Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 334
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 166(1): 167-80, 2016 Jun 30.
Article in English | MEDLINE | ID: mdl-27368103

ABSTRACT

Proliferating cells must cross a point of no return before they replicate their DNA and divide. This commitment decision plays a fundamental role in cancer and degenerative diseases and has been proposed to be mediated by phosphorylation of retinoblastoma (Rb) protein. Here, we show that inactivation of the anaphase-promoting complex/cyclosome (APC(Cdh1)) has the necessary characteristics to be the point of no return for cell-cycle entry. Our study shows that APC(Cdh1) inactivation is a rapid, bistable switch initiated shortly before the start of DNA replication by cyclin E/Cdk2 and made irreversible by Emi1. Exposure to stress between Rb phosphorylation and APC(Cdh1) inactivation, but not after APC(Cdh1) inactivation, reverted cells to a mitogen-sensitive quiescent state, from which they can later re-enter the cell cycle. Thus, APC(Cdh1) inactivation is the commitment point when cells lose the ability to return to quiescence and decide to progress through the cell cycle.


Subject(s)
Anaphase-Promoting Complex-Cyclosome/metabolism , Cdh1 Proteins/metabolism , Cell Cycle , Cell Cycle/drug effects , Cell Cycle Proteins/metabolism , Cell Line , Cell Line, Tumor , F-Box Proteins/metabolism , Humans , Mitogens/toxicity , Phosphorylation , Retinoblastoma Protein/metabolism
2.
Cell ; 159(7): 1681-97, 2014 Dec 18.
Article in English | MEDLINE | ID: mdl-25525883

ABSTRACT

Reprogramming to iPSCs resets the epigenome of somatic cells, including the reversal of X chromosome inactivation. We sought to gain insight into the steps underlying the reprogramming process by examining the means by which reprogramming leads to X chromosome reactivation (XCR). Analyzing single cells in situ, we found that hallmarks of the inactive X (Xi) change sequentially, providing a direct readout of reprogramming progression. Several epigenetic changes on the Xi occur in the inverse order of developmental X inactivation, whereas others are uncoupled from this sequence. Among the latter, DNA methylation has an extraordinary long persistence on the Xi during reprogramming, and, like Xist expression, is erased only after pluripotency genes are activated. Mechanistically, XCR requires both DNA demethylation and Xist silencing, ensuring that only cells undergoing faithful reprogramming initiate XCR. Our study defines the epigenetic state of multiple sequential reprogramming intermediates and establishes a paradigm for studying cell fate transitions during reprogramming.


Subject(s)
Cellular Reprogramming , Epigenesis, Genetic , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , X Chromosome/metabolism , Animals , Cdh1 Proteins/metabolism , DNA Methylation , Homeodomain Proteins/metabolism , Mice , Nanog Homeobox Protein , RNA, Long Noncoding/metabolism
3.
Cell ; 141(2): 268-79, 2010 Apr 16.
Article in English | MEDLINE | ID: mdl-20403323

ABSTRACT

One oscillation of Cyclin-dependent kinase (Cdk) activity, largely driven by periodic synthesis and destruction of cyclins, is tightly coupled to a single complete eukaryotic cell division cycle. Tight linkage of different steps in diverse cell-cycle processes to Cdk activity has been proposed to explain this coupling. Here, we demonstrate an intrinsically oscillatory module controlling nucleolar release and resequestration of the Cdc14 phosphatase, which is essential for mitotic exit in budding yeast. We find that this Cdc14 release oscillator functions at constant and physiological cyclin-Cdk levels, and is therefore independent of the Cdk oscillator. However, the frequency of the release oscillator is regulated by cyclin-Cdk activity. This observation together with its mechanism suggests that the intrinsically autonomous Cdc14 release cycles are locked at once-per-cell-cycle through entrainment by the Cdk oscillator in wild-type cells. This concept may have broad implications for the structure and evolution of eukaryotic cell-cycle control.


Subject(s)
Cell Cycle Proteins/metabolism , Cyclins/metabolism , Protein Tyrosine Phosphatases/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae/metabolism , Cdh1 Proteins , Cell Nucleolus/metabolism , Cyclin B/metabolism , Mitosis , Models, Biological , Protein Kinases/metabolism , Protein Serine-Threonine Kinases
4.
Nature ; 558(7709): 313-317, 2018 06.
Article in English | MEDLINE | ID: mdl-29875408

ABSTRACT

Mammalian cells integrate mitogen and stress signalling before the end of G1 phase to determine whether or not they enter the cell cycle1-4. Before cells can replicate their DNA in S phase, they have to activate cyclin-dependent kinases (CDKs), induce an E2F transcription program and inactivate the anaphase-promoting complex (APC/CCDH1, also known as the cyclosome), which is an E3 ubiquitin ligase that contains the co-activator CDH1 (also known as FZR, encoded by FZR1). It was recently shown that stress can return cells to quiescence after CDK2 activation and E2F induction but not after inactivation of APC/CCDH1, which suggests that APC/CCDH1 inactivation is the point of no return for cell-cycle entry 3 . Rapid inactivation of APC/CCDH1 requires early mitotic inhibitor 1 (EMI1)3,5, but the molecular mechanism that controls this cell-cycle commitment step is unknown. Here we show using human cell models that cell-cycle commitment is mediated by an EMI1-APC/CCDH1 dual-negative feedback switch, in which EMI1 is both a substrate and an inhibitor of APC/CCDH1. The inactivation switch triggers a transition between a state with low EMI1 levels and high APC/CCDH1 activity during G1 and a state with high EMI1 levels and low APC/CCDH1 activity during S and G2. Cell-based analysis, in vitro reconstitution and modelling data show that the underlying dual-negative feedback is bistable and represents a robust irreversible switch. Our study suggests that mammalian cells commit to the cell cycle by increasing CDK2 activity and EMI1 mRNA expression to trigger a one-way APC/CCDH1 inactivation switch that is mediated by EMI1 transitioning from acting as a substrate of APC/CCDH1 to being an inhibitor of APC/CCDH1.


Subject(s)
Cdh1 Proteins/antagonists & inhibitors , Cdh1 Proteins/metabolism , Cell Cycle Proteins/metabolism , Cell Cycle/physiology , F-Box Proteins/metabolism , Cell Cycle Proteins/genetics , Cyclin E/metabolism , Cyclin-Dependent Kinase 2/metabolism , F-Box Proteins/genetics , Feedback, Physiological , G1 Phase , HeLa Cells , Humans , S Phase
5.
Nature ; 553(7686): 91-95, 2018 01 04.
Article in English | MEDLINE | ID: mdl-29160310

ABSTRACT

Treatments that target immune checkpoints, such as the one mediated by programmed cell death protein 1 (PD-1) and its ligand PD-L1, have been approved for treating human cancers with durable clinical benefit. However, many patients with cancer fail to respond to compounds that target the PD-1 and PD-L1 interaction, and the underlying mechanism(s) is not well understood. Recent studies revealed that response to PD-1-PD-L1 blockade might correlate with PD-L1 expression levels in tumour cells. Hence, it is important to understand the mechanistic pathways that control PD-L1 protein expression and stability, which can offer a molecular basis to improve the clinical response rate and efficacy of PD-1-PD-L1 blockade in patients with cancer. Here we show that PD-L1 protein abundance is regulated by cyclin D-CDK4 and the cullin 3-SPOP E3 ligase via proteasome-mediated degradation. Inhibition of CDK4 and CDK6 (hereafter CDK4/6) in vivo increases PD-L1 protein levels by impeding cyclin D-CDK4-mediated phosphorylation of speckle-type POZ protein (SPOP) and thereby promoting SPOP degradation by the anaphase-promoting complex activator FZR1. Loss-of-function mutations in SPOP compromise ubiquitination-mediated PD-L1 degradation, leading to increased PD-L1 levels and reduced numbers of tumour-infiltrating lymphocytes in mouse tumours and in primary human prostate cancer specimens. Notably, combining CDK4/6 inhibitor treatment with anti-PD-1 immunotherapy enhances tumour regression and markedly improves overall survival rates in mouse tumour models. Our study uncovers a novel molecular mechanism for regulating PD-L1 protein stability by a cell cycle kinase and reveals the potential for using combination treatment with CDK4/6 inhibitors and PD-1-PD-L1 immune checkpoint blockade to enhance therapeutic efficacy for human cancers.


Subject(s)
B7-H1 Antigen/metabolism , Cullin Proteins/metabolism , Cyclin D/metabolism , Cyclin-Dependent Kinase 4/metabolism , Immunologic Surveillance , Neoplasms/immunology , Nuclear Proteins/metabolism , Repressor Proteins/metabolism , Tumor Escape/immunology , 14-3-3 Proteins/metabolism , Animals , B7-H1 Antigen/biosynthesis , Cdh1 Proteins/metabolism , Cell Cycle , Cell Line , Cyclin-Dependent Kinase 4/antagonists & inhibitors , Cyclin-Dependent Kinase 6/antagonists & inhibitors , Female , Humans , Lymphocytes, Tumor-Infiltrating/cytology , Lymphocytes, Tumor-Infiltrating/immunology , Male , Mice , Nuclear Proteins/chemistry , Phosphorylation , Programmed Cell Death 1 Receptor/metabolism , Prostatic Neoplasms/immunology , Proteasome Endopeptidase Complex/metabolism , Protein Stability , Proteolysis , Repressor Proteins/chemistry
6.
Dev Biol ; 482: 55-66, 2022 02.
Article in English | MEDLINE | ID: mdl-34922934

ABSTRACT

The coincidence of cell cycle exit and differentiation has been described in a wide variety of stem cells and organisms for decades, but the causal relationship is still unclear due to the complicated regulation of the cell cycle. Here, we used the planarian Dugesia japonica since they may possess a simple cell cycle regulation in which Cdh1 is one of the factors responsible for exiting the cell cycle. When cdh1 was functionally inhibited, the planarians could not maintain their tissue homeostasis and could not regenerate their missing body parts. While the knockdown of cdh1 caused pronounced accumulation of the stem cells, the progenitor and differentiated cells were decreased. Further analyses indicated that the stem cells with cdh1 knockdown did not undergo differentiation even though they received ERK signaling activation as an induction signal. These results suggested that stem cells could not acquire differentiation competence without cell cycle exit. Thus, we propose that cell cycle regulation determines the differentiation competence and that cell cycle exit to G0 enables stem cells to undergo differentiation.


Subject(s)
Cdh1 Proteins/genetics , Cell Cycle/physiology , Planarians/growth & development , Regeneration/genetics , Animals , Cdh1 Proteins/metabolism , Cell Differentiation/physiology , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , Planarians/cytology , RNA Interference , Regeneration/physiology , Stem Cells/cytology , Stem Cells/metabolism
7.
Brain ; 145(5): 1684-1697, 2022 06 03.
Article in English | MEDLINE | ID: mdl-34788397

ABSTRACT

FZR1, which encodes the Cdh1 subunit of the anaphase-promoting complex, plays an important role in neurodevelopment by regulating the cell cycle and by its multiple post-mitotic functions in neurons. In this study, evaluation of 250 unrelated patients with developmental and epileptic encephalopathies and a connection on GeneMatcher led to the identification of three de novo missense variants in FZR1. Whole-exome sequencing in 39 patient-parent trios and subsequent targeted sequencing in an additional cohort of 211 patients was performed to identify novel genes involved in developmental and epileptic encephalopathy. Functional studies in Drosophila were performed using three different mutant alleles of the Drosophila homologue of FZR1 fzr. All three individuals carrying de novo variants in FZR1 had childhood-onset generalized epilepsy, intellectual disability, mild ataxia and normal head circumference. Two individuals were diagnosed with the developmental and epileptic encephalopathy subtype myoclonic atonic epilepsy. We provide genetic-association testing using two independent statistical tests to support FZR1 association with developmental and epileptic encephalopathies. Further, we provide functional evidence that the missense variants are loss-of-function alleles using Drosophila neurodevelopment assays. Using three fly mutant alleles of the Drosophila homologue fzr and overexpression studies, we show that patient variants can affect proper neurodevelopment. With the recent report of a patient with neonatal-onset with microcephaly who also carries a de novo FZR1 missense variant, our study consolidates the relationship between FZR1 and developmental and epileptic encephalopathy and expands the associated phenotype. We conclude that heterozygous loss-of-function of FZR1 leads to developmental and epileptic encephalopathies associated with a spectrum of neonatal to childhood-onset seizure types, developmental delay and mild ataxia. Microcephaly can be present but is not an essential feature of FZR1-encephalopathy. In summary, our approach of targeted sequencing using novel gene candidates and functional testing in Drosophila will help solve undiagnosed myoclonic atonic epilepsy or developmental and epileptic encephalopathy cases.


Subject(s)
Cdh1 Proteins , Epilepsy, Generalized , Epilepsy , Microcephaly , Ataxia , Cdh1 Proteins/genetics , Child , Epilepsy/genetics , Epilepsy, Generalized/genetics , Humans , Loss of Function Mutation , Microcephaly/genetics , Phenotype
8.
Int J Mol Sci ; 24(4)2023 Feb 18.
Article in English | MEDLINE | ID: mdl-36835555

ABSTRACT

Cdh1p is one of the two substrate adaptor proteins of the anaphase promoting complex/cyclosome (APC/C), a ubiquitin ligase that regulates proteolysis during cell cycle. In this work, using a proteomic approach, we found 135 mitochondrial proteins whose abundance was significantly altered in the cdh1Δ mutant, with 43 up-regulated proteins and 92 down-regulated proteins. The group of significantly up-regulated proteins included subunits of the mitochondrial respiratory chain, enzymes from the tricarboxylic acid cycle and regulators of mitochondrial organization, suggesting a metabolic remodelling towards an increase in mitochondrial respiration. In accordance, mitochondrial oxygen consumption and Cytochrome c oxidase activity increased in Cdh1p-deficient cells. These effects seem to be mediated by the transcriptional activator Yap1p, a major regulator of the yeast oxidative stress response. YAP1 deletion suppressed the increased Cyc1p levels and mitochondrial respiration in cdh1Δ cells. In agreement, Yap1p is transcriptionally more active in cdh1Δ cells and responsible for the higher oxidative stress tolerance of cdh1Δ mutant cells. Overall, our results unveil a new role for APC/C-Cdh1p in the regulation of the mitochondrial metabolic remodelling through Yap1p activity.


Subject(s)
Cdh1 Proteins , Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae , Proteomics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Cdh1 Proteins/metabolism
9.
Development ; 146(15)2019 08 02.
Article in English | MEDLINE | ID: mdl-31315896

ABSTRACT

Tissue repair usually requires either polyploid cell growth or cell division, but the molecular mechanism promoting polyploidy and limiting cell division remains poorly understood. Here, we find that injury to the adult Drosophila epithelium causes cells to enter the endocycle through the activation of Yorkie-dependent genes (Myc and E2f1). Myc is even sufficient to induce the endocycle in the uninjured post-mitotic epithelium. As result, epithelial cells enter S phase but mitosis is blocked by inhibition of mitotic gene expression. The mitotic cell cycle program can be activated by simultaneously expressing the Cdc25-like phosphatase String (stg), while genetically depleting APC/C E3 ligase fizzy-related (fzr). However, forcing cells to undergo mitosis is detrimental to wound repair as the adult fly epithelium accumulates DNA damage, and mitotic errors ensue when cells are forced to proliferate. In conclusion, we find that wound-induced polyploidization enables tissue repair when cell division is not a viable option.


Subject(s)
DNA Damage/genetics , DNA-Binding Proteins/genetics , Drosophila Proteins/genetics , Epithelium/injuries , Mitosis/physiology , Transcription Factors/genetics , Wound Healing/physiology , Animals , Animals, Genetically Modified , Cdh1 Proteins/metabolism , Cell Cycle Proteins/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/physiology , Epithelial Cells/metabolism , Gene Expression Regulation/genetics , Mitosis/genetics , Polyploidy , Protein Tyrosine Phosphatases/metabolism , Wound Healing/genetics
10.
Exp Cell Res ; 404(2): 112632, 2021 07 15.
Article in English | MEDLINE | ID: mdl-33971196

ABSTRACT

Retinoblastoma protein (pRB) regulates cell cycle by utilizing different regions of its pocket domain for interacting with E2F family of transcription factors and with cellular and viral proteins containing an LxCxE motif. An LxCxE-like motif, LxCxD, is present in FZR1, an adaptor protein of the multi-subunit E3 ligase complex anaphase-promoting complex/cyclosome (APC/C). The APC/CFZR1 complex regulates the timely degradation of multiple cell cycle proteins for mitotic exit and maintains G1 state. We report that FZR1 interacts with pRB via its LxCxD motif. By using point mutations, we found that the cysteine residue in the FZR1 LxCxD motif is critical for direct interaction with pRb. The direct binding of the LxCxD motif of FZR1 to the pRB LxCxE binding pocket is confirmed by using human papillomavirus protein E7 as a competitor, both in vitro and in vivo. While mutation of the cysteine residue significantly disrupts FZR1 interaction with pRB, this motif does not affect FZR1 and core APC/C association. Expression of the FZR1 point mutant results in accumulation of S-phase kinase-associated protein 2 (SKP2) and Polo-like kinase 1 (PLK1), while p27Kip1 and p21Cip1 proteins are downregulated, indicating a G1 cell cycle defect. Consistently, cells containing point mutant FZR1 enter the S phase prematurely. Together our results suggest that the LxCxD motif of FZR1 is a critical determinant for the interaction between FZR1 and pRB and is important for G1 restriction.


Subject(s)
Anaphase-Promoting Complex-Cyclosome/metabolism , Cdh1 Proteins/metabolism , Cell Cycle/physiology , Retinoblastoma Protein/metabolism , Amino Acid Sequence/physiology , Anaphase-Promoting Complex-Cyclosome/genetics , Cell Cycle Proteins/genetics , Cell Division/physiology , Humans , Retinoblastoma Protein/genetics , Transcription Factors/metabolism
11.
Nucleic Acids Res ; 48(8): 4214-4229, 2020 05 07.
Article in English | MEDLINE | ID: mdl-32182338

ABSTRACT

Endoreplication, known as endocycle, is a variant of the cell cycle that differs from mitosis and occurs in specific tissues of different organisms. Endoreplicating cells generally undergo multiple rounds of genome replication without chromosome segregation. Previous studies demonstrated that Drosophila fizzy-related protein (Fzr) and its mammalian homolog Cdh1 function as key regulators of endoreplication entrance by activating the anaphase-promoting complex/cyclosome to initiate the ubiquitination and subsequent degradation of cell cycle factors such as Cyclin B (CycB). However, the molecular mechanism underlying Fzr-mediated endoreplication is not completely understood. In this study, we demonstrated that the transcription factor Myc acts downstream of Fzr during endoreplication in Drosophila salivary gland. Mechanistically, Fzr interacts with chromatin-associated histone H2B to enhance H2B ubiquitination in the Myc promoter and promotes Myc transcription. In addition to negatively regulating CycB transcription, the Fzr-ubiquitinated H2B (H2Bub)-Myc signaling cascade also positively regulates the transcription of the MCM6 gene that is involved in DNA replication by directly binding to specific motifs within their promoters. We further found that the Fzr-H2Bub-Myc signaling cascade regulating endoreplication progression is conserved between insects and mammalian cells. Altogether, our work uncovers a novel transcriptional cascade that is involved in Fzr-mediated endoreplication.


Subject(s)
Cdh1 Proteins/metabolism , DNA-Binding Proteins/metabolism , Drosophila Proteins/metabolism , Endoreduplication , Gene Expression Regulation , Transcription Factors/metabolism , Animals , Cell Line , Cyclin B/genetics , DNA Replication , DNA-Binding Proteins/genetics , Drosophila/genetics , Drosophila/metabolism , Drosophila Proteins/genetics , HEK293 Cells , Histones/metabolism , Humans , Minichromosome Maintenance Complex Component 6/genetics , Promoter Regions, Genetic , Salivary Glands/metabolism , Signal Transduction , Transcription Factors/genetics , Ubiquitination
12.
Proc Natl Acad Sci U S A ; 116(19): 9423-9432, 2019 05 07.
Article in English | MEDLINE | ID: mdl-31000600

ABSTRACT

The Hippo-YAP/TAZ signaling pathway plays a pivotal role in growth control during development and regeneration and its dysregulation is widely implicated in various cancers. To further understand the cellular and molecular mechanisms underlying Hippo signaling regulation, we have found that activities of core Hippo signaling components, large tumor suppressor (LATS) kinases and YAP/TAZ transcription factors, oscillate during mitotic cell cycle. We further identified that the anaphase-promoting complex/cyclosome (APC/C)Cdh1 E3 ubiquitin ligase complex, which plays a key role governing eukaryotic cell cycle progression, intrinsically regulates Hippo signaling activities. CDH1 recognizes LATS kinases to promote their degradation and, hence, YAP/TAZ regulation by LATS phosphorylation is under cell cycle control. As a result, YAP/TAZ activities peak in G1 phase. Furthermore, we show in Drosophila eye and wing development that Cdh1 is required in vivo to regulate the LATS homolog Warts with a conserved mechanism. Cdh1 reduction increased Warts levels, which resulted in reduction of the eye and wing sizes in a Yorkie dependent manner. Therefore, LATS degradation by APC/CCdh1 represents a previously unappreciated and evolutionarily conserved layer of Hippo signaling regulation.


Subject(s)
Anaphase-Promoting Complex-Cyclosome/metabolism , Antigens, CD/metabolism , Cadherins/metabolism , Cdh1 Proteins/metabolism , Drosophila Proteins/metabolism , G1 Phase/physiology , Intracellular Signaling Peptides and Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Signal Transduction/physiology , Anaphase-Promoting Complex-Cyclosome/genetics , Animals , Antigens, CD/genetics , Cadherins/genetics , Cdh1 Proteins/genetics , Drosophila Proteins/genetics , Drosophila melanogaster , HEK293 Cells , HeLa Cells , Hippo Signaling Pathway , Humans , Intracellular Signaling Peptides and Proteins/genetics , Protein Kinases/genetics , Protein Kinases/metabolism , Protein Serine-Threonine Kinases/genetics
13.
Zhonghua Yi Xue Yi Chuan Xue Za Zhi ; 39(11): 1279-1282, 2022 Nov 10.
Article in Zh | MEDLINE | ID: mdl-36317219

ABSTRACT

OBJECTIVE: To assess the association of genomic instability of epithelial cadherin 1 (CDH1) gene and clinicopathological characteristics of gastric cancer. METHODS: In total 120 paraffin-embedded gastric cancer tissue specimen were prepared, and genomic DNA was extracted. The genomic instability of the CDH1 gene was analyzed by immunohistochemistry and silver staining PCR-single-strand conformation polymorphism. RESULTS: The number of information individuals (heterozygotes) was 98 for the D16S752 locus. The detection rates for microsatellite instability (MSI) and loss of heterozygosity (LOH) at the D16S752 locus and the positive rate of CDH1 protein were 19.39%, 16.33% and 51.02%, respectively. The detection rate of MSI in TNM stages I or II was significantly higher than that in stages III or IV (P<0.05) while the detection rate of LOH was significantly lower than that in stages III or IV (P<0.05). The positive rate of CDH1 protein in TNM stages III or IV was significantly lower than that in stages I or II (P<0.05). The detection rate of MSI of cases with lymph node metastasis was significantly lower than that of without lymph node metastasis (P<0.05) while the detection rate of LOH was significantly higher than that without lymph node metastasis (P<0.05). The positive rate of CDH1 protein in patients with lymph node metastasis was significantly lower than that in patients without lymph node metastasis (P<0.05). The positive rate of CDH1 protein in MSI-positive group was significantly higher than that in MSI-negative group (P<0.05), and the positive rate of CDH1 protein in the LOH-positive group was significantly lower than that the LOH-negative group (P<0.05). CONCLUSION: The genomic instability of the CDH1 gene is associated with the progression of gastric cancer. MSI at the D16S752 locus may be used as a molecular marker for early gastric cancer, while LOH at this locus mostly occurs in advanced gastric cancer and can be regarded as an effective indicators for malignancy evaluation and prognosis.


Subject(s)
Stomach Neoplasms , Humans , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Lymphatic Metastasis , Cdh1 Proteins/genetics , Microsatellite Instability , Loss of Heterozygosity , Genomic Instability , Microsatellite Repeats , Antigens, CD/genetics , Cadherins/genetics
14.
Semin Cancer Biol ; 67(Pt 2): 80-91, 2020 12.
Article in English | MEDLINE | ID: mdl-32165320

ABSTRACT

The anaphase promoting complex/ cyclosome (APC/C), is an evolutionarily conserved protein complex essential for cellular division due to its role in regulating the mitotic transition from metaphase to anaphase. In this review, we highlight recent work that has shed light on our understanding of the role of APC/C coactivators, Cdh1 and Cdc20, in cancer initiation and development. We summarize the current state of knowledge regarding APC/C structure and function, as well as the distinct ways Cdh1 and Cdc20 are dysregulated in human cancer. We also discuss APC/C inhibitors, novel approaches for targeting the APC/C as a cancer therapy, and areas for future work.


Subject(s)
Anaphase-Promoting Complex-Cyclosome/metabolism , Antigens, CD/metabolism , Antineoplastic Agents/pharmacology , Cdc20 Proteins/metabolism , Cdh1 Proteins/metabolism , Neoplasms/pathology , Anaphase-Promoting Complex-Cyclosome/antagonists & inhibitors , Anaphase-Promoting Complex-Cyclosome/chemistry , Anaphase-Promoting Complex-Cyclosome/genetics , Antigens, CD/genetics , Carbamates/pharmacology , Cdc20 Proteins/genetics , Cdh1 Proteins/genetics , Diamines/pharmacology , Genomic Instability , Humans , Molecular Targeted Therapy/methods , Neoplasms/genetics
15.
Exp Cell Res ; 386(2): 111720, 2020 01 15.
Article in English | MEDLINE | ID: mdl-31738907

ABSTRACT

CHK1 and WEE1 play pivotal roles in G2/M checkpoint following exogenous DNA damage and regulation of DNA replication under normal cellular conditions. Here, we monitored and compared the cell cycle kinetics of mitosis-associated events after CHK1 and WEE1 inhibitor treatments in a human tongue cancer cell line (SAS). A fluorescent ubiquitination-based cell cycle indicator (Fucci) that reflects SCFSKP2 and APCCDH1 E3 ligase activities was used to monitor cell cycle progression. Numerous γH2AX-positive cells were observed within the S phase population of cells following CHK1 inhibitor treatment, and polyploid cells exhibiting DNA damage emerged via abortive mitosis (endomitosis) at 24 h post treatment. While WEE1 inhibitor-treated cells exhibited similar polyploidy via endomitosis at later time points, they possessed fewer γH2AX foci during S phase, and polyploid cells exhibiting DNA damage were scarce. Instead, mitosis duration greatly extended and was accompanied by an abnormal emission of Fucci red fluorescence. Kinetic analysis of Fucci fluorescence revealed that abnormal emission occurred at early M phase in a manner independent of green fluorescence degradation as a marker of APCCDH1 activation. When an inhibitor of the essential spindle checkpoint factor MPS1 was co-treated with a WEE1 inhibitor, the elongated mitosis duration and abnormal red fluorescence were abrogated, and WEE1-induced reduction of clonogenic survival was offset. We demonstrate novel differential effects on mitosis-associated events following CHK1 and WEE1 inhibitor treatments.


Subject(s)
Cell Cycle Proteins/genetics , Checkpoint Kinase 1/genetics , Epithelial Cells/drug effects , Gene Expression Regulation, Neoplastic , Protein Kinase Inhibitors/pharmacology , Protein-Tyrosine Kinases/genetics , Cdh1 Proteins/genetics , Cdh1 Proteins/metabolism , Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Checkpoint Kinase 1/antagonists & inhibitors , Checkpoint Kinase 1/metabolism , DNA Damage , Epithelial Cells/metabolism , Epithelial Cells/pathology , Flow Cytometry , G2 Phase Cell Cycle Checkpoints/drug effects , Genes, Reporter , HeLa Cells , Histones/genetics , Histones/metabolism , Humans , Mitosis/drug effects , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/metabolism , S Phase/drug effects , S-Phase Kinase-Associated Proteins/genetics , S-Phase Kinase-Associated Proteins/metabolism , Signal Transduction , Time-Lapse Imaging
16.
Mol Cell ; 50(5): 609-10, 2013 Jun 06.
Article in English | MEDLINE | ID: mdl-23746347

ABSTRACT

In this issue of Molecular Cell, He and colleagues (2013) unveil a high-resolution structure of a key regulatory interface in cell-cycle control: the destruction box sequence bound to the anaphase-promoting complex.


Subject(s)
Repressor Proteins/chemistry , Repressor Proteins/metabolism , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/metabolism , Ubiquitin-Protein Ligase Complexes/metabolism , Anaphase-Promoting Complex-Cyclosome , Animals , Cdh1 Proteins , Cell Cycle Proteins
17.
Mol Cell ; 50(5): 649-60, 2013 Jun 06.
Article in English | MEDLINE | ID: mdl-23707760

ABSTRACT

The anaphase-promoting complex/cyclosome (APC/C) regulates sister chromatid segregation and the exit from mitosis. Selection of most APC/C substrates is controlled by coactivator subunits (either Cdc20 or Cdh1) that interact with substrate destruction motifs--predominantly the destruction (D) box and KEN box degrons. How coactivators recognize D box degrons and how this is inhibited by APC/C regulatory proteins is not defined at the atomic level. Here, from the crystal structure of S. cerevisiae Cdh1 in complex with its specific inhibitor Acm1, which incorporates D and KEN box pseudosubstrate motifs, we describe the molecular basis for D box recognition. Additional interactions between Acm1 and Cdh1 identify a third protein-binding site on Cdh1 that is likely to confer coactivator-specific protein functions including substrate association. We provide a structural rationalization for D box and KEN box recognition by coactivators and demonstrate that many noncanonical APC/C degrons bind APC/C coactivators at the D box coreceptor.


Subject(s)
Repressor Proteins/chemistry , Repressor Proteins/metabolism , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/metabolism , Ubiquitin-Protein Ligase Complexes/metabolism , Amino Acid Motifs , Anaphase-Promoting Complex-Cyclosome , Animals , Binding Sites , Cdh1 Proteins , Cell Cycle Proteins , Crystallography, X-Ray , Models, Molecular , Protein Conformation , Protein Multimerization , Repressor Proteins/genetics , Saccharomyces cerevisiae Proteins/genetics
18.
Nucleic Acids Res ; 47(7): 3395-3406, 2019 04 23.
Article in English | MEDLINE | ID: mdl-30753559

ABSTRACT

Centromere identity is determined by the specific deposition of CENP-A, a histone H3 variant localizing exclusively at centromeres. Increased CENP-A expression, which is a frequent event in cancer, causes mislocalization, ectopic kinetochore assembly and genomic instability. Proteolysis regulates CENP-A expression and prevents its misincorporation across chromatin. How proteolysis restricts CENP-A localization to centromeres is not well understood. Here we report that, in Drosophila, CENP-ACID expression levels are regulated throughout the cell cycle by the combined action of SCFPpa and APC/CCdh1. We show that SCFPpa regulates CENP-ACID expression in G1 and, importantly, in S-phase preventing its promiscuous incorporation across chromatin during replication. In G1, CENP-ACID expression is also regulated by APC/CCdh1. We also show that Cal1, the specific chaperone that deposits CENP-ACID at centromeres, protects CENP-ACID from SCFPpa-mediated degradation but not from APC/CCdh1-mediated degradation. These results suggest that, whereas SCFPpa targets the fraction of CENP-ACID that is not in complex with Cal1, APC/CCdh1 mediates also degradation of the Cal1-CENP-ACID complex and, thus, likely contributes to the regulation of centromeric CENP-ACID deposition.


Subject(s)
Anaphase-Promoting Complex-Cyclosome/metabolism , Cdh1 Proteins/metabolism , Cell Cycle , Centromere Protein A/metabolism , Drosophila melanogaster/cytology , Drosophila melanogaster/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Cell Line , Centromere/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/enzymology , G1 Phase , S Phase
19.
J Cell Physiol ; 235(3): 2521-2531, 2020 03.
Article in English | MEDLINE | ID: mdl-31489637

ABSTRACT

Platelet endothelial cell adhesion molecule-1 (PECAM-1) is a member of the immunoglobulin superfamily and is expressed by hematopoietic and endothelial cells (ECs). Recent studies have shown that PECAM-1 plays a crucial role in promoting the development of the EC inflammatory response in the context of disturbed flow. However, the mechanistic pathways that control PECAM-1 protein stability remain largely unclear. Here, we identified PECAM-1 as a novel substrate of the APC/Cdh1 E3 ubiquitin ligase. Specifically, lentivirus-mediated Cdh1 depletion stabilized PECAM-1 in ECs. Conversely, overexpression of Cdh1 destabilized PECAM-1. The proteasome inhibitor MG132 blocked Cdh1-mediated PECAM-1 degradation. In addition, Cdh1 promoted K48-linked polyubiquitination of PECAM-1 in a destruction box-dependent manner. Furthermore, we demonstrated that compared with pulsatile shear stress (PS), oscillatory shear stress decreased the expression of Cdh1 and the ubiquitination of PECAM-1, therefore stabilizing PECAM-1 to promote inflammation in ECs. Hence, our study revealed a novel mechanism by which fluid flow patterns regulate EC homeostasis via Cdh1-dependent ubiquitination and subsequent degradation of PECAM-1.


Subject(s)
Antigens, CD/genetics , Cdh1 Proteins/genetics , Inflammation/genetics , Platelet Endothelial Cell Adhesion Molecule-1/genetics , Ubiquitin-Protein Ligases/genetics , Anaphase-Promoting Complex-Cyclosome/genetics , Cell Cycle/genetics , Endothelial Cells/metabolism , Endothelial Cells/pathology , HeLa Cells , Humans , Phosphorylation/genetics , Proteolysis , Ubiquitination/genetics
20.
J Cell Sci ; 131(14)2018 07 17.
Article in English | MEDLINE | ID: mdl-29898917

ABSTRACT

Somatic muscles are formed by the iterative fusion of myoblasts into muscle fibres. This process is driven by the recurrent recruitment of proteins to the cell membrane to induce F-actin nucleation at the fusion site. Although several proteins involved in myoblast fusion have been identified, knowledge about their subcellular regulation is rather elusive. We identified the anaphase-promoting complex (APC/C) adaptor Fizzy related (Fzr) as an essential regulator of heart and muscle development. We show that APC/CFzr regulates the fusion of myoblasts as well as the mitotic exit of pericardial cells, cardioblasts and myoblasts. Surprisingly, overproliferation is not causative for the observed fusion defects. Instead, fzr mutants exhibit smaller F-actin foci at the fusion site and display reduced membrane breakdown between adjacent myoblasts. We show that lack of APC/CFzr causes accumulation and mislocalisation of Rols and Duf, two proteins involved in the fusion process. Duf seems to serve as direct substrate of the APC/CFzr and its destruction depends on the presence of distinct degron sequences. These novel findings indicate that protein destruction and turnover constitute major events during myoblast fusion.


Subject(s)
Anaphase-Promoting Complex-Cyclosome/metabolism , Cdh1 Proteins/metabolism , Drosophila Proteins/metabolism , Drosophila/metabolism , Muscles/metabolism , Myoblasts/cytology , Myoblasts/metabolism , Myocardium/metabolism , Actins/genetics , Actins/metabolism , Anaphase-Promoting Complex-Cyclosome/genetics , Animals , Cdh1 Proteins/genetics , Cell Count , Cell Fusion , Cell Proliferation , Drosophila/genetics , Drosophila/growth & development , Drosophila Proteins/genetics , Female , Gene Expression Regulation, Developmental , Heart/growth & development , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , Muscle Development , Muscle Proteins/genetics , Muscle Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL