Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Cell Mol Life Sci ; 78(5): 1943-1955, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33145612

ABSTRACT

The Hippo signal transduction pathway is an essential regulator of organ size during developmental growth by controlling multiple cellular processes such as cell proliferation, cell death, differentiation, and stemness. Dysfunctional Hippo signaling pathway leads to dramatic tissue overgrowth. Here, we will briefly introduce the Hippo tumor suppressor pathway before focusing on one of its members and the unexpected twists that followed our quest of its functions in its multifarious actions beside the Hippo pathway: the STK38 kinase. In this review, we will precisely discuss the newly identified role of STK38 on regulating the nuclear export machinery by phosphorylating and activating, the major nuclear export receptor XPO1. Finally, we will phrase STK38's role on regulating the subcellular distribution of crucial cellular regulators such as Beclin1 and YAP1 with its implication in cancer.


Subject(s)
Karyopherins/metabolism , Neoplasms/metabolism , Protein Serine-Threonine Kinases/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Signal Transduction , Active Transport, Cell Nucleus , Animals , Beclin-1/metabolism , Cell Nucleus/metabolism , Humans , Phosphorylation , Exportin 1 Protein
2.
Cell Cycle ; 19(22): 2982-2995, 2020 11.
Article in English | MEDLINE | ID: mdl-33017560

ABSTRACT

The proper segregation of basic elements such as the compartmentalization of the genome and the shuttling of macromolecules between the nucleus and the cytoplasm is a crucial mechanism for homeostasis maintenance in eukaryotic cells. XPO1 (Exportin 1) is the major nuclear export receptor and is required for the export of proteins and RNAs out of the nucleus. STK38 (also known as NDR1) is a Hippo pathway serine/threonine kinase with multifarious functions in normal and cancer cells. In this review, we summarize the history of the discovery of the nucleo/cytoplasmic shuttling of proteins and focus on the major actor of nuclear export: XPO1. After describing the molecular events required for XPO1-mediated nuclear export of proteins, we introduce the Hippo pathway STK38 kinase, synthetize its regulation mechanisms as well as its biological functions in both normal and cancer cells, and finally its intersection with XPO1 biology. We discuss the recently identified mechanism of XPO1 activation by phosphorylation of XPO1_S1055 by STK38 and contextualize this finding according to the biological functions previously reported for both XPO1 and STK38, including the second identity of STK38 as an autophagy regulator. Finally, we phrase this newly identified activation mechanism into the general nuclear export machinery and examine the possible outcomes of nuclear export inhibition in cancer treatment.


Subject(s)
Cell Nucleus/metabolism , Hippo Signaling Pathway/metabolism , Karyopherins/metabolism , Neoplasms/metabolism , Protein Serine-Threonine Kinases/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Signal Transduction , Active Transport, Cell Nucleus , Autophagy , Cytoplasm/metabolism , Humans , Karyopherins/antagonists & inhibitors , Karyopherins/chemistry , Molecular Targeted Therapy/methods , Neoplasms/drug therapy , Phosphorylation , Protein Binding , Protein Domains , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/chemistry , Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors , Receptors, Cytoplasmic and Nuclear/chemistry , Tandem Repeat Sequences , Exportin 1 Protein
3.
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
5.
Oncotarget ; 7(28): 44142-44160, 2016 Jul 12.
Article in English | MEDLINE | ID: mdl-27283898

ABSTRACT

Oncogenic Ras signalling occurs frequently in many human cancers. However, no effective targeted therapies are currently available to treat patients suffering from Ras-driven tumours. Therefore, it is imperative to identify downstream effectors of Ras signalling that potentially represent promising new therapeutic options. Particularly, considering that autophagy inhibition can impair the survival of Ras-transformed cells in tissue culture and mouse models, an understanding of factors regulating the balance between autophagy and apoptosis in Ras-transformed human cells is needed. Here, we report critical roles of the STK38 protein kinase in oncogenic Ras transformation. STK38 knockdown impaired anoikis resistance, anchorage-independent soft agar growth, and in vivo xenograft growth of Ras-transformed human cells. Mechanistically, STK38 supports Ras-driven transformation through promoting detachment-induced autophagy. Even more importantly, upon cell detachment STK38 is required to sustain the removal of damaged mitochondria by mitophagy, a selective autophagic process, to prevent excessive mitochondrial reactive oxygen species production that can negatively affect cancer cell survival. Significantly, knockdown of PINK1 or Parkin, two positive regulators of mitophagy, also impaired anoikis resistance and anchorage-independent growth of Ras-transformed human cells, while knockdown of USP30, a negative regulator of PINK1/Parkin-mediated mitophagy, restored anchorage-independent growth of STK38-depleted Ras-transformed human cells. Therefore, our findings collectively reveal novel molecular players that determine whether Ras-transformed human cells die or survive upon cell detachment, which potentially could be exploited for the development of novel strategies to target Ras-transformed cells.


Subject(s)
Cell Transformation, Neoplastic/genetics , Mitophagy/genetics , Protein Serine-Threonine Kinases/genetics , ras Proteins/genetics , Animals , Anoikis/genetics , Apoptosis/genetics , Autophagy/genetics , Cell Line , Cell Line, Tumor , HCT116 Cells , HEK293 Cells , Humans , Mice, Nude , Protein Serine-Threonine Kinases/metabolism , RNA Interference , Transplantation, Heterologous , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , ras Proteins/metabolism
6.
Cell ; 144(2): 253-67, 2011 Jan 21.
Article in English | MEDLINE | ID: mdl-21241894

ABSTRACT

The study of macroautophagy in mammalian cells has described induction, vesicle nucleation, and membrane elongation complexes as key signaling intermediates driving autophagosome biogenesis. How these components are recruited to nascent autophagosomes is poorly understood, and although much is known about signaling mechanisms that restrain autophagy, the nature of positive inductive signals that can promote autophagy remain cryptic. We find that the Ras-like small G protein, RalB, is localized to nascent autophagosomes and is activated on nutrient deprivation. RalB and its effector Exo84 are required for nutrient starvation-induced autophagocytosis, and RalB activation is sufficient to promote autophagosome formation. Through direct binding to Exo84, RalB induces the assembly of catalytically active ULK1 and Beclin1-VPS34 complexes on the exocyst, which are required for isolation membrane formation and maturation. Thus, RalB signaling is a primary adaptive response to nutrient limitation that directly engages autophagocytosis through mobilization of the core vesicle nucleation machinery.


Subject(s)
Autophagy , Epithelial Cells/pathology , Phagosomes/metabolism , Signal Transduction , ral GTP-Binding Proteins/metabolism , Apoptosis Regulatory Proteins/metabolism , Beclin-1 , Cell Line , Class III Phosphatidylinositol 3-Kinases/metabolism , Epithelial Cells/microbiology , Humans , Membrane Proteins/metabolism , Multiprotein Complexes/metabolism , Salmonella typhimurium/physiology , Stress, Physiological , Vesicular Transport Proteins/metabolism
7.
Neurobiol Dis ; 41(1): 33-42, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20732423

ABSTRACT

In spinocerebellar ataxia-7 (SCA7), a polyglutamine (polyQ) expansion in the ataxin-7 protein leads to the formation of neuronal intranuclear inclusions (NIIs) and neurodegeneration. In this study, amyloid precursor-like protein 2 (APLP2) was identified as a partner protein for ataxin-7. APLP2, belonging to the APP gene family, undergoes secretase and caspase cleavages and has been implicated in the pathogenesis of Alzheimer's disease (AD). Activated caspase-3 cleaves APP family proteins to release N-terminal fragments (NTFs) and intracellular C-terminal domains (ICDs), which can translocate into the nucleus and induce neurotoxicity in AD. Here, we report abnormal nuclear relocation of APLP2 and detection of NTFs in NIIs in SCA7. The ICDs generated by caspase-3 cleavage of APLP2 accumulate in nuclei and contribute to a cumulative toxicity when coexpressed with mutated ataxin-7. Our data suggest that the interaction between APLP2 and ataxin-7 and proteolytic processing of APLP2 may contribute to the pathogenesis of SCA7.


Subject(s)
Amyloid beta-Protein Precursor/metabolism , Intranuclear Inclusion Bodies/metabolism , Nerve Tissue Proteins/metabolism , Peptide Fragments/metabolism , Spinocerebellar Ataxias/metabolism , Adult , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/toxicity , Animals , Ataxin-7 , Child , Humans , Intranuclear Inclusion Bodies/pathology , Mice , Mice, Transgenic , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/toxicity , Neurofibrillary Tangles/metabolism , Neurofibrillary Tangles/pathology , PC12 Cells , Peptide Fragments/genetics , Peptide Fragments/toxicity , Protein Processing, Post-Translational/genetics , Rats , Spinocerebellar Ataxias/etiology , Spinocerebellar Ataxias/pathology
8.
J Biol Chem ; 285(43): 33073-33081, 2010 Oct 22.
Article in English | MEDLINE | ID: mdl-20709745

ABSTRACT

Abnormalities in the process of endocytosis are classically linked to malignant transformation through the deficient down-regulation of signaling receptors. The present study describes a non-classical mechanism that does not require internalization by which endocytic proteins affect cell migration and basement membrane invasion. Specifically, we found that the endocytic adaptor epsin binds and regulates the biological properties of the signaling molecule RalBP1 (Ral-binding protein 1). Epsin interacted with the N terminus of RalBP1 via its characteristic epsin N-terminal homology (ENTH) domain. A combination of siRNA-mediated knock-down and transfection of siRNA-resistant constructs in fibrosarcoma cells demonstrated that impairment of the epsin-RalBP1 interaction led to cell migration and basement membrane invasion defects. We found the ENTH domain was necessary and sufficient to sustain normal cell migration and invasion. Because all the epsin endocytic motifs reside in the C-terminal part of the molecule, these results suggest that this novel regulatory circuit does not require endocytosis. In addition, cells depleted of epsin-RalBP1 complex displayed deficient activation of Rac1 and Arf6 suggesting a signaling function for this novel interaction. Further, overexpression of either epsin or RalBP1 enhanced migration and invasion of fibrosarcoma cells. Collectively, our results indicate that epsin regulates RalBP1 function in Rac1- and Arf6-dependent pathways to ultimately affect cell migration and invasion. We propose that the observed up-regulation of both epsin and RalBP1 in certain cancers contributes to their invasive characteristics.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Adaptor Proteins, Vesicular Transport/metabolism , Cell Movement , Fibrosarcoma/metabolism , GTPase-Activating Proteins/metabolism , ADP-Ribosylation Factor 6 , ADP-Ribosylation Factors/genetics , ADP-Ribosylation Factors/metabolism , ATP-Binding Cassette Transporters/genetics , Adaptor Proteins, Vesicular Transport/genetics , Amino Acid Motifs , Animals , Basement Membrane/metabolism , Basement Membrane/pathology , Cell Line, Tumor , Endocytosis/genetics , Fibrosarcoma/genetics , Fibrosarcoma/pathology , GTPase-Activating Proteins/genetics , Humans , Mice , NIH 3T3 Cells , Neoplasm Invasiveness/genetics , Neuropeptides/genetics , Neuropeptides/metabolism , Protein Structure, Tertiary , rac GTP-Binding Proteins/genetics , rac GTP-Binding Proteins/metabolism , rac1 GTP-Binding Protein/genetics , rac1 GTP-Binding Protein/metabolism
9.
PLoS Biol ; 7(11): e1000235, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19885391

ABSTRACT

Atypical protein kinase C (aPKC) isoforms have been implicated in cell polarisation and migration through association with Cdc42 and Par6. In distinct migratory models, the Exocyst complex has been shown to be involved in secretory events and migration. By RNA interference (RNAi) we show that the polarised delivery of the Exocyst to the leading edge of migrating NRK cells is dependent upon aPKCs. Reciprocally we demonstrate that aPKC localisation at the leading edge is dependent upon the Exocyst. The basis of this inter-dependence derives from two-hybrid, mass spectrometry, and co-immunoprecipitation studies, which demonstrate the existence of an aPKC-Exocyst interaction mediated by Kibra. Using RNAi and small molecule inhibitors, the aPKCs, Kibra, and the Exocyst are shown to be required for NRK cell migration and it is further demonstrated that they are necessary for the localized activation of JNK at the leading edge. The migration associated control of JNK by aPKCs determines JNK phosphorylation of the plasma membrane substrate Paxillin, but not the phosphorylation of the nuclear JNK substrate, c-jun. This plasma membrane localized JNK cascade serves to control the stability of focal adhesion complexes, regulating migration. The study integrates the polarising behaviour of aPKCs with the pro-migratory properties of the Exocyst complex, defining a higher order complex associated with the localised activation of JNK at the leading edge of migrating cells that determines migration rate.


Subject(s)
Cell Movement , Mitogen-Activated Protein Kinase 8/metabolism , Paxillin/metabolism , Protein Kinase C/metabolism , Animals , Blotting, Western , Carbazoles/pharmacology , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Membrane/metabolism , Cells, Cultured , Enzyme Activation , Enzyme Inhibitors/pharmacology , Exocytosis , Focal Adhesions/metabolism , Intracellular Signaling Peptides and Proteins , Membrane Proteins , Microscopy, Confocal , Microtubules/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase Kinases/metabolism , Phosphoproteins , Phosphorylation , Protein Binding , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/genetics , Proteins/genetics , Proteins/metabolism , RNA Interference , Rats , Two-Hybrid System Techniques
10.
Cell ; 127(1): 157-70, 2006 Oct 06.
Article in English | MEDLINE | ID: mdl-17018283

ABSTRACT

The monomeric RalGTPases, RalA and RalB are recognized as components of a regulatory framework supporting tumorigenic transformation. Specifically, RalB is required to suppress apoptotic checkpoint activation, the mechanistic basis of which is unknown. Reported effector proteins of RalB include the Sec5 component of the exocyst, an octameric protein complex implicated in tethering of vesicles to membranes. Surprisingly, we find that the RalB/Sec5 effector complex directly recruits and activates the atypical IkappaB kinase family member TBK1. In cancer cells, constitutive engagement of this pathway, via chronic RalB activation, restricts initiation of apoptotic programs typically engaged in the context of oncogenic stress. Although dispensable for survival in a nontumorigenic context, this pathway helps mount an innate immune response to virus exposure. These observations define the mechanistic contribution of RalGTPases to cancer cell survival and reveal the RalB/Sec5 effector complex as a component of TBK1-dependent innate immune signaling.


Subject(s)
Carrier Proteins/metabolism , Cell Survival , Immunity, Innate/physiology , Neoplasms/metabolism , Protein Serine-Threonine Kinases/metabolism , Signal Transduction/physiology , ral GTP-Binding Proteins/metabolism , Animals , Apoptosis/physiology , Carrier Proteins/genetics , Cell Transformation, Neoplastic , Enzyme Activation , HeLa Cells , Humans , Mice , Mice, Knockout , Multiprotein Complexes , Protein Serine-Threonine Kinases/genetics , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Vesicular Transport Proteins , ral GTP-Binding Proteins/genetics
11.
Trends Cell Biol ; 15(6): 327-32, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15953551

ABSTRACT

The Ras-like small G-proteins RalA and RalB have achieved some notoriety as components of one of a growing variety of candidate Ras effector pathways. Recent work has demonstrated that Ral GTPase activation is required to support both the initiation and maintenance of tumorigenic transformation of human cells. The mechanistic basis for this support remains to be defined. However, the discovery that the exocyst is a direct effector complex for activated Ral proteins suggests that mobilization of polarized exocytosis might be a basic component of the biological framework supporting tumorigenic progression.


Subject(s)
Exocytosis , Membrane Proteins/metabolism , Neoplasms/metabolism , Neoplasms/pathology , ral GTP-Binding Proteins/metabolism , Animals , Cell Transformation, Neoplastic , Humans , Protein Binding
12.
J Biol Chem ; 278(19): 17053-9, 2003 May 09.
Article in English | MEDLINE | ID: mdl-12624092

ABSTRACT

The exocyst complex is involved in the final stages of exocytosis, when vesicles are targeted to the plasma membrane and dock. The regulation of exocytosis is vital for a number of processes, for example, cell polarity, embryogenesis, and neuronal growth formation. Regulation of the exocyst complex in mammals was recently shown to be dependent upon binding of the small G protein, Ral, to Sec5, a central component of the exocyst. This interaction is thought to be necessary for anchoring the exocyst to secretory vesicles. We have determined the structure of the Ral-binding domain of Sec5 and shown that it adopts a fold that has not been observed in a G protein effector before. This fold belongs to the immunoglobulin superfamily in a subclass known as IPT domains. We have mapped the Ral binding site on this domain and found that it overlaps with protein-protein interaction sites on other IPT domains but that it is completely different from the G protein-geranyl-geranyl interaction face of the Ig-like domain of the Rho guanine nucleotide dissociation inhibitor. This mapping, along with available site-directed mutagenesis data, allows us to predict how Ral and Sec5 may interact.


Subject(s)
GTP Phosphohydrolases/metabolism , Membrane Proteins/analysis , ral GTP-Binding Proteins/metabolism , Amino Acid Sequence , Animals , Binding Sites , Escherichia coli , GTP Phosphohydrolases/chemistry , Membrane Proteins/chemistry , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Molecular Sequence Data , Protein Binding , Protein Conformation , Protein Structure, Tertiary , Sequence Alignment , Vesicular Transport Proteins , ral GTP-Binding Proteins/chemistry
13.
Mol Cell Biol ; 22(10): 3527-36, 2002 May.
Article in English | MEDLINE | ID: mdl-11971983

ABSTRACT

Spectrins, components of the membrane skeleton, are implicated in various cellular functions. Understanding the diversity of these functions requires better characterization of the interacting domains of spectrins, such as the SH3 domain. Yeast two-hybrid screening of a kidney cDNA library revealed that the SH3 domain of alpha II-spectrin binds specifically isoform A of low-molecular-weight phosphotyrosine phosphatase (LMW-PTP). The alpha II-spectrin SH3 domain does not interact with LMW-PTP B or C nor does LMW-PTP A interact with the alpha I-spectrin SH3 domain. The interaction of spectrin with LMW-PTP A led us to look for a tyrosine-phosphorylated residue in alpha II-spectrin. Western blotting showed that alpha II-spectrin is tyrosine phosphorylated in vivo. Using mutagenesis on recombinant peptides, we identified the residue Y1176 located in the calpain cleavage site of alpha II-spectrin, near the SH3 domain, as an in vitro substrate for Src kinase and LMW-PTP A. This Y1176 residue is also an in vivo target for kinases and phosphatases in COS cells. Phosphorylation of this residue decreases spectrin sensitivity to calpain in vitro. Similarly, the presence of phosphatase inhibitors in cell culture is associated with the absence of spectrin cleavage products. This suggests that the Y1176 phosphorylation state could modulate spectrin cleavage by calpain and may play an important role during membrane skeleton remodeling.


Subject(s)
Calpain/metabolism , Protein Tyrosine Phosphatases/metabolism , Spectrin/metabolism , Tyrosine/metabolism , Amino Acid Sequence , Animals , Cell Fractionation , Cell Line , Cytoskeleton/metabolism , Male , Molecular Sequence Data , Phosphorylation , Protein Isoforms , Protein Structure, Tertiary , Protein Tyrosine Phosphatases/chemistry , Protein Tyrosine Phosphatases/genetics , Rats , Rats, Sprague-Dawley , Sequence Alignment , Two-Hybrid System Techniques , Yeasts/genetics , Yeasts/metabolism
14.
Nat Cell Biol ; 4(1): 66-72, 2002 Jan.
Article in English | MEDLINE | ID: mdl-11740492

ABSTRACT

Delivery of cytoplasmic vesicles to discrete plasma-membrane domains is critical for establishing and maintaining cell polarity, neurite differentiation and regulated exocytosis. The exocyst is a multisubunit complex required for vectorial targeting of a subset of secretory vesicles. Mechanisms that regulate the activity of this complex in mammals are unknown. Here we show that Sec5, an integral component of the exocyst, is a direct target for activated Ral GTPases. Ral GTPases regulate targeting of basolateral proteins in epithelial cells, secretagogue-dependent exocytosis in neuroendocrine cells and assembly of exocyst complexes. These observations define Ral GTPases as critical regulators of vesicle trafficking.


Subject(s)
Exocytosis/physiology , GTPase-Activating Proteins , Membrane Proteins/physiology , Signal Transduction , ral GTP-Binding Proteins/physiology , Animals , Caco-2 Cells , Carrier Proteins/physiology , Cell Polarity/physiology , Fungal Proteins/physiology , HeLa Cells , Humans , PC12 Cells , Phospholipase D/physiology , Protein Transport/physiology , Rats , Secretory Vesicles/physiology , Vesicular Transport Proteins
SELECTION OF CITATIONS
SEARCH DETAIL
...