Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 121
Filter
1.
Front Immunol ; 12: 668602, 2021.
Article in English | MEDLINE | ID: mdl-34335569

ABSTRACT

Caspases are a family of cysteine proteases that play an essential role in inflammation, apoptosis, cell death, and development. Here we delve into the effects caused by heterologous expression of human caspase-1 in the yeast Saccharomyces cerevisiae and compare them to those of caspase-8. Overexpression of both caspases in the heterologous model led to their activation and caused mitochondrial hyperpolarization, damage to different organelles, and cell death. All these effects were dependent on their protease activity, and caspase-8 was more aggressive than caspase-1. Growth arrest could be at least partially explained by dysfunction of the actin cytoskeleton as a consequence of the processing of the yeast Bni1 formin, which we identify here as a likely direct substrate of both caspases. Through the modulation of the GAL1 promoter by using different galactose:glucose ratios in the culture medium, we have established a scenario in which caspase-1 is sufficiently expressed to become activated while yeast growth is not impaired. Finally, we used the yeast model to explore the role of death-fold domains (DD) of both caspases in their activity. Peculiarly, the DDs of either caspase showed an opposite involvement in its intrinsic activity, as the deletion of the caspase activation and recruitment domain (CARD) of caspase-1 enhanced its activity, whereas the deletion of the death effector domain (DED) of caspase-8 diminished it. We show that caspase-1 is able to efficiently process its target gasdermin D (GSDMD) when co-expressed in yeast. In sum, we propose that S. cerevisiae provides a manageable tool to explore caspase-1 activity and structure-function relationships.


Subject(s)
Caspase 1/biosynthesis , Caspase 8/biosynthesis , Mitochondria/enzymology , Saccharomyces cerevisiae/enzymology , Actin Cytoskeleton/enzymology , Actin Cytoskeleton/genetics , Caspase 1/genetics , Caspase 8/genetics , Enzyme Activation , Enzyme Induction , Galactokinase/genetics , Galactokinase/metabolism , Gene Expression Regulation, Fungal , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Microbial Viability , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , Mitochondria/genetics , Phosphate-Binding Proteins/genetics , Phosphate-Binding Proteins/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/growth & development , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Substrate Specificity
2.
Small GTPases ; 12(5-6): 429-439, 2021.
Article in English | MEDLINE | ID: mdl-33487105

ABSTRACT

Cell invasion is associated with numerous patho-physiologic states including cell development and metastatic dissemination. This process couples the activation of cell motility with the capacity to degrade the extracellular matrix, thereby permitting cells to pass through basal membranes. Invasion is sustained by the actions of invadosomes, an ensemble of subcellular structures with high functional homology. Invadosomes are 3D acto-adhesive structures that can also mediate local extracellular matrix degradation through the controlled delivery of proteases. Intracellular RHO GTPases play a central role in the regulation of invadosomes where their complex interplay regulates multiple invadosome functions. This review aims to provide an overview of the synergistic activities of the small GTPases in invadosome biology. This broad-based review also reinforces the importance of the spatiotemporal regulation of small GTPases and the impact of this process on invadosome dynamics.


Subject(s)
Actin Cytoskeleton/physiology , Cell Movement , Extracellular Matrix/metabolism , Monomeric GTP-Binding Proteins/metabolism , Podosomes/physiology , Actin Cytoskeleton/enzymology , Animals , Humans , Podosomes/enzymology
3.
Arterioscler Thromb Vasc Biol ; 40(10): 2346-2359, 2020 10.
Article in English | MEDLINE | ID: mdl-32787522

ABSTRACT

OBJECTIVE: AIBP (apolipoprotein A-I binding protein) is an effective and selective regulator of lipid rafts modulating many metabolic pathways originating from the rafts, including inflammation. The mechanism of action was suggested to involve stimulation by AIBP of cholesterol efflux, depleting rafts of cholesterol, which is essential for lipid raft integrity. Here we describe a different mechanism contributing to the regulation of lipid rafts by AIBP. Approach and Results: We demonstrate that modulation of rafts by AIBP may not exclusively depend on the rate of cholesterol efflux or presence of the key regulator of the efflux, ABCA1 (ATP-binding cassette transporter A-I). AIBP interacted with phosphatidylinositol 3-phosphate, which was associated with increased abundance and activation of Cdc42 and rearrangement of the actin cytoskeleton. Cytoskeleton rearrangement was accompanied with reduction of the abundance of lipid rafts, without significant changes in the lipid composition of the rafts. The interaction of AIBP with phosphatidylinositol 3-phosphate was blocked by AIBP substrate, NADPH (nicotinamide adenine dinucleotide phosphate), and both NADPH and silencing of Cdc42 interfered with the ability of AIBP to regulate lipid rafts and cholesterol efflux. CONCLUSIONS: Our findings indicate that an underlying mechanism of regulation of lipid rafts by AIBP involves PIP-dependent rearrangement of the cytoskeleton.


Subject(s)
Actin Cytoskeleton/enzymology , Cholesterol/metabolism , Membrane Microdomains/enzymology , Racemases and Epimerases/metabolism , ATP Binding Cassette Transporter 1/metabolism , Actin Cytoskeleton/genetics , Animals , HeLa Cells , Humans , Membrane Microdomains/genetics , Mice , Phosphatidylinositol 3-Kinase/metabolism , Signal Transduction , THP-1 Cells , cdc42 GTP-Binding Protein/genetics , cdc42 GTP-Binding Protein/metabolism
4.
J Cell Biol ; 219(6)2020 06 01.
Article in English | MEDLINE | ID: mdl-32311005

ABSTRACT

Microtubule-associated serine/threonine-protein kinase-like (MASTL) is a mitosis-accelerating kinase with emerging roles in cancer progression. However, possible cell cycle-independent mechanisms behind its oncogenicity remain ambiguous. Here, we identify MASTL as an activator of cell contractility and MRTF-A/SRF (myocardin-related transcription factor A/serum response factor) signaling. Depletion of MASTL increased cell spreading while reducing contractile actin stress fibers in normal and breast cancer cells and strongly impairing breast cancer cell motility and invasion. Transcriptome and proteome profiling revealed MASTL-regulated genes implicated in cell movement and actomyosin contraction, including Rho guanine nucleotide exchange factor 2 (GEF-H1, ARHGEF2) and MRTF-A target genes tropomyosin 4.2 (TPM4), vinculin (VCL), and nonmuscle myosin IIB (NM-2B, MYH10). Mechanistically, MASTL associated with MRTF-A and increased its nuclear retention and transcriptional activity. Importantly, MASTL kinase activity was not required for regulation of cell spreading or MRTF-A/SRF transcriptional activity. Taken together, we present a previously unknown kinase-independent role for MASTL as a regulator of cell adhesion, contractility, and MRTF-A/SRF activity.


Subject(s)
Actin Cytoskeleton/enzymology , Cell Adhesion/genetics , Microtubule-Associated Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Rho Guanine Nucleotide Exchange Factors/metabolism , Signal Transduction/genetics , Trans-Activators/metabolism , Actin Cytoskeleton/genetics , Actin Cytoskeleton/metabolism , Cell Cycle/genetics , Cell Line, Tumor , Cell Movement/genetics , Cell Nucleus/metabolism , Gene Expression Profiling , Humans , Integrins/genetics , Integrins/metabolism , Microtubule-Associated Proteins/deficiency , Microtubule-Associated Proteins/genetics , Nonmuscle Myosin Type IIB/genetics , Nonmuscle Myosin Type IIB/metabolism , Phosphorylation , Protein Binding , Protein Serine-Threonine Kinases/deficiency , Protein Serine-Threonine Kinases/genetics , Proteome/metabolism , RNA, Small Interfering , Rho Guanine Nucleotide Exchange Factors/genetics , Stress Fibers/genetics , Stress Fibers/metabolism , Trans-Activators/genetics , Transcriptome/genetics , Tropomyosin/genetics , Tropomyosin/metabolism , Vinculin/genetics , Vinculin/metabolism
5.
Am J Physiol Renal Physiol ; 318(6): F1377-F1390, 2020 06 01.
Article in English | MEDLINE | ID: mdl-32308020

ABSTRACT

Ste20-like kinase SLK is critical for embryonic development and may play an important role in wound healing, muscle homeostasis, cell migration, and tumor growth. Mice with podocyte-specific deletion of SLK show albuminuria and damage to podocytes as they age. The present study addressed the role of SLK in glomerular injury. We induced adriamycin nephrosis in 3- to 4-mo-old control and podocyte SLK knockout (KO) mice. Compared with control, SLK deletion exacerbated albuminuria and loss of podocytes, synaptopodin, and podocalyxin. Glomeruli of adriamycin-treated SLK KO mice showed diffuse increases in the matrix and sclerosis as well as collapse of the actin cytoskeleton. SLK can phosphorylate ezrin. The complex of phospho-ezrin, Na+/H+ exchanger regulatory factor 2, and podocalyxin in the apical domain of the podocyte is a key determinant of normal podocyte architecture. Deletion of SLK reduced glomerular ezrin and ezrin phosphorylation in adriamycin nephrosis. Also, deletion of SLK reduced the colocalization of ezrin and podocalyxin in the glomerulus. Cultured glomerular epithelial cells with KO of SLK showed reduced ezrin phosphorylation and podocalyxin expression as well as reduced F-actin. Thus, SLK deletion leads to podocyte injury as mice age and exacerbates injury in adriamycin nephrosis. The mechanism may at least in part involve ezrin phosphorylation as well as disruption of the cytoskeleton and podocyte apical membrane structure.


Subject(s)
Actin Cytoskeleton/enzymology , Doxorubicin , Glomerulosclerosis, Focal Segmental/enzymology , Nephrosis/enzymology , Podocytes/enzymology , Protein Serine-Threonine Kinases/deficiency , Actin Cytoskeleton/pathology , Actins/metabolism , Albuminuria/chemically induced , Albuminuria/enzymology , Albuminuria/genetics , Animals , Cells, Cultured , Cytoskeletal Proteins/metabolism , Disease Models, Animal , Gene Knockdown Techniques , Glomerulosclerosis, Focal Segmental/chemically induced , Glomerulosclerosis, Focal Segmental/genetics , Glomerulosclerosis, Focal Segmental/pathology , Mice, Knockout , Microfilament Proteins/metabolism , Nephrosis/chemically induced , Nephrosis/genetics , Nephrosis/pathology , Phosphoproteins/metabolism , Phosphorylation , Podocytes/pathology , Protein Serine-Threonine Kinases/genetics , Proteins/metabolism , Sodium-Hydrogen Exchangers/metabolism
6.
Exp Cell Res ; 390(2): 111961, 2020 05 15.
Article in English | MEDLINE | ID: mdl-32209306

ABSTRACT

N-alpha-acetyltransferase 80 (NAA80) was recently demonstrated to acetylate the N-terminus of actin, with NAA80 knockout cells showing actin cytoskeleton-related phenotypes, such as increased formation of membrane protrusions and accelerated migration. Here we report that NAA80 knockout cells additionally display fragmentation of the Golgi apparatus. We further employed rescue assays to demonstrate that this phenotype is connected to the ability of NAA80 to modify actin. Thus, re-expression of NAA80, which leads to re-establishment of actin's N-terminal acetyl group, rescued the Golgi fragmentation, whereas a catalytic dead NAA80 mutant could neither restore actin Nt-acetylation nor Golgi structure. The Golgi phenotype of NAA80 KO cells was shared by both migrating and non-migrating cells and live-cell imaging indicated increased Golgi dynamics in migrating NAA80 KO cells. Finally, we detected a drastic increase in the amount of F-actin in cells lacking NAA80, suggesting a causal relationship between this effect and the observed re-organization of Golgi structure. The findings further underscore the importance of actin Nt-acetylation and provide novel insight into its cellular roles, suggesting a mechanistic link between actin modification state and Golgi organization.


Subject(s)
Acetyltransferases/genetics , Actin Cytoskeleton/enzymology , Actins/genetics , Actins/metabolism , Golgi Apparatus/enzymology , Protein Processing, Post-Translational , Acetylation , Acetyltransferases/deficiency , Actin Cytoskeleton/ultrastructure , Cell Differentiation , Cell Line, Tumor , Cell Movement , Fibroblasts/metabolism , Fibroblasts/ultrastructure , Golgi Apparatus/ultrastructure , Humans , Phenotype , Time-Lapse Imaging
7.
Biosci Rep ; 40(1)2020 01 31.
Article in English | MEDLINE | ID: mdl-31844879

ABSTRACT

C3larvinA is a putative virulence factor produced by Paenibacillus larvae enterobacterial-repetitive-intergenic-consensus (ERIC) III/IV (strain 11-8051). Biochemical, functional and structural analyses of C3larvinA revealed that it belongs to the C3-like mono-ADP-ribosylating toxin subgroup. Mammalian RhoA was the target substrate for its transferase activity suggesting that it may be the biological target of C3larvinA. The kinetic parameters of the NAD+ substrate for the transferase (KM = 75 ± 10 µM) and glycohydrolase (GH) (KM = 107 ± 20 µM) reactions were typical for a C3-like bacterial toxin, including the Plx2A virulence factor from Paenibacillus larvae ERIC I. Upon cytoplasmic expression in yeast, C3larvinA caused a growth-defective phenotype indicating that it is an active C3-like toxin and is cytotoxic to eukaryotic cells. The catalytic variant of the Q187-X-E189 motif in C3larvinA showed no cytotoxicity toward yeast confirming that the cytotoxicity of this factor depends on its enzymatic activity. A homology consensus model of C3larvinA with NAD+ substrate was built on the structure of Plx2A, provided additional confirmation that C3larvinA is a member of the C3-like mono-ADP-ribosylating toxin subgroup. A homology model of C3larvinA with NADH and RhoA was built on the structure of the C3cer-NADH-RhoA complex which provided further evidence that C3larvinA is a C3-like toxin that shares an identical catalytic mechanism with C3cer from Bacillus cereus. C3larvinA induced actin cytoskeleton reorganization in murine macrophages, whereas in insect cells, vacuolization and bi-nucleated cells were observed. These cellular effects are consistent with C3larvinA disrupting RhoA function by covalent modification that is shared among C3-like bacterial toxins.


Subject(s)
ADP Ribose Transferases/metabolism , Bacterial Toxins/metabolism , Bees/microbiology , Paenibacillus larvae/enzymology , Virulence Factors/metabolism , rhoA GTP-Binding Protein/metabolism , ADP Ribose Transferases/chemistry , ADP Ribose Transferases/genetics , Actin Cytoskeleton/enzymology , Animals , Bacterial Toxins/chemistry , Bacterial Toxins/genetics , Host-Pathogen Interactions , Kinetics , Macrophages/enzymology , Mutation , Paenibacillus larvae/genetics , Paenibacillus larvae/pathogenicity , Protein Conformation , Saccharomyces cerevisiae/drug effects , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/growth & development , Sf9 Cells , Spodoptera , Structure-Activity Relationship , Substrate Specificity , Virulence , Virulence Factors/chemistry , Virulence Factors/genetics , rhoA GTP-Binding Protein/chemistry
8.
Nat Commun ; 10(1): 5200, 2019 11 15.
Article in English | MEDLINE | ID: mdl-31729365

ABSTRACT

The regulation of actin dynamics is essential for various cellular processes. Former evidence suggests a correlation between the function of non-conventional myosin motors and actin dynamics. Here we investigate the contribution of myosin 1b to actin dynamics using sliding motility assays. We observe that sliding on myosin 1b immobilized or bound to a fluid bilayer enhances actin depolymerization at the barbed end, while sliding on myosin II, although 5 times faster, has no effect. This work reveals a non-conventional myosin motor as another type of depolymerase and points to its singular interactions with the actin barbed end.


Subject(s)
Actins/chemistry , Actins/metabolism , Myosin Type I/metabolism , Actin Cytoskeleton/enzymology , Actin Cytoskeleton/metabolism , Actins/genetics , Animals , Humans , Myosin Type I/genetics , Myosin Type II/chemistry , Myosin Type II/genetics , Myosin Type II/metabolism , Polymerization , Rabbits
9.
Arterioscler Thromb Vasc Biol ; 38(10): 2423-2434, 2018 10.
Article in English | MEDLINE | ID: mdl-30354218

ABSTRACT

Objective- Actin cytoskeleton assembly and organization, as a result of focal adhesion (FA) formation during cell adhesion, are dependent on reactive oxygen species and the cellular redox environment. Poldip2 (polymerase δ-interacting protein 2), a novel regulator of NOX4 (NADPH oxidase 4), plays a significant role in reactive oxygen species production and cytoskeletal remodeling. Thus, we hypothesized that endogenous reactive oxygen species derived from Poldip2/NOX4 contribute to redox regulation of actin and cytoskeleton assembly during integrin-mediated cell adhesion. Approach and Results- Using vascular smooth muscle cells, we verified that hydrogen peroxide (H2O2) levels increase during integrin-mediated cell attachment as a result of activation of NOX4. Filamentous actin (F-actin) was oxidized by sulfenylation during cell attachment, with a peak at 3 hours (0.80±0.04 versus 0.08±0.13 arbitrary units at time zero), which was enhanced by overexpression of Poldip2. Depletion of Poldip2 or NOX4 using siRNA, or scavenging of endogenous H2O2 with catalase, inhibited F-actin oxidation by 78±26%, 99±1%, and 98±1%, respectively. To determine the consequence of F-actin oxidation, we examined the binding of F-actin to vinculin, a protein involved in FA complexes that regulates FA maturation. Vinculin binding during cell adhesion as well as migration capacity were inhibited after transfection with actin containing 2 oxidation-resistant point mutations (C272A and C374A). Silencing of Poldip2 or NOX4 also impaired actin-vinculin interaction, which disturbed maturation of FAs and inhibited cell migration. Conclusions- These results suggest that integrin engagement during cell attachment activates Poldip2/Nox4 to oxidize actin, which modulates FA assembly.


Subject(s)
Actin Cytoskeleton/enzymology , Carrier Proteins/metabolism , Cell Adhesion , Integrins/metabolism , Muscle, Smooth, Vascular/enzymology , Myocytes, Smooth Muscle/enzymology , NADPH Oxidase 4/metabolism , Nuclear Proteins/metabolism , Vinculin/metabolism , Actin Cytoskeleton/genetics , Animals , Carrier Proteins/genetics , Cell Movement , Cells, Cultured , Humans , Hydrogen Peroxide/metabolism , Muscle, Smooth, Vascular/ultrastructure , Myocytes, Smooth Muscle/ultrastructure , NADPH Oxidase 4/genetics , Nuclear Proteins/genetics , Oxidation-Reduction , Rats , Signal Transduction
10.
J Clin Invest ; 128(12): 5351-5367, 2018 12 03.
Article in English | MEDLINE | ID: mdl-30252678

ABSTRACT

MASTL, a Ser/Thr kinase that inhibits PP2A-B55 complexes during mitosis, is mutated in autosomal dominant thrombocytopenia. However, the connections between the cell-cycle machinery and this human disease remain unexplored. We report here that, whereas Mastl ablation in megakaryocytes prevented proper maturation of these cells, mice carrying the thrombocytopenia-associated mutation developed thrombocytopenia as a consequence of aberrant activation and survival of platelets. Activation of mutant platelets was characterized by hyperstabilized pseudopods mimicking the effect of PP2A inhibition and actin polymerization defects. These aberrations were accompanied by abnormal hyperphosphorylation of multiple components of the actin cytoskeleton and were rescued both in vitro and in vivo by inhibiting upstream kinases such as PKA, PKC, or AMPK. These data reveal an unexpected role of Mastl in actin cytoskeletal dynamics in postmitotic cells and suggest that the thrombocytopenia-associated mutation in MASTL is a pathogenic dominant mutation that mimics decreased PP2A activity resulting in altered phosphorylation of cytoskeletal regulatory pathways.


Subject(s)
Actin Cytoskeleton , Blood Platelets/enzymology , Chromosome Breakage , Chromosome Disorders , Microtubule-Associated Proteins , Mutation, Missense , Protein Serine-Threonine Kinases , Signal Transduction/genetics , Thrombocytopenia/congenital , Actin Cytoskeleton/enzymology , Actin Cytoskeleton/genetics , Amino Acid Substitution , Animals , Blood Platelets/pathology , Chromosome Disorders/enzymology , Chromosome Disorders/genetics , Chromosome Disorders/pathology , Mice , Mice, Transgenic , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Phosphorylation , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Thrombocytopenia/enzymology , Thrombocytopenia/genetics , Thrombocytopenia/pathology
11.
Proc Natl Acad Sci U S A ; 115(17): 4399-4404, 2018 04 24.
Article in English | MEDLINE | ID: mdl-29581253

ABSTRACT

Actin, one of the most abundant proteins in nature, participates in countless cellular functions ranging from organelle trafficking and pathogen motility to cell migration and regulation of gene transcription. Actin's cellular activities depend on the dynamic transition between its monomeric and filamentous forms, a process exquisitely regulated in cells by a large number of actin-binding and signaling proteins. Additionally, several posttranslational modifications control the cellular functions of actin, including most notably N-terminal (Nt)-acetylation, a prevalent modification throughout the animal kingdom. However, the biological role and mechanism of actin Nt-acetylation are poorly understood, and the identity of actin's N-terminal acetyltransferase (NAT) has remained a mystery. Here, we reveal that NAA80, a suggested NAT enzyme whose substrate specificity had not been characterized, is Nt-acetylating actin. We further show that actin Nt-acetylation plays crucial roles in cytoskeletal assembly in vitro and in cells. The absence of Nt-acetylation leads to significant differences in the rates of actin filament depolymerization and elongation, including elongation driven by formins, whereas filament nucleation by the Arp2/3 complex is mostly unaffected. NAA80-knockout cells display severely altered cytoskeletal organization, including an increase in the ratio of filamentous to globular actin, increased filopodia and lamellipodia formation, and accelerated cell motility. Together, the results demonstrate NAA80's role as actin's NAT and reveal a crucial role for actin Nt-acetylation in the control of cytoskeleton structure and dynamics.


Subject(s)
Acetyltransferases/metabolism , Actin Cytoskeleton/enzymology , Actin-Related Protein 2-3 Complex/metabolism , Cell Movement/physiology , N-Terminal Acetyltransferases/metabolism , Pseudopodia/enzymology , Acetylation , Acetyltransferases/genetics , Actin Cytoskeleton/genetics , Actin-Related Protein 2-3 Complex/genetics , HEK293 Cells , Humans , N-Terminal Acetyltransferases/genetics , Pseudopodia/genetics
12.
Mol Biol Cell ; 29(4): 419-434, 2018 02 15.
Article in English | MEDLINE | ID: mdl-29282284

ABSTRACT

An intact actomyosin network is essential for anchoring polarity proteins to the cell cortex and maintaining cell size asymmetry during asymmetric cell division of Drosophila neuroblasts (NBs). However, the mechanisms that control changes in actomyosin dynamics during asymmetric cell division remain unclear. We find that the actin-binding protein, Moesin, is essential for NB proliferation and mitotic progression in the developing brain. During metaphase, phosphorylated Moesin (p-Moesin) is enriched at the apical cortex, and loss of Moesin leads to defects in apical polarity maintenance and cortical stability. This asymmetric distribution of p-Moesin is determined by components of the apical polarity complex and Slik kinase. During later stages of mitosis, p-Moesin localization shifts more basally, contributing to asymmetric cortical extension and myosin basal furrow positioning. Our findings reveal Moesin as a novel apical polarity protein that drives cortical remodeling of dividing NBs, which is essential for polarity maintenance and initial establishment of cell size asymmetry.


Subject(s)
Asymmetric Cell Division/physiology , Drosophila Proteins/metabolism , Microfilament Proteins/metabolism , Neural Stem Cells/metabolism , Protein Serine-Threonine Kinases/metabolism , Actin Cytoskeleton/enzymology , Actomyosin/metabolism , Animals , Cell Membrane/enzymology , Cell Membrane/ultrastructure , Cell Polarity , Drosophila/enzymology , Drosophila Proteins/genetics , Metaphase , Phosphorylation , Protein Serine-Threonine Kinases/genetics
13.
Mol Cell ; 65(5): 885-899.e6, 2017 Mar 02.
Article in English | MEDLINE | ID: mdl-28238652

ABSTRACT

Loss of ER Ca2+ homeostasis triggers endoplasmic reticulum (ER) stress and drives ER-PM contact sites formation in order to refill ER-luminal Ca2+. Recent studies suggest that the ER stress sensor and mediator of the unfolded protein response (UPR) PERK regulates intracellular Ca2+ fluxes, but the mechanisms remain elusive. Here, using proximity-dependent biotin identification (BioID), we identified the actin-binding protein Filamin A (FLNA) as a key PERK interactor. Cells lacking PERK accumulate F-actin at the cell edges and display reduced ER-PM contacts. Following ER-Ca2+ store depletion, the PERK-FLNA interaction drives the expansion of ER-PM juxtapositions by regulating F-actin-assisted relocation of the ER-associated tethering proteins Stromal Interaction Molecule 1 (STIM1) and Extended Synaptotagmin-1 (E-Syt1) to the PM. Cytosolic Ca2+ elevation elicits rapid and UPR-independent PERK dimerization, which enforces PERK-FLNA-mediated ER-PM juxtapositions. Collectively, our data unravel an unprecedented role of PERK in the regulation of ER-PM appositions through the modulation of the actin cytoskeleton.


Subject(s)
Actin Cytoskeleton/enzymology , Actins/metabolism , Cell Membrane/enzymology , Endoplasmic Reticulum Stress , Endoplasmic Reticulum/enzymology , Filamins/metabolism , eIF-2 Kinase/metabolism , Animals , Calcium/metabolism , Calcium Signaling , Filamins/genetics , HEK293 Cells , HeLa Cells , Humans , Mice , Neoplasm Proteins/metabolism , Protein Multimerization , Protein Transport , RNA Interference , Signal Transduction , Stromal Interaction Molecule 1/metabolism , Synaptotagmin I/metabolism , Time Factors , Transfection , Unfolded Protein Response , eIF-2 Kinase/genetics
14.
J Cell Biol ; 216(1): 181-197, 2017 Jan 02.
Article in English | MEDLINE | ID: mdl-28007915

ABSTRACT

Podosomes represent a class of integrin-mediated cell-matrix adhesions formed by migrating and matrix-degrading cells. We demonstrate that in macrophage-like THP1 cells and fibroblasts stimulated to produce podosomes, down-regulation of the G-protein ARF1 or the ARF1 guanine nucleotide exchange factor, ARNO, by small, interfering RNA or pharmacological inhibitors led to striking podosome elimination. Concomitantly, treatments inducing podosome formation increased the level of guanosine triphosphate (GTP)-bound ARF1. ARNO was found to colocalize with the adhesive rings of podosomes, whereas ARF1 was localized to vesicular structures transiently contacting podosome rings. Inhibition of ARF1 led to an increase in RhoA-GTP levels and triggered assembly of myosin-IIA filaments in THP1 cells, whereas the suppression of myosin-IIA rescued podosome formation regardless of ARF1 inhibition. Finally, expression of constitutively active ARF1 in fibroblasts induced formation of putative podosome precursors: actin-rich puncta coinciding with matrix degradation sites and containing proteins of the podosome core but not of the adhesive ring. Thus, ARNO-ARF1 regulates formation of podosomes by inhibition of RhoA/myosin-II and promotion of actin core assembly.


Subject(s)
ADP-Ribosylation Factor 1/metabolism , GTPase-Activating Proteins/metabolism , Podosomes/enzymology , ADP-Ribosylation Factor 1/antagonists & inhibitors , ADP-Ribosylation Factor 1/genetics , Actin Cytoskeleton/enzymology , Actins/metabolism , Animals , Cell Line, Tumor , Enzyme Inhibitors/pharmacology , GTPase-Activating Proteins/genetics , Guanosine Triphosphate/metabolism , Humans , Mice , Microscopy, Fluorescence , Nonmuscle Myosin Type IIA/metabolism , Podosomes/drug effects , RNA Interference , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Signal Transduction , Time Factors , Transfection , rho-Associated Kinases/metabolism , rhoA GTP-Binding Protein/metabolism
15.
J Biol Chem ; 292(2): 488-504, 2017 Jan 13.
Article in English | MEDLINE | ID: mdl-27895123

ABSTRACT

We previously identified focal adhesion kinase (FAK) as an important regulator of ciliogenesis in multiciliated cells. FAK and other focal adhesion (FA) proteins associate with the basal bodies and their striated rootlets and form complexes named ciliary adhesions (CAs). CAs display similarities with FAs but are established in an integrin independent fashion and are responsible for anchoring basal bodies to the actin cytoskeleton during ciliogenesis as well as in mature multiciliated cells. FAK down-regulation leads to aberrant ciliogenesis due to impaired association between the basal bodies and the actin cytoskeleton, suggesting that FAK is an important regulator of the CA complex. However, the mechanism through which FAK functions in the complex is not clear, and in this study we examined the role of this protein in both ciliogenesis and ciliary function. We show that localization of FAK at CAs depends on interactions taking place at the amino-terminal (FERM) and carboxyl-terminal (FAT) domains and that both domains are required for proper ciliogenesis and ciliary function. Furthermore, we show that an interaction with another CA protein, paxillin, is essential for correct localization of FAK in multiciliated cells. This interaction is indispensable for both ciliogenesis and ciliary function. Finally, we provide evidence that despite the fact that FAK is in the active, open conformation at CAs, its kinase activity is dispensable for ciliogenesis and ciliary function revealing that FAK plays a scaffolding role in multiciliated cells. Overall these data show that the role of FAK at CAs displays similarities but also important differences compared with its role at FAs.


Subject(s)
Avian Proteins/metabolism , Basal Bodies/enzymology , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Actin Cytoskeleton/enzymology , Actin Cytoskeleton/genetics , Animals , Avian Proteins/genetics , Chickens , Cilia/enzymology , Cilia/genetics , Focal Adhesion Protein-Tyrosine Kinases/genetics , Paxillin/genetics , Paxillin/metabolism , Protein Domains , Xenopus Proteins/genetics , Xenopus Proteins/metabolism , Xenopus laevis
16.
Oncotarget ; 7(49): 80175-80189, 2016 Dec 06.
Article in English | MEDLINE | ID: mdl-27863394

ABSTRACT

Epithelial-mesenchymal transition (EMT) is one of the decisive steps regulating cancer invasion and metastasis. However, the molecular mechanisms underlying this transition require further clarification. MDA-9/syntenin (SDCBP) expression is elevated in breast cancer patient samples as well as cultured breast cancer cells. Silencing expression of MDA-9 in mesenchymal metastatic breast cancer cells triggered a change in cell morphology in both 2D- and 3D-cultures to a more epithelial-like phenotype, along with changes in EMT markers, cytoskeletal rearrangement and decreased invasion. Conversely, over expressing MDA-9 in epithelial non-metastatic breast cancer cells instigated a change in morphology to a more mesenchymal phenotype with corresponding changes in EMT markers, cytoskeletal rearrangement and an increase in invasion. We also found that MDA-9 upregulated active levels of known modulators of EMT, the small GTPases RhoA and Cdc42, via TGFß1. Reintroducing TGFß1 in MDA-9 silenced cells restored active RhoA and cdc42 levels, modulated cytoskeletal rearrangement and increased invasion. We further determined that MDA-9 interacts with TGFß1 via its PDZ1 domain. Finally, in vivo studies demonstrated that silencing the expression of MDA-9 resulted in decreased lung metastasis and TGFß1 re-expression partially restored lung metastases. Our findings provide evidence for the relevance of MDA-9 in mediating EMT in breast cancer and support the potential of MDA-9 as a therapeutic target against metastatic disease.


Subject(s)
Breast Neoplasms/enzymology , Epithelial-Mesenchymal Transition , Syntenins/metabolism , Transforming Growth Factor beta1/metabolism , cdc42 GTP-Binding Protein/metabolism , rhoA GTP-Binding Protein/metabolism , Actin Cytoskeleton/enzymology , Actin Cytoskeleton/pathology , Animals , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Movement , Cell Shape , Female , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/enzymology , Lung Neoplasms/genetics , Lung Neoplasms/secondary , Mice, Nude , Neoplasm Invasiveness , Phenotype , Protein Interaction Domains and Motifs , RNA Interference , Signal Transduction , Syntenins/genetics , Time Factors , Transfection , Transforming Growth Factor beta1/genetics , cdc42 GTP-Binding Protein/genetics , rhoA GTP-Binding Protein/genetics
17.
Cancer Lett ; 380(2): 505-512, 2016 10 01.
Article in English | MEDLINE | ID: mdl-27452906

ABSTRACT

Triple-negative breast cancers (TNBCs) are among the most aggressive cancers characterized by a high propensity to invade, metastasize and relapse. We previously reported that the TNBC-specific inhibitor, AMPI-109, significantly impairs the ability of TNBC cells to migrate and invade by reducing levels of the metastasis-promoting phosphatase, PRL-3. Here, we examined the mechanisms by which AMPI-109 and loss of PRL-3 impede cell migration and invasion. AMPI-109 treatment or knock down of PRL-3 expression were associated with deactivation of Src and ERK signaling and concomitant downregulation of RhoA and Rac1/2/3 GTPase protein levels. These cellular changes led to rearranged filamentous actin networks necessary for cell migration and invasion. Conversely, overexpression of PRL-3 promoted TNBC cell invasion by upregulating matrix metalloproteinase 10, which resulted in increased TNBC cell adherence to, and degradation of, the major basement membrane component laminin. Our data demonstrate that PRL-3 engages the focal adhesion pathway in TNBC cells as a key mechanism for promoting TNBC cell migration and invasion. Collectively, these data suggest that blocking PRL-3 activity may be an effective method for reducing the metastatic potential of TNBC cells.


Subject(s)
Actin Cytoskeleton/enzymology , Cell Adhesion , Cell Movement , Focal Adhesions/enzymology , Neoplasm Proteins/metabolism , Protein Tyrosine Phosphatases/metabolism , Triple Negative Breast Neoplasms/enzymology , Actin Cytoskeleton/drug effects , Actin Cytoskeleton/pathology , Antineoplastic Agents/pharmacology , Calcitriol/analogs & derivatives , Calcitriol/pharmacology , Cell Adhesion/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Focal Adhesions/drug effects , Focal Adhesions/pathology , Humans , Laminin/metabolism , Matrix Metalloproteinase 10/metabolism , Neoplasm Invasiveness , Neoplasm Metastasis , Neoplasm Proteins/genetics , Protein Tyrosine Phosphatases/genetics , RNA Interference , Signal Transduction , Time Factors , Transfection , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/pathology , rac GTP-Binding Proteins/metabolism , rhoA GTP-Binding Protein/metabolism , src-Family Kinases/metabolism
18.
Oncotarget ; 7(22): 31602-22, 2016 May 31.
Article in English | MEDLINE | ID: mdl-27203208

ABSTRACT

Loss of polarity and quiescence along with increased cellular invasiveness are associated with breast tumor progression. ROCK plays a central role in actin-cytoskeletal rearrangement. We used physiologically relevant 3D cultures of nonmalignant and cancer cells in gels made of laminin-rich extracellular matrix, to investigate ROCK function. Whereas expression levels of ROCK1 and ROCK2 were elevated in cancer cells compared to nonmalignant cells, this was not observed in 2D cultures. Malignant cells showed increased phosphorylation of MLC, corresponding to disorganized F-actin. Inhibition of ROCK signaling restored polarity, decreased disorganization of F-actin, and led to reduction of proliferation. Inhibition of ROCK also decreased EGFR and Integrinß1 levels, and consequently suppressed activation of Akt, MAPK and FAK as well as GLUT3 and LDHA levels. Again, ROCK inhibition did not inhibit these molecules in 2D. A triple negative breast cancer cell line, which lacks E-cadherin, had high levels of ROCK but was less sensitive to ROCK inhibitors. Exogenous overexpression of E-cadherin, however, rendered these cells strikingly sensitive to ROCK inhibition. Our results add to the growing literature that demonstrate the importance of context and tissue architecture in determining not only regulation of normal and malignant phenotypes but also drug response.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Cell Polarity/drug effects , Protein Kinase Inhibitors/pharmacology , Signal Transduction/drug effects , rho-Associated Kinases/antagonists & inhibitors , Actin Cytoskeleton/drug effects , Actin Cytoskeleton/enzymology , Antigens, CD , Breast Neoplasms/enzymology , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cadherins/genetics , Cadherins/metabolism , Cancer-Associated Fibroblasts/drug effects , Cancer-Associated Fibroblasts/enzymology , Cancer-Associated Fibroblasts/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Coculture Techniques , Dose-Response Relationship, Drug , Drug Resistance, Neoplasm , Female , Humans , Molecular Targeted Therapy , Phenotype , Transfection , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/enzymology , Triple Negative Breast Neoplasms/pathology , rho-Associated Kinases/metabolism , rhoA GTP-Binding Protein/metabolism
19.
J Cell Biol ; 212(4): 375-7, 2016 Feb 15.
Article in English | MEDLINE | ID: mdl-26880198

ABSTRACT

The mechanisms by which neuroligin adhesion molecules modulate synaptic plasticity remain unclear. In this issue, Liu et al. (2016. J. Cell Biol. http://dx.doi.org/10.1083/jcb.201509023) demonstrate that neuroligin 1 promotes actin assembly associated with synaptic strengthening independent of adhesion, suggesting additional ways for neuroligins to contribute to neuronal development and disease pathology.


Subject(s)
Actin Cytoskeleton/enzymology , Actin Depolymerizing Factors/metabolism , Cell Adhesion Molecules, Neuronal/metabolism , Cerebral Cortex/enzymology , Dendritic Spines/enzymology , Hippocampus/enzymology , Lim Kinases/metabolism , Neuronal Plasticity , Synapses/enzymology , Animals , Humans
20.
J Cell Biol ; 212(4): 449-63, 2016 Feb 15.
Article in English | MEDLINE | ID: mdl-26880202

ABSTRACT

Neuroligin (NLG) 1 is important for synapse development and function, but the underlying mechanisms remain unclear. It is known that at least some aspects of NLG1 function are independent of the presynaptic neurexin, suggesting that the C-terminal domain (CTD) of NLG1 may be sufficient for synaptic regulation. In addition, NLG1 is subjected to activity-dependent proteolytic cleavage, generating a cytosolic CTD fragment, but the significance of this process remains unknown. In this study, we show that the CTD of NLG1 is sufficient to (a) enhance spine and synapse number, (b) modulate synaptic plasticity, and (c) exert these effects via its interaction with spine-associated Rap guanosine triphosphatase-activating protein and subsequent activation of LIM-domain protein kinase 1/cofilin-mediated actin reorganization. Our results provide a novel postsynaptic mechanism by which NLG1 regulates synapse development and function.


Subject(s)
Actin Cytoskeleton/enzymology , Actin Depolymerizing Factors/metabolism , Cell Adhesion Molecules, Neuronal/metabolism , Cerebral Cortex/enzymology , Dendritic Spines/enzymology , Hippocampus/enzymology , Lim Kinases/metabolism , Neuronal Plasticity , Synapses/enzymology , Animals , Cell Adhesion Molecules, Neuronal/deficiency , Cell Adhesion Molecules, Neuronal/genetics , Cerebral Cortex/cytology , GTPase-Activating Proteins/metabolism , Genotype , HEK293 Cells , Hippocampus/cytology , Humans , In Vitro Techniques , Lim Kinases/deficiency , Lim Kinases/genetics , Mice, Knockout , Phenotype , Phosphorylation , Protein Binding , Protein Interaction Domains and Motifs , RNA Interference , Signal Transduction , Time Factors , Transfection , rap1 GTP-Binding Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...