Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Sci Total Environ ; 926: 171826, 2024 May 20.
Article in English | MEDLINE | ID: mdl-38521265

ABSTRACT

Territorial space conflicts (TSCs) in coastal zones stem from the intricate interaction between the land-sea dual system, significantly impacting the sustainable development of these areas. To accurately identify TSCs, our study proposed a theoretical framework based on a land-sea interaction perspective. We also assessed TSCs using the territorial space conflicts index and a social network analysis model. We demonstrated the proposed spatial conflicts assessment methodology through a case study of Wenzhou, a typical city on the east coast of China. Our results indicate that the distribution of TSCs gradually decreased from the coastal zone to the inland zone, with significant variation in the distribution of different conflict types across different zones. The findings also reveal that territorial space use had diverse impacts on the space conflict network, making it urgent to take targeted measures. In the future, it is crucial to comprehensively consider the overall pattern and distribution characteristics of current TSCs, as well as the spatial spillover effect of the overall network, to develop targeted coping strategies and regulation mechanisms that promote the integration and high-quality development of coastal territorial space systems. To maintain a sustainable coastal zone process, we proposed a set of optimization paths for alleviating TSCs and promoting the coordinated development of land and sea regions in China based on our study.

2.
Hepatology ; 2024 Jan 12.
Article in English | MEDLINE | ID: mdl-38214558

ABSTRACT

BACKGROUND AND AIMS: Evidence assessing the role of B cells and their antibodies, or lack thereof, in the spontaneous resolution of acute HCV infection is conflicting. Utilization of a strictly hepatotropic, HCV-related rodent hepacivirus (RHV) model circumvents many of the challenges facing the field in characterizing the immunological correlates of dichotomous infection outcomes. This study seeks to elucidate the importance of B cells in the clearance of acute RHV infection. APPROACH AND RESULTS: µMT mice were infected i.v. with RHV and found to develop chronic infection for over a year. Wild-type (WT) mice depleted of B cells also exhibited persistent viremia that resolved only upon B cell resurgence. The persistent infection developed by B1-8i and AID cre/cre mice revealed that antigen-specific, class-switched B cells or their antibodies were crucial for viral resolution. Virus-specific CD8 + and CD4 + T cells were characterized in these mice using newly developed major histocompatibility complex class I and II tetramers and ex vivo peptide stimulation. Immunoglobulin G (IgG) was purified from the serum of RHV- or lymphocytic choriomeningitis virus Armstrong-infected mice after viral clearance and passively transferred to AID cre/cre recipients, revealing viral clearance only in αRHV IgG recipients. Further, the transfer of αRHV IgG into B cell-depleted recipients also induced viral resolution. This ability of RHV-specific IgG to induce viral clearance was found to require the concomitant presence of CD8 + T cells. CONCLUSIONS: Our findings demonstrate a cooperative interdependence between immunoglobulins and the T cell compartment that is required for RHV resolution. Thus, HCV vaccine regimens should aim to simultaneously elicit robust HCV-specific antibody and T cell responses for optimal protective efficacy.

3.
bioRxiv ; 2023 Sep 01.
Article in English | MEDLINE | ID: mdl-37693595

ABSTRACT

The gut-brain axis, a bidirectional signaling network between the intestine and the central nervous system, is crucial to the regulation of host physiology and inflammation. Recent advances suggest a strong correlation between gut dysbiosis and neurological diseases, however, relatively little is known about how gut bacteria impact the brain. Here, we reveal that gut commensal bacteria can translocate directly to the brain when mice are fed an altered diet that causes dysbiosis and intestinal permeability, and that this also occurs without diet alteration in distinct murine models of neurological disease. The bacteria were not found in other systemic sites or the blood, but were detected in the vagus nerve. Unilateral cervical vagotomy significantly reduced the number of bacteria in the brain, implicating the vagus nerve as a conduit for translocation. The presence of bacteria in the brain correlated with microglial activation, a marker of neuroinflammation, and with neural protein aggregation, a hallmark of several neurodegenerative diseases. In at least one model, the presence of bacteria in the brain was reversible as a switch from high-fat to standard diet resulted in amelioration of intestinal permeability, led to a gradual loss of detectable bacteria in the brain, and reduced the number of neural protein aggregates. Further, in murine models of Alzheimer's disease, Parkinson's disease, and autism spectrum disorder, we observed gut dysbiosis, gut leakiness, bacterial translocation to the brain, and microglial activation. These data reveal a commensal bacterial translocation axis to the brain in models of diverse neurological diseases.

5.
mBio ; 12(6): e0274921, 2021 12 21.
Article in English | MEDLINE | ID: mdl-34749524

ABSTRACT

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has caused a historic pandemic of respiratory disease (coronavirus disease 2019 [COVID-19]), and current evidence suggests that severe disease is associated with dysregulated immunity within the respiratory tract. However, the innate immune mechanisms that mediate protection during COVID-19 are not well defined. Here, we characterize a mouse model of SARS-CoV-2 infection and find that early CCR2 signaling restricts the viral burden in the lung. We find that a recently developed mouse-adapted SARS-CoV-2 (MA-SARS-CoV-2) strain as well as the emerging B.1.351 variant trigger an inflammatory response in the lung characterized by the expression of proinflammatory cytokines and interferon-stimulated genes. Using intravital antibody labeling, we demonstrate that MA-SARS-CoV-2 infection leads to increases in circulating monocytes and an influx of CD45+ cells into the lung parenchyma that is dominated by monocyte-derived cells. Single-cell RNA sequencing (scRNA-Seq) analysis of lung homogenates identified a hyperinflammatory monocyte profile. We utilize this model to demonstrate that mechanistically, CCR2 signaling promotes the infiltration of classical monocytes into the lung and the expansion of monocyte-derived cells. Parenchymal monocyte-derived cells appear to play a protective role against MA-SARS-CoV-2, as mice lacking CCR2 showed higher viral loads in the lungs, increased lung viral dissemination, and elevated inflammatory cytokine responses. These studies have identified a potential CCR2-monocyte axis that is critical for promoting viral control and restricting inflammation within the respiratory tract during SARS-CoV-2 infection. IMPORTANCE SARS-CoV-2 has caused a historic pandemic of respiratory disease (COVID-19), and current evidence suggests that severe disease is associated with dysregulated immunity within the respiratory tract. However, the innate immune mechanisms that mediate protection during COVID-19 are not well defined. Here, we characterize a mouse model of SARS-CoV-2 infection and find that early CCR2-dependent infiltration of monocytes restricts the viral burden in the lung. We find that SARS-CoV-2 triggers an inflammatory response in the lung characterized by the expression of proinflammatory cytokines and interferon-stimulated genes. Using RNA sequencing and flow cytometry approaches, we demonstrate that SARS-CoV-2 infection leads to increases in circulating monocytes and an influx of CD45+ cells into the lung parenchyma that is dominated by monocyte-derived cells. Mechanistically, CCR2 signaling promoted the infiltration of classical monocytes into the lung and the expansion of monocyte-derived cells. Parenchymal monocyte-derived cells appear to play a protective role against MA-SARS-CoV-2, as mice lacking CCR2 showed higher viral loads in the lungs, increased lung viral dissemination, and elevated inflammatory cytokine responses. These studies have identified that the CCR2 pathway is critical for promoting viral control and restricting inflammation within the respiratory tract during SARS-CoV-2 infection.


Subject(s)
Lung/immunology , Pneumonia, Viral/prevention & control , Receptors, CCR2/immunology , SARS-CoV-2/immunology , Signal Transduction/immunology , Animals , COVID-19 , Cytokines/immunology , Disease Models, Animal , Female , Immunity, Innate , Inflammation , Lung/cytology , Lung/virology , Mice , Mice, Inbred C57BL , Monocytes/immunology , Pneumonia, Viral/immunology , Pneumonia, Viral/virology , Receptors, CCR2/genetics , Receptors, CCR2/metabolism , SARS-CoV-2/genetics , Viral Load , Virus Replication/immunology
6.
Mol Biol Cell ; 32(22): ar36, 2021 12 01.
Article in English | MEDLINE | ID: mdl-34668760

ABSTRACT

DNA replication stress activates the S-phase checkpoint that arrests the cell cycle, but it is poorly understood how cells recover from this arrest. Cyclin-dependent kinase (CDK) and protein phosphatase 2A (PP2A) are key cell cycle regulators, and Cdc55 is a regulatory subunit of PP2A in budding yeast. We found that yeast cells lacking functional PP2ACdc55 showed slow growth in the presence of hydroxyurea (HU), a DNA synthesis inhibitor, without obvious viability loss. Moreover, PP2A mutants exhibited delayed anaphase entry and sustained levels of anaphase inhibitor Pds1 after HU treatment. A DNA damage checkpoint Chk1 phosphorylates and stabilizes Pds1. We show that chk1Δ and mutation of the Chk1 phosphorylation sites in Pds1 largely restored efficient anaphase entry in PP2A mutants after HU treatment. In addition, deletion of SWE1, which encodes the inhibitory kinase for CDK or mutation of the Swe1 phosphorylation site in CDK (cdc28F19), also suppressed the anaphase entry delay in PP2A mutants after HU treatment. Our genetic data suggest that Swe1/CDK acts upstream of Pds1. Surprisingly, cdc55Δ showed significant suppression to the viability loss of S-phase checkpoint mutants during DNA synthesis block. Together, our results uncover a PP2A-Swe1-CDK-Chk1-Pds1 axis that promotes recovery from DNA replication stress.


Subject(s)
Anaphase/genetics , DNA Replication/physiology , Protein Phosphatase 2/metabolism , Saccharomyces cerevisiae/genetics , CDC2 Protein Kinase/genetics , CDC2 Protein Kinase/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Checkpoint Kinase 1/genetics , Checkpoint Kinase 1/metabolism , DNA Replication/drug effects , Green Fluorescent Proteins/genetics , Hydroxyurea/pharmacology , Microorganisms, Genetically-Modified , Mutation , Phosphorylation , Protein Phosphatase 2/genetics , Protein-Tyrosine Kinases/genetics , Protein-Tyrosine Kinases/metabolism , S Phase/genetics , Saccharomyces cerevisiae/drug effects , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Securin/genetics , Securin/metabolism
7.
bioRxiv ; 2021 May 04.
Article in English | MEDLINE | ID: mdl-33972938

ABSTRACT

SARS-CoV-2 has caused a historic pandemic of respiratory disease (COVID-19) and current evidence suggests severe disease is associated with dysregulated immunity within the respiratory tract. However, the innate immune mechanisms that mediate protection during COVID-19 are not well defined. Here we characterize a mouse model of SARS-CoV-2 infection and find that early CCR2-dependent infiltration of monocytes restricts viral burden in the lung. We find that a recently developed mouse-adapted MA-SARS-CoV-2 strain, as well as the emerging B. 1.351 variant, trigger an inflammatory response in the lung characterized by expression of pro-inflammatory cytokines and interferon-stimulated genes. scRNA-seq analysis of lung homogenates identified a hyper-inflammatory monocyte profile. Using intravital antibody labeling, we demonstrate that MA-SARS-CoV-2 infection leads to increases in circulating monocytes and an influx of CD45+ cells into the lung parenchyma that is dominated by monocyte-derived cells. We utilize this model to demonstrate that mechanistically, CCR2 signaling promotes infiltration of classical monocytes into the lung and expansion of monocyte-derived cells. Parenchymal monocyte-derived cells appear to play a protective role against MA-SARS-CoV-2, as mice lacking CCR2 showed higher viral loads in the lungs, increased lung viral dissemination, and elevated inflammatory cytokine responses. These studies have identified a CCR2-monocyte axis that is critical for promoting viral control and restricting inflammation within the respiratory tract during SARS-CoV-2 infection.

8.
Sci Rep ; 7(1): 11880, 2017 09 19.
Article in English | MEDLINE | ID: mdl-28928489

ABSTRACT

The interaction between chromosomes and spindle microtubules is essential for chromosome segregation. The kinetochore complex mediates this interaction. Previous studies indicate that the stability of kinetochore attachment is regulated by Aurora B/Ipl1 kinase and this regulation is conserved from yeast to mammalian cells. In budding yeast Saccharomyces cerevisiae, the ten-subunit Dam1/DASH complex bridges the interaction between kinetochores and microtubules, and some in vitro evidence indicates that the phosphorylation of Dam1 protein by Ipl1 kinase destabilizes this interaction. However, it is not clear if Dam1 phosphorylation is sufficient to regulate the stability of kinetochore attachment in vivo. Also, the significance of this regulation in response to chromosome detachment has not been fully investigated. Here we report that phospho-deficient dam1-3A mutants show stabilized kinetochore-microtubule attachment in vivo. This significantly delays the establishment of chromosome bipolar attachment after the disruption of kinetochore-microtubule interaction by a microtubule depolymerizing drug nocodazole. Moreover, dam1-3A cells show dramatic chromosome mis-segregation after treatment with nocodazole, presumably due to the combination of compromised bipolar attachment and premature spindle assembly checkpoint silencing in the mutant cells. Therefore, the regulation of Dam1 phosphorylation imposed by Ipl1 kinase is critical for faithful chromosome segregation.


Subject(s)
Aurora Kinase B/metabolism , Cell Cycle Proteins/metabolism , Chromosomes, Fungal/metabolism , Kinetochores/metabolism , Microtubule-Associated Proteins/metabolism , Microtubules/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Aurora Kinase B/genetics , Cell Cycle Proteins/genetics , Chromosome Segregation , Chromosomes, Fungal/genetics , Microtubule-Associated Proteins/genetics , Microtubules/genetics , Mutation , Phosphorylation , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics
9.
Genetics ; 205(3): 1169-1178, 2017 03.
Article in English | MEDLINE | ID: mdl-28040741

ABSTRACT

The spindle assembly checkpoint (SAC) monitors mistakes in kinetochore-microtubule interaction and its activation prevents anaphase entry. The SAC remains active until all chromosomes have achieved bipolar attachment which applies tension on kinetochores. Our previous data in budding yeast Saccharomyces cerevisiae show that Ipl1/Aurora B kinase and a centromere-associated protein, Sgo1, are required to prevent SAC silencing prior to tension generation, but we believe that this regulatory network is incomplete. Bub1 kinase is one of the SAC components, and Bub1-dependent H2A phosphorylation triggers centromere recruitment of Sgo1 by H2A in yeast and human cells. Although yeast cells lacking the kinase domain of Bub1 show competent SAC activation, we found that the mutant cells fail to maintain a prolonged checkpoint arrest in the presence of tensionless attachment. Mutation of the Bub1 phosphorylation site in H2A also results in premature SAC silencing in yeast cells. Previous data indicate that Sgo1 protein binds to PP2ARts1, and we found that rts1Δ mutants exhibited premature SAC silencing as well. We further revealed that sgo1 mutants with abolished binding to H2A or PP2ARts1 displayed premature SAC silencing. Together, our results suggest that, in budding yeast S. cerevisiae, the Bub1-H2A-Sgo1-PP2ARts1 axis prevents SAC silencing and helps prolonged checkpoint arrest prior to tension establishment at kinetochores.


Subject(s)
Histones/metabolism , M Phase Cell Cycle Checkpoints , Nuclear Proteins/metabolism , Protein Phosphatase 2/metabolism , Protein Serine-Threonine Kinases/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/genetics , Nuclear Proteins/genetics , Phosphorylation , Protein Phosphatase 2/genetics , Protein Processing, Post-Translational , Protein Serine-Threonine Kinases/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Signal Transduction
10.
Cell Rep ; 14(5): 1074-1085, 2016 Feb 09.
Article in English | MEDLINE | ID: mdl-26832405

ABSTRACT

The spindle assembly checkpoint (SAC) monitors chromosome attachment defects, and the assembly of SAC proteins at kinetochores is essential for its activation, but the SAC disassembly process remains unknown. We found that deletion of a 14-3-3 protein, Bmh1, or hyperactivation of Cdc14 early anaphase release (FEAR) allows premature SAC silencing in budding yeast, which depends on a kinetochore protein Fin1 that forms a complex with protein phosphatase PP1. Previous works suggest that FEAR-dependent Fin1 dephosphorylation promotes Bmh1-Fin1 dissociation, which enables kinetochore recruitment of Fin1-PP1. We found persistent kinetochore association of SAC protein Bub1 in fin1Δ mutants after anaphase entry. Therefore, we revealed a mechanism that clears SAC proteins from kinetochores. After anaphase entry, FEAR activation promotes kinetochore enrichment of Fin1-PP1, resulting in SAC disassembly at kinetochores. This mechanism is required for efficient SAC silencing after SAC is challenged, and untimely Fin1-kinetochore association causes premature SAC silencing and chromosome missegregation.


Subject(s)
Anaphase , Cytoskeletal Proteins/metabolism , Kinetochores/metabolism , M Phase Cell Cycle Checkpoints , Protein Serine-Threonine Kinases/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/cytology , Chromosome Segregation , Gene Silencing , Microbial Viability , Mutation/genetics , Saccharomyces cerevisiae/metabolism
11.
Cell Cycle ; 13(11): 1694-701, 2014.
Article in English | MEDLINE | ID: mdl-24776751

ABSTRACT

Chromosome bipolar attachment is achieved when sister kinetochores are attached by microtubules emanating from opposite spindle poles, and this process is essential for faithful chromosome segregation during anaphase. A fundamental question in cell biology is how cells ensure that chromosome segregation only occurs after bipolar attachment. It is well documented that unattached kinetochores activate the spindle assembly checkpoint (SAC) to delay chromosome segregation. Therefore, the silencing of the SAC is thought to trigger anaphase onset, but how correct chromosome attachment is coupled with SAC silencing and the subsequent anaphase onset is poorly understood. The establishment of chromosome bipolar attachment not only results in the occupancy of kinetochores by microtubules but also applies tension on sister kinetochores. A long-standing debate is whether the kinetochore attachment (occupancy) or the tension silences the SAC. Recent work in budding yeast reveals the SAC silencing network SSN that prevents SAC silencing prior to tension generation at kinetochores. Therefore, this signaling pathway ensures that SAC silencing and the subsequent anaphase onset occur only after chromosome bipolar attachment applies tension on chromosomes. This review will summarize the recent advances in the understanding of the SAC silencing process.


Subject(s)
Chromosome Segregation/physiology , Kinetochores/physiology , M Phase Cell Cycle Checkpoints/physiology , Models, Genetic , Signal Transduction/physiology , Dyneins/metabolism , M Phase Cell Cycle Checkpoints/genetics , Saccharomycetales
12.
Proc Natl Acad Sci U S A ; 110(52): 21036-41, 2013 Dec 24.
Article in English | MEDLINE | ID: mdl-24324173

ABSTRACT

Improper kinetochore attachments activate the spindle assembly checkpoint (SAC) to prevent anaphase onset, but it is poorly understood how this checkpoint is silenced to allow anaphase onset. Chromosome bipolar attachment applies tension on sister kinetochores, and the lack of tension delays anaphase onset. In budding yeast, the delay induced by tension defects depends on the intact SAC as well as increase in ploidy (Ipl1)/Aurora kinase and a centromere-associated protein ShuGOshin (Sgo1). Here we provide evidence indicating that Ipl1-dependent phosphorylation of the kinetochore protein Duo1 and Mps1 interacting (Dam1) prevents SAC silencing when tension is absent. The nonphosphorylatable dam1 mutant cells, as well as sgo1 mutant cells, are competent in SAC activation but unable to prevent SAC silencing in response to tension defects. We further found that phosphomimetic dam1 mutants exhibited delayed anaphase onset mainly due to the failure in SAC silencing, but destabilized kinetochore attachment likely plays a minor role in this delay. Because the tension resulting from bipolar attachment triggers the dephosphorylation of Dam1 by protein phosphatase 1, this dephosphorylation likely coordinates SAC silencing with chromosome bipolar attachment. Therefore, Sgo1, Ipl1 kinase, Dam1, and protein phosphatase 1 comprise the SAC silencing network that ensures the correct timing for anaphase onset.


Subject(s)
Anaphase/physiology , Cell Cycle Proteins/metabolism , Kinetochores/metabolism , M Phase Cell Cycle Checkpoints/physiology , Microtubule-Associated Proteins/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Signal Transduction/physiology , Aurora Kinases/metabolism , Biomechanical Phenomena , Blotting, Western , Kinetochores/physiology , Microscopy, Fluorescence , Nuclear Proteins/metabolism , Phosphorylation , Protein Phosphatase 1/metabolism , Saccharomycetales
13.
PLoS Genet ; 8(2): e1002492, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22319456

ABSTRACT

The attachment of sister kinetochores by microtubules emanating from opposite spindle poles establishes chromosome bipolar attachment, which generates tension on chromosomes and is essential for sister-chromatid segregation. Syntelic attachment occurs when both sister kinetochores are attached by microtubules from the same spindle pole and this attachment is unable to generate tension on chromosomes, but a reliable method to induce syntelic attachments is not available in budding yeast. The spindle checkpoint can sense the lack of tension on chromosomes as well as detached kinetochores to prevent anaphase onset. In budding yeast Saccharomyces cerevisiae, tension checkpoint proteins Aurora/Ipl1 kinase and centromere-localized Sgo1 are required to sense the absence of tension but are dispensable for the checkpoint response to detached kinetochores. We have found that the loss of function of a motor protein complex Cik1/Kar3 in budding yeast leads to syntelic attachments. Inactivation of either the spindle or tension checkpoint enables premature anaphase entry in cells with dysfunctional Cik1/Kar3, resulting in co-segregation of sister chromatids. Moreover, the abolished Kar3-kinetochore interaction in cik1 mutants suggests that the Cik1/Kar3 complex mediates chromosome movement along microtubules, which could facilitate bipolar attachment. Therefore, we can induce syntelic attachments in budding yeast by inactivating the Cik1/Kar3 complex, and this approach will be very useful to study the checkpoint response to syntelic attachments.


Subject(s)
Chromosomes/genetics , Kinetochores , M Phase Cell Cycle Checkpoints/genetics , Microtubule Proteins/genetics , Microtubule-Associated Proteins/genetics , Microtubules/genetics , Mitosis/genetics , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae/genetics , Anaphase/genetics , Aurora Kinases , Chromosome Segregation/genetics , Gene Expression Regulation, Fungal , Mutation , Nuclear Proteins/genetics , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Sister Chromatid Exchange/genetics
14.
Genetics ; 187(2): 397-407, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21135072

ABSTRACT

In budding yeast Saccharomyces cerevisiae, kinetochores are attached by microtubules during most of the cell cycle, but the duplication of centromeric DNA disassembles kinetochores, which results in a brief dissociation of chromosomes from microtubules. Kinetochore assembly is delayed in the presence of hydroxyurea, a DNA synthesis inhibitor, presumably due to the longer time required for centromeric DNA duplication. Some kinetochore mutants are sensitive to stressful DNA replication as these kinetochore proteins become essential for the establishment of the kinetochore-microtubule interaction after treatment with hydroxyurea. To identify more genes required for the efficient kinetochore-microtubule interaction under stressful DNA replication conditions, we carried out a genome-wide screen for yeast mutants sensitive to hydroxyurea. From this screen, cik1 and kar3 mutants were isolated. Kar3 is the minus-end-directed motor protein; Cik1 binds to Kar3 and is required for its motor function. After exposure to hydroxyurea, cik1 and kar3 mutant cells exhibit normal DNA synthesis kinetics, but they display a significant anaphase entry delay. Our results indicate that cik1 cells exhibit a defect in the establishment of chromosome bipolar attachment in the presence of hydroxyurea. Since Kar3 has been shown to drive the poleward chromosome movement along microtubules, our data support the possibility that this chromosome movement promotes chromosome bipolar attachment after stressful DNA replication.


Subject(s)
DNA Replication , Kinetochores/metabolism , Microtubule Proteins/genetics , Microtubule Proteins/metabolism , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Microtubules/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Anaphase/drug effects , DNA Replication/drug effects , Hydroxyurea/pharmacology , Intracellular Space/metabolism , Mutation , Nucleic Acid Synthesis Inhibitors/pharmacology , Protein Binding/drug effects , Protein Transport/drug effects
15.
Mol Biol Cell ; 20(16): 3671-9, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19570916

ABSTRACT

In the budding yeast Saccharomyces cerevisiae, Cdc14 is sequestered within the nucleolus before anaphase entry through its association with Net1/Cfi1, a nucleolar protein. Protein phosphatase PP2A(Cdc55) dephosphorylates Net1 and keeps it as a hypophosphorylated form before anaphase. Activation of the Cdc fourteen early anaphase release (FEAR) pathway after anaphase entry induces a brief Cdc14 release from the nucleolus. Some of the components in the FEAR pathway, including Esp1, Slk19, and Spo12, inactivate PP2A(Cdc55), allowing the phosphorylation of Net1 by mitotic cyclin-dependent kinase (Cdk) (Clb2-Cdk1). However, the function of another FEAR component, the Polo-like kinase Cdc5, remains elusive. Here, we show evidence indicating that Cdc5 promotes Cdc14 release primarily by stimulating the degradation of Swe1, the inhibitory kinase for mitotic Cdk. First, we found that deletion of SWE1 partially suppresses the FEAR defects in cdc5 mutants. In contrast, high levels of Swe1 impair FEAR activation. We also demonstrated that the accumulation of Swe1 in cdc5 mutants is responsible for the decreased Net1 phosphorylation. Therefore, we conclude that the down-regulation of Swe1 protein levels by Cdc5 promotes FEAR activation by relieving the inhibition on Clb2-Cdk1, the kinase for Net1 protein.


Subject(s)
Anaphase/physiology , Cell Cycle Proteins/metabolism , Protein Kinases/metabolism , Protein Tyrosine Phosphatases/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae/metabolism , CDC2 Protein Kinase/genetics , CDC2 Protein Kinase/metabolism , Cell Cycle Proteins/genetics , Cell Nucleolus/metabolism , Cyclin B/genetics , Cyclin B/metabolism , DNA, Ribosomal/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Protein Kinases/genetics , Protein Serine-Threonine Kinases , Protein Tyrosine Phosphatases/genetics , Protein-Tyrosine Kinases/genetics , Protein-Tyrosine Kinases/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Signal Transduction/physiology
16.
Cell Cycle ; 8(5): 700-4, 2009 Mar 01.
Article in English | MEDLINE | ID: mdl-19221480

ABSTRACT

The fluctuating activity of the cyclin-dependent kinases (Cdks) is critical for the periodic phosphorylation of a given Cdk substrate. Previous studies have been focus on the positive role of Cdk-dependent protein phosphorylation in cell cycle progression. Recent studies indicate that, in budding yeast, highly active S-phase cyclin-associated Cdk not only promotes DNA synthesis but also inhibits the initiation of chromosome segregation. The FEAR (Cdc14 early anaphase release) pathway alleviates the negative effect of the S-phase cyclin on anaphase by promoting the dephosphorylation of S-phase cyclin-specific substrates, revealing a new layer of regulation in the metaphase-to-anaphase transition.


Subject(s)
Anaphase , Cyclin-Dependent Kinases/metabolism , Metaphase , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Chromosome Segregation , Cyclin-Dependent Kinases/genetics , Gene Expression Regulation , Mitosis , Models, Biological , Phosphorylation , S Phase , Saccharomycetales/genetics , Saccharomycetales/metabolism
17.
PLoS Genet ; 4(11): e1000262, 2008 Nov.
Article in English | MEDLINE | ID: mdl-19023403

ABSTRACT

The kinetochore is a protein complex that assembles on centromeric DNA to mediate chromosome-microtubule interaction. Most eukaryotic cells form the spindle and establish kinetochore-microtubule interaction during mitosis, but budding yeast cells finish these processes in S-phase. It has long been noticed that the S-phase spindle in budding yeast is shorter than that in metaphase, but the biological significance of this short S-phase spindle structure remains unclear. We addressed this issue by using ask1-3, a temperature-sensitive kinetochore mutant that exhibits partially elongated spindles at permissive temperature in the presence of hydroxyurea (HU), a DNA synthesis inhibitor. After exposure to and removal of HU, ask1-3 cells show a delayed anaphase entry. This delay depends on the spindle checkpoint, which monitors kinetochore-microtubule interaction defects. Overproduction of microtubule-associated protein Ase1 or Cin8 also induces spindle elongation in HU-arrested cells. The spindle checkpoint-dependent anaphase entry delay is also observed after ASE1 or CIN8 overexpression in HU-arrested cells. Therefore, the shorter spindle in S-phase cells is likely to facilitate proper chromosome-microtubule interaction.


Subject(s)
Centromere/metabolism , Kinetochores/metabolism , Microtubules/metabolism , Saccharomycetales/genetics , Spindle Apparatus/metabolism , Anaphase , Cells, Cultured , Genes, Fungal , Hydroxyurea/pharmacology , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , S Phase , Saccharomycetales/metabolism
18.
Proc Natl Acad Sci U S A ; 105(42): 16177-82, 2008 Oct 21.
Article in English | MEDLINE | ID: mdl-18845678

ABSTRACT

The temporal phosphorylation of cell cycle-related proteins by cyclin-dependent kinases (Cdks) is critical for the correct order of cell cycle events. In budding yeast, CDC28 encodes the only Cdk and its association with various cyclins governs the temporal phosphorylation of Cdk substrates. S-phase Cdk substrates are phosphorylated earlier than mitotic Cdk substrates, which ensures the sequential order of DNA synthesis and mitosis. However, it remains unclear whether Cdk substrates are dephosphorylated in temporally distinct windows. Cdc14 is a conserved protein phosphatase responsible for the dephosphorylation of Cdk substrates. In budding yeast, FEAR (Cdc14 early anaphase release) and MEN (mitotic exit network) activate phosphatase Cdc14 by promoting its release from the nucleolus in early and late anaphase, respectively. Here, we show that the sequential Cdc14 release and the distinct degradation timing of different cyclins provides the molecular basis for the differential dephosphorylation windows of S-phase and mitotic cyclin substrates. Our data also indicate that FEAR-induced dephosphorylation of S-phase Cdk substrates facilitates anaphase progression, revealing an extra layer of mitotic regulation.


Subject(s)
Cyclin-Dependent Kinases/metabolism , Mitosis , Saccharomycetales/cytology , Saccharomycetales/enzymology , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cyclin-Dependent Kinases/genetics , Cyclins/metabolism , Mutation/genetics , Phosphoprotein Phosphatases/genetics , Phosphoprotein Phosphatases/metabolism , Phosphorylation , Protein Tyrosine Phosphatases/genetics , Protein Tyrosine Phosphatases/metabolism , Saccharomycetales/genetics , Substrate Specificity , Time Factors
19.
Cell Cycle ; 5(24): 2914-9, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17172870

ABSTRACT

In the presence of double strand breaks, DNA damage checkpoint halts cell cycle progression. However, cells ultimately escape the checkpoint arrest and reenter cell cycle in the presence of irreparable DNA damage. cdc5-ad was identified as a mutant that fails to adapt to the cell cycle arrest induced by DNA damage checkpoint. In budding yeast, Cdc5 protein kinase is a component of both MEN and FEAR pathways that are required for mitotic exit. It remains unclear whether the adaptation defect of cdc5-ad mutant cells is related to the function of Cdc5 in mitotic exit. Here we present evidence indicating that cdc5-ad mutant cells exhibit defects in mitotic exit. cdc5-ad mutant cells are sensitive to high dosage of Amn1, a negative regulator of MEN. It also shows synthetic growth defects with mutants in MEN pathway. Moreover, mutants in FEAR pathway exhibit defects in DNA damage adaptation. Thus, we conclude that the compromised mitotic exit pathway contributes to DNA damage adaptation defects in cdc5-ad mutant cells.


Subject(s)
Adaptation, Physiological , DNA Damage , Mitosis , Mutation/genetics , Saccharomycetales/cytology , Saccharomycetales/genetics , Cell Cycle Proteins/metabolism , Cell Cycle Proteins/toxicity , Cyclin B/metabolism , Gene Expression , Protein Kinases/metabolism , Protein Processing, Post-Translational , Protein Serine-Threonine Kinases , Protein-Tyrosine Kinases/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae Proteins/toxicity , Saccharomycetales/drug effects , Saccharomycetales/growth & development
SELECTION OF CITATIONS
SEARCH DETAIL
...