Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 64
Filter
Add more filters










Publication year range
1.
Nat Commun ; 15(1): 4023, 2024 May 13.
Article in English | MEDLINE | ID: mdl-38740816

ABSTRACT

Abscission is the final stage of cytokinesis, which cleaves the intercellular bridge (ICB) connecting two daughter cells. Abscission requires tight control of the recruitment and polymerization of the Endosomal Protein Complex Required for Transport-III (ESCRT-III) components. We explore the role of post-translational modifications in regulating ESCRT dynamics. We discover that SMYD2 methylates the lysine 6 residue of human CHMP2B, a key ESCRT-III component, at the ICB, impacting the dynamic relocation of CHMP2B to sites of abscission. SMYD2 loss-of-function (genetically or pharmacologically) causes CHMP2B hypomethylation, delayed CHMP2B polymerization and delayed abscission. This is phenocopied by CHMP2B lysine 6 mutants that cannot be methylated. Conversely, SMYD2 gain-of-function causes CHMP2B hypermethylation and accelerated abscission, specifically in cells undergoing cytokinetic challenges, thereby bypassing the abscission checkpoint. Additional experiments highlight the importance of CHMP2B methylation beyond cytokinesis, namely during ESCRT-III-mediated HIV-1 budding. We propose that lysine methylation signaling fine-tunes the ESCRT-III machinery to regulate the timing of cytokinetic abscission and other ESCRT-III dependent functions.


Subject(s)
Cytokinesis , Endosomal Sorting Complexes Required for Transport , Endosomal Sorting Complexes Required for Transport/metabolism , Endosomal Sorting Complexes Required for Transport/genetics , Humans , Methylation , HeLa Cells , Histone-Lysine N-Methyltransferase/metabolism , Histone-Lysine N-Methyltransferase/genetics , HIV-1/metabolism , HIV-1/genetics , HIV-1/physiology , Lysine/metabolism , Protein Processing, Post-Translational
2.
Cell Rep ; 43(4): 114034, 2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38568808

ABSTRACT

Escape from the bacterial-containing vacuole (BCV) is a key step of Shigella host cell invasion. Rab GTPases subverted to in situ-formed macropinosomes in the vicinity of the BCV have been shown to promote its rupture. The involvement of the BCV itself has remained unclear. We demonstrate that Rab35 is non-canonically entrapped at the BCV. Stimulated emission depletion imaging localizes Rab35 directly on the BCV membranes before vacuolar rupture. The bacterial effector IcsB, a lysine Nε-fatty acylase, is a key regulator of Rab35-BCV recruitment, and we show post-translational acylation of Rab35 by IcsB in its polybasic region. While Rab35 and IcsB are dispensable for the first step of BCV breakage, they are needed for the unwrapping of damaged BCV remnants from Shigella. This provides a framework for understanding Shigella invasion implicating re-localization of a Rab GTPase via its bacteria-dependent post-translational modification to support the mechanical unpeeling of the BCV.


Subject(s)
Bacterial Proteins , Protein Processing, Post-Translational , Shigella , Vacuoles , rab GTP-Binding Proteins , rab GTP-Binding Proteins/metabolism , Humans , Shigella/metabolism , Bacterial Proteins/metabolism , Vacuoles/metabolism , Vacuoles/microbiology , HeLa Cells
3.
Nat Commun ; 15(1): 1949, 2024 Mar 02.
Article in English | MEDLINE | ID: mdl-38431632

ABSTRACT

Cell division is completed by the abscission of the intercellular bridge connecting the daughter cells. Abscission requires the polymerization of an ESCRT-III cone close to the midbody to both recruit the microtubule severing enzyme spastin and scission the plasma membrane. Here, we found that the microtubule and the membrane cuts are two separate events that are regulated differently. Using HeLa cells, we uncovered that the F-actin disassembling protein Cofilin-1 controls the disappearance of a transient pool of branched F-actin which is precisely assembled at the tip of the ESCRT-III cone shortly before the microtubule cut. Functionally, Cofilin-1 and Arp2/3-mediated branched F-actin favor abscission by promoting local severing of the microtubules but do not participate later in the membrane scission event. Mechanistically, we propose that branched F-actin functions as a physical barrier that limits ESCRT-III cone elongation and thereby favors stable spastin recruitment. Our work thus reveals that F-actin controls the timely and local disassembly of microtubules required for cytokinetic abscission.


Subject(s)
Actins , Microtubules , Humans , Actins/metabolism , HeLa Cells , Spastin/metabolism , Microtubules/metabolism , Cytokinesis , Endosomal Sorting Complexes Required for Transport/metabolism , Actin Depolymerizing Factors/metabolism
4.
Dev Cell ; 58(22): 2477-2494.e8, 2023 Nov 20.
Article in English | MEDLINE | ID: mdl-37875118

ABSTRACT

Cilia protrude from the cell surface and play critical roles in intracellular signaling, environmental sensing, and development. Reduced actin-dependent contractility and intracellular trafficking are both required for ciliogenesis, but little is known about how these processes are coordinated. Here, we identified a Rac1- and Rab35-binding protein with a truncated BAR (Bin/amphiphysin/Rvs) domain that we named MiniBAR (also known as KIAA0355/GARRE1), which plays a key role in ciliogenesis. MiniBAR colocalizes with Rac1 and Rab35 at the plasma membrane and on intracellular vesicles trafficking to the ciliary base and exhibits fast pulses at the ciliary membrane. MiniBAR depletion leads to short cilia, resulting from abnormal Rac-GTP/Rho-GTP levels and increased acto-myosin-II-dependent contractility together with defective trafficking of IFT88 and ARL13B into cilia. MiniBAR-depleted zebrafish embryos display dysfunctional short cilia and hallmarks of ciliopathies, including left-right asymmetry defects. Thus, MiniBAR is a dual Rac and Rab effector that controls both actin cytoskeleton and membrane trafficking for ciliogenesis.


Subject(s)
Cytoskeletal Proteins , Zebrafish , Animals , Zebrafish/metabolism , Cytoskeletal Proteins/metabolism , Signal Transduction , Carrier Proteins/metabolism , Cilia/metabolism , Guanosine Triphosphate/metabolism , rab GTP-Binding Proteins/metabolism
5.
Dev Cell ; 58(19): 1917-1932.e6, 2023 10 09.
Article in English | MEDLINE | ID: mdl-37552987

ABSTRACT

Long ignored as a vestigial remnant of cytokinesis, the mammalian midbody (MB) is released post-abscission inside large extracellular vesicles called MB remnants (MBRs). Recent evidence suggests that MBRs can modulate cell proliferation and cell fate decisions. Here, we demonstrate that the MB matrix is the site of ribonucleoprotein assembly and is enriched in mRNAs that encode proteins involved in cell fate, oncogenesis, and pluripotency, which we are calling the MB granule. Both MBs and post-abscission MBRs are sites of spatiotemporally regulated translation, which is initiated when nascent daughter cells re-enter G1 and continues after extracellular release. MKLP1 and ARC are necessary for the localization and translation of RNA in the MB dark zone, whereas ESCRT-III is necessary to maintain translation levels in the MB. Our work reveals a unique translation event that occurs during abscission and within a large extracellular vesicle.


Subject(s)
Cytokinesis , RNA , Animals , Humans , Cell Differentiation , HeLa Cells , Mammals
6.
Biol Cell ; 115(4): e2200085, 2023 Apr.
Article in English | MEDLINE | ID: mdl-36597754

ABSTRACT

The human immunodeficiency virus type 1 (HIV-1) is an intracellular pathogen whose replication cycle strictly depends on the host cell molecular machinery. HIV-1 crosses twice the plasma membrane, to get in and to get out of the cell. Therefore, the first and the last line of intracellular component encountered by the virus is the cortical actin network. Here, we review the role of actin and actin-related proteins in HIV-1 entry, assembly, budding, and release. We first highlight the mechanisms controlling actin polymerization at the entry site that promote the clustering of HIV-1 receptors, a crucial step for the virus to fuse with the plasma membrane. Then, we describe how actin is transiently depolymerized locally to allow the capsid to cross the actin cortex, before migrating towards the nucleus. Finally, we review the role of several actin-binding proteins in actin remodeling events required for membrane deformation and curvature at the viral assembly site as well as for virus release. Strikingly, it appears that common actin-regulating pathways are involved in viral entry and exit. However, while the role of actin remodeling during entry is well understood, this is not the case during exit. We discuss remaining challenges regarding the actin-dependent mechanisms involved in HIV-1 entry and exit, and how they could be overcome.


Subject(s)
Actins , HIV-1 , Humans , Actins/metabolism , HIV-1/physiology , Actin Cytoskeleton/metabolism , Cell Line , Cell Membrane/metabolism
7.
Front Cell Dev Biol ; 10: 1046617, 2022.
Article in English | MEDLINE | ID: mdl-36506096

ABSTRACT

Cytokinetic abscission leads to the physical cut of the intercellular bridge (ICB) connecting the daughter cells and concludes cell division. In different animal cells, it is well established that the ESCRT-III machinery is responsible for the constriction and scission of the ICB. Here, we review the mechanical context of abscission. We first summarize the evidence that the ICB is initially under high tension and explain why, paradoxically, this can inhibit abscission in epithelial cells by impacting on ESCRT-III assembly. We next detail the different mechanisms that have been recently identified to release ICB tension and trigger abscission. Finally, we discuss whether traction-induced mechanical cell rupture could represent an ancient alternative mechanism of abscission and suggest future research avenues to further understand the role of mechanics in regulating abscission.

8.
Eur J Cell Biol ; 101(3): 151249, 2022.
Article in English | MEDLINE | ID: mdl-35716426

ABSTRACT

Actin is among the most abundant proteins in eukaryotic cells and assembles into dynamic filamentous networks regulated by many actin binding proteins. The actin cytoskeleton must be finely tuned, both in space and time, to fulfill key cellular functions such as cell division, cell shape changes, phagocytosis and cell migration. While actin oxidation by reactive oxygen species (ROS) at non-physiological levels are known for long to impact on actin polymerization and on the cellular actin cytoskeleton, growing evidence shows that direct and reversible oxidation/reduction of specific actin amino acids plays an important and physiological role in regulating the actin cytoskeleton. In this review, we describe which actin amino acid residues can be selectively oxidized and reduced in many different ways (e.g. disulfide bond formation, glutathionylation, carbonylation, nitration, nitrosylation and other oxidations), the cellular enzymes at the origin of these post-translational modifications, and the impact of actin redox modifications both in vitro and in vivo. We show that the regulated balance of oxidation and reduction of key actin amino acid residues contributes to the control of actin filament polymerization and disassembly at the subcellular scale and highlight how improper redox modifications of actin can lead to pathological conditions.


Subject(s)
Actin Cytoskeleton , Actins , Actin Cytoskeleton/metabolism , Actins/metabolism , Amino Acids/analysis , Amino Acids/metabolism , Microfilament Proteins/metabolism , Oxidation-Reduction
9.
Sci Adv ; 8(15): eabm5095, 2022 Apr 15.
Article in English | MEDLINE | ID: mdl-35417244

ABSTRACT

During cytokinesis, the intercellular bridge (ICB) connecting the daughter cells experiences pulling forces, which delay abscission by preventing the assembly of the ESCRT scission machinery. Abscission is thus triggered by tension release, but how ICB tension is controlled is unknown. Here, we report that caveolae, which are known to regulate membrane tension upon mechanical stress in interphase cells, are located at the midbody, at the abscission site, and at the ICB/cell interface in dividing cells. Functionally, the loss of caveolae delays ESCRT-III recruitment during cytokinesis and impairs abscission. This is the consequence of a twofold increase of ICB tension measured by laser ablation, associated with a local increase in myosin II activity at the ICB/cell interface. We thus propose that caveolae buffer membrane tension and limit contractibility at the ICB to promote ESCRT-III assembly and cytokinetic abscission. Together, this work reveals an unexpected connection between caveolae and the ESCRT machinery and the first role of caveolae in cell division.

10.
Nat Commun ; 13(1): 630, 2022 02 02.
Article in English | MEDLINE | ID: mdl-35110562

ABSTRACT

Broadly neutralizing antibodies (bNAbs) targeting the HIV-1 envelope glycoprotein (Env) are promising molecules for therapeutic or prophylactic interventions. Beyond neutralization, bNAbs exert Fc-dependent functions including antibody-dependent cellular cytotoxicity and activation of the complement. Here, we show that a subset of bNAbs targeting the CD4 binding site and the V1/V2 or V3 loops inhibit viral release from infected cells. We combined immunofluorescence, scanning electron microscopy, transmission electron microscopy and immunogold staining to reveal that some bNAbs form large aggregates of virions at the surface of infected cells. This activity correlates with the capacity of bNAbs to bind to Env at the cell surface and to neutralize cell-free viral particles. We further show that antibody bivalency is required for viral retention, and that aggregated virions are neutralized. We have thus identified an additional antiviral activity of bNAbs, which block HIV-1 release by tethering viral particles at the surface of infected cells.


Subject(s)
Antibodies, Neutralizing/immunology , HIV Antibodies/immunology , HIV-1/immunology , Virion/immunology , Antibody-Dependent Cell Cytotoxicity , Broadly Neutralizing Antibodies , Cell Line , Epitopes , HIV Infections/immunology , Host Microbial Interactions/immunology , Humans , T-Lymphocytes , env Gene Products, Human Immunodeficiency Virus/immunology
11.
Proc Natl Acad Sci U S A ; 118(47)2021 11 23.
Article in English | MEDLINE | ID: mdl-34785592

ABSTRACT

During osmotic changes of their environment, cells actively regulate their volume and plasma membrane tension that can passively change through osmosis. How tension and volume are coupled during osmotic adaptation remains unknown, as their quantitative characterization is lacking. Here, we performed dynamic membrane tension and cell volume measurements during osmotic shocks. During the first few seconds following the shock, cell volume varied to equilibrate osmotic pressures inside and outside the cell, and membrane tension dynamically followed these changes. A theoretical model based on the passive, reversible unfolding of the membrane as it detaches from the actin cortex during volume increase quantitatively describes our data. After the initial response, tension and volume recovered from hypoosmotic shocks but not from hyperosmotic shocks. Using a fluorescent membrane tension probe (fluorescent lipid tension reporter [Flipper-TR]), we investigated the coupling between tension and volume during these asymmetric recoveries. Caveolae depletion and pharmacological inhibition of ion transporters and channels, mTORCs, and the cytoskeleton all affected tension and volume responses. Treatments targeting mTORC2 and specific downstream effectors caused identical changes to both tension and volume responses, their coupling remaining the same. This supports that the coupling of tension and volume responses to osmotic shocks is primarily regulated by mTORC2.


Subject(s)
Cell Size , Membranes/metabolism , Osmosis/physiology , Actins/metabolism , Cell Membrane/metabolism , Cytoskeleton/metabolism , HeLa Cells , Humans , Membranes/drug effects , Models, Theoretical , Osmotic Pressure/physiology
12.
Curr Biol ; 31(10): 2203-2213.e5, 2021 05 24.
Article in English | MEDLINE | ID: mdl-33711249

ABSTRACT

The midbody at the center of the intercellular bridge connecting dividing cells recruits the machinery essential for the final steps of cytokinesis.1-5 Successive abscission on both sides of the midbody generates a free midbody remnant (MBR) that can be inherited and accumulated in many cancer, immortalized, and stem cells, both in culture and in vivo.6-12 Strikingly, this organelle was recently shown to contain information that induces cancer cell proliferation, influences cell polarity, and promotes dorso-ventral axis specification upon interaction with recipient cells.13-16 Yet the mechanisms by which the MBR is captured by either a daughter cell or a distant cell are poorly described.10,14 Here, we report that BST2/tetherin, a well-established restriction factor that blocks the release of numerous enveloped viruses from the surface of infected cells,17-20 plays an analogous role in retaining midbody remnants. We found that BST2 is enriched at the midbody during cytokinesis and localizes at the surface of MBRs in a variety of cells. Knocking out BST2 induces the detachment of MBRs from the cell surface, their accumulation in the extracellular medium, and their transfer to distant cells. Mechanistically, the localization of BST2 at the MBR membrane is both necessary and sufficient for the interaction between MBRs and the cell surface. We thus propose that BST2 tethers post-cytokinetic midbody remnants to the cell surface. This finding reveals new parallels between cytokinesis and viral biology21-26 that unexpectedly extend beyond the ESCRT-dependent abscission step.


Subject(s)
Antigens, CD , Bone Marrow Stromal Antigen 2 , Cytokinesis , Antigens, CD/genetics , Antigens, CD/physiology , Bone Marrow Stromal Antigen 2/physiology , Cell Membrane , GPI-Linked Proteins/physiology , HEK293 Cells , HeLa Cells , Hep G2 Cells , Humans , Organelles
13.
EMBO Rep ; 22(2): e50965, 2021 02 03.
Article in English | MEDLINE | ID: mdl-33393173

ABSTRACT

Proteins of the ADF/cofilin family play a central role in the disassembly of actin filaments, and their activity must be tightly regulated in cells. Recently, the oxidation of actin filaments by the enzyme MICAL1 was found to amplify the severing action of cofilin through unclear mechanisms. Using single filament experiments in vitro, we found that actin filament oxidation by MICAL1 increases, by several orders of magnitude, both cofilin binding and severing rates, explaining the dramatic synergy between oxidation and cofilin for filament disassembly. Remarkably, we found that actin oxidation bypasses the need for cofilin activation by dephosphorylation. Indeed, non-activated, phosphomimetic S3D-cofilin binds and severs oxidized actin filaments rapidly, in conditions where non-oxidized filaments are unaffected. Finally, tropomyosin Tpm1.8 loses its ability to protect filaments from cofilin severing activity when actin is oxidized by MICAL1. Together, our results show that MICAL1-induced oxidation of actin filaments suppresses their physiological protection from the action of cofilin. We propose that, in cells, direct post-translational modification of actin filaments by oxidation is a way to trigger their disassembly.


Subject(s)
Actin Depolymerizing Factors , Cofilin 1 , Microfilament Proteins , Mixed Function Oxygenases , Actin Cytoskeleton , Actins/genetics , Cofilin 1/genetics , Cytoskeleton
14.
Sci Rep ; 10(1): 16803, 2020 10 08.
Article in English | MEDLINE | ID: mdl-33033331

ABSTRACT

Tunneling nanotubes (TNTs) are F-actin rich structures that connect distant cells, allowing the transport of many cellular components, including vesicles, organelles and molecules. Rab GTPases are the major regulators of vesicle trafficking and also participate in actin cytoskeleton remodelling, therefore, we examined their role in TNTs. Rab35 functions with several proteins that are involved in vesicle trafficking such as ACAP2, MICAL-L1, ARF6 and EHD1, which are known to be involved in neurite outgrowth. Here we show that Rab35 promotes TNT formation and TNT-mediated vesicle transfer in a neuronal cell line. Furthermore, our data indicates that Rab35-GTP, ACAP2, ARF6-GDP and EHD1 act in a cascade mechanism to promote TNT formation. Interestingly, MICAL-L1 overexpression, shown to be necessary for the action of Rab35 on neurite outgrowth, showed no effect on TNTs, indicating that TNT formation and neurite outgrowth may be processed through similar but not identical pathways, further supporting the unique identity of these cellular protrusions.


Subject(s)
Nanotubes , Neurons/metabolism , rab GTP-Binding Proteins/metabolism , ADP-Ribosylation Factor 6 , ADP-Ribosylation Factors/metabolism , Animals , Blotting, Western , Cell Line , Cytoplasmic Vesicles/metabolism , Cytoplasmic Vesicles/ultrastructure , Flow Cytometry , GTPase-Activating Proteins/metabolism , Mice , Microscopy, Electron, Scanning , Microscopy, Fluorescence , Nanotubes/ultrastructure , Neurons/ultrastructure , Vesicular Transport Proteins/metabolism
15.
Nat Commun ; 11(1): 2835, 2020 06 05.
Article in English | MEDLINE | ID: mdl-32503983

ABSTRACT

Inherited peripheral neuropathies (IPNs) represent a broad group of disorders including Charcot-Marie-Tooth (CMT) neuropathies characterized by defects primarily arising in myelin, axons, or both. The molecular mechanisms by which mutations in nearly 100 identified IPN/CMT genes lead to neuropathies are poorly understood. Here we show that the Ras-related GTPase Rab35 controls myelin growth via complex formation with the myotubularin-related phosphatidylinositol (PI) 3-phosphatases MTMR13 and MTMR2, encoded by genes responsible for CMT-types 4B2 and B1 in humans, and found that it downregulates lipid-mediated mTORC1 activation, a pathway known to crucially regulate myelin biogenesis. Targeted disruption of Rab35 leads to hyperactivation of mTORC1 signaling caused by elevated levels of PI 3-phosphates and to focal hypermyelination in vivo. Pharmacological inhibition of phosphatidylinositol 3,5-bisphosphate synthesis or mTORC1 signaling ameliorates this phenotype. These findings reveal a crucial role for Rab35-regulated lipid turnover by myotubularins to repress mTORC1 activity and to control myelin growth.


Subject(s)
Mechanistic Target of Rapamycin Complex 1/metabolism , Myelin Sheath/metabolism , rab GTP-Binding Proteins/metabolism , Animals , Astrocytes , Charcot-Marie-Tooth Disease/genetics , Charcot-Marie-Tooth Disease/pathology , Down-Regulation , Gene Knock-In Techniques , HEK293 Cells , HeLa Cells , Humans , Lipid Metabolism/genetics , Mice, Transgenic , Mutation , Myelin Sheath/pathology , Primary Cell Culture , Protein Tyrosine Phosphatases, Non-Receptor/antagonists & inhibitors , Protein Tyrosine Phosphatases, Non-Receptor/genetics , Protein Tyrosine Phosphatases, Non-Receptor/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , rab GTP-Binding Proteins/genetics
16.
Nat Commun ; 11(1): 1941, 2020 04 22.
Article in English | MEDLINE | ID: mdl-32321914

ABSTRACT

Cytokinesis requires the constriction of ESCRT-III filaments on the side of the midbody, where abscission occurs. After ESCRT recruitment at the midbody, it is not known how the ESCRT-III machinery localizes to the abscission site. To reveal actors involved in abscission, we obtained the proteome of intact, post-abscission midbodies (Flemmingsome) and identified 489 proteins enriched in this organelle. Among these proteins, we further characterized a plasma membrane-to-ESCRT module composed of the transmembrane proteoglycan syndecan-4, ALIX and syntenin, a protein that bridges ESCRT-III/ALIX to syndecans. The three proteins are highly recruited first at the midbody then at the abscission site, and their depletion delays abscission. Mechanistically, direct interactions between ALIX, syntenin and syndecan-4 are essential for proper enrichment of the ESCRT-III machinery at the abscission site, but not at the midbody. We propose that the ESCRT-III machinery must be physically coupled to a membrane protein at the cytokinetic abscission site for efficient scission, uncovering common requirements in cytokinesis, exosome formation and HIV budding.


Subject(s)
Calcium-Binding Proteins/metabolism , Cell Cycle Proteins/metabolism , Cell Membrane/metabolism , Cytokinesis , Endosomal Sorting Complexes Required for Transport/metabolism , Organelles/metabolism , Syndecan-4/metabolism , Syntenins/metabolism , Calcium-Binding Proteins/genetics , Cell Cycle Proteins/genetics , Cell Membrane/genetics , Endosomal Sorting Complexes Required for Transport/genetics , Endosomes/genetics , Endosomes/metabolism , HeLa Cells , Humans , Organelles/genetics , Protein Binding , Syndecan-4/genetics , Syntenins/genetics
17.
Proc Natl Acad Sci U S A ; 117(8): 4169-4179, 2020 02 25.
Article in English | MEDLINE | ID: mdl-32029597

ABSTRACT

Abscission is the terminal step of cytokinesis leading to the physical separation of the daughter cells. In response to the abnormal presence of lagging chromatin between dividing cells, an evolutionarily conserved abscission/NoCut checkpoint delays abscission and prevents formation of binucleated cells by stabilizing the cytokinetic intercellular bridge (ICB). How this bridge is stably maintained for hours while the checkpoint is activated is poorly understood and has been proposed to rely on F-actin in the bridge region. Here, we show that actin polymerization is indeed essential for stabilizing the ICB when lagging chromatin is present, but not in normal dividing cells. Mechanistically, we found that a cytosolic pool of human methionine sulfoxide reductase B2 (MsrB2) is strongly recruited at the midbody in response to the presence of lagging chromatin and functions within the ICB to promote actin polymerization there. Consistently, in MsrB2-depleted cells, F-actin levels are decreased in ICBs, and dividing cells with lagging chromatin become binucleated as a consequence of unstable bridges. We further demonstrate that MsrB2 selectively reduces oxidized actin monomers and thereby counteracts MICAL1, an enzyme known to depolymerize actin filaments by direct oxidation. Finally, MsrB2 colocalizes and genetically interacts with the checkpoint components Aurora B and ANCHR, and the abscission delay upon checkpoint activation by nuclear pore defects also depends on MsrB2. Altogether, this work reveals that actin reduction by MsrB2 is a key component of the abscission checkpoint that favors F-actin polymerization and limits tetraploidy, a starting point for tumorigenesis.


Subject(s)
Actins/metabolism , Chromatin/metabolism , Cytokinesis/physiology , Drosophila Proteins/metabolism , Methionine Sulfoxide Reductases/metabolism , Microfilament Proteins/metabolism , Mitosis/physiology , Animals , Cell Line , Drosophila , Drosophila Proteins/genetics , Endosomal Sorting Complexes Required for Transport/metabolism , HeLa Cells , Humans , Methionine Sulfoxide Reductases/genetics , Microfilament Proteins/genetics , Mixed Function Oxygenases/genetics , Mixed Function Oxygenases/metabolism , Oxidation-Reduction
18.
Curr Biol ; 29(20): R1074-R1077, 2019 10 21.
Article in English | MEDLINE | ID: mdl-31639350

ABSTRACT

Cytokinesis leads to the physical separation of the daughter cells and requires the constriction of ESCRT filaments. How the ESCRT machinery is recruited in non-vertebrate organisms was puzzling, and is now shown to rely on a direct interaction between the ESCRT-associated protein Alix and the kinesin motor Pavarotti in Drosophila.


Subject(s)
Cytokinesis , Virus Release , Animals , Calcium-Binding Proteins/genetics , Cell Division , Drosophila , Drosophila Proteins , Endosomal Sorting Complexes Required for Transport , Microtubule-Associated Proteins
19.
EMBO Rep ; 20(10): e47625, 2019 10 04.
Article in English | MEDLINE | ID: mdl-31432619

ABSTRACT

Rab and Arl guanine nucleotide-binding (G) proteins regulate trafficking pathways essential for the formation, function and composition of primary cilia, which are sensory devices associated with Sonic hedgehog (Shh) signalling and ciliopathies. Here, using mammalian cells and zebrafish, we uncover ciliary functions for Rab35, a multitasking G protein with endocytic recycling, actin remodelling and cytokinesis roles. Rab35 loss via siRNAs, morpholinos or knockout reduces cilium length in mammalian cells and the zebrafish left-right organiser (Kupffer's vesicle) and causes motile cilia-associated left-right asymmetry defects. Consistent with these observations, GFP-Rab35 localises to cilia, as do GEF (DENND1B) and GAP (TBC1D10A) Rab35 regulators, which also regulate ciliary length and Rab35 ciliary localisation. Mammalian Rab35 also controls the ciliary membrane levels of Shh signalling regulators, promoting ciliary targeting of Smoothened, limiting ciliary accumulation of Arl13b and the inositol polyphosphate 5-phosphatase (INPP5E). Rab35 additionally regulates ciliary PI(4,5)P2 levels and interacts with Arl13b. Together, our findings demonstrate roles for Rab35 in regulating cilium length, function and membrane composition and implicate Rab35 in pathways controlling the ciliary levels of Shh signal regulators.


Subject(s)
Cilia/metabolism , Phosphatidylinositol 4,5-Diphosphate/metabolism , Zebrafish Proteins/metabolism , Zebrafish/metabolism , rab GTP-Binding Proteins/metabolism , Animals , Body Patterning , Cell Line , HEK293 Cells , Humans , Membranes/metabolism , Mice , Models, Biological , NIH 3T3 Cells , Nucleotides/metabolism , Protein Binding , Protein Transport , Telomerase/metabolism
20.
Nat Rev Mol Cell Biol ; 20(11): 663, 2019 11.
Article in English | MEDLINE | ID: mdl-31160684
SELECTION OF CITATIONS
SEARCH DETAIL
...