Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 64
Filter
1.
Mol Cell ; 53(1): 3-5, 2014 Jan 09.
Article in English | MEDLINE | ID: mdl-24411079

ABSTRACT

In this issue of Molecular Cell, Chiba and colleagues (Matsuzawa et al., 2014) identify Obg-like ATPase 1 (OLA1) as an additional member of the BRCA1/BARD1/γ-tubulin complex that is critically involved in centrosome amplification and microtubule aster formation.


Subject(s)
Adenosine Triphosphatases/metabolism , BRCA1 Protein/metabolism , Breast Neoplasms/metabolism , Centrosome/metabolism , GTP-Binding Proteins/metabolism , Female , Humans
2.
EMBO Rep ; 20(11): e48150, 2019 11 05.
Article in English | MEDLINE | ID: mdl-31544310

ABSTRACT

STK38 (also known as NDR1) is a Hippo pathway serine/threonine protein kinase with multifarious functions in normal and cancer cells. Using a context-dependent proximity-labeling assay, we identify more than 250 partners of STK38 and find that STK38 modulates its partnership depending on the cellular context by increasing its association with cytoplasmic proteins upon nutrient starvation-induced autophagy and with nuclear ones during ECM detachment. We show that STK38 shuttles between the nucleus and the cytoplasm and that its nuclear exit depends on both XPO1 (aka exportin-1, CRM1) and STK38 kinase activity. We further uncover that STK38 modulates XPO1 export activity by phosphorylating XPO1 on serine 1055, thus regulating its own nuclear exit. We expand our model to other cellular contexts by discovering that XPO1 phosphorylation by STK38 regulates also the nuclear exit of Beclin1 and YAP1, key regulator of autophagy and transcriptional effector, respectively. Collectively, our results reveal STK38 as an activator of XPO1, behaving as a gatekeeper of nuclear export. These observations establish a novel mechanism of XPO1-dependent cargo export regulation by phosphorylation of XPO1's C-terminal auto-inhibitory domain.


Subject(s)
Autophagy , Cell Nucleus/metabolism , Karyopherins/metabolism , Protein Serine-Threonine Kinases/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Carrier Proteins/metabolism , Chromatography, Liquid , Computational Biology/methods , Hippo Signaling Pathway , Humans , Phosphorylation , Protein Binding , Protein Interaction Mapping , Protein Transport , Signal Transduction , Tandem Mass Spectrometry , Exportin 1 Protein
3.
Semin Cancer Biol ; 48: 104-114, 2018 02.
Article in English | MEDLINE | ID: mdl-28579171

ABSTRACT

The NDR (nuclear Dbf2-related)/LATS (large tumour suppressor) family of kinases represents a subclass of the AGC (protein kinase A (PKA)/PKG/PKC-like) group of serine/threonine protein kinases. Members of the NDR/LATS family are vital components of conserved pathways controlling essential cellular processes, such as proliferation (cell cycle progression) and cell death. In particular, the central involvement of NDR/LATS as YAP/TAZ kinases in the Hippo tissue growth control pathway has gained much interest. In this review, we summarise the roles of mammalian NDR1/2 (aka STK38/STK38L) and LATS1/2 in immunity and cancer biology. We also discuss the activation mechanisms of NDR/LATS involving Ste20-like kinases and the MOB1 signal transducer, followed by an overview of NDR/LATS knockout mouse models. We further review the mutation and expression status of NDR/LATS in human cancers and their possible predictive and/or prognostic value in cancer treatment.


Subject(s)
Neoplasms/enzymology , Protein Serine-Threonine Kinases/physiology , Tumor Suppressor Proteins/physiology , Acyltransferases , Adaptor Proteins, Signal Transducing/metabolism , Animals , Humans , Mice, Knockout , Mutation , Neoplasms/immunology , Phosphoproteins/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Transcription Factors/metabolism , YAP-Signaling Proteins
4.
J Cell Sci ; 127(Pt 16): 3463-76, 2014 Aug 15.
Article in English | MEDLINE | ID: mdl-24928906

ABSTRACT

The morphology and polarized growth of cells depend on pathways that control the asymmetric distribution of regulatory factors. The evolutionarily conserved Ndr kinases play important roles in cell polarity and morphogenesis in yeast and invertebrates but it is unclear whether they perform a similar function in mammalian cells. Here, we analyze the function of mammalian Ndr1 and Ndr2 (also known as STK38 or STK38L, respectively) in the establishment of polarity in neurons. We show that they act downstream of the tumor suppressor Rassf5 and upstream of the polarity protein Par3 (also known as PARD3). Rassf5 and Ndr1 or Ndr2 are required during the polarization of hippocampal neurons to prevent the formation of supernumerary axons. Mechanistically, the Ndr kinases act by phosphorylating Par3 at Ser383 to inhibit its interaction with dynein, thereby polarizing the distribution of Par3 and reinforcing axon specification. Our results identify a novel Rassf5-Ndr-Par3 signaling cascade that regulates the transport of Par3 during the establishment of neuronal polarity. Their role in neuronal polarity suggests that Ndr kinases perform a conserved function as regulators of cell polarity.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cell Adhesion Molecules/metabolism , Cell Polarity , Neurons/metabolism , Protein Serine-Threonine Kinases/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Apoptosis Regulatory Proteins , Cell Adhesion Molecules/genetics , Cell Cycle Proteins , Mice , Mice, Knockout , Neurons/cytology , Neurons/enzymology , Phosphorylation , Protein Binding , Protein Serine-Threonine Kinases/genetics , Signal Transduction
5.
Semin Cell Dev Biol ; 23(7): 794-802, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22525225

ABSTRACT

Over the past decade Hippo kinase signalling has been established as an essential tumour suppressor pathway controlling tissue growth in flies and mammals. All members of the Hippo core signalling cassette are conserved from yeast to humans, whereby the yeast analogues of Hippo, Mats and Lats are central components of the mitotic exit network and septation initiation network in budding and fission yeast, respectively. Here, we discuss how far core Hippo signalling components in Drosophila melanogaster and mammals have reported similar mitotic functions as already established for their highly conserved yeast counterparts.


Subject(s)
Cell Division , G2 Phase , Signal Transduction , Yeasts/cytology , Yeasts/metabolism , Animals , Humans , Protein Serine-Threonine Kinases/metabolism , RNA Polymerase III/metabolism , Transcriptional Elongation Factors/metabolism
7.
Breast Cancer Res ; 14(6): 326, 2012 Dec 05.
Article in English | MEDLINE | ID: mdl-23216692

ABSTRACT

The proto-oncogenes YAP and TAZ have previously gained much attention as downstream effectors of Hippo tumour suppressor signalling. While the regulation of YAP/TAZ by MST/LATS kinases is reasonably well understood, the nature of factors functioning upstream of MST/LATS is yet to be elucidated in detail. A recent paper by Ma and co-workers defines a novel role for leukemia inhibitory factor receptor (LIFR) signalling upstream of the Hippo-YAP pathway in breast cancer metastasis. Moreover, a whole genome in vivo RNA interference screen by Lippmann and colleagues identified LIFR as a breast tumour suppressor. Here, we discuss the implications of these studies for breast cancer research and treatment.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Breast Neoplasms/genetics , Genes, Tumor Suppressor , Phosphoproteins/genetics , Protein Serine-Threonine Kinases/genetics , Receptors, OSM-LIF/genetics , Acyltransferases , Female , Hippo Signaling Pathway , Humans , MAP Kinase Kinase Kinases/metabolism , Protein Serine-Threonine Kinases/metabolism , RNA Interference , Signal Transduction/genetics , Transcription Factors/genetics , YAP-Signaling Proteins
8.
Biochem Soc Trans ; 40(1): 124-8, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22260677

ABSTRACT

The Hippo signal transduction cascade controls cell growth, proliferation and death, all of which are frequently deregulated in tumour cells. Since initial studies in Drosophila melanogaster were instrumental in defining Hippo signalling, the machinery was named after the central Ste20-like kinase Hippo. Moreover, given that loss of Hippo signalling components Hippo, Warts, and Mats resulted in uncontrolled tissue overgrowth, Hippo signalling was defined as a tumour-suppressor cascade. Significantly, all of the core factors of Hippo signalling have mammalian orthologues that functionally compensate for loss of their counterparts in Drosophila. Furthermore, studies in Drosophila and mammalian cell systems showed that Hippo signalling represents a kinase cascade that is tightly regulated by PPIs (protein-protein interactions). Several Hippo signalling molecules contain SARAH (Salvador/RASSF1A/Hippo) domains that mediate specific PPIs, thereby influencing the activities of MST1/2 (mammalian Ste20-like serine/threonine kinase 1/2) kinases, the human Hippo orthologues. Moreover, WW domains are present in several Hippo factors, and these domains also serve as interaction surfaces for regulatory PPIs in Hippo signalling. Finally, the kinase activities of LATS1/2 (large tumour-suppressor kinase 1/2), the human counterparts of Warts, are controlled by binding to hMOB1 (human Mps one binder protein 1), the human Mats. Therefore Hippo signalling is regulated by PPIs on several levels. In the present paper, I review the current understanding of how these regulatory PPIs are regulated and contribute to the functionality of Hippo signalling.


Subject(s)
Drosophila Proteins/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Nerve Tissue Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Adaptor Proteins, Signal Transducing/metabolism , Animals , Humans , Neoplasms/metabolism , Protein Binding , Protein Interaction Domains and Motifs , Protein Interaction Maps
9.
Nat Cell Biol ; 5(1): 64-70, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12510195

ABSTRACT

Von Hippel-Lindau (VHL) tumour suppressor gene inactivation is linked to the development of haemangioblastomas in the central nervous system and retina, often in association with other tumours, such as clear-cell carcinomas of the kidney and phaeochromocytomas. Here we show that the VHL protein (pVHL) is a microtubule-associated protein that can protect microtubules from depolymerization in vivo. Both the microtubule binding and stabilization functions of pVHL depend on amino acids 95-123 of pVHL, a mutational 'hot-spot' in VHL disease. From analysis of naturally occurring pVHL mutants, it seems that only point mutations such as pVHL(Y98H) and pVHL(Y112H) (that predispose to haemangioblastoma and phaeochromocytoma, but not to renal cell carcinoma) disrupt pVHL's microtubule-stabilizing function. Our data identify a role for pVHL in the regulation of microtubule dynamics and potentially provide a link between this function of pVHL and the pathogenesis of haemangioblastoma and phaeochromocytoma in the context of VHL disease.


Subject(s)
Genes, Tumor Suppressor , Ligases/metabolism , Microtubules/physiology , Microtubules/ultrastructure , Tumor Suppressor Proteins , Ubiquitin-Protein Ligases , von Hippel-Lindau Disease/genetics , Animals , COS Cells , Chlorocebus aethiops , HeLa Cells , Humans , Kinetics , Ligases/genetics , Microscopy, Fluorescence , Nocodazole/pharmacology , Recombinant Proteins/metabolism , Transfection , Von Hippel-Lindau Tumor Suppressor Protein
10.
Cell Signal ; 87: 110106, 2021 11.
Article in English | MEDLINE | ID: mdl-34363951

ABSTRACT

Monopolar spindle-one binder (MOBs) proteins are evolutionarily conserved and contribute to various cellular signalling pathways. Recently, we reported that hMOB2 functions in preventing the accumulation of endogenous DNA damage and a subsequent p53/p21-dependent G1/S cell cycle arrest in untransformed cells. However, the question of how hMOB2 protects cells from endogenous DNA damage accumulation remained enigmatic. Here, we uncover hMOB2 as a regulator of double-strand break (DSB) repair by homologous recombination (HR). hMOB2 supports the phosphorylation and accumulation of the RAD51 recombinase on resected single-strand DNA (ssDNA) overhangs. Physiologically, hMOB2 expression supports cancer cell survival in response to DSB-inducing anti-cancer compounds. Specifically, loss of hMOB2 renders ovarian and other cancer cells more vulnerable to FDA-approved PARP inhibitors. Reduced MOB2 expression correlates with increased overall survival in patients suffering from ovarian carcinoma. Taken together, our findings suggest that hMOB2 expression may serve as a candidate stratification biomarker of patients for HR-deficiency targeted cancer therapies, such as PARP inhibitor treatments.


Subject(s)
Neoplasms , Poly(ADP-ribose) Polymerase Inhibitors , Cell Line, Tumor , DNA Damage , DNA Repair , Homologous Recombination , Humans , Neoplasms/drug therapy , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors/therapeutic use
11.
PLoS One ; 16(8): e0254697, 2021.
Article in English | MEDLINE | ID: mdl-34424918

ABSTRACT

The PAF complex (PAFC) coordinates transcription elongation and mRNA processing and its CDC73/parafibromin subunit functions as a tumour suppressor. The NF2/Merlin tumour suppressor functions both at the cell cortex and nucleus and is a key mediator of contact inhibition but the molecular mechanisms remain unclear. In this study we have used affinity proteomics to identify novel Merlin interacting proteins and show that Merlin forms a complex with multiple proteins involved in RNA processing including the PAFC and the CHD1 chromatin remodeller. Tumour-derived inactivating mutations in both Merlin and the CDC73 PAFC subunit mutually disrupt their interaction and growth suppression by Merlin requires CDC73. Merlin interacts with the PAFC in a cell density-dependent manner and we identify a role for FAT cadherins in regulating the Merlin-PAFC interaction. Our results suggest that in addition to its function within the Hippo pathway, Merlin is part of a tumour suppressor network regulated by cell-cell adhesion which coordinates post-initiation steps of the transcription cycle of genes mediating contact inhibition.


Subject(s)
Cell Adhesion/genetics , DNA Helicases/genetics , DNA-Binding Proteins/genetics , Neoplasms/genetics , Neurofibromin 2/genetics , Tumor Suppressor Proteins/genetics , Cell Proliferation/genetics , Chromatin/genetics , Chromatin Assembly and Disassembly/genetics , Contact Inhibition/genetics , Gene Expression Regulation/genetics , HEK293 Cells , Humans , Neoplasms/pathology , Protein Binding/genetics , Protein Interaction Maps/genetics , Signal Transduction/genetics
12.
Biochim Biophys Acta ; 1784(1): 3-15, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17881309

ABSTRACT

The NDR (nuclear Dbf2-related) family of kinases is highly conserved from yeast to human, and has been classified as a subgroup of the AGC group of protein kinases based on the sequence of the catalytic domain. Like all other members of the AGC class of protein kinases, NDR kinases require the phosphorylation of conserved Ser/Thr residues for activation. Importantly, NDR family members have two unique stretches of primary sequence: an N-terminal regulatory (NTR) domain and an insert of several residues between subdomains VII and VIII of the kinase domain. The kinase domain insert functions as an auto-inhibitory sequence (AIS), while binding of the co-activator MOB (Mps-one binder) proteins to the NTR domain releases NDR kinases from inhibition of autophosphorylation. However, despite such advances in our understanding of the molecular activation mechanism(s) and physiological functions of NDR kinases in yeast and invertebrates, most biological NDR substrates still remain to be identified. Nevertheless, by showing that the centrosomal subpopulation of human NDR1/2 is required for proper centrosome duplication, the first biological role of human NDR1/2 kinases has been defined recently. How far NDR-driven centrosome overduplication could actually contribute to cellular transformation will also be discussed.


Subject(s)
Centrosome/physiology , Protein Serine-Threonine Kinases/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Cycle/physiology , Humans , Phosphorylation , Protein Serine-Threonine Kinases/chemistry , Signal Transduction
13.
Cell Signal ; 20(8): 1564-77, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18555663

ABSTRACT

The nuclear Dbf2-related protein kinases 1 and 2 (NDR1/2) are closely-related AGC family kinases that are strongly conserved through evolution. In mammals, they are activated inter alia by phosphorylation of an hydrophobic domain threonine-residue [NDR1(Thr-444)/NDR2(Thr-442)] by an extrinsic protein kinase followed by autophosphorylation of a catalytic domain serine-residue [NDR1(Ser-281)/NDR2(Ser-282)]. We examined NDR1/2 expression and regulation in primary cultures of neonatal rat cardiac myocytes and in perfused adult rat hearts. In myocytes, transcripts for NDR2, but not NDR1, were induced by the hypertrophic agonist, endothelin-1. NDR1(Thr-444) and NDR2(Thr-442) were rapidly phosphorylated (maximal in 15-30 min) in myocytes exposed to some phosphoprotein Ser-/Thr-phosphatase 1/2 inhibitors (calyculin A, okadaic acid) and, to a lesser extent, by hyperosmotic shock, low concentrations of H(2)O(2), or chelerythrine. In myocytes adenovirally-transduced to express FLAG-NDR2 (which exhibited a mainly-cytoplasmic localisation), the same agents increased FLAG-NDR2 activity as assessed by in vitro protein kinase assays, indicative of FLAG-NDR2(Ser-282/Thr-442) phosphorylation. Calyculin A-induced phosphorylation of NDR1(Thr-444)/NDR2(Thr-442) and activation of FLAG-NDR2 were inhibited by staurosporine, but not by other protein kinase inhibitors tested. In ex vivo rat hearts, NDR1(Thr-444)/NDR2(Thr-442) were phosphorylated in response to ischaemia-reperfusion or calyculin A. From a pathological viewpoint, we conclude that activities of NDR1 and NDR2 are responsive to cytotoxic stresses in heart preparations and this may represent a previously-unidentified response to myocardial ischaemia in vivo.


Subject(s)
Enzyme Inhibitors/pharmacology , Myocardium/enzymology , Myocytes, Cardiac/enzymology , Phosphoprotein Phosphatases/antagonists & inhibitors , Protein Serine-Threonine Kinases/metabolism , Animals , Cells, Cultured , Cloning, Molecular , Enzyme Activation , Female , Marine Toxins , Myocardial Reperfusion Injury/enzymology , Osmotic Pressure , Oxazoles/pharmacology , Oxidative Stress , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/genetics , Rats , Threonine/metabolism
14.
Mol Cell Biol ; 26(15): 5784-96, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16847331

ABSTRACT

Inactivation of the von Hippel-Lindau (VHL) tumor suppressor gene is linked to the development of tumors of the eyes, kidneys, and central nervous system. VHL encodes two gene products, pVHL30 and pVHL19, of which one, pVHL30, associates functionally with microtubules (MTs) to regulate their stability. Here we report that pVHL30 is a novel substrate of glycogen synthase kinase 3 (GSK3) in vitro and in vivo. Phosphorylation of pVHL on serine 68 (S68) by GSK3 requires a priming phosphorylation event at serine 72 (S72) mediated in vitro by casein kinase I. Functional analysis of pVHL species carrying nonphosphorylatable or phosphomimicking mutations at S68 and/or S72 reveals a central role for these phosphorylation events in the regulation of pVHL's MT stabilization (but not binding) activity. Taken together, our results identify pVHL as a novel priming-dependent substrate of GSK3 and suggest a dual-kinase mechanism in the control of pVHL's MT stabilization function. Since GSK3 is a component of multiple signaling pathways that are altered in human cancer, our results further imply that normal operation of the GSK3-pVHL axis may be a critical aspect of pVHL's tumor suppressor mechanism through the regulation of MT dynamics.


Subject(s)
Glycogen Synthase Kinase 3/metabolism , Peptide Fragments/metabolism , Protein Isoforms/metabolism , Von Hippel-Lindau Tumor Suppressor Protein/metabolism , Amino Acid Sequence , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Line , Glycogen Synthase Kinase 3/genetics , Humans , Mice , Microtubules/metabolism , Molecular Sequence Data , Mutation , Peptide Fragments/genetics , Phosphorylation , Protein Binding , Protein Isoforms/genetics , Sequence Alignment , Serine/metabolism , Signal Transduction/physiology , Von Hippel-Lindau Tumor Suppressor Protein/genetics
15.
Methods Mol Biol ; 1893: 305-317, 2019.
Article in English | MEDLINE | ID: mdl-30565143

ABSTRACT

The Hippo tissue growth control and regeneration pathway is a main regulator of the YAP/TAZ effectors. In this regard, the LATS/NDR serine/threonine protein kinases can function as central components of the Hippo core module. More specifically, LATS/NDR-mediated phosphorylation of YAP/TAZ on different residues can regulate the subcellular localization and/or stability of YAP/TAZ. Therefore, the assessment of LATS/NDR activities can serve as readout for the activity status of the Hippo pathway. Here, we describe our preferred methodology regarding the measurement of the activities of LATS/NDR kinases.


Subject(s)
Enzyme Assays , Protein Serine-Threonine Kinases/metabolism , Enzyme Activation , Enzyme Assays/methods , Immunoprecipitation
16.
Cells ; 8(6)2019 06 10.
Article in English | MEDLINE | ID: mdl-31185650

ABSTRACT

The family of MOBs (monopolar spindle-one-binder proteins) is highly conserved in the eukaryotic kingdom. MOBs represent globular scaffold proteins without any known enzymatic activities. They can act as signal transducers in essential intracellular pathways. MOBs have diverse cancer-associated cellular functions through regulatory interactions with members of the NDR/LATS kinase family. By forming additional complexes with serine/threonine protein kinases of the germinal centre kinase families, other enzymes and scaffolding factors, MOBs appear to be linked to an even broader disease spectrum. Here, we review our current understanding of this emerging protein family, with emphases on post-translational modifications, protein-protein interactions, and cellular processes that are possibly linked to cancer and other diseases. In particular, we summarise the roles of MOBs as core components of the Hippo tissue growth and regeneration pathway.


Subject(s)
Neoplasms/pathology , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Animals , Apoptosis , Cell Cycle Checkpoints , DNA Repair , Humans , Neoplasms/metabolism , Protein Interaction Maps , Protein Processing, Post-Translational , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/classification
17.
Methods Mol Biol ; 1893: 319-331, 2019.
Article in English | MEDLINE | ID: mdl-30565144

ABSTRACT

The Hippo tumor suppressor pathway is fundamental to the coordination of death, growth, proliferation, and differentiation on the cellular level. At the molecular level, a highly conserved Hippo core cassette is central for the regulation of effector activities such as the co-transcriptional activity of YAP. In particular, the mammalian MST1/2 serine/threonine protein kinases (termed Hippo kinase in Drosophila melanogaster) can act as central signal transducers as part of the Hippo core cassette. In this chapter we describe in vitro kinase assays using recombinant MST1/2 kinases and recombinant MST1/2 kinase substrate.


Subject(s)
Enzyme Assays , Protein Serine-Threonine Kinases/metabolism , Recombinant Fusion Proteins/metabolism , Blotting, Western , Enzyme Activation
18.
Biochim Biophys Acta Mol Cell Res ; 1866(10): 1556-1566, 2019 10.
Article in English | MEDLINE | ID: mdl-31326538

ABSTRACT

Chaperone-assisted selective autophagy (CASA) initiated by the cochaperone Bcl2-associated athanogene 3 (BAG3) represents an important mechanism for the disposal of misfolded and damaged proteins in mammalian cells. Under mechanical stress, the cochaperone cooperates with the small heat shock protein HSPB8 and the cytoskeleton-associated protein SYNPO2 to degrade force-unfolded forms of the actin-crosslinking protein filamin. This is essential for muscle maintenance in flies, fish, mice and men. Here, we identify the serine/threonine protein kinase 38 (STK38), which is part of the Hippo signaling network, as a novel interactor of BAG3. STK38 was previously shown to facilitate cytoskeleton assembly and to promote mitophagy as well as starvation and detachment induced autophagy. Significantly, our study reveals that STK38 exerts an inhibitory activity on BAG3-mediated autophagy. Inhibition relies on a disruption of the functional interplay of BAG3 with HSPB8 and SYNPO2 upon binding of STK38 to the cochaperone. Of note, STK38 attenuates CASA independently of its kinase activity, whereas previously established regulatory functions of STK38 involve target phosphorylation. The ability to exert different modes of regulation on central protein homeostasis (proteostasis) machineries apparently allows STK38 to coordinate the execution of diverse macroautophagy pathways and to balance cytoskeleton assembly and degradation.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Apoptosis Regulatory Proteins/metabolism , Autophagy/physiology , Protein Serine-Threonine Kinases/metabolism , Proteostasis/physiology , Adaptor Proteins, Signal Transducing/genetics , Apoptosis Regulatory Proteins/genetics , Cytoskeleton/metabolism , Filamins/metabolism , HEK293 Cells , HeLa Cells , Heat-Shock Proteins/metabolism , Humans , Microfilament Proteins , Mitophagy , Molecular Chaperones/metabolism , Protein Binding , Proteomics , Signal Transduction , Stress, Mechanical
19.
J Exp Clin Cancer Res ; 38(1): 158, 2019 Apr 12.
Article in English | MEDLINE | ID: mdl-30979377

ABSTRACT

BACKGROUND: RASSF1A, a tumor suppressor gene, is frequently inactivated in lung cancer leading to a YAP-dependent epithelial-mesenchymal transition (EMT). Such effects are partly due to the inactivation of the anti-migratory RhoB GTPase via the inhibitory phosphorylation of GEF-H1, the GDP/GTP exchange factor for RhoB. However, the kinase responsible for RhoB/GEF-H1 inactivation in RASSF1A-depleted cells remained unknown. METHODS: NDR1/2 inactivation by siRNA or shRNA effects on epithelial-mesenchymal transition, invasion, xenograft formation and growth in SCID-/- Beige mice, apoptosis, proliferation, cytokinesis, YAP/TAZ activation were investigated upon RASSF1A loss in human bronchial epithelial cells (HBEC). RESULTS: We demonstrate here that depletion of the YAP-kinases NDR1/2 reverts migration and metastatic properties upon RASSF1A loss in HBEC. We show that NDR2 interacts directly with GEF-H1 (which contains the NDR phosphorylation consensus motif HXRXXS/T), leading to GEF-H1 phosphorylation. We further report that the RASSF1A/NDR2/GEF-H1/RhoB/YAP axis is involved in proper cytokinesis in human bronchial cells, since chromosome proper segregation are NDR-dependent upon RASSF1A or GEF-H1 loss in HBEC. CONCLUSION: To summarize, our data support a model in which, upon RASSF1A silencing, NDR2 gets activated, phosphorylates and inactivates GEF-H1, leading to RhoB inactivation. This cascade induced by RASSF1A loss in bronchial cells is responsible for metastasis properties, YAP activation and cytokinesis defects.


Subject(s)
Cell Movement/genetics , Cytokinesis/genetics , Gene Silencing , Genes, Suppressor , Protein Serine-Threonine Kinases/metabolism , Tumor Suppressor Proteins/genetics , Animals , Biomarkers, Tumor , Cell Cycle Proteins , Cell Line, Tumor , Disease Models, Animal , Epithelial-Mesenchymal Transition/genetics , Heterografts , Humans , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/mortality , Lung Neoplasms/pathology , Mice , Neoplasm Metastasis , Nuclear Proteins/metabolism , Phenotype , Phosphorylation , Prognosis , Transcription Factors/metabolism , rhoB GTP-Binding Protein/metabolism
20.
Nat Commun ; 10(1): 5755, 2019 12 17.
Article in English | MEDLINE | ID: mdl-31848340

ABSTRACT

Autophagy perturbation represents an emerging therapeutic strategy in cancer. Although LATS1 and LATS2 kinases, core components of the mammalian Hippo pathway, have been shown to exert tumor suppressive activities, here we report a pro-survival role of LATS1 but not LATS2 in hepatocellular carcinoma (HCC) cells. Specifically, LATS1 restricts lethal autophagy in HCC cells induced by sorafenib, the standard of care for advanced HCC patients. Notably, autophagy regulation by LATS1 is independent of its kinase activity. Instead, LATS1 stabilizes the autophagy core-machinery component Beclin-1 by promoting K27-linked ubiquitination at lysine residues K32 and K263 on Beclin-1. Consequently, ubiquitination of Beclin-1 negatively regulates autophagy by promoting inactive dimer formation of Beclin-1. Our study highlights a functional diversity between LATS1 and LATS2, and uncovers a scaffolding role of LATS1 in mediating a cross-talk between the Hippo signaling pathway and autophagy.


Subject(s)
Autophagy/immunology , Carcinoma, Hepatocellular/pathology , Cell Survival/immunology , Liver Neoplasms/pathology , Protein Serine-Threonine Kinases/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Autophagy/drug effects , Beclin-1/metabolism , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/immunology , Carcinoma, Hepatocellular/mortality , Cell Line, Tumor , Cell Proliferation , Cell Survival/drug effects , Datasets as Topic , Disease-Free Survival , Drug Resistance, Neoplasm/immunology , Hippo Signaling Pathway , Humans , Kaplan-Meier Estimate , Liver/pathology , Liver Neoplasms/drug therapy , Liver Neoplasms/immunology , Liver Neoplasms/mortality , Lysine/metabolism , Mice , Mice, Knockout , Organoids , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/immunology , Protein Stability , Signal Transduction/drug effects , Signal Transduction/immunology , Sorafenib/pharmacology , Sorafenib/therapeutic use , Tumor Suppressor Proteins/immunology , Ubiquitination , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL