmotic stress-induced YAP activation enhances cellular stress adaptation. Our findings reveal a critical role for NLK-mediated Ser128 phosphorylation in YAP regulation and a crosstalk between osmotic stress and the Hippo pathway.


Subject(s)
Intracellular Signaling Peptides and Proteins/metabolism , Nuclear Proteins/metabolism , Osmotic Pressure , Protein Serine-Threonine Kinases/metabolism , Serine/metabolism , Transcription Factors/metabolism , Cell Cycle , Cell Cycle Proteins , Cell Nucleolus , Cytoplasm/metabolism , Enzyme Activation , HEK293 Cells , Humans , Intracellular Signaling Peptides and Proteins/chemistry , Phosphorylation , Protein Binding , Protein Serine-Threonine Kinases/chemistry , Protein Transport , Serine/chemistry , Signal Transduction
6.
Elife ; 82019 05 21.
Article in English | MEDLINE | ID: mdl-31112131

ABSTRACT

The mammalian target of rapamycin complex 1 (mTORC1) regulates cell growth, metabolism, and autophagy. Extensive research has focused on pathways that activate mTORC1 like growth factors and amino acids; however, much less is known about signaling cues that directly inhibit mTORC1 activity. Here, we report that G-protein coupled receptors (GPCRs) paired to Gαs proteins increase cyclic adenosine 3'5' monophosphate (cAMP) to activate protein kinase A (PKA) and inhibit mTORC1. Mechanistically, PKA phosphorylates the mTORC1 component Raptor on Ser 791, leading to decreased mTORC1 activity. Consistently, in cells where Raptor Ser 791 is mutated to Ala, mTORC1 activity is partially rescued even after PKA activation. Gαs-coupled GPCRs stimulation leads to inhibition of mTORC1 in multiple cell lines and mouse tissues. Our results uncover a signaling pathway that directly inhibits mTORC1, and suggest that GPCRs paired to Gαs proteins may be potential therapeutic targets for human diseases with hyperactivated mTORC1.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , Mechanistic Target of Rapamycin Complex 1/antagonists & inhibitors , Protein Processing, Post-Translational , Receptors, G-Protein-Coupled/metabolism , Regulatory-Associated Protein of mTOR/metabolism , Signal Transduction , Animals , Cell Line , Humans , Phosphorylation
7.
Bio Protoc ; 7(14)2017 Jul 20.
Article in English | MEDLINE | ID: mdl-29104896

ABSTRACT

This protocol describes a method to directly measure LATS activity by an in vitro kinase assay using YAP as a substrate.

8.
Nat Rev Gastroenterol Hepatol ; 13(6): 324-37, 2016 06.
Article in English | MEDLINE | ID: mdl-27147489

ABSTRACT

The Hippo pathway is a signalling cascade conserved from Drosophila melanogaster to mammals. The mammalian core kinase components comprise MST1 and MST2, SAV1, LATS1 and LATS2 and MOB1A and MOB1B. The transcriptional co-activators YAP1 and TAZ are the downstream effectors of the Hippo pathway and regulate target gene expression. Hippo signalling has crucial roles in the control of organ size, tissue homeostasis and regeneration, and dysregulation of the Hippo pathway can lead to uncontrolled cell growth and malignant transformation. Mammalian intestine consists of a stem cell compartment as well as differentiated cells, and its ability to regenerate rapidly after injury makes it an excellent model system to study tissue homeostasis, regeneration and tumorigenesis. Several studies have established the important role of the Hippo pathway in these processes. In addition, crosstalk between Hippo and other signalling pathways provides tight, yet versatile, regulation of tissue homeostasis. In this Review, we summarize studies on the role of the Hippo pathway in the intestine on these physiological processes and the underlying mechanisms responsible, and discuss future research directions and potential therapeutic strategies targeting Hippo signalling in intestinal disease.


Subject(s)
Intestine, Large/physiology , Intestine, Small/physiology , Protein Serine-Threonine Kinases/metabolism , Regeneration/physiology , Signal Transduction/physiology , Acyltransferases , Adaptor Proteins, Signal Transducing/metabolism , Animals , Bone Morphogenetic Proteins/metabolism , Carcinogenesis/metabolism , Cell Adhesion/physiology , Colonic Neoplasms/physiopathology , Drosophila melanogaster , Hedgehog Proteins/metabolism , Hippo Signaling Pathway , Homeostasis/physiology , Humans , Irritable Bowel Syndrome/physiopathology , Liver Neoplasms/physiopathology , Mammals , Organ Size , Phosphoproteins/metabolism , Receptors, G-Protein-Coupled/physiology , Receptors, Notch/metabolism , Stress, Physiological/physiology , Transcription Factors/metabolism , Wnt Signaling Pathway/physiology , YAP-Signaling Proteins
9.
Trends Mol Med ; 21(4): 212-22, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25702974

ABSTRACT

The Hippo signaling pathway is important for controlling organ size and tissue homeostasis. Originally identified in Drosophila melanogaster, the core components of the Hippo pathway are highly conserved in mammals. The Hippo pathway can be modulated by a wide range of stimuli, including G protein-coupled receptor (GPCR) signaling, changes in the actin cytoskeleton, cell-cell contact, and cell polarity. When activated, the Hippo pathway functions as a tumor suppressor to limit cell growth. However, dysregulation by genetic inactivation of core pathway components or amplification or gene fusion of its downstream effectors results in increased cell proliferation and decreased apoptosis and differentiation. Unsurprisingly, this can lead to tissue overgrowth, tumorigenesis, and many other diseases.


Subject(s)
Drosophila Proteins/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Neoplasms/genetics , Nuclear Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Trans-Activators/metabolism , Animals , Apoptosis , Carcinogenesis , Cell Proliferation , Drosophila Proteins/genetics , Drosophila melanogaster , Gene Expression Regulation, Developmental , Gene Expression Regulation, Neoplastic , Hippo Signaling Pathway , Homeostasis , Humans , Intracellular Signaling Peptides and Proteins/genetics , Mammals , Neoplasms/pathology , Nuclear Proteins/genetics , Organ Size , Protein Serine-Threonine Kinases/genetics , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Trans-Activators/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , YAP-Signaling Proteins
10.
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