Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 63
Filter
1.
EMBO J ; 42(24): e114557, 2023 Dec 11.
Article in English | MEDLINE | ID: mdl-37987147

ABSTRACT

Motile cells encounter microenvironments with locally heterogeneous mechanochemical composition. Individual compositional parameters, such as chemokines and extracellular matrix pore sizes, are well known to provide guidance cues for pathfinding. However, motile cells face diverse cues at the same time, raising the question of how they respond to multiple and potentially competing signals on their paths. Here, we reveal that amoeboid cells require nuclear repositioning, termed nucleokinesis, for adaptive pathfinding in heterogeneous mechanochemical micro-environments. Using mammalian immune cells and the amoeba Dictyostelium discoideum, we discover that frequent, rapid and long-distance nucleokinesis is a basic component of amoeboid pathfinding, enabling cells to reorientate quickly between locally competing cues. Amoeboid nucleokinesis comprises a two-step polarity switch and is driven by myosin-II forces that readjust the nuclear to the cellular path. Impaired nucleokinesis distorts path adaptions and causes cellular arrest in the microenvironment. Our findings establish that nucleokinesis is required for amoeboid cell navigation. Given that many immune cells, amoebae, and some cancer cells utilize an amoeboid migration strategy, these results suggest that nucleokinesis underlies cellular navigation during unicellular biology, immunity, and disease.


Subject(s)
Amoeba , Dictyostelium , Animals , Cell Movement , Extracellular Matrix , Mammals
2.
Mol Hum Reprod ; 28(10)2022 09 29.
Article in English | MEDLINE | ID: mdl-35944223

ABSTRACT

In the ovary, proliferation and differentiation of granulosa cells (GCs) drive follicular growth. Our immunohistochemical study in a non-human primate, the Rhesus monkey, showed that the mitochondrial activity marker protein cytochrome c oxidase subunit 4 (COX4) increases in GCs in parallel to follicle size, and furthermore, its intracellular localization changes. This suggested that there is mitochondrial biogenesis and trafficking, and implicates the actions of gonadotropins, which regulate follicular growth and ovulation. Human KGN cells, i.e. granulosa tumour cells, were therefore used to study these possibilities. To robustly elevate cAMP, and thereby mimic the actions of gonadotropins, we used forskolin (FSK). FSK increased the cell size and the amount of mitochondrial DNA of KGN cells within 24 h. As revealed by MitoTracker™ experiments and ultrastructural 3D reconstruction, FSK treatment induced the formation of elaborate mitochondrial networks. H89, a protein kinase A (PKA) inhibitor, reduced the network formation. A proteomic analysis indicated that FSK elevated the levels of regulators of the cytoskeleton, among others (data available via ProteomeXchange with identifier PXD032160). The steroidogenic enzyme CYP11A1 (Cytochrome P450 Family 11 Subfamily A Member 1), located in mitochondria, was more than 3-fold increased by FSK, implying that the cAMP/PKA-associated structural changes occur in parallel with the acquisition of steroidogenic competence of mitochondria in KGN cells. In summary, the observations show increases in mitochondria and suggest intracellular trafficking of mitochondria in GCs during follicular growth, and indicate that they may partially be under the control of gonadotropins and cAMP. In line with this, increased cAMP in KGN cells profoundly affected mitochondrial dynamics in a PKA-dependent manner and implicated cytoskeletal changes.


Subject(s)
Cholesterol Side-Chain Cleavage Enzyme , Electron Transport Complex IV , Animals , Cholesterol Side-Chain Cleavage Enzyme/genetics , Cholesterol Side-Chain Cleavage Enzyme/metabolism , Colforsin/pharmacology , Cyclic AMP-Dependent Protein Kinases/genetics , Cyclic AMP-Dependent Protein Kinases/metabolism , DNA, Mitochondrial , Electron Transport Complex IV/metabolism , Female , Gonadotropins/metabolism , Gonadotropins/pharmacology , Granulosa Cells/metabolism , Mitochondria/metabolism , Proteomics
3.
J Cell Sci ; 129(18): 3462-72, 2016 09 15.
Article in English | MEDLINE | ID: mdl-27505897

ABSTRACT

Chemotactic responses of eukaryotic cells require a signal processing system that translates an external gradient of attractant into directed motion. To challenge the response system to its limits, we increased the size of Dictyostelium discoideum cells by using electric-pulse-induced fusion. Large cells formed multiple protrusions at different sites along the gradient of chemoattractant, independently turned towards the gradient and competed with each other. Finally, these cells succeeded to re-establish polarity by coordinating front and tail activities. To analyse the responses, we combined two approaches, one aimed at local responses by visualising the dynamics of Ras activation at the front regions of reorientating cells, the other at global changes of polarity by monitoring front-to-tail-directed actin flow. Asymmetric Ras activation in turning protrusions underscores that gradients can be sensed locally and translated into orientation. Different to cells of normal size, the polarity of large cells is not linked to an increasing front-to-tail gradient of the PIP3-phosphatase PTEN. But even in large cells, the front communicates with the tail through an actin flow that might act as carrier of a protrusion inhibitor.


Subject(s)
Actins/metabolism , Cell Size , Chemotaxis , Dictyostelium/cytology , Dictyostelium/metabolism , PTEN Phosphohydrolase/metabolism , Rheology , ras Proteins/metabolism , Cell Size/drug effects , Cell Surface Extensions/metabolism , Chemotactic Factors/pharmacology , Chemotaxis/drug effects , Cyclic AMP/metabolism , Dictyostelium/drug effects , Diffusion , Pinocytosis/drug effects
4.
Proc Natl Acad Sci U S A ; 111(1): E25-33, 2014 Jan 07.
Article in English | MEDLINE | ID: mdl-24347642

ABSTRACT

The Cdc42- and Rac-interactive binding motif (CRIB) of coronin binds to Rho GTPases with a preference for GDP-loaded Rac. Mutation of the Cdc42- and Rac-interactive binding motif abrogates Rac binding. This results in increased 1evels of activated Rac in coronin-deficient Dictyostelium cells (corA(-)), which impacts myosin II assembly. corA(-) cells show increased accumulation of myosin II in the cortex of growth-phase cells. Myosin II assembly is regulated by myosin heavy chain kinase-mediated phosphorylation of its tail. Kinase activity depends on the activation state of the p21-activated kinase a. The myosin II defect of corA(-) mutant is alleviated by dominant-negative p21-activated kinase a. It is rescued by wild-type coronin, whereas coronin carrying a mutated Cdc42- and Rac-interactive binding motif failed to rescue the myosin defect in corA(-) mutant cells. Ectopically expressed myosin heavy chain kinases affinity purified from corA(-) cells show reduced kinase activity. We propose that coronin through its affinity for GDP-Rac regulates the availability of GTP-Rac for activation of downstream effectors.


Subject(s)
4-Butyrolactone/analogs & derivatives , Gene Expression Regulation , cdc42 GTP-Binding Protein/metabolism , rac GTP-Binding Proteins/metabolism , 4-Butyrolactone/genetics , 4-Butyrolactone/metabolism , Amino Acid Motifs , Amino Acid Sequence , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cytoskeleton/metabolism , Dictyostelium/genetics , Dictyostelium/metabolism , Microscopy, Fluorescence , Models, Molecular , Molecular Sequence Data , Mutation , Phosphorylation , Protein Interaction Mapping , Protein Structure, Tertiary , Protozoan Proteins/metabolism , Signal Transduction
5.
Biochem Biophys Res Commun ; 467(4): 730-5, 2015 Nov 27.
Article in English | MEDLINE | ID: mdl-26482849

ABSTRACT

Dictyostelium discoideum cells resemble in many aspects human leukocytes and serve as a model to study actin cytoskeleton dynamics and cell migration of highly motile cells. Dictyostelium cells deficient in the actin-binding protein filamin (ddFLN) showed a surprisingly subtle change in phenotype with no or only minor effects in single cell motility. These findings were in contrast to the strong actin-crosslinking activities measured for filamin in vitro. In the present study, we set out to revisit the role of ddFLN in cell migration. For this purpose, we examined migration of wild-type, ddFLN-null and ddFLN-overexpressing cells under different conditions. In addition to cyclic-AMP chemotaxis assays using micropipettes, we explored cell migration under more confined conditions: an under-agarose 2D assay and a 3D assay employing a collagen matrix that was adapted from assays for leukocytes. Using 3D migration conditions, cells deficient in ddFLN displayed only a minor impairment of motility, similar to the results obtained for migration in 2D. However, cells overexpressing ddFLN showed a remarkable decrease in the speed of migration in particular in 3D environments. We suggest that these results are in line with an increased stiffening of the cortex due to the crosslinking activity of overexpressed ddFLN. Our conclusion is that the absolute level of ddFLN is critical for efficient migration. Furthermore, our results show that under conditions of increased mechanical stress, Dictyostelium cells, like leukocytes, switch to a bleb-based mode of movement.


Subject(s)
Chemotaxis , Dictyostelium/physiology , Dictyostelium/cytology , Filamins/physiology
6.
PLoS Pathog ; 9(9): e1003598, 2013 Sep.
Article in English | MEDLINE | ID: mdl-24068924

ABSTRACT

The causative agent of Legionnaires' disease, Legionella pneumophila, uses the Icm/Dot type IV secretion system (T4SS) to form in phagocytes a distinct "Legionella-containing vacuole" (LCV), which intercepts endosomal and secretory vesicle trafficking. Proteomics revealed the presence of the small GTPase Ran and its effector RanBP1 on purified LCVs. Here we validate that Ran and RanBP1 localize to LCVs and promote intracellular growth of L. pneumophila. Moreover, the L. pneumophila protein LegG1, which contains putative RCC1 Ran guanine nucleotide exchange factor (GEF) domains, accumulates on LCVs in an Icm/Dot-dependent manner. L. pneumophila wild-type bacteria, but not strains lacking LegG1 or a functional Icm/Dot T4SS, activate Ran on LCVs, while purified LegG1 produces active Ran(GTP) in cell lysates. L. pneumophila lacking legG1 is compromised for intracellular growth in macrophages and amoebae, yet is as cytotoxic as the wild-type strain. A downstream effect of LegG1 is to stabilize microtubules, as revealed by conventional and stimulated emission depletion (STED) fluorescence microscopy, subcellular fractionation and Western blot, or by microbial microinjection through the T3SS of a Yersinia strain lacking endogenous effectors. Real-time fluorescence imaging indicates that LCVs harboring wild-type L. pneumophila rapidly move along microtubules, while LCVs harboring ΔlegG1 mutant bacteria are stalled. Together, our results demonstrate that Ran activation and RanBP1 promote LCV formation, and the Icm/Dot substrate LegG1 functions as a bacterial Ran activator, which localizes to LCVs and promotes microtubule stabilization, LCV motility as well as intracellular replication of L. pneumophila.


Subject(s)
Bacterial Proteins/metabolism , GTPase-Activating Proteins/metabolism , Legionella pneumophila/physiology , Macrophages/microbiology , Microtubules/metabolism , Phagosomes/metabolism , ran GTP-Binding Protein/metabolism , Animals , Bacterial Proteins/genetics , Cell Line , Enzyme Activation , GTPase-Activating Proteins/antagonists & inhibitors , GTPase-Activating Proteins/genetics , Gene Silencing , Humans , Legionella pneumophila/genetics , Legionella pneumophila/immunology , Legionella pneumophila/ultrastructure , Legionnaires' Disease/immunology , Legionnaires' Disease/metabolism , Legionnaires' Disease/microbiology , Legionnaires' Disease/pathology , Macrophages/immunology , Macrophages/metabolism , Macrophages/ultrastructure , Mice , Microtubule Proteins/chemistry , Microtubule Proteins/metabolism , Microtubules/ultrastructure , Mutation , Phagocytosis , Phagosomes/enzymology , Phagosomes/ultrastructure , Polymerization , Protein Stability , Protein Transport , Virus Replication , ran GTP-Binding Protein/antagonists & inhibitors , ran GTP-Binding Protein/genetics
7.
Cell Microbiol ; 16(7): 977-92, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24397557

ABSTRACT

The environmental bacterium Legionella pneumophila causes a severe pneumonia termed Legionnaires' disease. L. pneumophila employs a conserved mechanism to replicate within a specific vacuole in macrophages or protozoa such as the social soil amoeba Dictyostelium discoideum. Pathogen-host interactions depend on the Icm/Dot type IV secretion system (T4SS), which translocates approximately 300 different effector proteins into host cells. Here we analyse the effects of L. pneumophila on migration and chemotaxis of amoebae, macrophages or polymorphonuclear neutrophils (PMN). Using under-agarose assays, L. pneumophila inhibited in a dose- and T4SS-dependent manner the migration of D. discoideum towards folate as well as starvation-induced aggregation of the social amoebae. Similarly, L. pneumophila impaired migration of murine RAW 264.7 macrophages towards the cytokines CCL5 and TNFα, or of primary human PMN towards the peptide fMLP respectively. L. pneumophila lacking the T4SS-translocated activator of the small eukaryotic GTPase Ran, Lpg1976/LegG1, hyper-inhibited the migration of D. discoideum, macrophages or PMN. The phenotype was reverted by plasmid-encoded LegG1 to an extent observed for mutant bacteria lacking a functional Icm/Dot T4SS.Similarly, LegG1 promoted random migration of L. pneumophila-infected macrophages and A549 epithelial cells in a Ran-dependent manner, or upon 'microbial microinjection' into HeLa cells by a Yersinia strain lacking endogenous effectors. Single-cell tracking and real-time analysis of L. pneumophila-infected phagocytes revealed that the velocity and directionality of the cells were decreased, and cell motility as well as microtubule dynamics was impaired. Taken together, these findings indicate that the L. pneumophila Ran activator LegG1 and consequent microtubule polymerization are implicated in Icm/Dot-dependent inhibition of phagocyte migration.


Subject(s)
Bacterial Proteins/metabolism , Cell Movement , Legionella pneumophila/physiology , Macrophages/microbiology , Neutrophils/microbiology , Animals , Bacterial Proteins/genetics , Bacterial Secretion Systems , Cell Line , Dictyostelium/microbiology , Enzyme Activators/metabolism , Host-Pathogen Interactions , Humans , Macrophages/physiology , Mice , Microtubules/metabolism , Neutrophils/physiology , Protein Transport , ran GTP-Binding Protein/metabolism
8.
BMC Cell Biol ; 15: 25, 2014 Jul 01.
Article in English | MEDLINE | ID: mdl-24986648

ABSTRACT

BACKGROUND: Nuclear Dbf-related/large tumor suppressor (NDR/LATS) kinases have been shown recently to control pathways that regulate mitotic exit, cytokinesis, cell growth, morphological changes and apoptosis. LATS kinases are core components of the Hippo signaling cascade and important tumor suppressors controlling cell proliferation and organ size in flies and mammals, and homologs are also present in yeast and Dictyostelium discoideum. Ras proto-oncogens regulate many biological functions, including differentiation, proliferation and apoptosis. Dysfunctions of LATS kinases or Ras GTPases have been implicated in the development of a variety of cancers in humans. RESULTS: In this study we used the model organism Dictyostelium discoideum to analyze the functions of NdrC, a homolog of the mammalian LATS2 protein, and present a novel regulatory mechanism for this kinase. Deletion of the ndrC gene caused impaired cell division and loss of centrosome integrity. A yeast two-hybrid analysis, using activated Ras proteins as bait, revealed NdrC as an interactor and identified its Ras-binding domain. Further in vitro pull-down assays showed that NdrC binds RasG and RasB, and to a lesser extent RasC and Rap1. In cells lacking NdrC, the levels of activated RasB and RasG are up-regulated, suggesting a functional connection between RasB, RasG, and NdrC. CONCLUSIONS: Dictyostelium discoideum NdrC is a LATS2-homologous kinase that is important for the regulation of cell division. NdrC contains a Ras-binding domain and interacts preferentially with RasB and RasG. Changed levels of both, RasB or RasG, have been shown previously to interfere with cell division. Since a defect in cell division is exhibited by NdrC-null cells, RasG-null cells, and cells overexpressing activated RasB, we propose a model for the regulation of cytokinesis by NdrC that involves the antagonistic control by RasB and RasG.


Subject(s)
Dictyostelium/cytology , Dictyostelium/enzymology , Protein Serine-Threonine Kinases/metabolism , Protozoan Proteins/metabolism , Tumor Suppressor Proteins/metabolism , ras Proteins/metabolism , Amino Acid Sequence , Animals , Cell Division , Dictyostelium/chemistry , Humans , Molecular Sequence Data , Protein Serine-Threonine Kinases/analysis , Protozoan Proteins/analysis , Signal Transduction , Tumor Suppressor Proteins/analysis , ras Proteins/analysis
9.
Cell Commun Signal ; 12: 37, 2014 Jun 15.
Article in English | MEDLINE | ID: mdl-24930026

ABSTRACT

BACKGROUND: The receptor for activated C-kinase 1 (RACK1) is a conserved protein belonging to the WD40 repeat family of proteins. It folds into a beta propeller with seven blades which allow interactions with many proteins. Thus it can serve as a scaffolding protein and have roles in several cellular processes. RESULTS: We identified the product of the Dictyostelium discoideum gpbB gene as the Dictyostelium RACK1 homolog. The protein is mainly cytosolic but can also associate with cellular membranes. DdRACK1 binds to phosphoinositides (PIPs) in protein-lipid overlay and liposome-binding assays. The basis of this activity resides in a basic region located in the extended loop between blades 6 and 7 as revealed by mutational analysis. Similar to RACK1 proteins from other organisms DdRACK1 interacts with G protein subunits alpha, beta and gamma as shown by yeast two-hybrid, pulldown, and immunoprecipitation assays. Unlike the Saccharomyces cerevisiae and Cryptococcus neoformans RACK1 proteins it does not appear to take over Gß function in D. discoideum as developmental and other defects were not rescued in Gß null mutants overexpressing GFP-DdRACK1. Overexpression of GFP-tagged DdRACK1 and a mutant version (DdRACK1mut) which carried a charge-reversal mutation in the basic region in wild type cells led to changes during growth and development. CONCLUSION: DdRACK1 interacts with heterotrimeric G proteins and can through these interactions impact on processes specifically regulated by these proteins.


Subject(s)
Dictyostelium/metabolism , GTP-Binding Proteins/metabolism , Gene Expression Regulation, Developmental , Protozoan Proteins/metabolism , Amino Acid Sequence , Dictyostelium/growth & development , GTP-Binding Proteins/chemistry , GTP-Binding Proteins/genetics , Molecular Sequence Data , Organ Specificity , Phosphatidylinositols/metabolism , Protein Binding , Protein Multimerization , Protein Subunits/chemistry , Protein Subunits/genetics , Protein Subunits/metabolism , Protein Transport , Protozoan Proteins/chemistry , Protozoan Proteins/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism
10.
Traffic ; 12(3): 301-12, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21134080

ABSTRACT

Dictyostelium discoideum cells are professional phagocytes that provide an easily accessible system to gain insights into the mechanisms and the regulatory machinery controlling phagocytosis. Here, we describe a novel function for nuclear Dbf2-related (NDR) family kinases in phagocytosis of D. discoideum. Deletion of one of the four NDR kinases of D. discoideum, NdrA, resulted in impaired cell growth caused by reduced phagocytosis rates. Detailed analysis of NdrA-null cells revealed that the formation of phagocytic cups was delayed. Microscopic investigations showed that NdrA localizes to centrosomes, and NdrA was also identified in isolated centrosome preparations. The localization of NdrA is regulated during the cell cycle. In prophase, NdrA disappears from the centrosome and forms a cloud-like structure around the spindle, which is totally absent during later stages until completion of mitosis. Our results suggest that a signal which originates from the NdrA kinase at the centrosome affects the efficiency of phagocytosis. We assume that in NdrA-null cells the defects in phagocytosis may be caused by an impairment of vesicle trafficking, which is involved in providing new membrane at the sites of particle uptake.


Subject(s)
Dictyostelium/enzymology , Phagocytosis/physiology , Protein Serine-Threonine Kinases/metabolism , Protozoan Proteins/metabolism , Cell Cycle/physiology , Centrosome/enzymology , Dictyostelium/genetics , Fluorescent Antibody Technique , Gene Expression Regulation, Enzymologic , Phagocytosis/genetics , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/genetics , Protein Transport , Protozoan Proteins/genetics
11.
Biochem Biophys Res Commun ; 431(3): 490-5, 2013 Feb 15.
Article in English | MEDLINE | ID: mdl-23333327

ABSTRACT

Kinesins are ATP-dependent molecular motors that mediate unidirectional intracellular transport along microtubules. Dictyostelium discoideum has 13 different kinesin isoforms including two members of the kinesin-7 family, Kif4 and Kif11. While Kif4 is structurally and functionally related to centromere-associated CENP-E proteins involved in the transport of chromosomes to the poles during mitosis, the function of the unusually short CENP-E variant Kif11 is unclear. Here we show that orthologs of short CENP-E variants are present in plants and fungi, and analyze functional properties of the Dictyostelium CENP-E version, Kif11. Gene knockout mutants reveal that Kif11 is not required for mitosis or development. Imaging of GFP-labeled Kif11 expressing Dictyostelium cells indicates that Kif11 is a plus-end directed motor that accumulates at microtubule plus ends. By multiple motor gliding assays, we show that Kif11 moves with an average velocity of 38nm/s, thus defining Kif11 as a very slow motor. The activity of the Kif11 motor appears to be modulated via interactions with the non-catalytic tail region. Our work highlights a subclass of kinesin-7-like motors that function outside of a role in mitosis.


Subject(s)
Adenosine Triphosphatases/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Dictyostelium/metabolism , Kinesins/metabolism , Adenosine Triphosphatases/classification , Adenosine Triphosphatases/genetics , Chromosomal Proteins, Non-Histone/classification , Chromosomal Proteins, Non-Histone/genetics , Dictyostelium/genetics , Gene Knockout Techniques , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Kinesins/classification , Kinesins/genetics , Mitosis , Phylogeny , Protein Structure, Secondary
12.
Biophys J ; 103(6): 1170-8, 2012 Sep 19.
Article in English | MEDLINE | ID: mdl-22995489

ABSTRACT

In a motile eukaryotic cell, front protrusion and tail retraction are superimposed on each other. To single out mechanisms that result in front to tail or in tail to front transition, we separated the two processes in time using cells that oscillate between a full front and a full tail state. State transitions were visualized by total internal reflection fluorescence microscopy using as a front marker PIP3 (phosphatidylinositol [3,4,5] tris-phosphate), and as a tail marker the tumor-suppressor PTEN (phosphatase tensin homolog) that degrades PIP3. Negative fluctuations in the PTEN layer of the membrane gated a local increase in PIP3. In a subset of areas lacking PTEN (PTEN holes), PIP3 was amplified until a propagated wave was initiated. Wave propagation implies that a PIP3 signal is transmitted by a self-sustained process, such that the temporal and spatial profiles of the signal are maintained during passage of the wave across the entire expanse of the cell membrane. Actin clusters were remodeled into a ring along the perimeter of the expanding PIP3 wave. The reverse transition of PIP3 to PTEN was linked to the previous site of wave initiation: where PIP3 decayed first, the entry of PTEN was primed.


Subject(s)
Cell Polarity , PTEN Phosphohydrolase/metabolism , Phosphatidylinositol Phosphates/metabolism , Actins/chemistry , Dictyostelium/cytology , Dictyostelium/enzymology , Kinetics , Microscopy, Fluorescence , Protein Multimerization , Protein Structure, Quaternary , Stochastic Processes
13.
Cell Mol Life Sci ; 68(2): 303-13, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20640912

ABSTRACT

Dictyostelium discoideum harbors a short (CRN12) and a long coronin (CRN7) composed of one and two beta-propellers, respectively. They are primarily present in the cell cortex and cells lacking CRN12 (corA⁻) or CRN7 (corB⁻) have defects in actin driven processes. We compared the characteristics of a mutant cell line (corA⁻/corB⁻) lacking CRN12 and CRN7 with the single mutants focusing on cytokinesis, phagocytosis, chemotaxis and development. Cytokinesis, uptake of small particles, and developmental defects were not enhanced in the corA⁻/corB⁻ strain as compared to the single mutants, whereas motility and phagocytosis of yeast particles were more severely impaired. It appears that although both proteins affect the same processes they do not act in a redundant manner. Rather, they often act antagonistically, which is in accordance with their proposed roles in the actin cytoskeleton where CRN12 acts in actin disassembly whereas CRN7 stabilizes actin filaments and protects them from disassembly.


Subject(s)
Actins , Cytoskeleton , Dictyostelium/metabolism , Microfilament Proteins , Actins/metabolism , Animals , Chemotaxis/physiology , Cytokinesis/physiology , Cytoskeleton/metabolism , Dictyostelium/growth & development , Dictyostelium/ultrastructure , Escherichia coli , Gene Deletion , Legionella pneumophila , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , Phagocytosis/physiology , Protozoan Proteins/genetics , Protozoan Proteins/metabolism , Saccharomyces cerevisiae , Transfection
14.
Nucleus ; 13(1): 144-154, 2022 12.
Article in English | MEDLINE | ID: mdl-35298348

ABSTRACT

Dictyostelium amoebae perform a semi-closed mitosis, in which the nuclear envelope is fenestrated at the insertion sites of the mitotic centrosomes and around the central spindle during karyokinesis. During late telophase the centrosome relocates to the cytoplasmic side of the nucleus, the central spindle disassembles and the nuclear fenestrae become closed. Our data indicate that Dictyostelium spastin (DdSpastin) is a microtubule-binding and severing type I membrane protein that plays a role in this process. Its mitotic localization is in agreement with a requirement for the removal of microtubules that would hinder closure of the fenestrae. Furthermore, DdSpastin interacts with the HeH/ LEM-family protein Src1 in BioID analyses as well as the inner nuclear membrane protein Sun1, and shows subcellular co-localizations with Src1, Sun1, the ESCRT component CHMP7 and the IST1-like protein filactin, suggesting that the principal pathway of mitotic nuclear envelope remodeling is conserved between animals and Dictyostelium amoebae.


Subject(s)
Dictyostelium , Nuclear Envelope , Animals , Cell Nucleus Division , Dictyostelium/metabolism , Mitosis , Nuclear Envelope/metabolism , Spastin/metabolism
15.
Cells ; 11(11)2022 05 29.
Article in English | MEDLINE | ID: mdl-35681473

ABSTRACT

Directional cell migration and the establishment of polarity play an important role in development, wound healing, and host cell defense. While actin polymerization provides the driving force at the cell front, the microtubule network assumes a regulatory function, in coordinating front protrusion and rear retraction. By using Dictyostelium discoideum cells as a model for amoeboid movement in different 2D and 3D environments, the position of the centrosome relative to the nucleus was analyzed using live-cell microscopy. Our results showed that the centrosome was preferentially located rearward of the nucleus under all conditions tested for directed migration, while the nucleus was oriented toward the expanding front. When cells are hindered from straight movement by obstacles, the centrosome is displaced temporarily from its rearward location to the side of the nucleus, but is reoriented within seconds. This relocalization is supported by the presence of intact microtubules and their contact with the cortex. The data suggest that the centrosome is responsible for coordinating microtubules with respect to the nucleus. In summary, we have analyzed the orientation of the centrosome during different modes of migration in an amoeboid model and present evidence that the basic principles of centrosome positioning and movement are conserved between Dictyostelium and human leukocytes.


Subject(s)
Dictyostelium , Cell Movement , Cell Nucleus , Centrosome , Humans , Microtubules
16.
J Biol Chem ; 285(12): 9249-61, 2010 Mar 19.
Article in English | MEDLINE | ID: mdl-20071332

ABSTRACT

Dictyostelium discoideum Coronin7 (DdCRN7) together with human Coronin7 (CRN7) and Pod-1 of Drosophila melanogaster and Caenorhabditis elegans belong to the coronin family of WD-repeat domain-containing proteins. Coronin7 proteins are characterized by two WD-repeat domains that presumably fold into two beta-propeller structures. DdCRN7 shares highest homology with human CRN7, a protein with roles in membrane trafficking. DdCRN7 is present in the cytosol and accumulates in cell surface projections during movement and phago- and pinocytosis. Cells lacking CRN7 have altered chemotaxis and phagocytosis. Furthermore, loss of CRN7 affects the infection process by the pathogen Legionella pneumophila and allows a more efficient internalization of bacteria. To provide a mechanism for CNR7 action, we studied actin-related aspects. We could show that CRN7 binds directly to F-actin and protects actin filaments from depolymerization. CRN7 also associated with F-actin in vivo. It was present in the Triton X-100-insoluble cytoskeleton, colocalized with F-actin, and its distribution was sensitive to drugs affecting the actin cytoskeleton. We propose that the CRN7 role in chemotaxis and phagocytosis is through its effect on the actin cytoskeleton.


Subject(s)
Actins/chemistry , Dictyostelium/metabolism , Microfilament Proteins/chemistry , Cell Adhesion , Cell Movement , Chemotaxis , Cyclic AMP/metabolism , Cytoskeleton/metabolism , Glutathione Transferase/metabolism , Humans , Legionella pneumophila/metabolism , Models, Biological , Mutation , Phagocytosis , Pinocytosis , Protein Structure, Tertiary
17.
Biochem Biophys Res Commun ; 404(4): 1016-22, 2011 Jan 28.
Article in English | MEDLINE | ID: mdl-21187070

ABSTRACT

Posttranslational histone modifications play an important role in modulating gene expression and chromatin structure. Here we report the identification of histone H3K79 dimethylation in the simple eukaryote Dictyostelium discoideum. We have deleted the D. discoideum Dot1/KMT4 homologue and demonstrate that it is the sole enzyme responsible for histone H3K79me2. Cells lacking Dot1 are reduced in growth and delayed in development, but do not show apparent changes in cell cycle regulation. Furthermore, our results indicate that Dot1 contributes to UV damage resistance and DNA repair in D. discoideum. In summary, the data support the view that the machinery controlling the setting of histone marks is evolutionary highly conserved and provide evidence that D. discoideum is a suitable model system to analyze these modifications and their functions during development and differentiation.


Subject(s)
DNA Damage , DNA Repair , Dictyostelium/growth & development , Histone-Lysine N-Methyltransferase/metabolism , Amino Acid Sequence , Dictyostelium/enzymology , Dictyostelium/genetics , Histone-Lysine N-Methyltransferase/genetics , Histones/metabolism , Methylation , Molecular Sequence Data
18.
BMC Biol ; 8: 154, 2010 Dec 29.
Article in English | MEDLINE | ID: mdl-21190565

ABSTRACT

BACKGROUND: The uptake of particles by actin-powered invagination of the plasma membrane is common to protozoa and to phagocytes involved in the immune response of higher organisms. The question addressed here is how a phagocyte may use geometric cues to optimize force generation for the uptake of a particle. We survey mechanisms that enable a phagocyte to remodel actin organization in response to particles of complex shape. RESULTS: Using particles that consist of two lobes separated by a neck, we found that Dictyostelium cells transmit signals concerning the curvature of a surface to the actin system underlying the plasma membrane. Force applied to a concave region can divide a particle in two, allowing engulfment of the portion first encountered. The phagosome membrane that is bent around the concave region is marked by a protein containing an inverse Bin-Amphiphysin-Rvs (I-BAR) domain in combination with an Src homology (SH3) domain, similar to mammalian insulin receptor tyrosine kinase substrate p53. Regulatory proteins enable the phagocyte to switch activities within seconds in response to particle shape. Ras, an inducer of actin polymerization, is activated along the cup surface. Coronin, which limits the lifetime of actin structures, is reversibly recruited to the cup, reflecting a program of actin depolymerization. The various forms of myosin-I are candidate motor proteins for force generation in particle uptake, whereas myosin-II is engaged only in retracting a phagocytic cup after a switch to particle release. Thus, the constriction of a phagocytic cup differs from the contraction of a cleavage furrow in mitosis. CONCLUSIONS: Phagocytes scan a particle surface for convex and concave regions. By modulating the spatiotemporal pattern of actin organization, they are capable of switching between different modes of interaction with a particle, either arresting at a concave region and applying force in an attempt to sever the particle there, or extending the cup along the particle surface to identify the very end of the object to be ingested. Our data illustrate the flexibility of regulatory mechanisms that are at the phagocyte's disposal in exploring an environment of irregular geometry.


Subject(s)
Organelle Shape/physiology , Phagocytes/physiology , Phagocytosis/physiology , Actin Cytoskeleton/chemistry , Actin Cytoskeleton/metabolism , Actins/metabolism , Dictyostelium/cytology , Dictyostelium/genetics , Dictyostelium/metabolism , Dictyostelium/physiology , Fluorescence Recovery After Photobleaching , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Models, Biological , Myosins/metabolism , Organisms, Genetically Modified , Phagocytes/metabolism , Phagocytosis/genetics , Phosphatidylinositols/metabolism , Phosphatidylinositols/physiology , Physical Phenomena , Protein Multimerization/physiology , ras Proteins/metabolism , ras Proteins/physiology
19.
Front Cell Dev Biol ; 9: 742310, 2021.
Article in English | MEDLINE | ID: mdl-34869330

ABSTRACT

Actin dynamics plays a crucial role in regulating essential cell functions and thereby is largely responsible to a considerable extent for cellular energy consumption. Certain pathological conditions in humans, like neurological disorders such as Alzheimer's disease or amyotrophic lateral sclerosis (ALS) as well as variants of nemaline myopathy are associated with cytoskeletal abnormalities, so-called actin-cofilin rods. Actin-cofilin rods are aggregates consisting mainly of actin and cofilin, which are formed as a result of cellular stress and thereby help to ensure the survival of cells under unfavorable conditions. We have used Dictyostelium discoideum, an established model system for cytoskeletal research to study formation and principles of cytoplasmic actin rod assembly in response to energy depletion. Experimentally, depletion of ATP was provoked by addition of either sodium azide, dinitrophenol, or 2-deoxy-glucose, and the formation of rod assembly was recorded by live-cell imaging. Furthermore, we show that hyperosmotic shock induces actin-cofilin rods, and that a drop in the intracellular pH accompanies this condition. Our data reveal that acidification of the cytoplasm can induce the formation of actin-cofilin rods to varying degrees and suggest that a local reduction in cellular pH may be a cause for the formation of cytoplasmic rods. We hypothesize that local phase separation mechanistically triggers the assembly of actin-cofilin rods and thereby influences the material properties of actin structures.

20.
Cell Motil Cytoskeleton ; 66(11): 929-39, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19479821

ABSTRACT

During a REMI screen for proteins regulating cytokinesis in Dictyostelium discoideum we isolated a mutant forming multinucleate cells. The gene affected in this mutant encoded a kinase, SepA, which is an ortholog of Cdc7, a serine-threonine kinase essential for septum formation in Schizosaccharomyces pombe. Localization of SepA-GFP in live cells and its presence in isolated centrosomes indicated that SepA, like its upstream regulator Spg1, is associated with centrosomes. Knockout mutants of SepA showed a severe cytokinesis defect and a delay in development. In multinucleate SepA-null cells nuclear division proceeded normally and synchronously. However, often cleavage furrows were either missing or atypical: they were extremely asymmetric and constriction was impaired. Cortexillin-I, a marker localizing strictly to the furrow in wild-type cells, demonstrated that large, crescent-shaped furrows expanded and persisted long after the spindle regressed and nuclei returned to the interphase state. Outside the furrow the filamentous actin system of the cell cortex showed strong ruffling activity. These data suggest that SepA is involved in the spatial and temporal control system organizing cortical activities in mitotic and postmitotic cells.


Subject(s)
Cytokinesis/physiology , Dictyostelium/ultrastructure , Protein Serine-Threonine Kinases/metabolism , Actins/metabolism , Actins/ultrastructure , Animals , Cell Cycle Proteins/genetics , Cell Surface Extensions/ultrastructure , Centrosome/metabolism , Centrosome/ultrastructure , Dictyostelium/chemistry , Microscopy, Confocal , Protein Serine-Threonine Kinases/genetics , Schizosaccharomyces pombe Proteins/genetics , Spindle Apparatus/physiology , Spindle Apparatus/ultrastructure
SELECTION OF CITATIONS
SEARCH DETAIL