Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
2.
J Cell Sci ; 134(6)2021 03 26.
Article in English | MEDLINE | ID: mdl-33602740

ABSTRACT

Movement of the cell nucleus typically involves the cytoskeleton and either polymerization-based pushing forces or motor-based pulling forces. In the fission yeast Schizosaccharomyces pombe, nuclear movement and positioning are thought to depend on microtubule polymerization-based pushing forces. Here, we describe a novel, microtubule-independent, form of nuclear movement in fission yeast. Microtubule-independent nuclear movement is directed towards growing cell tips, and it is strongest when the nucleus is close to a growing cell tip, and weakest when the nucleus is far from that tip. Microtubule-independent nuclear movement requires actin cables but does not depend on actin polymerization-based pushing or myosin V-based pulling forces. The vesicle-associated membrane protein (VAMP)-associated proteins (VAPs) Scs2 and Scs22, which are critical for endoplasmic reticulum-plasma membrane contact sites in fission yeast, are also required for microtubule-independent nuclear movement. We also find that in cells in which microtubule-based pushing forces are present, disruption of actin cables leads to increased fluctuations in interphase nuclear positioning and subsequent altered septation. Our results suggest two non-exclusive mechanisms for microtubule-independent nuclear movement, which may help illuminate aspects of nuclear positioning in other cells.


Subject(s)
Schizosaccharomyces pombe Proteins , Schizosaccharomyces , Cell Nucleus , Interphase , Microtubules , Schizosaccharomyces pombe Proteins/genetics
3.
Curr Biol ; 29(13): 2199-2207.e10, 2019 07 08.
Article in English | MEDLINE | ID: mdl-31287970

ABSTRACT

Microtubule (MT) nucleation depends on the γ-tubulin complex (γ-TuC), in which multiple copies of the heterotetrameric γ-tubulin small complex (γ-TuSC) associate to form a ring-like structure (in metazoans, γ-tubulin ring complex; γ-TuRC) [1-7]. Additional conserved regulators of the γ-TuC include the small protein Mzt1 (MOZART1 in human; GIP1/1B and GIP2/1A in plants) [8-13] and proteins containing a Centrosomin Motif 1 (CM1) domain [10, 14-19]. Many insights into γ-TuC regulators have come from in vivo analysis in fission yeast Schizosaccharomyces pombe. The S. pombe CM1 protein Mto1 recruits the γ-TuC to microtubule-organizing centers (MTOCs) [14, 20-22], and analysis of Mto1[bonsai], a truncated version of Mto1 that cannot localize to MTOCs, has shown that Mto1 also has a role in γ-TuC activation [23]. S. pombe Mzt1 interacts with γ-TuSC and is essential for γ-TuC function and localization to MTOCs [11, 12]. However, the mechanisms by which Mzt1 functions remain unclear. Here we describe reconstitution of MT nucleation using purified recombinant Mto1[bonsai], the Mto1 partner protein Mto2, γ-TuSC, and Mzt1. Multiple copies of the six proteins involved coassemble to form a 34-40S ring-like "MGM" holocomplex that is a potent MT nucleator in vitro. Using purified MGM and subcomplexes, we investigate the role of Mzt1 in MT nucleation. Our results suggest that Mzt1 is critical to stabilize Alp6, the S. pombe homolog of human γ-TuSC protein GCP3, in an "interaction-competent" form within the γ-TuSC. This is essential for MGM to become a functional nucleator.


Subject(s)
Microtubule-Associated Proteins/genetics , Microtubules/metabolism , Schizosaccharomyces pombe Proteins/genetics , Schizosaccharomyces/genetics , Microtubule-Associated Proteins/metabolism , Microtubule-Organizing Center/metabolism , Schizosaccharomyces/metabolism , Schizosaccharomyces pombe Proteins/metabolism , Tubulin/metabolism
4.
Cell Rep ; 26(6): 1654-1667.e7, 2019 02 05.
Article in English | MEDLINE | ID: mdl-30726745

ABSTRACT

NDR/LATS kinases regulate multiple aspects of cell polarity and morphogenesis from yeast to mammals. Fission yeast NDR/LATS kinase Orb6 has been proposed to control cell polarity by regulating the Cdc42 guanine nucleotide exchange factor Gef1. Here, we show that Orb6 regulates polarity largely independently of Gef1 and that Orb6 positively regulates exocytosis. Through Orb6 inhibition in vivo and quantitative global phosphoproteomics, we identify Orb6 targets, including proteins involved in membrane trafficking. We confirm Sec3 and Sec5, conserved components of the exocyst complex, as substrates of Orb6 both in vivo and in vitro, and we show that Orb6 kinase activity is important for exocyst localization to cell tips and for exocyst activity during septum dissolution after cytokinesis. We further find that Orb6 phosphorylation of Sec3 contributes to exocyst function in concert with exocyst protein Exo70. We propose that Orb6 contributes to polarized growth by regulating membrane trafficking at multiple levels.


Subject(s)
Cell Cycle Proteins/genetics , Exocytosis/genetics , Gene Expression Regulation, Fungal , Phosphoproteins/genetics , Protein Serine-Threonine Kinases/genetics , Schizosaccharomyces pombe Proteins/genetics , Schizosaccharomyces/genetics , Vesicular Transport Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Polarity , Cytokinesis/genetics , Phosphoproteins/classification , Phosphoproteins/metabolism , Phosphorylation , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Serine-Threonine Kinases/metabolism , Proteomics/methods , Rho Guanine Nucleotide Exchange Factors/genetics , Rho Guanine Nucleotide Exchange Factors/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Schizosaccharomyces/metabolism , Schizosaccharomyces pombe Proteins/metabolism , Vesicular Transport Proteins/metabolism
5.
J Cell Sci ; 131(14)2018 07 19.
Article in English | MEDLINE | ID: mdl-29930085

ABSTRACT

The conserved Rho-family GTPase Cdc42 plays a central role in eukaryotic cell polarity. The rod-shaped fission yeast Schizosaccharomyces pombe has two Cdc42 guanine nucleotide exchange factors (GEFs), Scd1 and Gef1, but little is known about how they are coordinated in polarized growth. Although the microtubule cytoskeleton is normally not required for polarity maintenance in fission yeast, we show here that when scd1 function is compromised, disruption of microtubules or the polarity landmark proteins Tea1, Tea4 or Pom1 leads to disruption of polarized growth. Instead, cells adopt an isotropic-like pattern of growth, which we term PORTLI growth. Surprisingly, PORTLI growth is caused by spatially inappropriate activity of Gef1. Although most Cdc42 GEFs are membrane associated, we find that Gef1 is a broadly distributed cytosolic protein rather than a membrane-associated protein at cell tips like Scd1. Microtubules and the Tea1-Tea4-Pom1 axis counteract inappropriate Gef1 activity by regulating the localization of the Cdc42 GTPase-activating protein Rga4. Our results suggest a new model of fission yeast cell polarity regulation, involving coordination of 'local' (Scd1) and 'global' (Gef1) Cdc42 GEFs via microtubules and microtubule-dependent polarity landmarks.


Subject(s)
Guanine Nucleotide Exchange Factors/metabolism , Microtubule-Associated Proteins/metabolism , Microtubules/metabolism , Protein Kinases/metabolism , Rho Guanine Nucleotide Exchange Factors/metabolism , Schizosaccharomyces pombe Proteins/metabolism , Schizosaccharomyces/cytology , Schizosaccharomyces/metabolism , Cell Polarity , Guanine Nucleotide Exchange Factors/genetics , Microtubule-Associated Proteins/genetics , Microtubules/genetics , Protein Kinases/genetics , Rho Guanine Nucleotide Exchange Factors/genetics , Schizosaccharomyces/genetics , Schizosaccharomyces pombe Proteins/genetics , cdc42 GTP-Binding Protein/genetics , cdc42 GTP-Binding Protein/metabolism
6.
Mol Cell Proteomics ; 14(1): 243-50, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25368411

ABSTRACT

Stable Isotope Labeling by Amino Acids (SILAC) is a commonly used method in quantitative proteomics. Because of compatibility with trypsin digestion, arginine and lysine are the most widely used amino acids for SILAC labeling. We observed that Schizosaccharomyces pombe (fission yeast) cannot be labeled with a specific form of arginine, (13)C(6) (15)N(4)-arginine (Arg-10), which limits the exploitation of SILAC technology in this model organism. We hypothesized that in the fission yeast the guanidinium group of (13)C(6) (15)N(4)-arginine is catabolized by arginase and urease activity to (15)N1-labeled ammonia that is used as a precursor for general amino acid biosynthesis. We show that disruption of Ni(2+)-dependent urease activity, through deletion of the sole Ni(2+) transporter Nic1, blocks this recycling in ammonium-supplemented EMMG medium to enable (13)C(6) (15)N(4)-arginine labeling for SILAC strategies in S. pombe. Finally, we employed Arg-10 in a triple-SILAC experiment to perform quantitative comparison of G1 + S, M, and G2 cell cycle phases in S. pombe.


Subject(s)
Arginine/metabolism , Cation Transport Proteins/genetics , Isotope Labeling/methods , Schizosaccharomyces pombe Proteins/genetics , Carbon Isotopes , Cell Cycle , Nitrogen Isotopes , Proteomics/methods , Schizosaccharomyces/genetics , Schizosaccharomyces/metabolism
7.
J Cell Sci ; 126(Pt 21): 5052-61, 2013 Nov 01.
Article in English | MEDLINE | ID: mdl-23986474

ABSTRACT

The chemical genetic strategy in which mutational enlargement of the ATP-binding site sensitises of a protein kinase to bulky ATP analogues has proved to be an elegant tool for the generation of conditional analogue-sensitive kinase alleles in a variety of model organisms. Here, we describe a novel substitution mutation in the kinase domain that can enhance the sensitivity of analogue-sensitive kinases. Substitution of a methionine residue to phenylalanine in the +2 position after HRDLKxxN motif of the subdomain VIb within the kinase domain markedly increased the sensitivities of the analogue-sensitive kinases to ATP analogues in three out of five S. pombe kinases (i.e. Plo1, Orb5 and Wee1) that harbor this conserved methionine residue. Kinome alignment established that a methionine residue is found at this site in 5-9% of kinases in key model organisms, suggesting that a broader application of this structural modification may enhance ATP analogue sensitivity of analogue-sensitive kinases in future studies. We also show that the enhanced sensitivity of the wee1.as8 allele in a cdc25.22 background can be exploited to generate highly synchronised mitotic and S phase progression at 36°C. Proof-of-principle experiments show how this novel synchronisation technique will prove of great use in the interrogation of the mitotic or S-phase functions through temperature sensitivity mutation of molecules of interest in fission yeast.


Subject(s)
Adenosine Triphosphate/metabolism , Casein Kinase II/genetics , Cell Cycle Proteins/genetics , Mitosis , Mutation, Missense , Nuclear Proteins/genetics , Protein Serine-Threonine Kinases/genetics , Protein-Tyrosine Kinases/genetics , Schizosaccharomyces pombe Proteins/genetics , Schizosaccharomyces/enzymology , Adenosine Triphosphate/analogs & derivatives , Amino Acid Substitution , Casein Kinase II/chemistry , Casein Kinase II/metabolism , Cell Cycle Proteins/chemistry , Cell Cycle Proteins/metabolism , Conserved Sequence , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/metabolism , Methionine/genetics , Methionine/metabolism , Nuclear Proteins/chemistry , Nuclear Proteins/metabolism , Phenylalanine/genetics , Phenylalanine/metabolism , Phosphoprotein Phosphatases/genetics , Phosphoprotein Phosphatases/metabolism , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/metabolism , Protein Structure, Tertiary , Protein-Tyrosine Kinases/chemistry , Protein-Tyrosine Kinases/metabolism , Schizosaccharomyces/cytology , Schizosaccharomyces/genetics , Schizosaccharomyces pombe Proteins/chemistry , Schizosaccharomyces pombe Proteins/metabolism
8.
J Biol Chem ; 285(15): 11427-32, 2010 Apr 09.
Article in English | MEDLINE | ID: mdl-20178992

ABSTRACT

Eukaryotic Holliday junction (HJ) resolvases have attracted much attention recently with the identification of at least three distinct proteins that can cleave model HJs in vitro. However, the specific DNA structure(s) that these proteins act upon in the cell is unknown. Here, we describe a system in budding yeast to directly and quantitatively monitor in vivo HJ resolution. We found that Yen1 acts redundantly with Mus81, but not Slx1, to resolve a model HJ in vivo. This functional overlap specifically extends to the repair/bypass of lesions that impede the progression of replication forks but not to the repair of double-strand breaks induced by ionizing radiation. Together, these results suggest a direct role for Yen1 in the response to DNA damage and implicate overlapping HJ resolution functions of Yen1 with Mus81 during replication fork repair.


Subject(s)
DNA, Cruciform/metabolism , DNA-Binding Proteins/metabolism , Endonucleases/metabolism , Gene Expression Regulation, Fungal , Holliday Junction Resolvases/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Cloning, Molecular , DNA Damage , DNA Repair , DNA Replication , DNA-Binding Proteins/genetics , Dimerization , Endonucleases/genetics , Escherichia coli/metabolism , Escherichia coli Proteins/metabolism , Holliday Junction Resolvases/genetics , Models, Biological , Plasmids/metabolism , Radiation, Ionizing , Saccharomyces cerevisiae Proteins/genetics
9.
Nucleic Acids Res ; 38(6): 1889-901, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20047969

ABSTRACT

In budding yeast the DNA helicase Mph1 prevents genome rearrangements during ectopic homologous recombination (HR) by suppressing the formation of crossovers (COs). Here we show that during ectopic HR repair, the anti-CO function of Mph1 is intricately associated with the mismatch repair (MMR) factor, MutSalpha. In particular, during HR repair using a completely homologous substrate, we reveal an MMR-independent function of MutSalpha in generating COs that is specifically antagonized by Mph1, but not Sgs1. In contrast, both Mph1 and MutSalpha are required to efficiently suppress COs in the presence of a homeologous substrate. Mph1 acts redundantly with Sgs1 in this respect since mph1Delta sgs1Delta double mutant cells pheno-copy MutSalpha mutants and completely fail to discriminate homologous and homeologous sequences during HR repair. However, this defect of mph1Delta sgs1Delta cells is not due to an inability to carry out MMR but rather is accompanied by elevated levels of gene conversion (GC) and bi-directional GC tracts specifically in non-crossover products. Models describing how Mph1, MutSalpha and Sgs1 act in concert to suppress genome rearrangements during ectopic HR repair are discussed.


Subject(s)
DEAD-box RNA Helicases/metabolism , DNA Repair , DNA-Binding Proteins/metabolism , MutS Homolog 2 Protein/metabolism , Recombination, Genetic , Saccharomyces cerevisiae Proteins/metabolism , DEAD-box RNA Helicases/genetics , DNA Mismatch Repair , DNA-Binding Proteins/genetics , Gene Conversion , MutS Homolog 2 Protein/genetics , Mutation , RecQ Helicases/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL