Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
J Biol Chem ; 299(8): 104929, 2023 08.
Article in English | MEDLINE | ID: mdl-37330173

ABSTRACT

Homologous recombination (HR) is a major pathway for the repair of DNA double-strand breaks, the most severe form of DNA damage. The Rad51 protein is central to HR, but multiple auxiliary factors regulate its activity. The heterodimeric Swi5-Sfr1 complex is one such factor. It was previously shown that two sites within the intrinsically disordered domain of Sfr1 are critical for the interaction with Rad51. Here, we show that phosphorylation of five residues within this domain regulates the interaction of Swi5-Sfr1 with Rad51. Biochemical reconstitutions demonstrated that a phosphomimetic mutant version of Swi5-Sfr1 is defective in both the physical and functional interaction with Rad51. This translated to a defect in DNA repair, with the phosphomimetic mutant yeast strain phenocopying a previously established interaction mutant. Interestingly, a strain in which Sfr1 phosphorylation was blocked also displayed sensitivity to DNA damage. Taken together, we propose that controlled phosphorylation of Sfr1 is important for the role of Swi5-Sfr1 in promoting Rad51-dependent DNA repair.


Subject(s)
DNA Repair , Rad51 Recombinase , Schizosaccharomyces pombe Proteins , DNA Breaks, Double-Stranded , DNA Damage , Homologous Recombination , Rad51 Recombinase/metabolism , Schizosaccharomyces/enzymology , Schizosaccharomyces/genetics , Schizosaccharomyces pombe Proteins/genetics , Schizosaccharomyces pombe Proteins/metabolism , Mutation , Phosphorylation
2.
Curr Genet ; 67(6): 919-936, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34296348

ABSTRACT

Here, we report the development of methodologies that enable genetic modification of a Basidiomycota yeast, Naganishia liquifaciens. The gene targeting method employs electroporation with PCR products flanked by an 80 bp sequence homologous to the target. The method, combined with a newly devised CRISPR-Cas9 system, routinely achieves 80% gene targeting efficiency. We further explored the genetic requirement for this homologous recombination (HR)-mediated gene targeting. The absence of Ku70, a major component of the non-homologous end joining (NHEJ) pathway of DNA double-strand break repair, almost completely eliminated inaccurate integration of the marker. Gene targeting with short homology (80 bp) was almost exclusively dependent on Rad52, an essential component of HR in the Ascomycota yeasts Saccharomyces cerevisiae and Schizosaccharomyces pombe. By contrast, the RecA homolog Rad51, which performs homology search and strand exchange in HR, plays a relatively minor role in gene targeting, regardless of the homology length (80 bp or 1 kb). The absence of both Rad51 and Rad52, however, completely eliminated gene targeting. Unlike Ascomycota yeasts, the absence of Rad52 in N. liquefaciens conferred only mild sensitivity to ionizing radiation. These traits associated with the absence of Rad52 are reminiscent of findings in mice.


Subject(s)
Basidiomycota/genetics , Fungal Proteins/metabolism , Gene Targeting , Rad51 Recombinase/metabolism , Rad52 DNA Repair and Recombination Protein/metabolism , CRISPR-Cas Systems , Gene Editing , Genetic Complementation Test , Genetic Engineering , Genetic Loci , Homologous Recombination , Intramolecular Oxidoreductases/genetics , Intramolecular Oxidoreductases/metabolism , Ku Autoantigen/genetics , Transformation, Genetic
3.
Curr Opin Genet Dev ; 71: 19-26, 2021 12.
Article in English | MEDLINE | ID: mdl-34246071

ABSTRACT

Homologous recombination (HR) is a universal phenomenon conserved from viruses to humans. The mechanisms of HR are essentially the same in humans and simple unicellular eukaryotes like yeast. Two highly diverged yeast species, Saccharomyces cerevisiae and Schizosaccharomyces pombe, have proven exceptionally useful in understanding the fundamental mechanisms of eukaryotic HR by serving as a source for unique biological insights and also complementing each other. Here, we will review the features of S. pombe HR mechanisms in comparison to S. cerevisiae and other model organisms. Particular emphasis will be put on the biochemical characterization of HR mechanisms uncovered using S. pombe proteins.


Subject(s)
Schizosaccharomyces pombe Proteins , Schizosaccharomyces , Eukaryotic Cells , Homologous Recombination/genetics , Humans , Saccharomyces cerevisiae , Schizosaccharomyces/genetics , Schizosaccharomyces pombe Proteins/genetics
4.
Nucleic Acids Res ; 49(12): 6832-6848, 2021 07 09.
Article in English | MEDLINE | ID: mdl-34157114

ABSTRACT

Rad51 is the key protein in homologous recombination that plays important roles during DNA replication and repair. Auxiliary factors regulate Rad51 activity to facilitate productive recombination, and prevent inappropriate, untimely or excessive events, which could lead to genome instability. Previous genetic analyses identified a function for Rrp1 (a member of the Rad5/16-like group of SWI2/SNF2 translocases) in modulating Rad51 function, shared with the Rad51 mediator Swi5-Sfr1 and the Srs2 anti-recombinase. Here, we show that Rrp1 overproduction alleviates the toxicity associated with excessive Rad51 levels in a manner dependent on Rrp1 ATPase domain. Purified Rrp1 binds to DNA and has a DNA-dependent ATPase activity. Importantly, Rrp1 directly interacts with Rad51 and removes it from double-stranded DNA, confirming that Rrp1 is a translocase capable of modulating Rad51 function. Rrp1 affects Rad51 binding at centromeres. Additionally, we demonstrate in vivo and in vitro that Rrp1 possesses E3 ubiquitin ligase activity with Rad51 as a substrate, suggesting that Rrp1 regulates Rad51 in a multi-tiered fashion.


Subject(s)
DNA-Binding Proteins/metabolism , Rad51 Recombinase/metabolism , Schizosaccharomyces pombe Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Adenosine Triphosphatases/metabolism , DNA, Fungal/metabolism , DNA-Binding Proteins/isolation & purification , DNA-Binding Proteins/physiology , Genomic Instability , Schizosaccharomyces/genetics , Schizosaccharomyces/metabolism , Schizosaccharomyces pombe Proteins/isolation & purification , Schizosaccharomyces pombe Proteins/physiology
5.
DNA Repair (Amst) ; 104: 103114, 2021 08.
Article in English | MEDLINE | ID: mdl-34111757

ABSTRACT

DNA is the molecule that stores the chemical instructions necessary for life and its stability is therefore of the utmost importance. Despite this, DNA is damaged by both exogenous and endogenous factors at an alarming frequency. The most severe type of DNA damage is a double-strand break (DSB), in which a scission occurs in both strands of the double helix, effectively dividing a single normal chromosome into two pathological chromosomes. Homologous recombination (HR) is a universal DSB repair mechanism that solves this problem by identifying another region of the genome that shares high sequence similarity with the DSB site and using it as a template for repair. Rad51 possess the enzymatic activity that is essential for this repair but several auxiliary factors are required for Rad51 to fulfil its function. It is becoming increasingly clear that many HR factors are subjected to post-translational modification. Here, we review what is known about how these modifications affect HR. We first focus on cases where there is experimental evidence to support a function for the modification, then discuss speculative cases where a function can be inferred. Finally, we contemplate why such modifications might be necessary.


Subject(s)
Protein Processing, Post-Translational , Recombinational DNA Repair , Acetylation , Animals , Eukaryota , Humans , Phosphorylation , Rad51 Recombinase/metabolism , Sumoylation , Ubiquitination
6.
Proc Natl Acad Sci U S A ; 118(11)2021 03 16.
Article in English | MEDLINE | ID: mdl-33836577

ABSTRACT

The Mre11-Rad50-Nbs1 complex (MRN) is important for repairing DNA double-strand breaks (DSBs) by homologous recombination (HR). The endonuclease activity of MRN is critical for resecting 5'-ended DNA strands at DSB ends, producing 3'-ended single-strand DNA, a prerequisite for HR. This endonuclease activity is stimulated by Ctp1, the Schizosaccharomyces pombe homolog of human CtIP. Here, with purified proteins, we show that Ctp1 phosphorylation stimulates MRN endonuclease activity by inducing the association of Ctp1 with Nbs1. The highly conserved extreme C terminus of Ctp1 is indispensable for MRN activation. Importantly, a polypeptide composed of the conserved 15 amino acids at the C terminus of Ctp1 (CT15) is sufficient to stimulate Mre11 endonuclease activity. Furthermore, the CT15 equivalent from CtIP can stimulate human MRE11 endonuclease activity, arguing for the generality of this stimulatory mechanism. Thus, we propose that Nbs1-mediated recruitment of CT15 plays a pivotal role in the activation of the Mre11 endonuclease by Ctp1/CtIP.


Subject(s)
DNA-Binding Proteins/metabolism , Exodeoxyribonucleases/metabolism , Peptides/metabolism , Schizosaccharomyces pombe Proteins/metabolism , Schizosaccharomyces/metabolism , Amino Acid Sequence , Casein Kinase II/metabolism , Conserved Sequence , DNA Breaks, Double-Stranded , Phosphorylation
7.
Elife ; 102021 01 25.
Article in English | MEDLINE | ID: mdl-33493431

ABSTRACT

Homologous recombination (HR) is essential for maintaining genome stability. Although Rad51 is the key protein that drives HR, multiple auxiliary factors interact with Rad51 to potentiate its activity. Here, we present an interdisciplinary characterization of the interactions between Rad51 and these factors. Through structural analysis, we identified an evolutionarily conserved acidic patch of Rad51. The neutralization of this patch completely abolished recombinational DNA repair due to defects in the recruitment of Rad51 to DNA damage sites. This acidic patch was found to be important for the interaction with Rad55-Rad57 and essential for the interaction with Rad52. Furthermore, biochemical reconstitutions demonstrated that neutralization of this acidic patch also impaired the interaction with Rad54, indicating that a single motif is important for the interaction with multiple auxiliary factors. We propose that this patch is a fundamental motif that facilitates interactions with auxiliary factors and is therefore essential for recombinational DNA repair.


The DNA molecule contains the chemical instructions necessary for life. Its physical integrity is therefore vital, yet it is also under constant threat from external and internal factors. As a result, organisms have evolved an arsenal of mechanisms to repair damaged DNA. For instance, when the two complementary strands that form the DNA molecule are broken at the same location, the cell triggers a mechanism known as homologous recombination. A protein known as Rad51 orchestrates this process, helped by an array of other proteins that include Rad55-Rad57, Rad52, and Rad54. These physically bind to Rad51 and activate it in different ways. However, exactly how these interactions take place remained unclear. To find out more, Afshar et al. examined models of the structure of Rad51, revealing that three of the protein's building blocks create a prominent, negatively charged patch that could be important for DNA repair. Yeast cells were then genetically manipulated to produce a modified version of Rad51 in which the three building blocks were neutralised. These organisms were unable to repair their DNA. Further biochemical tests showed that the modified protein could no longer attach well to Rad55-Rad57 or Rad54, and could not stick to Rad52 at all. In fact, without its negatively charged patch, Rad51 could not find the ends of broken DNA strands, a process which is normally aided by Rad55-Rad57 and Rad52. Taken together, these results suggest that the helper proteins all interact with Rad51 in the same place, even though they play different roles. Faulty DNA repair processes have been linked to devastating consequences such as cell death or cancer. Understanding the details of DNA repair in yeast can serve as a template for research in more complex organisms, opening the possibility of applications for human health.


Subject(s)
Homologous Recombination/genetics , Rad51 Recombinase/genetics , Recombinational DNA Repair/genetics , Schizosaccharomyces pombe Proteins/genetics , Schizosaccharomyces/genetics , Rad51 Recombinase/metabolism , Schizosaccharomyces/metabolism , Schizosaccharomyces pombe Proteins/metabolism
8.
Microbiol Resour Announc ; 9(47)2020 Nov 19.
Article in English | MEDLINE | ID: mdl-33214294

ABSTRACT

The draft genome sequence of the deep-sea yeast Naganishia liquefaciens strain N6, isolated from the Japan Trench, is reported here. This strain was previously classified into a Cryptococcus clade. Phylogenetic analysis using the presented sequence suggests that strain N6 is in the clade of the genus Naganishia.

9.
Nat Commun ; 11(1): 2950, 2020 06 11.
Article in English | MEDLINE | ID: mdl-32528002

ABSTRACT

During homologous recombination, Rad51 forms a nucleoprotein filament on single-stranded DNA to promote DNA strand exchange. This filament binds to double-stranded DNA (dsDNA), searches for homology, and promotes transfer of the complementary strand, producing a new heteroduplex. Strand exchange proceeds via two distinct three-strand intermediates, C1 and C2. C1 contains the intact donor dsDNA whereas C2 contains newly formed heteroduplex DNA. Here, we show that the conserved DNA binding motifs, loop 1 (L1) and loop 2 (L2) in site I of Rad51, play distinct roles in this process. L1 is involved in formation of the C1 complex whereas L2 mediates the C1-C2 transition, producing the heteroduplex. Another DNA binding motif, site II, serves as the DNA entry position for initial Rad51 filament formation, as well as for donor dsDNA incorporation. Our study provides a comprehensive molecular model for the catalytic process of strand exchange mediated by eukaryotic RecA-family recombinases.


Subject(s)
DNA/metabolism , Rad51 Recombinase/chemistry , Rad51 Recombinase/metabolism , Adenosine Triphosphate/metabolism , Binding Sites/genetics , DNA/genetics , DNA Damage/genetics , DNA Damage/physiology , DNA Repair/genetics , DNA Repair/physiology , DNA, Single-Stranded/genetics , Homologous Recombination/genetics , Homologous Recombination/physiology , Humans , Mutation/genetics , Nucleic Acid Heteroduplexes/genetics , Nucleic Acid Heteroduplexes/metabolism , Protein Structure, Secondary , Rad51 Recombinase/genetics , Saccharomyces cerevisiae/genetics , Schizosaccharomyces/genetics
10.
Proc Natl Acad Sci U S A ; 117(22): 12062-12070, 2020 06 02.
Article in English | MEDLINE | ID: mdl-32414915

ABSTRACT

Homologous recombination (HR) is a universal mechanism operating in somatic and germ-line cells, where it contributes to the maintenance of genome stability and ensures the faithful distribution of genetic material, respectively. The ability to identify and exchange the strands of two homologous DNA molecules lies at the heart of HR and is mediated by RecA-family recombinases. Dmc1 is a meiosis-specific RecA homolog in eukaryotes, playing a predominant role in meiotic HR. However, Dmc1 cannot function without its two major auxiliary factor complexes, Swi5-Sfr1 and Hop2-Mnd1. Through biochemical reconstitutions, we demonstrate that Swi5-Sfr1 and Hop2-Mnd1 make unique contributions to stimulate Dmc1-driven strand exchange in a synergistic manner. Mechanistically, Swi5-Sfr1 promotes establishment of the Dmc1 nucleoprotein filament, whereas Hop2-Mnd1 defines a critical, rate-limiting step in initiating strand exchange. Following execution of this function, we propose that Swi5-Sfr1 then promotes strand exchange with Hop2-Mnd1. Thus, our findings elucidate distinct yet complementary roles of two auxiliary factors in Dmc1-driven strand exchange, providing mechanistic insights into some of the most critical steps in meiotic HR.


Subject(s)
Cell Cycle Proteins/metabolism , DNA-Binding Proteins/metabolism , Homologous Recombination/physiology , Rad51 Recombinase/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Transcription Factors/metabolism , DNA/metabolism , Meiosis/physiology , Rec A Recombinases/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Schizosaccharomyces/genetics , Schizosaccharomyces/metabolism
11.
Elife ; 92020 03 24.
Article in English | MEDLINE | ID: mdl-32204793

ABSTRACT

Although Rad51 is the key protein in homologous recombination (HR), a major DNA double-strand break repair pathway, several auxiliary factors interact with Rad51 to promote productive HR. We present an interdisciplinary characterization of the interaction between Rad51 and Swi5-Sfr1, a conserved auxiliary factor. Two distinct sites within the intrinsically disordered N-terminus of Sfr1 (Sfr1N) were found to cooperatively bind Rad51. Deletion of this domain impaired Rad51 stimulation in vitro and rendered cells sensitive to DNA damage. By contrast, amino acid-substitution mutants, which had comparable biochemical defects, could promote DNA repair, suggesting that Sfr1N has another role in addition to Rad51 binding. Unexpectedly, the DNA repair observed in these mutants was dependent on Rad55-Rad57, another auxiliary factor complex hitherto thought to function independently of Swi5-Sfr1. When combined with the finding that they form a higher-order complex, our results imply that Swi5-Sfr1 and Rad55-Rad57 can collaboratively stimulate Rad51 in Schizosaccharomyces pombe.


The DNA within cells contains the instructions necessary for life and it must be carefully maintained. DNA is constantly being damaged by radiation and other factors so cells have evolved an arsenal of mechanisms that repair this damage. An enzyme called Rad51 drives one such DNA repair process known as homologous recombination. A pair of regulatory proteins known as the Swi5-Sfr1 complex binds to Rad51 and activates it. The complex can be thought of as containing two modules with distinct roles: one comprising the first half of the Sfr1 protein and that is capable of binding to Rad51, and a second consisting of the rest of Sfr1 bound to Swi5, which is responsible for activating Rad51. Here, Argunhan, Sakakura et al. used genetic and biochemical approaches to study how this first module, known as "Sfr1N", interacts with Rad51 in a microbe known as fission yeast. The experiments showed that both modules of Swi5-Sfr1 were important for Rad51 to drive homologous recombination. Swi5-Sfr1 complexes carrying mutations in the region of Sfr1N that binds to Rad51 were unable to activate Rad51 in a test tube. However, fission yeast cells containing the same mutations were able to repair their DNA without problems. This was due to the presence of another pair of proteins known as the Rad55-Rad57 complex that also bound to Swi5-Sfr1. The findings of Argunhan, Sakakura et al. suggest that the Swi5-Sfr1 and Rad55-Rad57 complexes work together to activate Rad51. Many genetically inherited diseases and cancers have been linked to mutations in DNA repair proteins. The fundamental mechanisms of DNA repair are very similar from yeast to humans and other animals, therefore, understanding the details of DNA repair in yeast may ultimately benefit human health in the future.


Subject(s)
DNA Damage/physiology , DNA Repair/physiology , Rad51 Recombinase/metabolism , Schizosaccharomyces/metabolism , Escherichia coli , Gene Expression Regulation, Fungal , Magnetic Resonance Spectroscopy , Protein Binding , Protein Domains , Rad51 Recombinase/genetics , Schizosaccharomyces/genetics , Schizosaccharomyces pombe Proteins/genetics , Schizosaccharomyces pombe Proteins/metabolism
12.
J Vis Exp ; (144)2019 02 13.
Article in English | MEDLINE | ID: mdl-30829327

ABSTRACT

The DNA strand exchange reaction mediated by Rad51 is a critical step of homologous recombination. In this reaction, Rad51 forms a nucleoprotein filament on single-stranded DNA (ssDNA) and captures double-stranded DNA (dsDNA) non-specifically to interrogate it for a homologous sequence. After encountering homology, Rad51 catalyzes DNA strand exchange to mediate pairing of the ssDNA with the complementary strand of the dsDNA. This reaction is highly regulated by numerous accessary proteins in vivo. Although conventional biochemical assays have been successfully employed to examine the role of such accessory protein in vitro, kinetic analysis of intermediate formation and its progression into a final product has proven challenging due to the unstable and transient nature of the reaction intermediates. To observe these reaction steps directly in solution, fluorescence resonance energy transfer (FRET)-based real-time observation systems of this reaction were established. Kinetic analysis of real-time observations shows that the DNA strand exchange reaction mediated by Rad51 obeys a three-step reaction model involving the formation of a three-strand DNA intermediate, maturation of this intermediate, and the release of ssDNA from the mature intermediate. The Swi5-Sfr1 complex, an accessary protein conserved in eukaryotes, strongly enhances the second and third steps of this reaction. The FRET-based assays presented here enable us to uncover the molecular mechanisms through which recombination accessary proteins stimulate the DNA strand exchange activity of Rad51. The primary goal of this protocol is to enhance the repertoire of techniques available to researchers in the field of homologous recombination, particularly those working with proteins from species other than Schizosaccharomyces pombe, so that the evolutionary conservation of the findings presented herein can be determined.


Subject(s)
Biosensing Techniques/methods , DNA, Single-Stranded/genetics , DNA/chemistry , Rad51 Recombinase/metabolism
13.
Cell Cycle ; 17(3): 273-274, 2018.
Article in English | MEDLINE | ID: mdl-29233050
14.
Curr Genet ; 64(2): 423-427, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29071381

ABSTRACT

The meiotic cell cycle provides a unique model to study the relationship between recombinational DNA repair and the cell cycle, since homologous recombination, induced by programmed DNA double-strand breaks (DSBs), is integrated as an essential step during meiosis. The pachytene checkpoint, which is situated towards the end of meiotic prophase I, coordinates homologous recombination and cell cycle progression, similar to the DNA damage checkpoint mechanisms operating in vegetative cells. However, there are a number of features unique to meiosis, making the system optimized for the purpose of meiosis. Our recent work highlights the involvement of three major cell cycle kinases, Dbf4-dependent Cdc7 kinase, Polo kinase and CDK, in coordinating homologous recombination and the meiotic cell cycle. In this review, we will discuss the unique interplay between meiotic cell cycle control and homologous recombination during meiosis I.


Subject(s)
DNA Damage/genetics , Meiosis/genetics , Meiotic Prophase I/genetics , Saccharomyces cerevisiae/genetics , Cell Cycle Proteins/genetics , DNA Breaks, Double-Stranded , DNA-Binding Proteins/genetics , Phosphorylation , Protein Serine-Threonine Kinases/genetics , Saccharomyces cerevisiae Proteins/genetics
15.
EMBO J ; 36(17): 2488-2509, 2017 09 01.
Article in English | MEDLINE | ID: mdl-28694245

ABSTRACT

The synaptonemal complex (SC) is a proteinaceous macromolecular assembly that forms during meiotic prophase I and mediates adhesion of paired homologous chromosomes along their entire lengths. Although prompt disassembly of the SC during exit from prophase I is a landmark event of meiosis, the underlying mechanism regulating SC destruction has remained elusive. Here, we show that DDK (Dbf4-dependent Cdc7 kinase) is central to SC destruction. Upon exit from prophase I, Dbf4, the regulatory subunit of DDK, directly associates with and is phosphorylated by the Polo-like kinase Cdc5. In parallel, upregulated CDK1 activity also targets Dbf4. An enhanced Dbf4-Cdc5 interaction pronounced phosphorylation of Dbf4 and accelerated SC destruction, while reduced/abolished Dbf4 phosphorylation hampered destruction of SC proteins. SC destruction relieved meiotic inhibition of the ubiquitous recombinase Rad51, suggesting that the mitotic recombination machinery is reactivated following prophase I exit to repair any persisting meiotic DNA double-strand breaks. Taken together, we propose that the concerted action of DDK, Polo-like kinase, and CDK1 promotes efficient SC destruction at the end of prophase I to ensure faithful inheritance of the genome.


Subject(s)
Cell Cycle Proteins/metabolism , Fungal Proteins/metabolism , Meiosis/physiology , Protein Kinases/metabolism , Synaptonemal Complex/metabolism , Phosphorylation , Saccharomycetales/metabolism
16.
FEBS Lett ; 591(14): 2035-2047, 2017 07.
Article in English | MEDLINE | ID: mdl-28423184

ABSTRACT

Homologous recombination (HR) is the process whereby two DNA molecules that share high sequence similarity are able to recombine to generate hybrid DNA molecules. Throughout evolution, the ability of HR to identify highly similar DNA sequences has been adopted for numerous biological phenomena including DNA repair, meiosis, telomere maintenance, ribosomal DNA amplification and immunological diversity. Although Rad51 and Dmc1 are the key proteins that promote HR in mitotic and meiotic cells, respectively, accessory proteins that allow Rad51 and Dmc1 to effectively fulfil their functions have been identified in all examined model systems. In this Review, we discuss the roles of the highly conserved Swi5-Sfr1 accessory complex in yeast, mice and humans, and explore similarities and differences between these species.


Subject(s)
Homologous Recombination , Nuclear Proteins/metabolism , Animals , Conserved Sequence , Humans , Recombinases/metabolism , Schizosaccharomyces/genetics , Schizosaccharomyces/metabolism , Species Specificity
17.
Microb Cell ; 3(3): 126-128, 2016 Feb 19.
Article in English | MEDLINE | ID: mdl-28357343

ABSTRACT

The synaptonemal complex (SC) is a meiosis-specific chromosomal structure in which homologous chromosomes are intimately linked through arrays of specialized proteins called transverse filaments (TF). Widely conserved in eukaryote meiosis, the SC forms during prophase I and is essential for accurate segregation of homologous chromosomes at meiosis I. However, the basic mechanism overlooking formation and regulation of the SC has been poorly understood. By using the budding yeast Saccharomyces cerevisiae, we recently showed that SC formation is controlled through the attachment of multiple molecules of small ubiquitin-like modifier (SUMO) to a regulator of TF assembly. Intriguingly, this SUMOylation is activated by TF, implicating the involvement of a positive feedback loop in the control of SC assembly. We discuss the implication of this finding and possible involvement of a similar mechanism in regulating other processes.

18.
J Cell Biol ; 211(4): 785-93, 2015 Nov 23.
Article in English | MEDLINE | ID: mdl-26598615

ABSTRACT

During meiotic prophase I, proteinaceous structures called synaptonemal complexes (SCs) connect homologous chromosomes along their lengths via polymeric arrays of transverse filaments (TFs). Thus, control of TF polymerization is central to SC formation. Using budding yeast, we show that efficiency of TF polymerization closely correlates with the extent of SUMO conjugation to Ecm11, a component of SCs. HyperSUMOylation of Ecm11 leads to highly aggregative TFs, causing frequent assembly of extrachromosomal structures. In contrast, hypoSUMOylation leads to discontinuous, fragmented SCs, indicative of defective TF polymerization. We further show that the N terminus of the yeast TF, Zip1, serves as an activator for Ecm11 SUMOylation. Coexpression of the Zip1 N terminus and Gmc2, a binding partner of Ecm11, is sufficient to induce robust polySUMOylation of Ecm11 in nonmeiotic cells. Because TF assembly is mediated through N-terminal head-to-head associations, our results suggest that mutual activation between TF assembly and Ecm11 polySUMOylation acts as a positive feedback loop that underpins SC assembly.


Subject(s)
Fungal Proteins/metabolism , Saccharomycetales/metabolism , Sumoylation , Synaptonemal Complex/metabolism , Cell Cycle Proteins/metabolism , Endopeptidases/metabolism , Feedback, Physiological , Nuclear Proteins/metabolism , Protein Interaction Domains and Motifs , Protein Multimerization
19.
FEBS Open Bio ; 3: 302-4, 2013.
Article in English | MEDLINE | ID: mdl-23951551

ABSTRACT

A newly identified human locus on chromosome 15 was recently associated with zinc accumulation. Based on a prior report of a threefold difference in zinc accumulation between fumble(1) heterozygous mutants and control fly strains, it was suggested that phosphopantothenoylcysteine decarboxylase might affect zinc status through its effects on vitamin B5 (pantothenate) metabolism. We report here that outcrossed fumble(1) heterozygous mutant flies with low zinc content have been recovered, suggesting that pantothenate metabolism did not alter zinc homeostasis in fumble(1) heterozygous flies. We show instead that the Drosophila condition of low body zinc accumulation is an X-chromosome-linked recessive trait.

20.
PLoS One ; 8(6): e65875, 2013.
Article in English | MEDLINE | ID: mdl-23762445

ABSTRACT

Meiotic recombination plays an essential role in the proper segregation of chromosomes at meiosis I in many sexually reproducing organisms. Meiotic recombination is initiated by the scheduled formation of genome-wide DNA double-strand breaks (DSBs). The timing of DSB formation is strictly controlled because unscheduled DSB formation is detrimental to genome integrity. Here, we investigated the role of DNA damage checkpoint mechanisms in the control of meiotic DSB formation using budding yeast. By using recombination defective mutants in which meiotic DSBs are not repaired, the effect of DNA damage checkpoint mutations on DSB formation was evaluated. The Tel1 (ATM) pathway mainly responds to unresected DSB ends, thus the sae2 mutant background in which DSB ends remain intact was employed. On the other hand, the Mec1 (ATR) pathway is primarily used when DSB ends are resected, thus the rad51 dmc1 double mutant background was employed in which highly resected DSBs accumulate. In order to separate the effect caused by unscheduled cell cycle progression, which is often associated with DNA damage checkpoint defects, we also employed the ndt80 mutation which permanently arrests the meiotic cell cycle at prophase I. In the absence of Tel1, DSB formation was reduced in larger chromosomes (IV, VII, II and XI) whereas no significant reduction was found in smaller chromosomes (III and VI). On the other hand, the absence of Rad17 (a critical component of the ATR pathway) lead to an increase in DSB formation (chromosomes VII and II were tested). We propose that, within prophase I, the Tel1 pathway facilitates DSB formation, especially in bigger chromosomes, while the Mec1 pathway negatively regulates DSB formation. We also identified prophase I exit, which is under the control of the DNA damage checkpoint machinery, to be a critical event associated with down-regulating meiotic DSB formation.


Subject(s)
Chromosomes, Fungal/genetics , DNA Breaks, Double-Stranded , Meiosis/genetics , Mutation/genetics , Recombination, Genetic/genetics , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae/genetics , Cell Cycle Proteins/genetics , DNA, Fungal/genetics , DNA-Binding Proteins/genetics , Intracellular Signaling Peptides and Proteins/genetics , Protein Serine-Threonine Kinases/genetics , Rad51 Recombinase/genetics , Saccharomyces cerevisiae/growth & development
SELECTION OF CITATIONS
SEARCH DETAIL
...