Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.947
Filter
Add more filters

Publication year range
1.
Mol Cell ; 81(9): 1951-1969.e6, 2021 05 06.
Article in English | MEDLINE | ID: mdl-33761311

ABSTRACT

The initiation of DNA replication involves cell cycle-dependent assembly and disassembly of protein complexes, including the origin recognition complex (ORC) and CDC6 AAA+ ATPases. We report that multiple short linear protein motifs (SLiMs) within intrinsically disordered regions (IDRs) in ORC1 and CDC6 mediate cyclin-CDK-dependent and independent protein-protein interactions, conditional on the cell cycle phase. A domain within the ORC1 IDR is required for interaction between the ORC1 and CDC6 AAA+ domains in G1, whereas the same domain prevents CDC6-ORC1 interaction during mitosis. Then, during late G1, this domain facilitates ORC1 destruction by a SKP2-cyclin A-CDK2-dependent mechanism. During G1, the CDC6 Cy motif cooperates with cyclin E-CDK2 to promote ORC1-CDC6 interactions. The CDC6 IDR regulates self-interaction by ORC1, thereby controlling ORC1 protein levels. Protein phosphatase 1 binds directly to a SLiM in the ORC1 IDR, causing ORC1 de-phosphorylation upon mitotic exit, increasing ORC1 protein, and promoting pre-RC assembly.


Subject(s)
ATPases Associated with Diverse Cellular Activities/metabolism , Cell Cycle Proteins/metabolism , Cell Nucleus/metabolism , DNA Replication , Intrinsically Disordered Proteins/metabolism , Mitosis , Nuclear Proteins/metabolism , Origin Recognition Complex/metabolism , AAA Domain , ATPases Associated with Diverse Cellular Activities/genetics , Cell Cycle Proteins/genetics , Cell Nucleus/genetics , Cyclin A/genetics , Cyclin A/metabolism , Cyclin E/genetics , Cyclin E/metabolism , G1 Phase , HeLa Cells , Humans , Intrinsically Disordered Proteins/genetics , Nuclear Proteins/genetics , Origin Recognition Complex/genetics , Phosphorylation , Protein Binding , Protein Interaction Domains and Motifs , Protein Phosphatase 1/genetics , Protein Phosphatase 1/metabolism , Protein Stability , S-Phase Kinase-Associated Proteins/genetics , S-Phase Kinase-Associated Proteins/metabolism
2.
Development ; 150(5)2023 03 01.
Article in English | MEDLINE | ID: mdl-36805640

ABSTRACT

The stem cell pools at the shoot apex and root tip give rise to all the above- and below-ground tissues of a plant. Previous studies in Arabidopsis identified a TSO1-MYB3R1 transcriptional module that controls the number and size of the stem cell pools at the shoot apex and root tip. As TSO1 and MYB3R1 are homologous to components of an animal cell cycle regulatory complex, DREAM, Arabidopsis mutants of TSO1 and MYB3R1 provide valuable tools for investigations into the link between cell cycle regulation and stem cell maintenance in plants. In this study, an Arabidopsis cyclin A gene, CYCA3;4, was identified as a member of the TSO1-MYB3R1 regulatory module and cyca3;4 mutations suppressed the tso1-1 mutant phenotype specifically in the shoot. The work reveals how the TSO1-MYB3R1 module is integrated with the cell cycle machinery to control cell division at the shoot meristem.


Subject(s)
Arabidopsis Proteins , Arabidopsis , Animals , Arabidopsis/genetics , Meristem/metabolism , Arabidopsis Proteins/metabolism , Cyclin A/genetics , Cyclin A/metabolism , Mutation , Fertility , Gene Expression Regulation, Plant , Plant Shoots/metabolism
3.
EMBO Rep ; 25(2): 524-543, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38253688

ABSTRACT

Metabolites derived from the intestinal microbiota play an important role in maintaining skeletal muscle growth, function, and metabolism. Here, we found that D-malate (DMA) is produced by mouse intestinal microorganisms and its levels increase during aging. Moreover, we observed that dietary supplementation of 2% DMA inhibits metabolism in mice, resulting in reduced muscle mass, strength, and the number of blood vessels, as well as the skeletal muscle fiber type I/IIb ratio. In vitro assays demonstrate that DMA decreases the proliferation of vascular endothelial cells and suppresses the formation of blood vessels. In vivo, we further demonstrated that boosting angiogenesis by muscular VEGFB injection rescues the inhibitory effects of D-malate on muscle mass and fiber area. By transcriptomics analysis, we identified that the mechanism underlying the effects of DMA depends on the elevated intracellular acetyl-CoA content and increased Cyclin A acetylation rather than redox balance. This study reveals a novel mechanism by which gut microbes impair muscle angiogenesis and may provide a therapeutic target for skeletal muscle dysfunction in cancer or aging.


Subject(s)
Endothelial Cells , Microbiota , Mice , Animals , Endothelial Cells/metabolism , Acetylation , Cyclin A/metabolism , Angiogenesis , Malates/metabolism , Muscle, Skeletal/metabolism , Aging
4.
Mol Cell ; 65(2): 231-246, 2017 Jan 19.
Article in English | MEDLINE | ID: mdl-28017591

ABSTRACT

Chromatin featuring the H3 variant CENP-A at the centromere is critical for its mitotic function and epigenetic maintenance. Assembly of centromeric chromatin is restricted to G1 phase through inhibitory action of Cdk1/2 kinases in other phases of the cell cycle. Here, we identify the two key targets sufficient to maintain cell-cycle control of CENP-A assembly. We uncovered a single phosphorylation site in the licensing factor M18BP1 and a cyclin A binding site in the CENP-A chaperone, HJURP, that mediated specific inhibitory phosphorylation. Simultaneous expression of mutant proteins lacking these residues results in complete uncoupling from the cell cycle. Consequently, CENP-A assembly is fully recapitulated under high Cdk activities, indistinguishable from G1 assembly. We find that Cdk-mediated inhibition is exerted by sequestering active factors away from the centromere. Finally, we show that displacement of M18BP1 from the centromere is critical for the assembly mechanism of CENP-A.


Subject(s)
Autoantigens/metabolism , Centromere/metabolism , Chromatin Assembly and Disassembly , Chromatin/metabolism , Chromosomal Proteins, Non-Histone/metabolism , G1 Phase Cell Cycle Checkpoints , Autoantigens/genetics , CDC2 Protein Kinase , Centromere/genetics , Centromere Protein A , Chromatin/genetics , Chromosomal Proteins, Non-Histone/genetics , Cyclin A/genetics , Cyclin A/metabolism , Cyclin-Dependent Kinase 2/genetics , Cyclin-Dependent Kinase 2/metabolism , Cyclin-Dependent Kinases/genetics , Cyclin-Dependent Kinases/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , HEK293 Cells , HeLa Cells , Humans , Mutation , Phosphorylation , Protein Binding , Protein Interaction Domains and Motifs , Signal Transduction , Transfection
5.
J Biol Chem ; 299(3): 102957, 2023 03.
Article in English | MEDLINE | ID: mdl-36717077

ABSTRACT

Cyclin A and CDC25A are both activators of cyclin-dependent kinases (CDKs): cyclin A acts as an activating subunit of CDKs and CDC25A a phosphatase of the inhibitory phosphorylation sites of the CDKs. In this study, we uncovered an inverse relationship between the two CDK activators. As cyclin A is an essential gene, we generated a conditional silencing cell line using a combination of CRISPR-Cas9 and degron-tagged cyclin A. Destruction of cyclin A promoted an acute accumulation of CDC25A. The increase of CDC25A after cyclin A depletion occurred throughout the cell cycle and was independent on cell cycle delay caused by cyclin A deficiency. Moreover, we determined that the inverse relationship with cyclin A was specific for CDC25A and not for other CDC25 family members or kinases that regulate the same sites in CDKs. Unexpectedly, the upregulation of CDC25A was mainly caused by an increase in transcriptional activity instead of a change in the stability of the protein. Reversing the accumulation of CDC25A severely delayed G2-M in cyclin A-depleted cells. Taken together, these data provide evidence of a compensatory mechanism involving CDC25A that ensures timely mitotic entry at different levels of cyclin A.


Subject(s)
Cyclin A , Cyclin-Dependent Kinases , cdc25 Phosphatases , cdc25 Phosphatases/genetics , cdc25 Phosphatases/metabolism , Cell Cycle , Cell Division , Cyclin A/metabolism , Cyclin-Dependent Kinases/metabolism , Phosphorylation
6.
EMBO J ; 39(11): e104419, 2020 06 02.
Article in English | MEDLINE | ID: mdl-32350921

ABSTRACT

Two mitotic cyclin types, cyclin A and B, exist in higher eukaryotes, but their specialised functions in mitosis are incompletely understood. Using degron tags for rapid inducible protein removal, we analyse how acute depletion of these proteins affects mitosis. Loss of cyclin A in G2-phase prevents mitotic entry. Cells lacking cyclin B can enter mitosis and phosphorylate most mitotic proteins, because of parallel PP2A:B55 phosphatase inactivation by Greatwall kinase. The final barrier to mitotic establishment corresponds to nuclear envelope breakdown, which requires a decisive shift in the balance of cyclin-dependent kinase Cdk1 and PP2A:B55 activity. Beyond this point, cyclin B/Cdk1 is essential for phosphorylation of a distinct subset of mitotic Cdk1 substrates that are essential to complete cell division. Our results identify how cyclin A, cyclin B and Greatwall kinase coordinate mitotic progression by increasing levels of Cdk1-dependent substrate phosphorylation.


Subject(s)
CDC2 Protein Kinase/metabolism , Cyclin A/metabolism , Cyclin B/metabolism , Mitosis , Protein Phosphatase 2/metabolism , CDC2 Protein Kinase/genetics , Cell Line , Cyclin A/genetics , Cyclin B/genetics , Humans , Protein Phosphatase 2/genetics
7.
Cell ; 138(2): 352-65, 2009 Jul 23.
Article in English | MEDLINE | ID: mdl-19592082

ABSTRACT

Cyclins are regulatory subunits of cyclin-dependent kinases. Cyclin A, the first cyclin ever cloned, is thought to be an essential component of the cell-cycle engine. Mammalian cells encode two A-type cyclins, testis-specific cyclin A1 and ubiquitously expressed cyclin A2. Here, we tested the requirement for cyclin A function using conditional knockout mice lacking both A-type cyclins. We found that acute ablation of cyclin A in fibroblasts did not affect cell proliferation, but led to prolonged expression of another cyclin, cyclin E, across the cell cycle. However, combined ablation of all A- and E-type cyclins extinguished cell division. In contrast, cyclin A function was essential for cell-cycle progression of hematopoietic and embryonic stem cells. Expression of cyclin A is particularly high in these compartments, which might render stem cells dependent on cyclin A, whereas in fibroblasts cyclins A and E play redundant roles in cell proliferation.


Subject(s)
Cyclin A/metabolism , Embryo, Mammalian/cytology , Embryonic Stem Cells/metabolism , Fibroblasts/metabolism , Hematopoietic Stem Cells/metabolism , Animals , Cyclin A/genetics , Cyclin E/genetics , Cyclin E/metabolism , Mice , Mice, Knockout
8.
Int J Mol Sci ; 25(12)2024 Jun 12.
Article in English | MEDLINE | ID: mdl-38928190

ABSTRACT

The study of the physiological and pathophysiological processes under extreme conditions facilitates a better understanding of the state of a healthy organism and can also shed light on the pathogenesis of diseases. In recent years, it has become evident that gravitational stress affects both the whole organism and individual cells. We have previously demonstrated that simulated microgravity inhibits proliferation, induces apoptosis, changes morphology, and alters the surface marker expression of megakaryoblast cell line MEG-01. In the present work, we investigate the expression of cell cycle cyclins in MEG-01 cells. We performed several experiments for 24 h, 72 h, 96 h and 168 h. Flow cytometry and Western blot analysis demonstrated that the main change in the levels of cyclins expression occurs under conditions of simulated microgravity after 96 h. Thus, the level of cyclin A expression showed an increase in the RPM group during the first 4 days, followed by a decrease, which, together with the peak of cyclin D, may indicate inhibition of the cell cycle in the G2 phase, before mitosis. In addition, based on the data obtained by PCR analysis, we were also able to see that both cyclin A and cyclin B expression showed a peak at 72 h, followed by a gradual decrease at 96 h. STED microscopy data also confirmed that the main change in cyclin expression of MEG-01 cells occurs at 96 h, under simulated microgravity conditions, compared to static control. These results suggested that the cell cycle disruption induced by RPM-simulated microgravity in MEG-01 cells may be associated with the altered expression of the main regulators of the cell cycle. Thus, these data implicate the development of cellular stress in MEG-01 cells, which may be important for proliferating human cells exposed to microgravity in real space.


Subject(s)
Cell Cycle , Cyclins , Weightlessness Simulation , Humans , Cell Line , Cyclins/metabolism , Cyclins/genetics , Megakaryocyte Progenitor Cells/metabolism , Megakaryocyte Progenitor Cells/cytology , Cyclin A/metabolism , Cyclin A/genetics , Cell Proliferation , Cyclin B/metabolism , Cyclin B/genetics
9.
J Biol Chem ; 298(9): 102319, 2022 09.
Article in English | MEDLINE | ID: mdl-35926712

ABSTRACT

B-Myb is a highly conserved member of the vertebrate Myb family of transcription factors that plays a critical role in cell-cycle progression and proliferation. Myb proteins activate Myb-dependent promoters by interacting specifically with Myb-binding site (MBS) sequences using their DNA-binding domain (DBD). Transactivation of MBS promoters by B-Myb is repressed by its negative regulatory domain (NRD), and phosphorylation of the NRD by Cdk2-CyclinA relieves the repression to activate B-Myb-dependent promoters. However, the structural mechanisms underlying autoinhibition and activation of B-Myb-mediated transcription have been poorly characterized. Here, we determined that a region in the B-Myb NRD (residues 510-600) directly associates with the DBD and inhibits binding of the DBD to the MBS DNA sequence. We demonstrate using biophysical assays that phosphorylation of the NRD at T515, T518, and T520 is sufficient to disrupt the interaction between the NRD and the DBD, which results in increased affinity for MBS DNA and increased B-Myb-dependent promoter activation in cell assays. Our biochemical characterization of B-Myb autoregulation and the activating effects of phosphorylation provide insight into how B-Myb functions as a site-specific transcription factor.


Subject(s)
Cell Cycle Proteins , Cyclin-Dependent Kinase 2 , DNA , Trans-Activators , Cell Cycle Proteins/chemistry , Cell Cycle Proteins/metabolism , Cyclin A/metabolism , Cyclin-Dependent Kinase 2/genetics , Cyclin-Dependent Kinase 2/metabolism , DNA/metabolism , Humans , Phosphorylation , Protein Domains , Trans-Activators/chemistry , Trans-Activators/metabolism , Transcriptional Activation
10.
Nat Chem Biol ; 17(4): 456-464, 2021 04.
Article in English | MEDLINE | ID: mdl-33526892

ABSTRACT

Cyclin-dependent kinases (CDKs) are the master regulators of the eukaryotic cell cycle. To become activated, CDKs require both regulatory phosphorylation and binding of a cognate cyclin subunit. We studied the activation process of the G1/S kinase Cdk2 in solution and developed a thermodynamic model that describes the allosteric coupling between regulatory phosphorylation, cyclin binding and inhibitor binding. The results explain why monomeric Cdk2 lacks activity despite sampling an active-like state, reveal that regulatory phosphorylation enhances allosteric coupling with the cyclin subunit and show that this coupling underlies differential recognition of Cdk2 and Cdk4 inhibitors. We identify an allosteric hub that has diverged between Cdk2 and Cdk4 and show that this hub controls the strength of allosteric coupling. The altered allosteric wiring of Cdk4 leads to compromised activity toward generic peptide substrates and comparative specialization toward its primary substrate retinoblastoma (RB).


Subject(s)
Allosteric Regulation/physiology , Cyclin-Dependent Kinase 2/metabolism , Allosteric Site/genetics , Cell Cycle/physiology , Cell Cycle Proteins/metabolism , Cyclin A/metabolism , Cyclin-Dependent Kinase 2/antagonists & inhibitors , Cyclin-Dependent Kinases/antagonists & inhibitors , Cyclin-Dependent Kinases/metabolism , Cyclins/metabolism , Humans , Microtubule-Associated Proteins/metabolism , Models, Biological , Phosphorylation/physiology , Protein Kinase Inhibitors/metabolism , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins/metabolism , Tumor Suppressor Proteins/metabolism
11.
PLoS Genet ; 16(8): e1008988, 2020 08.
Article in English | MEDLINE | ID: mdl-32841231

ABSTRACT

Achieving complete and precise genome duplication requires that each genomic segment be replicated only once per cell division cycle. Protecting large eukaryotic genomes from re-replication requires an overlapping set of molecular mechanisms that prevent the first DNA replication step, the DNA loading of MCM helicase complexes to license replication origins, after S phase begins. Previous reports have defined many such origin licensing inhibition mechanisms, but the temporal relationships among them are not clear, particularly with respect to preventing re-replication in G2 and M phases. Using a combination of mutagenesis, biochemistry, and single cell analyses in human cells, we define a new mechanism that prevents re-replication through hyperphosphorylation of the essential MCM loading protein, Cdt1. We demonstrate that Cyclin A/CDK1 can hyperphosphorylate Cdt1 to inhibit MCM re-loading in G2 phase. The mechanism of inhibition is to block Cdt1 binding to MCM independently of other known Cdt1 inactivation mechanisms such as Cdt1 degradation during S phase or Geminin binding. Moreover, our findings suggest that Cdt1 dephosphorylation at the mitosis-to-G1 phase transition re-activates Cdt1. We propose that multiple distinct, non-redundant licensing inhibition mechanisms act in a series of sequential relays through each cell cycle phase to ensure precise genome duplication.


Subject(s)
DNA Replication/genetics , Genome, Human/genetics , Replication Origin/genetics , Segmental Duplications, Genomic/genetics , CDC2 Protein Kinase/genetics , Cell Cycle Proteins/genetics , Cyclin A/genetics , G2 Phase/genetics , Geminin/genetics , Genes, Duplicate/genetics , HEK293 Cells , Humans , Minichromosome Maintenance Proteins/genetics , Phosphorylation/genetics , S Phase/genetics
12.
Int J Mol Sci ; 24(2)2023 Jan 13.
Article in English | MEDLINE | ID: mdl-36675143

ABSTRACT

Camptothecin (CPT), first isolated from Chinese tree Camptotheca acuminate, produces rapid and prolonged inhibition of DNA synthesis and induction of DNA damage by targeting topoisomerase I (top1), which is highly activated in cancer cells. CPT thus exhibits remarkable anticancer activities in various cancer types, and is a promising therapeutic agent for the treatment of cancers. However, it remains to be uncovered underlying its cytotoxicity toward germ cells. In this study we found that CPT, a cell cycle-specific anticancer agent, reduced fecundity and exhibited significant cytotoxicity toward GSCs and two-cell cysts. We showed that CPT induced GSC loss and retarded two-cell cysts differentiation in a niche- or apoptosis-independent manner. Instead, CPT induced ectopic expression of a differentiation factor, bag of marbles (Bam), and regulated the expression of cyclin A, which contributed to GSC loss. In addition, CPT compromised two-cell cysts differentiation by decreasing the expression of Bam and inducing cell arrest at G1/S phase via cyclin A, eventually resulting in two-cell accumulation. Collectively, this study demonstrates, for the first time in vivo, that the Bam-cyclin A axis is involved in CPT-mediated germline stem cell loss and two-cell cysts differentiation defects via inducing cell cycle arrest, which could provide information underlying toxicological effects of CPT in the productive system, and feature its potential to develop as a pharmacology-based germline stem cell regulation agent.


Subject(s)
Cysts , Drosophila Proteins , Animals , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Cyclins/metabolism , Drosophila Proteins/metabolism , Cell Differentiation , Cyclin A/metabolism , Camptothecin/pharmacology , Camptothecin/metabolism , Cell Cycle Checkpoints , Germ Cells/metabolism , Cysts/metabolism
13.
Biochem Biophys Res Commun ; 627: 5-11, 2022 10 30.
Article in English | MEDLINE | ID: mdl-36007335

ABSTRACT

CDK2 forms a complex with cyclin A and cyclin E to promote the progress of cell cycle, but when cyclin A and cyclin E are dissociated from the complex and degraded by the ubiquitin proteasome pathway, the fate of the inactive CDK2 is unclear. In this study, we found that the inactive CDK2 protein was degraded by autophagy-lysosome pathway. In the classic model of G0/G1 phase arrest induced by serum starvation, we found that the mRNA level in CDK2 did not change but the protein level decreased. Subsequently, using PI3K and AKT inhibitors and gene knockout methods, it was found that CDK2 degradation was mediated by the inhibition of PI3Kα/AKTT308. In addition, P62/SQSTM1 was found to bind to the inactivated CDK2 protein to help it enter autophagy-lysosome degradation in a CTSB-dependent manner. Taken together, these results confirm that the PI3Kα/AKTT308 inhibition leads to degradation of CDK2 protein in the autophagy-lysosome pathway. These data reveal a new molecular mechanism of CDK2 protein degradation and provide a new strategy and method for regulating CDK2 protein.


Subject(s)
Cyclin E , Proto-Oncogene Proteins c-akt , Autophagy/genetics , Cyclin A/metabolism , Cyclin E/metabolism , Cyclin-Dependent Kinase 2/genetics , Cyclin-Dependent Kinase 2/metabolism , Lysosomes/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Sequestosome-1 Protein/metabolism
14.
Stem Cells ; 39(12): 1671-1687, 2021 12.
Article in English | MEDLINE | ID: mdl-34460135

ABSTRACT

The study of proliferation regulation in human pluripotent stem cells is crucial to gain insights into understanding the physiology of these cells. However, redox regulation of the pluripotent cell cycle remains largely unexplored. Here, using human embryonic stem cells (hESCs) as well as human induced pluripotent stem cells (hiPSCs), we demonstrate that the level of reactive oxygen species (ROS) in pluripotent cells oscillates in accordance with the cell cycle progression with the peak occurring at transition from S to G2 /M phase of the cycle. A decrease of this level by antioxidants leads to hindered S-phase initiation and progression but does not affect the early-G1 -phase or mitosis. Cells exposed to antioxidants in the early-G1 -phase accumulate the phosphorylated retinoblastoma protein and overcome the restriction point but are unable to accumulate the main regulators of the S phase-CYCLIN A and GEMININ. Based on the previous findings that CYCLIN A stability is affected by redox homeostasis disturbances in somatic cells, we compared the responses to antioxidant treatments in hESCs and in their differentiated fibroblast-like progeny cells (difESCs). In difESCs, similar to hESCs, a decrease in ROS level results in the disruption of S-phase initiation accompanied by a deficiency of the CYCLIN A level. Moreover, in antioxidant-treated cells, we revealed the accumulation of DNA breaks, which was accompanied by activation of the apoptosis program in pluripotent cells. Thus, we conclude that maintaining the physiological ROS level is essential for promotion of proliferation and accurate DNA synthesis in pluripotent cells and their differentiated descendants.


Subject(s)
Induced Pluripotent Stem Cells , Pluripotent Stem Cells , Antioxidants/metabolism , Cell Cycle/physiology , Cell Proliferation , Cyclin A/metabolism , Humans , Induced Pluripotent Stem Cells/metabolism , Mitosis , Pluripotent Stem Cells/metabolism , Reactive Oxygen Species/metabolism
15.
Nutr Cancer ; 74(5): 1734-1744, 2022.
Article in English | MEDLINE | ID: mdl-34353198

ABSTRACT

Diabetes mellitus (DM) is associated with an increased risk and progression of cholangiocarcinoma (CCA). High glucose underlying the association between DM and CCA by modulating the intracellular signaling has been demonstrated. However, the effects of DM and hyperglycemia on cell cycle machineries and progression of CCA remain elucidated. CCA cells, KKU-213A and KKU-213B were cultured in normal (NG, 5.6 mM) or high glucose (HG, 25 mM) resembling euglycemia and hyperglycemia. Western blotting was used to determine expressions of cell cycle machineries in CCA cells. The expression of cyclin A in CCA tissues from patients with or without hyperglycemia was determined by immunohistochemistry. Pan-cyclin dependent kinases (CDKs) inhibitor and silencing of cyclin A expression were investigated as a possible modality targeting CCA treatment in patients with DM. High glucose induced expression of cell cycle machinery proteins in both CCA cells. Among these, cyclin A was consistently and significantly upregulated. Nuclear cyclin A was significantly increased in tumor tissues from CCA patients with hyperglycemia and was significantly associated with post-operative survival of shorter than 5 mo. Silencing cyclin A expression sensitized CCA cells to pan-CDKs inhibitor, suggesting the combined treatment as an alternative approach for treatment of CCA patients with DM.


Subject(s)
Bile Duct Neoplasms , Cholangiocarcinoma , Diabetes Mellitus , Hyperglycemia , Bile Duct Neoplasms/drug therapy , Bile Duct Neoplasms/metabolism , Bile Ducts, Intrahepatic/metabolism , Bile Ducts, Intrahepatic/pathology , Cell Line, Tumor , Cell Proliferation , Cholangiocarcinoma/drug therapy , Cholangiocarcinoma/metabolism , Cyclin A/metabolism , Cyclin A/pharmacology , Cyclins/metabolism , Glucose/pharmacology , Humans , Protein Kinase Inhibitors/pharmacology , Up-Regulation
16.
Mol Biol Rep ; 49(11): 10421-10429, 2022 Nov.
Article in English | MEDLINE | ID: mdl-36104587

ABSTRACT

BACKGROUND: Bilirubin, as an essential constituent of cellular signaling pathways, may have a role in cell growth and apoptosis in breast cancer, although the biochemical relevance is still unclear. The purpose of the present study is to recognize the mechanism underlying bilirubin-induced apoptosis in breast cancer cell lines. METHODS AND RESULTS: To detect the cell viability, MTT assay was carried out. Apoptosis was assessed by flow cytometry analysis and caspase activities were determined by colorimetric method. The expression of AhR, cyclin D1, cyclin A, p53, p27, Bcl-2, and Bax were examined using real-time PCR. The cell viability has been reduced by bilirubin in a dose-dependent manner and an intrinsic apoptotic response has been occurred that was evidenced by the elevation of caspase-3 and - 9 activities. Bilirubin induced cell arrest in cell-cycle progression, which was associated with the induction of AhR expression, down-regulation of cyclin D1, cyclin A, and upregulation of p53 and p27 expression. Following bilirubin treatment, Bcl-2 was decreased and Bax protein was increased in both cell lines. CONCLUSIONS: To discuss, bilirubin, as a naturally occurring antiproliferative molecule, mediates growth inhibition by induction of cell cycle arrest and apoptosis in MCF-7 and MDA-MB-468 breast cancer cells. It is associated with the suppression of cyclin A, D1, and Bcl-2; induction of p53, p27, and Bax together with the activation of caspase-3 and - 9.


Subject(s)
Breast Neoplasms , Cyclin D1 , Humans , Female , G1 Phase Cell Cycle Checkpoints , bcl-2-Associated X Protein/metabolism , Caspase 3/genetics , Caspase 3/metabolism , Cyclin D1/genetics , Cyclin D1/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Bilirubin/pharmacology , Cell Line, Tumor , Apoptosis , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Cyclin A/metabolism
17.
PLoS Genet ; 15(7): e1008253, 2019 07.
Article in English | MEDLINE | ID: mdl-31291240

ABSTRACT

Endoreplication is a cell cycle variant that entails cell growth and periodic genome duplication without cell division, and results in large, polyploid cells. Cells switch from mitotic cycles to endoreplication cycles during development, and also in response to conditional stimuli during wound healing, regeneration, aging, and cancer. In this study, we use integrated approaches in Drosophila to determine how mitotic cycles are remodeled into endoreplication cycles, and how similar this remodeling is between induced and developmental endoreplicating cells (iECs and devECs). Our evidence suggests that Cyclin A / CDK directly activates the Myb-MuvB (MMB) complex to induce transcription of a battery of genes required for mitosis, and that repression of CDK activity dampens this MMB mitotic transcriptome to promote endoreplication in both iECs and devECs. iECs and devECs differed, however, in that devECs had reduced expression of E2F1-dependent genes that function in S phase, whereas repression of the MMB transcriptome in iECs was sufficient to induce endoreplication without a reduction in S phase gene expression. Among the MMB regulated genes, knockdown of AurB protein and other subunits of the chromosomal passenger complex (CPC) induced endoreplication, as did knockdown of CPC-regulated cytokinetic, but not kinetochore, proteins. Together, our results indicate that the status of a CycA-Myb-MuvB-AurB network determines the decision to commit to mitosis or switch to endoreplication in both iECs and devECs, and suggest that regulation of different steps of this network may explain the known diversity of polyploid cycle types in development and disease.


Subject(s)
Drosophila Proteins/genetics , Drosophila/genetics , Endoreduplication , Animals , Aurora Kinase B/metabolism , Cell Cycle Proteins/metabolism , Cyclin A/metabolism , Drosophila/metabolism , Drosophila Proteins/metabolism , Female , Gene Expression Profiling , Mitosis , Polyploidy , Proto-Oncogene Proteins c-myb/metabolism
18.
Int J Mol Sci ; 23(15)2022 Aug 07.
Article in English | MEDLINE | ID: mdl-35955918

ABSTRACT

Lidocaine injection is a common treatment for tendon injuries. However, the evidence suggests that lidocaine is toxic to tendon cells. This study investigated the effects of lidocaine on cultured tendon cells, focusing on the molecular mechanisms underlying cell proliferation and extracellular matrix (ECM) production. Tendon cells cultured from rat Achilles tendons were treated with 0.5, 1.0, or 1.5 mg/mL lidocaine for 24 h. Cell proliferation was evaluated by Cell Counting Kit 8 (CCK-8) assay and bromodeoxyuridine (BrdU) assay. Cell apoptosis was assessed by Annexin V and propidium iodide (PI) stain. Cell cycle progression and cell mitosis were assessed through flow cytometry and immunofluorescence staining, respectively. The expression of cyclin E, cyclin A, cyclin-dependent kinase 2 (CDK2), p21, p27, p53, matrix metalloproteinases-2 (MMP-2), matrix metalloproteinases-9 (MMP-9), type I collagen, and type III collagen were examined through Western blotting, and the enzymatic activity of MMP-9 was determined through gelatin zymography. Lidocaine reduced cell proliferation and reduced G1/S transition and cell mitosis. Lidocaine did not have a significant negative effect on cell apoptosis. Lidocaine significantly inhibited cyclin A and CDK2 expression but promoted p21, p27, and p53 expression. Furthermore, the expression of MMP-2 and MMP-9 increased, whereas that of type I and type III collagen decreased. Lidocaine also increased the enzymatic activity of MMP-9. Our findings support the premise that lidocaine inhibits tendon cell proliferation by changing the expression of cell-cycle-related proteins and reduces ECM production by altering levels of MMPs and collagens.


Subject(s)
Collagen Type III , Matrix Metalloproteinase 9 , Animals , Cell Cycle Proteins/metabolism , Cell Proliferation , Collagen Type III/genetics , Cyclin A/metabolism , Cyclin-Dependent Kinase 2/metabolism , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Down-Regulation , Extracellular Matrix/metabolism , Lidocaine/pharmacology , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Rats , Tendons/metabolism , Tumor Suppressor Protein p53/metabolism
19.
Biochem Biophys Res Commun ; 549: 91-97, 2021 04 16.
Article in English | MEDLINE | ID: mdl-33667714

ABSTRACT

Mammalian cyclin A-CDK (cyclin-dependent kinase) activity during mitotic exit is regulated by two redundant pathways, cyclin degradation and CDK inhibitors (CKIs). Ectopic expression of a destruction box-truncated (thereby stabilized) mutant of cyclin A in the mouse embryonic fibroblasts nullizygous for three CKIs (p21, p27, and p107) results in constitutive activation ("hyperactivation") of cyclin A-CDK and induces rapid tetraploidization, suggesting loss of the two redundant pathways causes genomic instability. To elucidate the mechanism underlying teraploidization by hyperactive cyclin A-CDK, we first examined if the induction of tetraploidization depends on specific cell cycle stage(s). Arresting the cell cycle at either S phase or M phase blocked the induction of tetraploidization, which was restored by subsequent release from the arrest. These results suggest that both S- and M-phase progressions are necessary for the tetraploidization by hyperactive cyclin A-CDK and that the tetraploidization is not caused by chromosome endoreduplication but by mitotic failure. We also observed that the induction of tetraploidization is associated with excessive duplication of centrosomes, which was suppressed by S-phase but not M-phase block, suggesting that hyperactive cyclin A-CDK promotes centrosome overduplication during S phase. Time-lapse microscopy revealed that hyperactive cyclin A-CDK can lead cells to bypass cell division and enter pseudo-G1 state. These observations implicate that hyperactive cyclin A-CDK causes centrosome overduplication, which leads to mitotic slippage and subsequent tetraploidization.


Subject(s)
Centrosome/metabolism , Chromosomes, Mammalian/metabolism , Cyclin A/metabolism , Cyclin-Dependent Kinases/metabolism , Polyploidy , Animals , Cell Cycle Proteins/metabolism , Cyclin A/genetics , Embryo, Mammalian/cytology , Fibroblasts/metabolism , Humans , Mice , Mitosis , Mutation/genetics , S Phase
20.
Mol Cell ; 52(1): 37-51, 2013 Oct 10.
Article in English | MEDLINE | ID: mdl-24076217

ABSTRACT

The mechanisms that underlie and dictate the different biological outcomes of E2F-1 activity have yet to be elucidated. We describe the residue-specific methylation of E2F-1 by the asymmetric dimethylating protein arginine methyltransferase 1 (PRMT1) and symmetric dimethylating PRMT5 and relate the marks to different functional consequences of E2F-1 activity. Methylation by PRMT1 hinders methylation by PRMT5, which augments E2F-1-dependent apoptosis, whereas PRMT5-dependent methylation favors proliferation by antagonizing methylation by PRMT1. The ability of E2F-1 to prompt apoptosis in DNA damaged cells coincides with enhanced PRMT1 methylation. In contrast, cyclin A binding to E2F-1 impedes PRMT1 methylation and augments PRMT5 methylation, thus ensuring that E2F-1 is locked into its cell-cycle progression mode. The Tudor domain protein p100-TSN reads the symmetric methylation mark, and binding of p100-TSN downregulates E2F-1 apoptotic activity. Our results define an exquisite level of precision in the reader-writer interplay that governs the biological outcome of E2F-1 activity.


Subject(s)
Apoptosis , Cell Proliferation , E2F1 Transcription Factor/metabolism , Protein Processing, Post-Translational , Protein-Arginine N-Methyltransferases/metabolism , Repressor Proteins/metabolism , Amino Acid Motifs , Arginine , Cell Line, Tumor , Chromatin Assembly and Disassembly , Cyclin A/metabolism , DNA Damage , E2F1 Transcription Factor/genetics , Gene Expression Regulation , Humans , Methylation , Promoter Regions, Genetic , Protein Binding , Protein-Arginine N-Methyltransferases/genetics , RNA Interference , Repressor Proteins/genetics , Signal Transduction , Transcription, Genetic , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL