Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 82
Filter
Add more filters

Publication year range
1.
Cell ; 185(23): 4376-4393.e18, 2022 11 10.
Article in English | MEDLINE | ID: mdl-36318920

ABSTRACT

The function of biomolecular condensates is often restricted by condensate dissolution. Whether condensates can be suppressed without condensate dissolution is unclear. Here, we show that upstream regulators of the Hippo signaling pathway form functionally antagonizing condensates, and their coalescence into a common phase provides a mode of counteracting the function of biomolecular condensates without condensate dissolution. Specifically, the negative regulator SLMAP forms Hippo-inactivating condensates to facilitate pathway inhibition by the STRIPAK complex. In response to cell-cell contact or osmotic stress, the positive regulators AMOT and KIBRA form Hippo-activating condensates to facilitate pathway activation. The functionally antagonizing SLMAP and AMOT/KIBRA condensates further coalesce into a common phase to inhibit STRIPAK function. These findings provide a paradigm for restricting the activity of biomolecular condensates without condensate dissolution, shed light on the molecular principles of multiphase organization, and offer a conceptual framework for understanding upstream regulation of the Hippo signaling pathway.


Subject(s)
Hippo Signaling Pathway , Protein Serine-Threonine Kinases , Signal Transduction
2.
Nat Rev Mol Cell Biol ; 24(12): 895-911, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37626124

ABSTRACT

Complex physiological processes control whether stem cells self-renew, differentiate or remain quiescent. Two decades of research have placed the Hippo pathway, a highly conserved kinase signalling cascade, and its downstream molecular effectors YAP and TAZ at the nexus of this decision. YAP and TAZ translate complex biological cues acting on stem cells - from mechanical forces to cellular metabolism - into genome-wide effects to mediate stem cell functions. While aberrant YAP/TAZ activity drives stem cell dysfunction in ageing, tumorigenesis and disease, therapeutic targeting of Hippo signalling and YAP/TAZ can boost stem cell activity to enhance regeneration. In this Review, we discuss how YAP/TAZ control the self-renewal, fate and plasticity of stem cells in different contexts, how dysregulation of YAP/TAZ in stem cells leads to disease, and how therapeutic modalities targeting YAP/TAZ may benefit regenerative medicine and cancer therapy.


Subject(s)
Cell Self Renewal , Signal Transduction , Transcriptional Coactivator with PDZ-Binding Motif Proteins , YAP-Signaling Proteins , Humans , Carcinogenesis , Signal Transduction/physiology , Stem Cells/metabolism , YAP-Signaling Proteins/metabolism , Transcriptional Coactivator with PDZ-Binding Motif Proteins/metabolism
3.
Cell ; 164(3): 406-19, 2016 Jan 28.
Article in English | MEDLINE | ID: mdl-26824654

ABSTRACT

The Hippo signaling pathway functions through Yorkie to control tissue growth and homeostasis. How this pathway regulates non-developmental processes remains largely unexplored. Here, we report an essential role for Hippo signaling in innate immunity whereby Yorkie directly regulates the transcription of the Drosophila IκB homolog, Cactus, in Toll receptor-mediated antimicrobial response. Loss of Hippo pathway tumor suppressors or activation of Yorkie in fat bodies, the Drosophila immune organ, leads to elevated cactus mRNA levels, decreased expression of antimicrobial peptides, and vulnerability to infection by Gram-positive bacteria. Furthermore, Gram-positive bacteria acutely activate Hippo-Yorkie signaling in fat bodies via the Toll-Myd88-Pelle cascade through Pelle-mediated phosphorylation and degradation of the Cka subunit of the Hippo-inhibitory STRIPAK PP2A complex. Our studies elucidate a Toll-mediated Hippo signaling pathway in antimicrobial response, highlight the importance of regulating IκB/Cactus transcription in innate immunity, and identify Gram-positive bacteria as extracellular stimuli of Hippo signaling under physiological settings.


Subject(s)
Drosophila melanogaster/immunology , Immunity, Innate , Signal Transduction , Animals , DNA-Binding Proteins/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/growth & development , Drosophila melanogaster/metabolism , Drosophila melanogaster/microbiology , Escherichia coli/physiology , Fat Body/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Larva/metabolism , Male , Pectobacterium carotovorum/physiology , Phosphoproteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Staphylococcus aureus/physiology , Toll-Like Receptors/metabolism
4.
Genes Dev ; 36(21-24): 1119-1128, 2022.
Article in English | MEDLINE | ID: mdl-36522128

ABSTRACT

The Hippo-YAP signaling pathway plays a critical role in development, homeostasis, regeneration, and tumorigenesis by converging on YAP, a coactivator for the TEAD family DNA-binding transcription factors, to regulate downstream transcription programs. Given its pivotal role as the nuclear effector of the Hippo pathway, YAP is indispensable in multiple developmental and tissue contexts. Here we report that the essentiality of YAP in liver and lung development can be genetically bypassed by simultaneous inactivation of the TEAD corepressor VGLL4. This striking antagonistic epistasis suggests that the major physiological function of YAP is to antagonize VGLL4. We further show that the YAP-VGLL4 antagonism plays a widespread role in regulating Hippo pathway output beyond normal development, as inactivation of Vgll4 dramatically enhanced intrahepatic cholangiocarcinoma formation in Nf2-deficient livers and ameliorated CCl4-induced damage in normal livers. Interestingly, Vgll4 expression is temporally regulated in development and regeneration and, in certain contexts, provides a better indication of overall Hippo pathway output than YAP phosphorylation. Together, these findings highlight the central importance of VGLL4-mediated transcriptional repression in Hippo pathway regulation and inform potential strategies to modulate Hippo signaling in cancer and regenerative medicine.


Subject(s)
Hippo Signaling Pathway , Transcription Factors , Transcription Factors/genetics , Transcription Factors/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , YAP-Signaling Proteins , TEA Domain Transcription Factors
5.
Genes Dev ; 35(7-8): 495-511, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33766984

ABSTRACT

Epithelioid hemangioendothelioma (EHE) is a poorly understood and devastating vascular cancer. Sequencing of EHE has revealed a unique gene fusion between the Hippo pathway nuclear effector TAZ (WWTR1) and the brain-enriched transcription factor CAMTA1 in ∼90% of cases. However, it remains unclear whether the TAZ-CAMTA1 gene fusion is a driver of EHE, and potential targeted therapies are unknown. Here, we show that TAZ-CAMTA1 expression in endothelial cells is sufficient to drive the formation of vascular tumors with the distinctive features of EHE, and inhibition of TAZ-CAMTA1 results in the regression of these vascular tumors. We further show that activated TAZ resembles TAZ-CAMTA1 in driving the formation of EHE-like vascular tumors, suggesting that constitutive activation of TAZ underlies the pathological features of EHE. We show that TAZ-CAMTA1 initiates an angiogenic and regenerative-like transcriptional program in endothelial cells, and disruption of the TAZ-CAMTA1-TEAD interaction or ectopic expression of a dominant negative TEAD in vivo inhibits TAZ-CAMTA1-mediated transformation. Our study provides the first genetic model of a TAZ fusion oncoprotein driving its associated human cancer, pinpointing TAZ-CAMTA1 as the key driver and a valid therapeutic target of EHE.


Subject(s)
Calcium-Binding Proteins/metabolism , Carcinogenesis/genetics , Endothelial Cells/pathology , Gene Expression Regulation, Neoplastic , Hemangioendothelioma, Epithelioid/genetics , Hemangioendothelioma, Epithelioid/pathology , Intracellular Signaling Peptides and Proteins/metabolism , Trans-Activators/metabolism , Animals , Calcium-Binding Proteins/genetics , Cell Line, Tumor , Gene Fusion , Humans , Intracellular Signaling Peptides and Proteins/genetics , Mice , Trans-Activators/genetics , Transcriptional Coactivator with PDZ-Binding Motif Proteins
6.
Trends Biochem Sci ; 2024 May 09.
Article in English | MEDLINE | ID: mdl-38729842

ABSTRACT

Decades of work in developmental genetics has given us a deep mechanistic understanding of the fundamental signaling pathways underlying animal development. However, little is known about how these pathways emerged and changed over evolutionary time. Here, we review our current understanding of the evolutionary emergence of the Hippo pathway, a conserved signaling pathway that regulates tissue size in animals. This pathway has deep evolutionary roots, emerging piece by piece in the unicellular ancestors of animals, with a complete core pathway predating the origin of animals. Recent functional studies in close unicellular relatives of animals and early-branching animals suggest an ancestral function Hippo pathway of cytoskeletal regulation, which was subsequently co-opted to regulate proliferation and animal tissue size.

7.
Cell ; 154(6): 1342-55, 2013 Sep 12.
Article in English | MEDLINE | ID: mdl-24012335

ABSTRACT

Although Merlin/NF2 was discovered two decades ago as a tumor suppressor underlying Neurofibromatosis type II, its precise molecular mechanism remains poorly understood. Recent studies in Drosophila revealed a potential link between Merlin and the Hippo pathway by placing Merlin genetically upstream of the kinase Hpo/Mst. In contrast to the commonly depicted linear model of Merlin functioning through Hpo/Mst, here we show that in both Drosophila and mammals, Merlin promotes downstream Hippo signaling without activating the intrinsic kinase activity of Hpo/Mst. Instead, Merlin directly binds and recruits the effector kinase Wts/Lats to the plasma membrane. Membrane recruitment, in turn, promotes Wts phosphorylation by the Hpo-Sav kinase complex. We further show that disruption of the actin cytoskeleton promotes Merlin-Wts interactions, which implicates Merlin in actin-mediated regulation of Hippo signaling. Our findings elucidate an important molecular function of Merlin and highlight the plasma membrane as a critical subcellular compartment for Hippo signal transduction.


Subject(s)
Cell Membrane/metabolism , Drosophila melanogaster/metabolism , Neurofibromin 2/metabolism , Signal Transduction , Amino Acid Sequence , Animals , Biological Evolution , Cytoskeleton/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/cytology , Intracellular Signaling Peptides and Proteins/metabolism , Mice , Molecular Sequence Data , Protein Serine-Threonine Kinases/metabolism , Sequence Alignment
8.
Genes Dev ; 32(11-12): 781-793, 2018 06 01.
Article in English | MEDLINE | ID: mdl-29891559

ABSTRACT

Autosomal dominant polycystic kidney disease (ADPKD) is an inherited disorder caused by mutations in PKD1 or PKD2 and affects one in 500-1000 humans. Limited treatment is currently available for ADPKD. Here we identify the Hippo signaling effector YAP and its transcriptional target, c-Myc, as promoters of cystic kidney pathogenesis. While transgenic overexpression of YAP promotes proliferation and tubule dilation in mouse kidneys, loss of YAP/TAZ or c-Myc suppresses cystogenesis in a mouse ADPKD model resulting from Pkd1 deficiency. Through a comprehensive kinase inhibitor screen based on a novel three-dimensional (3D) culture of Pkd1 mutant mouse kidney cells, we identified a signaling pathway involving the RhoGEF (guanine nucleotide exchange factor) LARG, the small GTPase RhoA, and the RhoA effector Rho-associated kinase (ROCK) as a critical signaling module between PKD1 and YAP. Further corroborating its physiological importance, inhibition of RhoA signaling suppresses cystogenesis in 3D culture of Pkd1 mutant kidney cells as well as Pkd1 mutant mouse kidneys in vivo. Taken together, our findings implicate the RhoA-YAP-c-Myc signaling axis as a critical mediator and potential drug target in ADPKD.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Kidney/physiopathology , Phosphoproteins/metabolism , Polycystic Kidney Diseases/genetics , Polycystic Kidney Diseases/physiopathology , Proto-Oncogene Proteins c-myc/metabolism , Signal Transduction , rho GTP-Binding Proteins/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Cell Cycle Proteins , Cell Line , Cells, Cultured , Disease Models, Animal , HEK293 Cells , Humans , Kidney/cytology , Kidney/pathology , Mice , Phosphoproteins/genetics , Polycystic Kidney Diseases/pathology , Proto-Oncogene Proteins c-myc/genetics , YAP-Signaling Proteins , rho GTP-Binding Proteins/genetics , rhoA GTP-Binding Protein
9.
Cell ; 139(4): 757-69, 2009 Nov 13.
Article in English | MEDLINE | ID: mdl-19914168

ABSTRACT

TGF-beta and BMP receptor kinases activate Smad transcription factors by C-terminal phosphorylation. We have identified a subsequent agonist-induced phosphorylation that plays a central dual role in Smad transcriptional activation and turnover. As receptor-activated Smads form transcriptional complexes, they are phosphorylated at an interdomain linker region by CDK8 and CDK9, which are components of transcriptional mediator and elongation complexes. These phosphorylations promote Smad transcriptional action, which in the case of Smad1 is mediated by the recruitment of YAP to the phosphorylated linker sites. An effector of the highly conserved Hippo organ size control pathway, YAP supports Smad1-dependent transcription and is required for BMP suppression of neural differentiation of mouse embryonic stem cells. The phosphorylated linker is ultimately recognized by specific ubiquitin ligases, leading to proteasome-mediated turnover of activated Smad proteins. Thus, nuclear CDK8/9 drive a cycle of Smad utilization and disposal that is an integral part of canonical BMP and TGF-beta pathways.


Subject(s)
Bone Morphogenetic Proteins/metabolism , Cyclin-Dependent Kinase 8/metabolism , Cyclin-Dependent Kinase 9/metabolism , Smad Proteins/genetics , Transcriptional Activation , Transforming Growth Factor beta/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Cycle Proteins , Cell Line , Contact Inhibition , Embryo, Mammalian/cytology , Humans , Mice , Organ Size , Phosphoproteins/metabolism , Phosphorylation , Protein Structure, Tertiary , Signal Transduction , Smad Proteins/chemistry , Smad1 Protein/genetics , YAP-Signaling Proteins
10.
Dev Biol ; 487: 1-9, 2022 07.
Article in English | MEDLINE | ID: mdl-35405135

ABSTRACT

The development of a functional organ requires not only patterning mechanisms that confer proper identities to individual cells, but also growth-regulatory mechanisms that specify the final size of the organ. At the turn of the 21st century, comprehensive genetic screens in model organisms had successfully uncovered the major signaling pathways that mediate pattern formation in metazoans. In contrast, signaling pathways dedicated to growth control were less explored. The past two decades has witnessed the emergence of the Hippo signaling pathway as a central mediator of organ size control through coordinated regulation of cell proliferation and apoptosis. Here I reflect on the early discoveries in Drosophila that elucidated the core kinase cascade and transcriptional machinery of the Hippo pathway, highlight its deep evolutionary conservation from humans to unicellular relatives of metazoan, and discuss the complex regulation of Hippo signaling by upstream inputs. This historical perspective underscores the importance of model organisms in uncovering fundamental and universal mechanisms of life processes.


Subject(s)
Drosophila Proteins , Animals , Drosophila/genetics , Drosophila Proteins/metabolism , Hippo Signaling Pathway , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Protein Serine-Threonine Kinases/genetics
11.
Genes Dev ; 29(14): 1493-506, 2015 Jul 15.
Article in English | MEDLINE | ID: mdl-26193883

ABSTRACT

Mutations in Adenomatous polyposis coli (APC) underlie familial adenomatous polyposis (FAP), an inherited cancer syndrome characterized by the widespread development of colorectal polyps. APC is best known as a scaffold protein in the ß-catenin destruction complex, whose activity is antagonized by canonical Wnt signaling. Whether other effector pathways mediate APC's tumor suppressor function is less clear. Here we report that activation of YAP, the downstream effector of the Hippo signaling pathway, is a general hallmark of tubular adenomas from FAP patients. We show that APC functions as a scaffold protein that facilitates the Hippo kinase cascade by interacting with Sav1 and Lats1. Consistent with the molecular link between APC and the Hippo signaling pathway, genetic analysis reveals that YAP is absolutely required for the development of APC-deficient adenomas. These findings establish Hippo-YAP signaling as a critical effector pathway downstream from APC, independent from its involvement in the ß-catenin destruction complex.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Adenomatous Polyposis Coli Protein/genetics , Adenomatous Polyposis Coli/physiopathology , Phosphoproteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Adenoma/enzymology , Adenoma/physiopathology , Adenomatous Polyposis Coli/enzymology , Adenomatous Polyposis Coli/genetics , Adenomatous Polyposis Coli Protein/metabolism , Animals , Carcinogenesis/genetics , Carcinogenesis/pathology , Cell Cycle Proteins , Cells, Cultured , Hippo Signaling Pathway , Humans , Intestines/physiopathology , Mice , Transcription Factors , YAP-Signaling Proteins , beta Catenin/metabolism
12.
Genes Dev ; 29(13): 1416-31, 2015 Jul 01.
Article in English | MEDLINE | ID: mdl-26108669

ABSTRACT

The Mst-Lats kinase cascade is central to the Hippo tumor-suppressive pathway that controls organ size and tissue homeostasis. The adaptor protein Mob1 promotes Lats activation by Mst, but the mechanism remains unknown. Here, we show that human Mob1 binds to autophosphorylated docking motifs in active Mst2. This binding enables Mob1 phosphorylation by Mst2. Phosphorylated Mob1 undergoes conformational activation and binds to Lats1. We determine the crystal structures of phospho-Mst2-Mob1 and phospho-Mob1-Lats1 complexes, revealing the structural basis of both phosphorylation-dependent binding events. Further biochemical and functional analyses demonstrate that Mob1 mediates Lats1 activation through dynamic scaffolding and allosteric mechanisms. Thus, Mob1 acts as a phosphorylation-regulated coupler of kinase activation by virtue of its ability to engage multiple ligands. We propose that stepwise, phosphorylation-triggered docking interactions of nonkinase elements enhance the specificity and robustness of kinase signaling cascades.


Subject(s)
Adaptor Proteins, Signal Transducing , Models, Molecular , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/physiology , Signal Transduction , Adaptor Proteins, Signal Transducing/chemistry , Adaptor Proteins, Signal Transducing/metabolism , Amino Acid Motifs , Amino Acid Sequence , Animals , Binding Sites , Cells, Cultured , Crystallization , Drosophila melanogaster , Hippo Signaling Pathway , Humans , Molecular Sequence Data , Phosphorylation , Protein Binding , Protein Serine-Threonine Kinases/metabolism , Protein Structure, Quaternary , Sequence Alignment , Serine-Threonine Kinase 3
13.
Genes Dev ; 29(12): 1285-97, 2015 Jun 15.
Article in English | MEDLINE | ID: mdl-26109051

ABSTRACT

The Hippo signaling pathway converges on YAP to regulate growth, differentiation, and regeneration. Previous studies with overexpressed proteins have shown that YAP is phosphorylated by its upstream kinase, Lats1/2, on multiple sites, including an evolutionarily conserved 14-3-3-binding site whose phosphorylation is believed to inhibit YAP by excluding it from the nucleus. Indeed, nuclear localization of YAP or decreased YAP phosphorylation at this site (S168 in Drosophila, S127 in humans, and S112 in mice) is widely used in current literature as a surrogate of YAP activation even though the physiological importance of this phosphorylation event in regulating endogenous YAP activity has not been defined. Here we address this question by introducing a Yap(S112A) knock-in mutation in the endogenous Yap locus. The Yap(S112A) mice are surprisingly normal despite nuclear localization of the mutant YAP protein in vivo and profound defects in cytoplasmic translocation in vitro. Interestingly, the mutant Yap(S112A) mice show a compensatory decrease in YAP protein levels due to increased phosphorylation at a mammalian-specific phosphodegron site on YAP. These findings reveal a robust homeostatic mechanism that maintains physiological levels of YAP activity and caution against the assumptive use of YAP localization alone as a surrogate of YAP activity.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Homeostasis/physiology , Phosphoproteins/genetics , Phosphoproteins/metabolism , Protein Serine-Threonine Kinases/physiology , Signal Transduction/physiology , Animals , Cell Cycle Proteins , Cell Nucleus/metabolism , Cells, Cultured , Cytoplasm/metabolism , Feedback, Physiological , Gene Expression Regulation, Developmental , Gene Knock-In Techniques , Hippo Signaling Pathway , Homeostasis/genetics , Liver/pathology , Mice , Mice, Inbred C57BL , Mutation , Phosphorylation , Protein Binding , Protein Transport/genetics , YAP-Signaling Proteins
14.
Genes Dev ; 29(12): 1271-84, 2015 Jun 15.
Article in English | MEDLINE | ID: mdl-26109050

ABSTRACT

YAP (Yes-associated protein) and TAZ (transcriptional coactivator with PDZ-binding motif) are major downstream effectors of the Hippo pathway that influences tissue homeostasis, organ size, and cancer development. Aberrant hyperactivation of YAP/TAZ causes tissue overgrowth and tumorigenesis, whereas their inactivation impairs tissue development and regeneration. Dynamic and precise control of YAP/TAZ activity is thus important to ensure proper physiological regulation and homeostasis of the cells. Here, we show that YAP/TAZ activation results in activation of their negative regulators, LATS1/2 (large tumor suppressor 1/2) kinases, to constitute a negative feedback loop of the Hippo pathway in both cultured cells and mouse tissues. YAP/TAZ in complex with the transcription factor TEAD (TEA domain family member) directly induce LATS2 expression. Furthermore, YAP/TAZ also stimulate the kinase activity of LATS1/2 through inducing NF2 (neurofibromin 2). This feedback regulation is responsible for the transient activation of YAP upon lysophosphatidic acid (LPA) stimulation and the inhibition of YAP-induced cell migration. Thus, this LATS-mediated feedback loop provides an efficient mechanism to establish the robustness and homeostasis of YAP/TAZ regulation.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Feedback, Physiological/physiology , Homeostasis/physiology , Neurofibromin 2/metabolism , Phosphoproteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Transcription Factors/metabolism , Acyltransferases , Adaptor Proteins, Signal Transducing/genetics , Animals , Cell Cycle Proteins , Cell Movement , Cells, Cultured , DNA-Binding Proteins/metabolism , Enzyme Activation/physiology , Female , Gene Expression Regulation , HEK293 Cells , Hippo Signaling Pathway , Homeostasis/genetics , Humans , Liver/metabolism , Male , Mice , Phosphoproteins/genetics , Protein Serine-Threonine Kinases/genetics , Transcription Factors/genetics , Tumor Suppressor Proteins/genetics , YAP-Signaling Proteins
15.
Genes Dev ; 28(5): 432-7, 2014 Mar 01.
Article in English | MEDLINE | ID: mdl-24589775

ABSTRACT

Despite recent progress, the physiological role of Hippo signaling in mammary gland development and tumorigenesis remains poorly understood. Here we show that the Hippo pathway is functionally dispensable in virgin mammary glands but specifically required during pregnancy. In contrast to many other tissues, hyperactivation of YAP in mammary epithelia does not induce hyperplasia but leads to defects in terminal differentiation. Interestingly, loss of YAP causes no obvious defects in virgin mammary glands but potently suppresses oncogene-induced mammary tumors. The selective requirement for YAP in oncogenic growth highlights the potential of YAP inhibitors as molecular targeted therapies against breast cancers.


Subject(s)
Carcinogenesis/pathology , Cell Differentiation , Mammary Glands, Animal/cytology , Mammary Glands, Animal/pathology , Mammary Neoplasms, Animal/pathology , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Animals , Female , Hippo Signaling Pathway , Mammary Glands, Animal/growth & development , Mice, Inbred C57BL , Mice, Transgenic , Oncogenes/genetics , Pregnancy , Time
16.
J Biol Chem ; 295(7): 1889-1897, 2020 02 14.
Article in English | MEDLINE | ID: mdl-31862735

ABSTRACT

Sphingolipids (SLs) are structurally diverse lipids that are defined by the presence of a long-chain base (LCB) backbone. Typically, LCBs contain a single Δ4E double bond (DB) (mostly d18:1), whereas the dienic LCB sphingadienine (d18:2) contains a second DB at the Δ14Z position. The enzyme introducing the Δ14Z DB is unknown. We analyzed the LCB plasma profile in a gender-, age-, and BMI-matched subgroup of the CoLaus cohort (n = 658). Sphingadienine levels showed a significant association with gender, being on average ∼30% higher in females. A genome-wide association study (GWAS) revealed variants in the fatty acid desaturase 3 (FADS3) gene to be significantly associated with the plasma d18:2/d18:1 ratio (p = -log 7.9). Metabolic labeling assays, FADS3 overexpression and knockdown approaches, and plasma LCB profiling in FADS3-deficient mice confirmed that FADS3 is a bona fide LCB desaturase and required for the introduction of the Δ14Z double bond. Moreover, we showed that FADS3 is required for the conversion of the atypical cytotoxic 1-deoxysphinganine (1-deoxySA, m18:0) to 1-deoxysphingosine (1-deoxySO, m18:1). HEK293 cells overexpressing FADS3 were more resistant to m18:0 toxicity than WT cells. In summary, using a combination of metabolic profiling and GWAS, we identified FADS3 to be essential for forming Δ14Z DB containing LCBs, such as d18:2 and m18:1. Our results unravel FADS3 as a Δ14Z LCB desaturase, thereby disclosing the last missing enzyme of the SL de novo synthesis pathway.


Subject(s)
Fatty Acid Desaturases/genetics , Genome-Wide Association Study , Sphingolipids/genetics , Animals , Fatty Acid Desaturases/blood , HEK293 Cells , Humans , Lipids/genetics , Mice , Sphingolipids/blood , Sphingosine/analogs & derivatives , Sphingosine/metabolism , Spine/metabolism
17.
Development ; 145(4)2018 02 21.
Article in English | MEDLINE | ID: mdl-29467233

ABSTRACT

Genetic studies in Drosophila have been instrumental in characterizing the Hippo pathway, which converges on the co-activator Yorkie to regulate target gene transcription. A routinely used strategy to interrogate upstream regulators of Yorkie involves the examination of selected Hippo target genes upon loss or gain of function of a suspected pathway regulator. A caveat with this strategy is that aberrant expression of a given Hippo target per se does not distinguish whether it is caused by changes in Yorkie or Yorkie-independent inputs converging on the same target gene. Building on previous findings that the DNA-binding transcription factor Scalloped mediates both Yorkie overexpression and loss-of-function phenotypes yet is itself dispensable for normal eye development, we describe a simple strategy to distinguish these possibilities by analyzing double-mutant clones of scalloped and a suspected Yorkie regulator. We provide proof of principle that this strategy can be used effectively to validate canonical Yorkie regulators and to exclude proteins that impact target expression independent of Yorkie. The described methodology and reagents should facilitate efforts to assess the expanding repertoire of proteins implicated in regulation of Yorkie activity.


Subject(s)
Drosophila Proteins/metabolism , Epistasis, Genetic , Intracellular Signaling Peptides and Proteins/metabolism , Nuclear Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Trans-Activators/metabolism , Transcription Factors/metabolism , Animals , Cloning, Organism , Drosophila/genetics , Drosophila Proteins/genetics , Gene Expression Regulation, Developmental , Intracellular Signaling Peptides and Proteins/genetics , Nuclear Proteins/genetics , Protein Serine-Threonine Kinases/genetics , Reproducibility of Results , Signal Transduction , Trans-Activators/genetics , Transcription Factors/genetics , YAP-Signaling Proteins
18.
Genes Dev ; 27(11): 1223-32, 2013 Jun 01.
Article in English | MEDLINE | ID: mdl-23752589

ABSTRACT

The Hippo tumor suppressor pathway plays an important role in tissue homeostasis that ensures development of functional organs at proper size. The YAP transcription coactivator is a major effector of the Hippo pathway and is phosphorylated and inactivated by the Hippo pathway kinases Lats1/2. It has recently been shown that YAP activity is regulated by G-protein-coupled receptor signaling. Here we demonstrate that cyclic adenosine monophosphate (cAMP), a second messenger downstream from Gαs-coupled receptors, acts through protein kinase A (PKA) and Rho GTPases to stimulate Lats kinases and YAP phosphorylation. We also show that inactivation of YAP is crucial for PKA-induced adipogenesis. In addition, PKA activation in Drosophila inhibits the expression of Yorki (Yki, a YAP ortholog) target genes involved in cell proliferation and death. Taken together, our study demonstrates that Hippo-YAP is a key signaling branch of cAMP and PKA and reveals new insight into mechanisms of PKA in regulating a broad range of cellular functions.


Subject(s)
Cell Differentiation , Cyclic AMP-Dependent Protein Kinases/metabolism , Drosophila Proteins/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Acyltransferases , Adipogenesis , Animals , Cell Line , Cell Proliferation , Cyclic AMP/metabolism , Drosophila Proteins/antagonists & inhibitors , Drosophila melanogaster/enzymology , Drosophila melanogaster/metabolism , Enzyme Activation , Humans , Mice , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/metabolism , Phosphorylation , Second Messenger Systems/physiology , Serine-Threonine Kinase 3 , Trans-Activators/antagonists & inhibitors , Trans-Activators/metabolism , Transcription Factors/antagonists & inhibitors , Transcription Factors/metabolism , Tumor Suppressor Proteins/metabolism , YAP-Signaling Proteins , rho GTP-Binding Proteins/metabolism
19.
PLoS Biol ; 15(3): e2000949, 2017 03.
Article in English | MEDLINE | ID: mdl-28248965

ABSTRACT

During development, progenitor expansion, lineage allocation, and implementation of differentiation programs need to be tightly coordinated so that different cell types are generated in the correct numbers for appropriate tissue size and function. Pancreatic dysfunction results in some of the most debilitating and fatal diseases, including pancreatic cancer and diabetes. Several transcription factors regulating pancreas lineage specification have been identified, and Notch signalling has been implicated in lineage allocation, but it remains unclear how these processes are coordinated. Using a combination of genetic approaches, organotypic cultures of embryonic pancreata, and genomics, we found that sphingosine-1-phosphate (S1p), signalling through the G protein coupled receptor (GPCR) S1pr2, plays a key role in pancreas development linking lineage allocation and specification. S1pr2 signalling promotes progenitor survival as well as acinar and endocrine specification. S1pr2-mediated stabilisation of the yes-associated protein (YAP) is essential for endocrine specification, thus linking a regulator of progenitor growth with specification. YAP stabilisation and endocrine cell specification rely on Gαi subunits, revealing an unexpected specificity of selected GPCR intracellular signalling components. Finally, we found that S1pr2 signalling posttranscriptionally attenuates Notch signalling levels, thus regulating lineage allocation. Both S1pr2-mediated YAP stabilisation and Notch attenuation are necessary for the specification of the endocrine lineage. These findings identify S1p signalling as a novel key pathway coordinating cell survival, lineage allocation, and specification and linking these processes by regulating YAP levels and Notch signalling. Understanding lineage allocation and specification in the pancreas will shed light in the origins of pancreatic diseases and may suggest novel therapeutic approaches.


Subject(s)
Cell Lineage , Lysophospholipids/metabolism , Pancreas/cytology , Signal Transduction , Sphingosine/analogs & derivatives , Acinar Cells/cytology , Adaptor Proteins, Signal Transducing/metabolism , Animals , Body Patterning , Cell Cycle Proteins , Cell Differentiation , Cell Survival , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Mice , Models, Biological , Phosphoproteins/metabolism , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Protein Subunits/metabolism , Receptors, Lysosphingolipid/metabolism , Receptors, Notch/metabolism , Sphingosine/metabolism , Stem Cells/cytology , YAP-Signaling Proteins
20.
Circ Res ; 123(1): 43-56, 2018 06 22.
Article in English | MEDLINE | ID: mdl-29794022

ABSTRACT

RATIONALE: Microvascular inflammation and endothelial dysfunction secondary to unchecked activation of endothelium play a critical role in the pathophysiology of sepsis and organ failure. The intrinsic signaling mechanisms responsible for dampening excessive activation of endothelial cells are not completely understood. OBJECTIVE: To determine the central role of YAP (Yes-associated protein), the major transcriptional coactivator of the Hippo pathway, in modulating the strength and magnitude of endothelial activation and vascular inflammation. METHODS AND RESULTS: Endothelial-specific YAP knockout mice showed increased basal expression of E-selectin and ICAM (intercellular adhesion molecule)-1 in endothelial cells, a greater number of adherent neutrophils in postcapillary venules and increased neutrophil counts in bronchoalveolar lavage fluid. Lipopolysaccharide challenge of these mice augmented NF-κB (nuclear factor-κB) activation, expression of endothelial adhesion proteins, neutrophil and monocyte adhesion to cremaster muscle venules, transendothelial neutrophil migration, and lung inflammatory injury. Deletion of YAP in endothelial cells also markedly augmented the inflammatory response and cardiovascular dysfunction in a polymicrobial sepsis model induced by cecal ligation and puncture. YAP functioned by interacting with the E3 ubiquitin-protein ligase TLR (Toll-like receptor) signaling adaptor TRAF6 (tumor necrosis factor receptor-associated factor 6) to ubiquitinate TRAF6, and thus promoted TRAF6 degradation and modification resulting in inhibition of NF-κB activation. TRAF6 depletion in endothelial cells rescued the augmented inflammatory phenotype in mice with endothelial cell-specific deletion of YAP. CONCLUSIONS: YAP modulates the activation of endothelial cells and suppresses vascular inflammation through preventing TRAF6-mediated NF-κB activation and is hence essential for limiting the severity of sepsis-induced inflammation and organ failure.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Endothelial Cells/physiology , Endothelium, Vascular/physiopathology , Phosphoproteins/physiology , TNF Receptor-Associated Factor 6/metabolism , Vasculitis/etiology , Adaptor Proteins, Signal Transducing/deficiency , Adaptor Proteins, Signal Transducing/genetics , Animals , Capillary Permeability , Cell Adhesion , Cell Cycle Proteins , E-Selectin/metabolism , Endothelial Cells/cytology , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Intercellular Adhesion Molecule-1/metabolism , Leukocyte Count , Mice , Mice, Knockout , Microvessels , Monocytes/physiology , NF-kappa B/metabolism , Neutrophils/cytology , Phosphoproteins/deficiency , Phosphoproteins/genetics , Sepsis/complications , Ubiquitin-Protein Ligases/metabolism , Ubiquitination , Venules/cytology , YAP-Signaling Proteins
SELECTION OF CITATIONS
SEARCH DETAIL