Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
Gastroenterology ; 153(1): 263-276.e8, 2017 07.
Article in English | MEDLINE | ID: mdl-28363639

ABSTRACT

BACKGROUND & AIMS: Patients with acute liver failure (ALF) have defects in innate immune responses to microbes (immune paresis) and are susceptible to sepsis. Cytotoxic T-lymphocyte-associated protein 4 (CTLA4), which interacts with the membrane receptor B7 (also called CD80 and CD86), is a negative regulator of T-cell activation. We collected T cells from patients with ALF and investigated whether inhibitory signals down-regulate adaptive immune responses in patients with ALF. METHODS: We collected peripheral blood mononuclear cells from patients with ALF and controls from September 2013 through September 2015 (45 patients with ALF, 20 patients with acute-on-chronic liver failure, 15 patients with cirrhosis with no evidence of acute decompensation, 20 patients with septic shock but no cirrhosis or liver disease, and 20 healthy individuals). Circulating CD4+ T cells were isolated and analyzed by flow cytometry. CD4+ T cells were incubated with antigen, or agonist to CD3 and dendritic cells, with or without antibody against CTLA4; T-cell proliferation and protein expression were quantified. We measured levels of soluble B7 molecules in supernatants of isolated primary hepatocytes, hepatic sinusoidal endothelial cells, and biliary epithelial cells from healthy or diseased liver tissues. We also measured levels of soluble B7 serum samples from patients and controls, and mice with acetaminophen-induced liver injury using enzyme-linked immunosorbent assays. RESULTS: Peripheral blood samples from patients with ALF had a higher proportion of CD4+ CTLA4+ T cells than controls; patients with infections had the highest proportions. CD4+ T cells from patients with ALF had a reduced proliferative response to antigen or CD3 stimulation compared to cells from controls; incubation of CD4+ T cells from patients with ALF with an antibody against CTLA4 increased their proliferative response to antigen and to CD3 stimulation, to the same levels as cells from controls. CD4+ T cells from controls up-regulated expression of CTLA4 after 24-48 hours culture with sera from patients with ALF; these sera were found to have increased concentrations of soluble B7 compared to sera from controls. Necrotic human primary hepatocytes exposed to acetaminophen, but not hepatic sinusoidal endothelial cells and biliary epithelial cells from patients with ALF, secreted high levels of soluble B7. Sera from mice with acetaminophen-induced liver injury contained high levels of soluble B7 compared to sera from mice without liver injury. Plasma exchange reduced circulating levels of soluble B7 in patients with ALF and expression of CTLA4 on T cells. CONCLUSIONS: Peripheral CD4+ T cells from patients with ALF have increased expression of CTLA4 compared to individuals without ALF; these cells have a reduced response to antigen and CD3 stimulation. We found sera of patients with ALF and from mice with liver injury to have high concentrations of soluble B7, which up-regulates CTLA4 expression by T cells and reduces their response to antigen. Plasma exchange reduces levels of B7 in sera from patients with ALF and might be used to restore antimicrobial responses to patients.


Subject(s)
Adaptive Immunity , B7-1 Antigen/blood , CD4-Positive T-Lymphocytes/metabolism , CTLA-4 Antigen/metabolism , Liver Failure, Acute/immunology , Acetaminophen/toxicity , Acute-On-Chronic Liver Failure/immunology , Adult , Animals , Antibodies/pharmacology , B7-1 Antigen/metabolism , CD3 Complex/pharmacology , CD4 Lymphocyte Count , CD4-Positive T-Lymphocytes/immunology , CTLA-4 Antigen/immunology , Cell Proliferation , Cells, Cultured , Chemical and Drug Induced Liver Injury/blood , Coculture Techniques , Dendritic Cells , Hepatocytes/metabolism , Humans , Liver Cirrhosis/immunology , Lymphocyte Activation , Mice , Middle Aged , Shock, Septic/immunology
2.
Genes Dev ; 24(22): 2505-16, 2010 Nov 15.
Article in English | MEDLINE | ID: mdl-20971813

ABSTRACT

During female meiosis, bivalent chromosomes are thought to be held together from birth until ovulation by sister chromatid cohesion mediated by cohesin complexes whose ring structure depends on kleisin subunits, either Rec8 or Scc1. Because cohesion is established at DNA replication in the embryo, its maintenance for such a long time may require cohesin turnover. To address whether Rec8- or Scc1-containing cohesin holds bivalents together and whether it turns over, we created mice whose kleisin subunits can be cleaved by TEV protease. We show by microinjection experiments and confocal live-cell imaging that Rec8 cleavage triggers chiasmata resolution during meiosis I and sister centromere disjunction during meiosis II, while Scc1 cleavage triggers sister chromatid disjunction in the first embryonic mitosis, demonstrating a dramatic transition from Rec8- to Scc1-containing cohesin at fertilization. Crucially, activation of an ectopic Rec8 transgene during the growing phase of Rec8(TEV)(/TEV) oocytes does not prevent TEV-mediated bivalent destruction, implying little or no cohesin turnover for ≥2 wk during oocyte growth. We suggest that the inability of oocytes to regenerate cohesion may contribute to age-related meiosis I errors.


Subject(s)
Cell Cycle Proteins/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Chromosomes/metabolism , Nuclear Proteins/metabolism , Oocytes/growth & development , Oocytes/metabolism , Phosphoproteins/metabolism , Animals , Cells, Cultured , Centromere/genetics , Chromosomes/genetics , Endopeptidases/metabolism , Female , Mice , Nuclear Proteins/genetics , Phosphoproteins/genetics , Cohesins
3.
J Cell Biol ; 172(6): 847-60, 2006 Mar 13.
Article in English | MEDLINE | ID: mdl-16533945

ABSTRACT

Separase is a protease whose liberation from its inhibitory chaperone Securin triggers sister chromatid disjunction at anaphase onset in yeast by cleaving cohesin's kleisin subunit. We have created conditional knockout alleles of the mouse Separase and Securin genes. Deletion of both copies of Separase but not Securin causes embryonic lethality. Loss of Securin reduces Separase activity because deletion of just one copy of the Separase gene is lethal to embryos lacking Securin. In embryonic fibroblasts, Separase depletion blocks sister chromatid separation but does not prevent other aspects of mitosis, cytokinesis, or chromosome replication. Thus, fibroblasts lacking Separase become highly polyploid. Hepatocytes stimulated to proliferate in vivo by hepatectomy also become unusually large and polyploid in the absence of Separase but are able to regenerate functional livers. Separase depletion in bone marrow causes aplasia and the presumed death of hematopoietic cells other than erythrocytes. Destruction of sister chromatid cohesion by Separase may be a universal feature of mitosis in eukaryotic cells.


Subject(s)
Cell Cycle Proteins/genetics , Cell Cycle/genetics , Chromosome Segregation/genetics , DNA Replication Timing/genetics , Endopeptidases/genetics , Mitosis/genetics , Anaphase/genetics , Animals , Carrier Proteins/genetics , Cell Line , Chromosomal Proteins, Non-Histone/genetics , Embryonic Development/genetics , Female , Fibroblasts , Genes, Lethal/genetics , Hematopoietic Stem Cells/metabolism , Hepatocytes , Liver Regeneration/genetics , Male , Mice , Mice, Knockout , Nuclear Proteins/genetics , Polyploidy , Securin , Separase , Cohesins
4.
PLoS Biol ; 3(3): e86, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15737064

ABSTRACT

Cohesion between sister chromatids is essential for their bi-orientation on mitotic spindles. It is mediated by a multisubunit complex called cohesin. In yeast, proteolytic cleavage of cohesin's alpha kleisin subunit at the onset of anaphase removes cohesin from both centromeres and chromosome arms and thus triggers sister chromatid separation. In animal cells, most cohesin is removed from chromosome arms during prophase via a separase-independent pathway involving phosphorylation of its Scc3-SA1/2 subunits. Cohesin at centromeres is refractory to this process and persists until metaphase, whereupon its alpha kleisin subunit is cleaved by separase, which is thought to trigger anaphase. What protects centromeric cohesin from the prophase pathway? Potential candidates are proteins, known as shugoshins, that are homologous to Drosophila MEI-S332 and yeast Sgo1 proteins, which prevent removal of meiotic cohesin complexes from centromeres at the first meiotic division. A vertebrate shugoshin-like protein associates with centromeres during prophase and disappears at the onset of anaphase. Its depletion by RNA interference causes HeLa cells to arrest in mitosis. Most chromosomes bi-orient on a metaphase plate, but precocious loss of centromeric cohesin from chromosomes is accompanied by loss of all sister chromatid cohesion, the departure of individual chromatids from the metaphase plate, and a permanent cell cycle arrest, presumably due to activation of the spindle checkpoint. Remarkably, expression of a version of Scc3-SA2 whose mitotic phosphorylation sites have been mutated to alanine alleviates the precocious loss of sister chromatid cohesion and the mitotic arrest of cells lacking shugoshin. These data suggest that shugoshin prevents phosphorylation of cohesin's Scc3-SA2 subunit at centromeres during mitosis. This ensures that cohesin persists at centromeres until activation of separase causes cleavage of its alpha kleisin subunit. Centromeric cohesion is one of the hallmarks of mitotic chromosomes. Our results imply that it is not an intrinsically stable property, because it can easily be destroyed by mitotic kinases, which are kept in check by shugoshin.


Subject(s)
Cell Cycle Proteins/physiology , Centromere/physiology , Fungal Proteins/physiology , Mitosis/physiology , Nuclear Proteins/physiology , Amino Acid Sequence , Anaphase/physiology , Animals , Base Sequence , Chromosomal Proteins, Non-Histone , DNA Primers , HeLa Cells , Humans , Mice , Molecular Sequence Data , Peptide Fragments/chemistry , Proteins/chemistry , Proteins/genetics , RNA, Small Interfering/genetics , Sister Chromatid Exchange , Cohesins
5.
Nat Cell Biol ; 18(2): 225-233, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26751286

ABSTRACT

Zygotic epigenetic reprogramming entails genome-wide DNA demethylation that is accompanied by Tet methylcytosine dioxygenase 3 (Tet3)-driven oxidation of 5-methylcytosine (5mC) to 5-hydroxymethylcytosine (5hmC; refs 1-4). Here we demonstrate using detailed immunofluorescence analysis and ultrasensitive LC-MS-based quantitative measurements that the initial loss of paternal 5mC does not require 5hmC formation. Small-molecule inhibition of Tet3 activity, as well as genetic ablation, impedes 5hmC accumulation in zygotes without affecting the early loss of paternal 5mC. Instead, 5hmC accumulation is dependent on the activity of zygotic Dnmt3a and Dnmt1, documenting a role for Tet3-driven hydroxylation in targeting de novo methylation activities present in the early embryo. Our data thus provide further insights into the dynamics of zygotic reprogramming, revealing an intricate interplay between DNA demethylation, de novo methylation and Tet3-driven hydroxylation.


Subject(s)
5-Methylcytosine/metabolism , Cellular Reprogramming , Cytosine/analogs & derivatives , DNA Methylation , Epigenesis, Genetic , Zygote/metabolism , Animals , Biomarkers/metabolism , Chromatography, Liquid , Cytosine/metabolism , DNA (Cytosine-5-)-Methyltransferase 1 , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methyltransferase 3A , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Dioxygenases , Embryo Culture Techniques , Fertilization in Vitro , Fluorescent Antibody Technique , Gene Expression Regulation, Developmental , Kinetics , Mass Spectrometry , Mice , Mice, Knockout , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism
6.
J Cell Biol ; 204(6): 891-900, 2014 Mar 17.
Article in English | MEDLINE | ID: mdl-24637322

ABSTRACT

Female meiosis is driven by the activities of two major kinases, cyclin-dependent kinase 1 (Cdk1) and mitogen-activated protein kinase (MAPK). To date, the role of MAPK in control of meiosis is thought to be restricted to maintaining metaphase II arrest through stabilizing Cdk1 activity. In this paper, we find that MAPK and Cdk1 play compensatory roles to suppress the anaphase-promoting complex/cyclosome (APC/C) activity early in prometaphase, thereby allowing accumulation of APC/C substrates essential for meiosis I. Furthermore, inhibition of MAPK around the onset of APC/C activity at the transition from meiosis I to meiosis II led to accelerated completion of meiosis I and an increase in aneuploidy at metaphase II. These effects appear to be mediated via a Cdk1/MAPK-dependent stabilization of the spindle assembly checkpoint, which when inhibited leads to increased APC/C activity. These findings demonstrate new roles for MAPK in the regulation of meiosis in mammalian oocytes.


Subject(s)
Anaphase-Promoting Complex-Cyclosome/metabolism , CDC2 Protein Kinase/metabolism , Meiotic Prophase I , Mitogen-Activated Protein Kinases/metabolism , Animals , Cells, Cultured , Chromatids/metabolism , Chromosome Segregation , Female , Kinetochores/metabolism , M Phase Cell Cycle Checkpoints , Mad2 Proteins/metabolism , Mice , Mice, Knockout , Oocytes/enzymology , Prometaphase , Securin/metabolism
7.
J Cell Biol ; 206(7): 843-53, 2014 Sep 29.
Article in English | MEDLINE | ID: mdl-25246615

ABSTRACT

In mitosis, the Greatwall kinase (called microtubule-associated serine/threonine kinase like [Mastl] in mammals) is essential for prometaphase entry or progression by suppressing protein phosphatase 2A (PP2A) activity. PP2A suppression in turn leads to high levels of Cdk1 substrate phosphorylation. We have used a mouse model with an oocyte-specific deletion of Mastl to show that Mastl-null oocytes resume meiosis I and reach metaphase I normally but that the onset and completion of anaphase I are delayed. Moreover, after the completion of meiosis I, Mastl-null oocytes failed to enter meiosis II (MII) because they reassembled a nuclear structure containing decondensed chromatin. Our results show that Mastl is required for the timely activation of anaphase-promoting complex/cyclosome to allow meiosis I exit and for the rapid rise of Cdk1 activity that is needed for the entry into MII in mouse oocytes.


Subject(s)
Anaphase-Promoting Complex-Cyclosome/metabolism , CDC2 Protein Kinase/metabolism , Meiosis , Microtubule-Associated Proteins/physiology , Protein Serine-Threonine Kinases/physiology , Anaphase , Animals , CDC2 Protein Kinase/genetics , Enzyme Induction , Female , HEK293 Cells , Humans , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Fluorescence , Oocytes/enzymology , Phosphorylation , Protein Phosphatase 2/metabolism , Protein Processing, Post-Translational , Single-Cell Analysis
8.
Cell Div ; 8(1): 12, 2013 Aug 22.
Article in English | MEDLINE | ID: mdl-23967866

ABSTRACT

BACKGROUND: Sister chromatid cohesion mediated by the cohesin complex is essential for accurate chromosome segregation during mitosis and meiosis. Loading of cohesin onto chromosomes is dependent on another protein complex called kollerin, containing Nipbl/Scc2 and Mau2/Scc4. Nipbl is an evolutionarily conserved large protein whose haploinsufficiency in humans causes a developmental disorder called Cornelia de Lange syndrome. Although the function of Nipbl homologues for chromosome cohesion in meiotic cells of non-vertebrate models has been elucidated, Nipbl has not been characterized so far in mammalian spermatocytes or oocytes. FINDINGS: Here we describe our analyses on the expression and localization of Nipbl in nuclei of mouse spermatocytes and oocytes at different stages of meiotic prophase. In both spermatocytes and oocytes we found that Nipbl is associated with the axial/lateral element of the synaptonemal complex (AE/LE) to which cohesin also localizes. Interestingly, Nipbl in spermatocytes, but not in oocytes, dissociates from the AE/LE at mid-pachytene stage coincident with completion of DNA double-strand break repair. CONCLUSIONS: Our data propose that cohesin loading activity is maintained during early stages of meiotic prophase in mammalian spermatocytes and oocytes.

9.
Methods Mol Biol ; 957: 47-57, 2013.
Article in English | MEDLINE | ID: mdl-23138943

ABSTRACT

Distinct meiotic cohesin complexes play fundamental roles in various meiosis-specific chromosomal events in spatiotemporally different manners during mammalian meiotic prophase. Immunostaining is one of the essential methods to study meiotic cohesin dynamics. For the study of cohesins in the meiotic prophase of oocytes, ovaries should be taken from the embryos during a very limited period before birth. Here we focus on some technical tips concerning the preparation of oocyte chromosome spreads for immunostaining. Further, we describe a method for chromosome fluorescence in situ hybridization (FISH) against immunostained oocytes.


Subject(s)
Cell Cycle Proteins/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Cytogenetic Analysis/methods , Embryo, Mammalian/cytology , Meiosis/genetics , Oocytes/cytology , Oocytes/metabolism , Animals , Centrifugation , Chromosomes, Mammalian/genetics , Female , In Situ Hybridization, Fluorescence , Mice , Ovary/cytology , Ovary/embryology , Prophase/genetics , Cohesins
10.
Mol Cell Biol ; 33(8): 1561-70, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23401851

ABSTRACT

Posttranscriptional regulatory mechanisms are crucial for protein synthesis during spermatogenesis and are often organized by the chromatoid body. Here, we identify the RNA methyltransferase NSun2 as a novel component of the chromatoid body and, further, show that NSun2 is essential for germ cell differentiation in the mouse testis. In NSun2-depleted testes, genes encoding Ddx4, Miwi, and Tudor domain-containing (Tdr) proteins are repressed, indicating that RNA-processing and posttranscriptional pathways are impaired. Loss of NSun2 specifically blocked meiotic progression of germ cells into the pachytene stage, as spermatogonial and Sertoli cells were unaffected in knockout mice. We observed the same phenotype when we simultaneously deleted NSun2 and Dnmt2, the only other cytosine-5 RNA methyltransferase characterized to date, indicating that Dnmt2 was not functionally redundant with NSun2 in spermatogonial stem cells or Sertoli cells. Specific NSun2- and Dnmt2-methylated tRNAs decreased in abundance when both methyltransferases were deleted, suggesting that RNA methylation pathways play an essential role in male germ cell differentiation.


Subject(s)
DNA (Cytosine-5-)-Methyltransferases/metabolism , Methyltransferases/metabolism , Spermatogenesis , Spermatozoa/metabolism , Testis/cytology , Animals , Argonaute Proteins/genetics , Cell Differentiation , DEAD-box RNA Helicases/genetics , DNA (Cytosine-5-)-Methyltransferases/genetics , Gene Expression Profiling , Infertility, Male/genetics , Male , Meiotic Prophase I , Methylation , Methyltransferases/genetics , Mice , Mice, Knockout , Pachytene Stage/genetics , Protein Processing, Post-Translational , RNA/genetics , RNA/metabolism , RNA Processing, Post-Transcriptional , RNA, Transfer/genetics , RNA, Transfer/metabolism , Ribonucleoproteins, Small Nuclear/genetics , Sertoli Cells/metabolism , Spermatogenesis/genetics , Spermatogonia/metabolism , Testis/enzymology
11.
J Cell Sci ; 122(Pt 15): 2686-98, 2009 Aug 01.
Article in English | MEDLINE | ID: mdl-19625504

ABSTRACT

Proteolytic activity of separase is required for chiasma resolution during meiosis I in mouse oocytes. Rec8, the meiosis-specific alpha-kleisin subunit of cohesin, is a key target of separase in yeast. Is the equivalent protein also a target in mammals? We show here that separase cleaves mouse Rec8 at three positions in vitro but only when the latter is hyper-phosphorylated. Expression of a Rec8 variant (Rec8-N) that cannot be cleaved in vitro at these sites causes sterility in male mice. Their seminiferous tubules lack a normal complement of 2 C secondary spermatocytes and 1 C spermatids and contain instead a high proportion of cells with enlarged nuclei. Chromosome spreads reveal that Rec8-N expression has no effect in primary spermatocytes but produces secondary spermatocytes and spermatids with a 4 C DNA content, suggesting that the first and possibly also the second meiotic division is abolished. Expression of Rec8-N in oocytes causes chromosome segregation to be asynchronous and delays its completion by 2-3 hours during anaphase I, probably due to inefficient proteolysis of Rec8-N by separase. Despite this effect, chromosome segregation must be quite accurate as Rec8-N does not greatly reduce female fertility. Our data is consistent with the notion that Rec8 cleavage is important and probably crucial for the resolution of chiasmata in males and females.


Subject(s)
Cell Cycle Proteins/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Endopeptidases/metabolism , Meiosis/physiology , Nuclear Proteins/physiology , Phosphoproteins/physiology , Animals , Blotting, Western , Cell Cycle Proteins/genetics , Chromosome Segregation , Chromosomes, Artificial, Bacterial , Endopeptidases/genetics , Female , Genes, myc/physiology , Infertility, Male , Male , Mice , Mice, Transgenic , Mutagenesis, Site-Directed , Oocytes/cytology , Oocytes/metabolism , Peptide Fragments/metabolism , Protein Subunits , Separase , Spermatogenesis , Cohesins
12.
Curr Biol ; 19(5): 369-80, 2009 Mar 10.
Article in English | MEDLINE | ID: mdl-19249208

ABSTRACT

BACKGROUND: Missegregation of chromosomes during meiosis in human females causes aneuploidy, including trisomy 21, and is thought also to be the major cause of age-related infertility. Most errors are thought to occur at the first meiotic division. The high frequency of errors raises questions as to whether the surveillance mechanism known as the spindle assembly checkpoint (SAC) that controls the anaphase-promoting complex or cyclosome (APC/C) operates effectively in oocytes. Experimental approaches hitherto used to inactivate the SAC in oocytes suffer from a number of drawbacks. RESULTS: Bub1 protein was depleted specifically in oocytes with a Zp3-Cre transgene to delete exons 7 and 8 from a floxed BUB1(F) allele. Loss of Bub1 greatly accelerates resolution of chiasmata and extrusion of polar bodies. It also causes defective biorientation of bivalents, massive chromosome missegregation at meiosis I, and precocious loss of cohesion between sister centromeres. By using a quantitative assay for APC/C-mediated securin destruction, we show that the APC/C is activated in an exponential fashion, with activity peaking 12-13 hr after GVBD, and that this process is advanced by 5 hr in oocytes lacking Bub1. Importantly, premature chiasmata resolution does not occur in Bub1-deficient oocytes also lacking either the APC/C's Apc2 subunit or separase. Finally, we show that Bub1's kinase domain is not required to delay APC/C activation. CONCLUSIONS: We conclude that far from being absent or ineffective, the SAC largely determines the timing of APC/C and hence separase activation in oocytes, delaying it for about 5 hr.


Subject(s)
Oocytes/physiology , Protein Serine-Threonine Kinases/metabolism , Spindle Apparatus/metabolism , Ubiquitin-Protein Ligase Complexes/metabolism , Anaphase-Promoting Complex-Cyclosome , Animals , Apc2 Subunit, Anaphase-Promoting Complex-Cyclosome , Cell Cycle Proteins/metabolism , Chromosome Segregation , Endopeptidases/metabolism , Enzyme Activation , Female , Humans , Male , Meiosis/physiology , Mice , Mice, Transgenic , Oocytes/cytology , Pregnancy , Protein Serine-Threonine Kinases/genetics , Separase , Ubiquitin-Protein Ligase Complexes/genetics
13.
Cell ; 126(1): 135-46, 2006 Jul 14.
Article in English | MEDLINE | ID: mdl-16839882

ABSTRACT

In yeast, resolution of chiasmata in meiosis I requires proteolytic cleavage along chromosome arms of cohesin's Rec8 subunit by separase. Since activation of separase by the anaphase-promoting complex (APC/C) is supposedly not required for meiosis I in Xenopus oocytes, it has been suggested that animal cells might resolve chiasmata by a separase-independent mechanism related to the so-called "prophase pathway" that removes cohesin from chromosome arms during mitosis. By expressing Cre recombinase from a zona pellucida promoter, we have deleted a floxed allele of separase specifically in mouse oocytes. This prevents removal of Rec8 from chromosome arms and resolution of chiasmata. It also hinders extrusion of the first polar body (PBE) and causes female sterility. mRNA encoding wild-type but not catalytically inactive separase restores chiasma resolution. Both types of mRNA restore PBE. Proteolytic activity of separase is therefore essential for Rec8's removal from chromosome arms and for chiasma resolution but not for PBE.


Subject(s)
Cell Cycle Proteins/physiology , Chromosomes/genetics , Endopeptidases/physiology , Meiosis/genetics , Nuclear Proteins/genetics , Oocytes/metabolism , Peptide Hydrolases/genetics , Phosphoproteins/genetics , Animals , Cell Cycle Proteins/genetics , Cells, Cultured , Chromosome Segregation/genetics , Cytokinesis/genetics , Down-Regulation/genetics , Endopeptidases/genetics , Female , Gene Deletion , Genes, cdc/physiology , Humans , Male , Metaphase/genetics , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Transgenic , Oocytes/cytology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Separase
14.
Genes Dev ; 18(1): 88-98, 2004 Jan 01.
Article in English | MEDLINE | ID: mdl-14724179

ABSTRACT

The anaphase-promoting complex or cyclosome (APC/C) is an ubiquitin protein ligase that together with Cdc20 and Cdh1 targets mitotic proteins for degradation by the proteosome. APC-Cdc20 activity during mitosis triggers anaphase by destroying securin and cyclins. APC-Cdh1 promotes degradation of cyclins and other proteins during G(1). We show that loss of APC/C during embryogenesis is early lethal before embryonic day E6.5 (E6.5). To investigate the role of APC/C in quiescent cells, we conditionally inactivated the subunit Apc2 in mice. Deletion of Apc2 in quiescent hepatocytes caused re-entry into the cell cycle and arrest in metaphase, resulting in liver failure. Re-entry into the cell cycle either occurred without any proliferative stimulus or could be easily induced. We demonstrate that the APC has an additional function to prevent hepatocytes from unscheduled re-entry into the cell cycle.


Subject(s)
Cell Division/genetics , Hepatocytes/cytology , Ubiquitin-Protein Ligase Complexes/physiology , Anaphase-Promoting Complex-Cyclosome , Animals , Apc2 Subunit, Anaphase-Promoting Complex-Cyclosome , Caenorhabditis elegans/cytology , Caenorhabditis elegans Proteins , Drosophila melanogaster/cytology , Humans , Mice , Mice, Knockout , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae Proteins , Schizosaccharomyces/cytology , Ubiquitin-Protein Ligase Complexes/deficiency , Ubiquitin-Protein Ligase Complexes/genetics
15.
Bioorg Med Chem Lett ; 12(20): 2807-10, 2002 Oct 21.
Article in English | MEDLINE | ID: mdl-12270151

ABSTRACT

Bioassay-guided separation by use of the fission yeast expressing NES of Rev, a HIV-1 viral regulatory protein, resulted in isolation of valtrate (1) as a new Rev-transport inhibitor from the nucleus to cytoplasm from Valerianae Radix. Valtrate (1) also inhibited the p-24 production of HIV-1 virus without showing any cytotoxicity against the host MT-4 cells.


Subject(s)
HIV Reverse Transcriptase , Iridoids/isolation & purification , Iridoids/pharmacology , Reverse Transcriptase Inhibitors/isolation & purification , Reverse Transcriptase Inhibitors/pharmacology , Valerian/chemistry , Biotin , Cell Line , Electrophoresis, Polyacrylamide Gel , Fatty Acids, Unsaturated/isolation & purification , Fatty Acids, Unsaturated/pharmacology , HIV Core Protein p24/biosynthesis , Humans , Reverse Transcriptase Inhibitors/chemistry , Saccharomyces cerevisiae/enzymology , Saccharomyces cerevisiae/genetics
SELECTION OF CITATIONS
SEARCH DETAIL