Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 686
Filter
1.
Anal Chem ; 95(2): 1643-1651, 2023 01 17.
Article in English | MEDLINE | ID: mdl-36580602

ABSTRACT

Cellular senescence is a stable cell cycle arrest in response to stress or other damage stimuli to maintain tissue homeostasis. However, the accumulation of senescent cells can lead to the progression of various senescence-related disorders. In this paper, we describe the development of a ß-galactosidase-activatable near-infrared (NIR) senoprobe, NBGal, for the detection of senescent cells based on the use of the FDA-approved Nile blue (NB) fluorophore. NBGal was validated in chemotherapeutic-induced senescence cancer models in vitro using SK-Mel 103 and 4T1 cell lines. In vivo monitoring of cellular senescence was evaluated in orthotopic triple-negative breast cancer-bearing mice treated with palbociclib to induce senescence. In all cases, NBGal exhibited a selective tracking of senescent cells mainly ascribed to the overexpressed ß-galactosidase enzyme responsible for hydrolyzing the NBGal probe generating the highly emissive NB fluorophore. In this way, NBGal has proven to be a qualitative, rapid, and minimally invasive probe that allows the direct detection of senescent cells in vivo.


Subject(s)
Cellular Senescence , Mice , Animals , Cell Cycle Checkpoints/physiology , Cell Line , beta-Galactosidase/metabolism
2.
Cell Cycle ; 22(1): 1-37, 2023 01.
Article in English | MEDLINE | ID: mdl-36005738

ABSTRACT

In proliferating cells and tissues a number of checkpoints (G1/S and G2/M) preceding cell division (M-phase) require the signal provided by growth factors present in serum. IGFs (I and II) have been demonstrated to constitute key intrinsic components of the peptidic active fraction of mammalian serum. In vivo genetic ablation studies have shown that the cellular signal triggered by the IGFs through their cellular receptors represents a non-replaceable requirement for cell growth and cell cycle progression. Retroactive and current evaluation of published literature sheds light on the intracellular circuitry activated by these factors providing us with a better picture of the pleiotropic mechanistic actions by which IGFs regulate both cell size and mitogenesis under developmental growth as well as in malignant proliferation. The present work aims to summarize the cumulative knowledge learned from the IGF ligands/receptors and their intracellular signaling transducers towards control of cell size and cell-cycle with particular focus to their actionable circuits in human cancer. Furthermore, we bring novel perspectives on key functional discriminants of the IGF growth-mitogenic pathway allowing re-evaluation on some of its signal components based upon established evidences.


Subject(s)
Cell Cycle Checkpoints , Insulin-Like Growth Factor I , Receptor, Insulin , Somatomedins , Animals , Humans , Cell Cycle/genetics , Cell Cycle/physiology , Cell Cycle Checkpoints/genetics , Cell Cycle Checkpoints/physiology , Cell Proliferation , Insulin-Like Growth Factor I/metabolism , Mammals/metabolism , Receptor, IGF Type 1/genetics , Receptor, Insulin/genetics , Receptor, Insulin/metabolism , Receptors, Somatomedin/genetics
3.
Int J Mol Sci ; 23(14)2022 Jul 11.
Article in English | MEDLINE | ID: mdl-35887001

ABSTRACT

JIB-04, a pan-histone lysine demethylase (KDM) inhibitor, targets drug-resistant cells, along with colorectal cancer stem cells (CSCs), which are crucial for cancer recurrence and metastasis. Despite the advances in CSC biology, the effect of JIB-04 on liver CSCs (LCSCs) and the malignancy of hepatocellular carcinoma (HCC) has not been elucidated yet. Here, we showed that JIB-04 targeted KDMs, leading to the growth inhibition and cell cycle arrest of HCC, and abolished the viability of LCSCs. JIB-04 significantly attenuated CSC tumorsphere formation, growth, relapse, migration, and invasion in vitro. Among KDMs, the deficiency of KDM4B, KDM4D, and KDM6B reduced the viability of the tumorspheres, suggesting their roles in the function of LCSCs. RNA sequencing revealed that JIB-04 affected various cancer-related pathways, especially the PI3K/AKT pathway, which is crucial for HCC malignancy and the maintenance of LCSCs. Our results revealed KDM6B-dependent AKT2 expression and the downregulation of E2F-regulated genes via JIB-04-induced inhibition of the AKT2/FOXO3a/p21/RB axis. A ChIP assay demonstrated JIB-04-induced reduction in H3K27me3 at the AKT2 promoter and the enrichment of KDM6B within this promoter. Overall, our results strongly suggest that the inhibitory effect of JIB-04 on HCC malignancy and the maintenance of LCSCs is mediated via targeting the KDM6B-AKT2 pathway, indicating the therapeutic potential of JIB-04.


Subject(s)
Antineoplastic Agents , Carcinoma, Hepatocellular , Cell Cycle Checkpoints , Histone Demethylases , Jumonji Domain-Containing Histone Demethylases , Liver Neoplasms , Aminopyridines , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/metabolism , Cell Cycle Checkpoints/drug effects , Cell Cycle Checkpoints/physiology , Cell Line, Tumor , Histone Demethylases/antagonists & inhibitors , Histone Demethylases/metabolism , Histone Demethylases/pharmacology , Histones/metabolism , Humans , Hydrazones , Jumonji Domain-Containing Histone Demethylases/pharmacology , Jumonji Domain-Containing Histone Demethylases/therapeutic use , Liver Neoplasms/drug therapy , Liver Neoplasms/metabolism , Lysine/metabolism , Neoplasm Recurrence, Local/metabolism , Neoplastic Stem Cells/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism
4.
Cell Death Dis ; 13(7): 627, 2022 07 20.
Article in English | MEDLINE | ID: mdl-35853868

ABSTRACT

Histone acetylation is essential for initiating and maintaining a permissive chromatin conformation and gene transcription. Dysregulation of histone acetylation can contribute to tumorigenesis and metastasis. Using inducible cre-recombinase and CRISPR/Cas9-mediated deletion, we investigated the roles of the histone lysine acetyltransferase TIP60 (KAT5/HTATIP) in human cells, mouse cells, and mouse embryos. We found that loss of TIP60 caused complete cell growth arrest. In the absence of TIP60, chromosomes failed to align in a metaphase plate during mitosis. In some TIP60 deleted cells, endoreplication occurred instead. In contrast, cell survival was not affected. Remarkably, the cell growth arrest caused by loss of TIP60 was independent of the tumor suppressors p53, INK4A and ARF. TIP60 was found to be essential for the acetylation of H2AZ, specifically at lysine 7. The mRNA levels of 6236 human and 8238 mouse genes, including many metabolism genes, were dependent on TIP60. Among the top 50 differentially expressed genes, over 90% were downregulated in cells lacking TIP60, supporting a role for TIP60 as a key co-activator of transcription. We propose a primary role of TIP60 in H2AZ lysine 7 acetylation and transcriptional activation, and that this fundamental role is essential for cell proliferation. Growth arrest independent of major tumor suppressors suggests TIP60 as a potential anti-cancer drug target.


Subject(s)
Histones , Lysine Acetyltransferase 5 , Lysine , Tumor Suppressor Protein p53 , Acetylation , Animals , Cell Cycle Checkpoints/physiology , Cyclin-Dependent Kinase Inhibitor p16/genetics , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Histone Acetyltransferases/genetics , Histone Acetyltransferases/metabolism , Histones/genetics , Histones/metabolism , Humans , Lysine/metabolism , Lysine Acetyltransferase 5/deficiency , Lysine Acetyltransferase 5/genetics , Lysine Acetyltransferase 5/metabolism , Mice , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
5.
Biochem J ; 479(2): 185-206, 2022 01 28.
Article in English | MEDLINE | ID: mdl-35098993

ABSTRACT

In the last 20 years, a growing army of systems biologists has employed quantitative experimental methods and theoretical tools of data analysis and mathematical modeling to unravel the molecular details of biological control systems with novel studies of biochemical clocks, cellular decision-making, and signaling networks in time and space. Few people know that one of the roots of this new paradigm in cell biology can be traced to a serendipitous discovery by an obscure Russian biochemist, Boris Belousov, who was studying the oxidation of citric acid. The story is told here from an historical perspective, tracing its meandering path through glycolytic oscillations, cAMP signaling, and frog egg development. The connections among these diverse themes are drawn out by simple mathematical models (nonlinear differential equations) that share common structures and properties.


Subject(s)
Biological Clocks/physiology , Cell Cycle Checkpoints/physiology , Cyclic AMP/metabolism , Signal Transduction/physiology , Systems Biology/methods , Amoeba/metabolism , Animals , Anura/embryology , Citric Acid , Glycolysis/physiology , Models, Biological , Ovum/growth & development , Oxidation-Reduction , Yeasts/metabolism
6.
Cell Rep ; 38(2): 110211, 2022 01 11.
Article in English | MEDLINE | ID: mdl-35021076

ABSTRACT

Oncogene-induced senescence (OIS) is a form of stable cell-cycle arrest arising in response to oncogenic stimulation. OIS must be bypassed for transformation, but the mechanisms of OIS establishment and bypass remain poorly understood, especially at the post-transcriptional level. Here, we show that the RNA-binding protein UNR/CSDE1 enables OIS in primary mouse keratinocytes. Depletion of CSDE1 leads to senescence bypass, cell immortalization, and tumor formation, indicating that CSDE1 behaves as a tumor suppressor. Unbiased high-throughput analyses uncovered that CSDE1 promotes OIS by two independent molecular mechanisms: enhancement of the stability of senescence-associated secretory phenotype (SASP) factor mRNAs and repression of Ybx1 mRNA translation. Importantly, depletion of YBX1 from immortal keratinocytes rescues senescence and uncouples proliferation arrest from the SASP, revealing multilayered mechanisms exerted by CSDE1 to coordinate senescence. Our data highlight the relevance of post-transcriptional control in the regulation of senescence.


Subject(s)
Cellular Senescence/physiology , DNA-Binding Proteins/metabolism , RNA-Binding Proteins/metabolism , Animals , Cell Cycle Checkpoints/genetics , Cell Cycle Checkpoints/physiology , Cell Line , Cell Proliferation/physiology , Cellular Senescence/genetics , DNA-Binding Proteins/physiology , Female , Gene Expression/genetics , Gene Expression Regulation/genetics , Humans , Keratinocytes/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Nude , Oncogenes/genetics , Primary Cell Culture , RNA Processing, Post-Transcriptional/physiology , RNA-Binding Proteins/physiology , Senescence-Associated Secretory Phenotype/genetics , Senescence-Associated Secretory Phenotype/physiology , Signal Transduction/physiology , Y-Box-Binding Protein 1/metabolism
7.
Mech Ageing Dev ; 201: 111619, 2022 01.
Article in English | MEDLINE | ID: mdl-34995645

ABSTRACT

Frailty of the locomotory organs has become a widespread problem in the geriatric population. The major factor leading to frailty is an age-associated decrease in muscular mass and a reduced number of muscular cells and myofibers. In aged muscular tissues, muscular satellite cells (MuSCs) are reduced due to abnormalities in their self-renewal and the induction of apoptosis. However, the molecular mechanisms connecting aging-associated physiological changes and the reduction of MuSCs are largely unknown. NIMA-related kinase 2 (Nek2), a member of the Nek family of serine/threonine kinases, was found to be downregulated in aged MuSCs/progenitors. Further, Nek2 downregulation was found to inhibit self-renewal and apoptotic cell death by activating the p53-dependent checkpoint. Attenuated NEK2 expression was also observed in the muscular tissues of elderly donors, and its function was confirmed to be conserved in humans. Overall, this study proposes a novel mechanism for inducing muscular atrophy to understand aging-associated muscular diseases.


Subject(s)
Aging , Apoptosis/physiology , Cell Self Renewal/physiology , NIMA-Related Kinases/metabolism , Sarcopenia , Satellite Cells, Skeletal Muscle , Aging/pathology , Aging/physiology , Animals , Cell Cycle Checkpoints/physiology , Cell Cycle Proteins/metabolism , Cells, Cultured , Down-Regulation , Humans , Mice , NIMA-Related Kinases/physiology , Sarcopenia/metabolism , Sarcopenia/pathology , Satellite Cells, Skeletal Muscle/pathology , Satellite Cells, Skeletal Muscle/physiology
8.
Basic Clin Pharmacol Toxicol ; 130(2): 254-267, 2022 Feb.
Article in English | MEDLINE | ID: mdl-34845850

ABSTRACT

BACKGROUND: A fibrotic liver may have an impaired regenerative capacity. Because liver transplantation is donor limited, understanding the regenerative ability of a fibrotic liver is important. METHODS: A two-thirds partial hepatectomy (PH) was performed in C57Bl/6 mice with or without carbon tetrachloride (CCl4 ) treatment. Liver regeneration in the fibrotic liver after PH was assessed by the intrahepatic expression of the cell cycle regulators p53, p21, cyclin D1, c-Fos and CDK2 using Western blot analysis. In addition, the expression of PGC-1α and the cell proliferation-related proteins PCNA and phosphate histone H3 was determined by Western blot and immunohistochemical staining analyses. Histone epigenetic modification of the PGC-1α promoter was investigated through chromatin immunoprecipitation (ChIP) and reverse transcription-quantitative polymerase chain reaction (RT-qPCR) assays. The impact of PGC-1α on liver regeneration after PH was further evaluated in PGC-1α-knockout mice. RESULTS: A decreased expression of PGC-1α and liver regeneration-related genes in the fibrotic liver was detected after a PH. Histone acetylation at the PGC-1α promoter led to increases in PGC-1α expression and the survival rate in the fibrotic group after a PH. PGC-1α-mediated liver regeneration was further demonstrated in PGC-1αf/f albcre+/0 mice. CONCLUSION: Targeting PGC-1α may represent a strategy to improve the treatment of PH in patients with liver fibrosis.


Subject(s)
Hepatectomy/methods , Liver Cirrhosis/therapy , Liver Regeneration/physiology , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Animals , Carbon Tetrachloride , Cell Cycle Checkpoints/physiology , Cell Line , Humans , Liver Cirrhosis/genetics , Liver Regeneration/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Promoter Regions, Genetic
9.
Prostate ; 82(2): 182-192, 2022 02.
Article in English | MEDLINE | ID: mdl-34672379

ABSTRACT

BACKGROUND: Despite multiple treatment advances for castration-resistant prostate cancer (CRPC), there are currently no curative therapies and patients ultimately to succumb to the disease. Docetaxel (DTX) is the standard first-line chemotherapy for patients with metastatic CRPC; however, drug resistance is inevitable and often develops rapidly, leading to disease progression in nearly all patients. In contrast, when DTX is deployed with androgen deprivation therapy in castration-sensitive disease, more durable responses and improved outcomes are observed, suggesting that aberrant androgen receptor (AR) signaling accelerates DTX resistance in CRPC. In this study, we demonstrate that AR dysregulates the mitotic checkpoint, a critical pathway involved in the anticancer action of DTX. METHODS: Androgen-dependent and independent cell lines were used to evaluate the role of AR in DTX resistance. Impact of drug treatment on cell viability, survival, and cell-cycle distribution were determined by plate-based viability assay, clonogenic assay, and cell-cycle analysis by flow cytometry, respectively. Mitotic checkpoint kinase signal transduction and apoptosis activation was evaluated by Western blotting. Pathway gene expression analysis was evaluated by RT-PCR. A Bliss independence model was used to calculate synergy scores for drug combination studies. RESULTS: Activation of AR in hormone-sensitive cells induces a rescue phenotype by increasing cell viability and survival and attenuating G2/M arrest in response to DTX. Analysis of mitotic checkpoint signaling shows that AR negatively regulates spindle checkpoint signaling, resulting in premature mitotic progression and evasion of apoptosis. This phenotype is characteristic of mitotic slippage and is also observed in CRPC cell lines where we demonstrate involvement of AR splice variant AR-v7 in dysregulation of checkpoint signaling. Our findings suggest that DTX resistance is mediated through mechanisms that drive premature mitotic exit. Using pharmacologic inhibitors of anaphase-promoting complex/cyclosome and polo-like kinase 1, we show that blocking mitotic exit induces mitotic arrest, apoptosis, and synergistically inhibits cell survival in combination with DTX. CONCLUSION: Our results suggest that targeting the mechanisms of dysregulated mitotic checkpoint signaling in AR-reactivated tumors has significant clinical potential to extend treatment benefit with DTX and improve outcomes in patients with lethal prostate cancer.


Subject(s)
Androgen Receptor Antagonists/pharmacology , Cell Cycle Checkpoints , Docetaxel/pharmacology , Prostatic Neoplasms, Castration-Resistant , Signal Transduction/drug effects , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Cycle Checkpoints/drug effects , Cell Cycle Checkpoints/physiology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Drug Resistance, Neoplasm/drug effects , Humans , Male , Prostatic Neoplasms, Castration-Resistant/drug therapy , Prostatic Neoplasms, Castration-Resistant/metabolism , Prostatic Neoplasms, Castration-Resistant/pathology , Receptors, Androgen/metabolism
10.
Plant J ; 109(3): 490-507, 2022 02.
Article in English | MEDLINE | ID: mdl-34741364

ABSTRACT

Being sessile organisms, plants are ubiquitously exposed to stresses that can affect the DNA replication process or cause DNA damage. To cope with these problems, plants utilize DNA damage response (DDR) pathways, consisting of both highly conserved and plant-specific elements. As a part of this DDR, cell cycle checkpoint control mechanisms either pause the cell cycle, to allow DNA repair, or lead cells into differentiation or programmed cell death, to prevent the transmission of DNA errors in the organism through mitosis or to its offspring via meiosis. The two major DDR cell cycle checkpoints control either the replication process or the G2/M transition. The latter is largely overseen by the plant-specific SOG1 transcription factor, which drives the activity of cyclin-dependent kinase inhibitors and MYB3R proteins, which are rate limiting for the G2/M transition. By contrast, the replication checkpoint is controlled by different players, including the conserved kinase WEE1 and likely the transcriptional repressor RBR1. These checkpoint mechanisms are called upon during developmental processes, in retrograde signaling pathways, and in response to biotic and abiotic stresses, including metal toxicity, cold, salinity, and phosphate deficiency. Additionally, the recent expansion of research from Arabidopsis to other model plants has revealed species-specific aspects of the DDR. Overall, it is becoming evidently clear that the DNA damage checkpoint mechanisms represent an important aspect of the adaptation of plants to a changing environment, hence gaining more knowledge about this topic might be helpful to increase the resilience of plants to climate change.


Subject(s)
Absorption, Physiological/genetics , Arabidopsis/genetics , Arabidopsis/physiology , Cell Cycle Checkpoints/genetics , Cell Cycle Checkpoints/physiology , DNA Damage/genetics , Stress, Physiological/genetics , Absorption, Physiological/physiology , DNA Damage/physiology , Gene Expression Regulation, Plant , Genes, Plant , Stress, Physiological/physiology , Transcription Factors
11.
Mol Cell Biol ; 42(2): e0031021, 2022 02 17.
Article in English | MEDLINE | ID: mdl-34898277

ABSTRACT

RASSF6, a member of the tumor suppressor Ras-association domain family (RASSF) proteins, regulates cell cycle arrest and apoptosis via p53 and plays a tumor suppressor role. We previously reported that RASSF6 blocks MDM2-mediated p53 degradation and enhances p53 expression. In this study, we demonstrated that RASSF6 has nuclear localization and nuclear export signals and that DNA damage triggers the nuclear accumulation of RASSF6. We found that RASSF6 directly binds to BAF53, the component of SWI/SNF complex. DNA damage induces CDK9-mediated phosphorylation of BAF53, which enhances the interaction with RASSF6 and increases the amount of RASSF6 in the nucleus. Subsequently, RASSF6 augments the interaction between BAF53 and BAF60a, another component of the SWI/SNF complex, and further promotes the interaction of BAF53 and BAF60a with p53. BAF53 silencing or BAF60a silencing attenuates RASSF6-mediated p53 target gene transcription and apoptosis. Thus, RASSF6 is involved in the regulation of DNA damage-induced complex formation, including BAF53, BAF60a, and p53.


Subject(s)
Actins/metabolism , Apoptosis Regulatory Proteins/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Cyclin-Dependent Kinase 9/metabolism , DNA Damage/genetics , DNA-Binding Proteins/metabolism , Transcription, Genetic/genetics , Tumor Suppressor Protein p53/metabolism , Actins/genetics , Apoptosis/physiology , Apoptosis Regulatory Proteins/genetics , Cell Cycle Checkpoints/genetics , Cell Cycle Checkpoints/physiology , Chromosomal Proteins, Non-Histone/genetics , Cyclin-Dependent Kinase 9/genetics , DNA Damage/physiology , DNA-Binding Proteins/genetics , Humans , Monomeric GTP-Binding Proteins/genetics , Tumor Suppressor Proteins/metabolism
12.
Nat Commun ; 12(1): 6562, 2021 11 12.
Article in English | MEDLINE | ID: mdl-34772946

ABSTRACT

Quiescent neural stem cells (NSCs) in the adult mouse brain are the source of neurogenesis that regulates innate and adaptive behaviors. Adult NSCs in the subventricular zone are derived from a subpopulation of embryonic neural stem-progenitor cells (NPCs) that is characterized by a slower cell cycle relative to the more abundant rapid cycling NPCs that build the brain. Yet, how slow cell cycle can cause the establishment of adult NSCs remains largely unknown. Here, we demonstrate that Notch and an effector Hey1 form a module that is upregulated by cell cycle arrest in slowly dividing NPCs. In contrast to the oscillatory expression of the Notch effectors Hes1 and Hes5 in fast cycling progenitors, Hey1 displays a non-oscillatory stationary expression pattern and contributes to the long-term maintenance of NSCs. These findings reveal a novel division of labor in Notch effectors where cell cycle rate biases effector selection and cell fate.


Subject(s)
Adult Stem Cells/metabolism , Cell Cycle Checkpoints/physiology , Cell Cycle Proteins/metabolism , Neurogenesis/physiology , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Brain/cytology , Cell Cycle/genetics , Cell Cycle/physiology , Cell Cycle Checkpoints/genetics , Cell Cycle Proteins/genetics , Embryonic Stem Cells , Gene Expression , Lateral Ventricles/metabolism , Mice , Nervous System , Neurogenesis/genetics , Receptor, Notch1 , Repressor Proteins/metabolism
13.
J Zhejiang Univ Sci B ; 22(10): 839-855, 2021 Oct 15.
Article in English | MEDLINE | ID: mdl-34636187

ABSTRACT

B cell lymphoma 2 (Bcl-2) is an important antiapoptotic gene that plays a dual role in the maintenance of the dynamic balance between the survival and death of cancer cells. In our previous study, Bcl-2 was shown to delay the G0/G1 to S phase entry by regulating the mitochondrial metabolic pathways to produce lower levels of adenosine triphosphate (ATP) and reactive oxygen species (ROS). However, the detailed molecular mechanisms or pathways by which Bcl-2 regulates the cell cycle remain unknown. Here, we compared the effects of Bcl-2 overexpression with an empty vector control in the NIH3T3 cell line synchronized by serum starvation, and evaluated the effects using proteomic analysis. The effect of Bcl-2 on cell cycle regulation was detected by monitoring Bcl-2 and p27 expression. The result of subsequent proteomic analysis of Bcl-2 overexpressing cells identified 169 upregulated and 120 downregulated proteins with a 1.5-fold change. These differentially expressed proteins were enriched in a number of signaling pathways predominantly involving the ribosome and oxidative phosphorylation, according to the data of Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses. These results indicated that Bcl-2 potentially acts at the translation level to influence proteins or enzymes of the respiratory chain or in the ribosome, and thereby regulates the cell cycle. Additionally, differentially expressed proteins involved in oxidative phosphorylation were determined to account for most of the effects of Bcl-2 on the cell cycle mediated by the mitochondrial pathway investigated in our previous study. These results can provide assistance for additional in-depth studies on the regulation of the cell cycle by Bcl-2. The results of the proteomic analysis determined the mechanism of Bcl-2-dependent delay of the cell cycle progression. In summary, the results of this study provide a novel mechanistic basis for identifying the key proteins or pathways for designing and developing precisely targeted cancer drugs.


Subject(s)
Cell Cycle Checkpoints/physiology , Proteomics/methods , Proto-Oncogene Proteins c-bcl-2/physiology , Animals , Cluster Analysis , Mice , NIH 3T3 Cells
14.
Oxid Med Cell Longev ; 2021: 9955717, 2021.
Article in English | MEDLINE | ID: mdl-34650666

ABSTRACT

Dynamic cytoskeletal rearrangements underlie the changes that occur during cell division in proliferating cells. MICAL2 has been reported to possess reactive oxygen species- (ROS-) generating properties and act as an important regulator of cytoskeletal dynamics. However, whether it plays a role in gastric cancer cell proliferation is not known. In the present study, we found that MICAL2 was highly expressed in gastric cancer tissues, and this high expression level was associated with carcinogenesis and poor overall survival in gastric cancer patients. The knockdown of MICAL2 led to cell cycle arrest in the S phase and attenuated cell proliferation. Concomitant with S-phase arrest, a decrease in CDK6 and cyclin D protein levels was observed. Furthermore, MICAL2 knockdown attenuated intracellular ROS generation, while MICAL2 overexpression led to a decrease in the p-YAP/YAP ratio and promoted YAP nuclear localization and cell proliferation, effects that were reversed by pretreatment with the ROS scavenger N-acetyl-L-cysteine (NAC) and SOD-mimetic drug tempol. We further found that MICAL2 induced Cdc42 activation, and activated Cdc42 mediated the effect of MICAL2 on YAP dephosphorylation and nuclear translocation. Collectively, our results showed that MICAL2 has a promotive effect on gastric cancer cell proliferation through ROS generation and Cdc42 activation, both of which independently contribute to YAP dephosphorylation and its nuclear translocation.


Subject(s)
Cell Cycle Proteins/metabolism , Cell Proliferation/physiology , Microfilament Proteins/metabolism , Oxidoreductases/metabolism , Stomach Neoplasms/metabolism , Transcription Factors/metabolism , Carcinogenesis/metabolism , Cell Cycle Checkpoints/physiology , Cell Line, Tumor , Cell Movement/physiology , Gene Expression Regulation, Neoplastic/physiology , Humans
15.
Elife ; 102021 08 04.
Article in English | MEDLINE | ID: mdl-34346309

ABSTRACT

The abscission checkpoint regulates the ESCRT membrane fission machinery and thereby delays cytokinetic abscission to protect genomic integrity in response to residual mitotic errors. The checkpoint is maintained by Aurora B kinase, which phosphorylates multiple targets, including CHMP4C, a regulatory ESCRT-III subunit necessary for this checkpoint. We now report the discovery that cytoplasmic abscission checkpoint bodies (ACBs) containing phospho-Aurora B and tri-phospho-CHMP4C develop during an active checkpoint. ACBs are derived from mitotic interchromatin granules, transient mitotic structures whose components are housed in splicing-related nuclear speckles during interphase. ACB formation requires CHMP4C, and the ESCRT factor ALIX also contributes. ACB formation is conserved across cell types and under multiple circumstances that activate the checkpoint. Finally, ACBs retain a population of ALIX, and their presence correlates with delayed abscission and delayed recruitment of ALIX to the midbody where it would normally promote abscission. Thus, a cytoplasmic mechanism helps regulate midbody machinery to delay abscission.


When a cell divides, it must first carefully duplicate its genetic information and package these copies into compartments housed in the two new cells. Errors in this process lead to genetic mistakes that trigger cancer or other harmful biological events. Quality control checks exist to catch errors before it is too late. This includes a final 'abscission' checkpoint right before the end of division, when the two new cells are still connected by a thin membrane bridge. If cells fail to pass this 'no cut' checkpoint, they delay severing their connection until the mistake is fixed. A group of proteins called ESCRTs is responsible for splitting the two cells apart if nothing is amiss. The abscission checkpoint blocks this process by altering certain proteins in the ESCRT complex, but exactly how this works is not yet clear. To find out more, Williams et al. imaged ESCRT factors in a new experimental system in which the abscission checkpoint is active in many cells. This showed that, in this context, certain ESCRT components were rerouted from the thread of membrane between the daughter cells to previously unknown structures, which Williams et al. named abscission checkpoint bodies. These entities also sequestered other factors that participate in the abscission checkpoint and factors that contribute to gene expression. These results are key to better understand how cells regulate their division; in particular, they provide a new framework to explore when this process goes wrong and contributes to cancer.


Subject(s)
Cell Cycle Checkpoints/physiology , Cell Division/physiology , Endosomal Sorting Complexes Required for Transport/metabolism , Gene Expression Regulation/physiology , Cell Line , Endosomal Sorting Complexes Required for Transport/genetics , Humans , RNA Interference , RNA, Small Interfering , Single-Cell Analysis
16.
PLoS Comput Biol ; 17(8): e1009008, 2021 08.
Article in English | MEDLINE | ID: mdl-34379640

ABSTRACT

Modeling biochemical reactions by means of differential equations often results in systems with a large number of variables and parameters. As this might complicate the interpretation and generalization of the obtained results, it is often desirable to reduce the complexity of the model. One way to accomplish this is by replacing the detailed reaction mechanisms of certain modules in the model by a mathematical expression that qualitatively describes the dynamical behavior of these modules. Such an approach has been widely adopted for ultrasensitive responses, for which underlying reaction mechanisms are often replaced by a single Hill function. Also time delays are usually accounted for by using an explicit delay in delay differential equations. In contrast, however, S-shaped response curves, which by definition have multiple output values for certain input values and are often encountered in bistable systems, are not easily modeled in such an explicit way. Here, we extend the classical Hill function into a mathematical expression that can be used to describe both ultrasensitive and S-shaped responses. We show how three ubiquitous modules (ultrasensitive responses, S-shaped responses and time delays) can be combined in different configurations and explore the dynamics of these systems. As an example, we apply our strategy to set up a model of the cell cycle consisting of multiple bistable switches, which can incorporate events such as DNA damage and coupling to the circadian clock in a phenomenological way.


Subject(s)
Cell Cycle/physiology , Models, Biological , Animals , Cell Cycle/genetics , Cell Cycle Checkpoints/genetics , Cell Cycle Checkpoints/physiology , Cell Cycle Proteins/genetics , Cell Cycle Proteins/physiology , Computational Biology , Computer Simulation , DNA Damage , Humans , Kinetics , Systems Biology
17.
PLoS Genet ; 17(7): e1009650, 2021 07.
Article in English | MEDLINE | ID: mdl-34288923

ABSTRACT

Quiescence, an actively-maintained reversible state of cell cycle arrest, is not well understood. PTEN is one of the most frequently lost tumor suppressors in human cancers and regulates quiescence of stem cells and cancer cells. The sole PTEN ortholog in Caenorhabditis elegans is daf-18. In a C. elegans loss-of-function mutant for daf-18, primordial germ cells (PGCs) divide inappropriately in L1 larvae hatched into starvation conditions, in a TOR-dependent manner. Here, we further investigated the role of daf-18 in maintaining PGC quiescence in L1 starvation. We found that maternal or zygotic daf-18 is sufficient to maintain cell cycle quiescence, that daf-18 acts in the germ line and soma, and that daf-18 affects timing of PGC divisions in fed animals. Importantly, our results also implicate daf-18 in repression of germline zygotic gene activation, though not in germline fate specification. However, TOR is less important to germline zygotic gene expression, suggesting that in the absence of food, daf-18/PTEN prevents inappropriate germline zygotic gene activation and cell division by distinct mechanisms.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Cell Cycle Checkpoints/physiology , Germ Cells/metabolism , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/physiology , Cell Division/genetics , Cell Proliferation/genetics , Larva/genetics , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Signal Transduction/genetics , Transcriptional Activation/genetics , Zygote/metabolism
18.
Biomed Pharmacother ; 141: 111795, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34098217

ABSTRACT

Cancer recurrence poses a significant challenge. At the cellular level, recurrence takes place as a result of reactivation of dormant cancer cells residing at G0 phase. The aim of the study was to identify compounds that can trap prostate and lung cancer cells in G0 phase from a new Chinese herb recipe, Astringent recipe, consisting of Radix Paeoniae Alba, Agrimonia pilosa Ledeb, Fructus Mume, Fritillaria thunbergii Miq., Ganoderma Lucidum Karst, and Astragalus membranaceus (Fisch.) Bunge. Astringent recipe impeded cell cycle progression in prostate and lung cancer cells by rounding them up at G0 phase by flow cytometric analysis of cancer cells stained with Hoechst 33342 and Pyronin Y, respectively, for DNA and RNA. The anti-cancer efficacy of the recipe was found to be attributable to Agrimonia pilosa Ledeb. Further study established that agrimol B, a polyphenol derived from Agrimonia pilosa Ledeb, contributed to the activity of the herb. The action of agrimol B on the cancer cells was likely derived from its effect on c-MYC, SKP2 and p27 by immunoblotting and immunofluorescence. Oral administration of Agrimonia pilosa Ledeb or agrimol B reduced growth of prostate cancer cell xenograft in animal. In conclusion, Agrimol B can enrich for prostate and lung cancer cells in G0 state and influence key regulators that govern G0 status.


Subject(s)
Agrimonia , Antineoplastic Agents, Phytogenic/pharmacology , Butanones/pharmacology , G1 Phase Cell Cycle Checkpoints/drug effects , Phenols/pharmacology , Plant Extracts/pharmacology , Tumor Burden/drug effects , A549 Cells , Animals , Antineoplastic Agents, Phytogenic/isolation & purification , Butanones/isolation & purification , Cell Cycle Checkpoints/drug effects , Cell Cycle Checkpoints/physiology , Dose-Response Relationship, Drug , Ellagic Acid/pharmacology , G1 Phase Cell Cycle Checkpoints/physiology , HEK293 Cells , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Phenols/isolation & purification , Plant Extracts/isolation & purification , Tumor Burden/physiology
19.
Development ; 148(13)2021 07 01.
Article in English | MEDLINE | ID: mdl-34164654

ABSTRACT

Understanding the mechanisms of embryonic cell cycles is a central goal of developmental biology, as the regulation of the cell cycle must be closely coordinated with other events during early embryogenesis. Quantitative imaging approaches have recently begun to reveal how the cell cycle oscillator is controlled in space and time, and how it is integrated with mechanical signals to drive morphogenesis. Here, we discuss how the Drosophila embryo has served as an excellent model for addressing the molecular and physical mechanisms of embryonic cell cycles, with comparisons to other model systems to highlight conserved and species-specific mechanisms. We describe how the rapid cleavage divisions characteristic of most metazoan embryos require chemical waves and cytoplasmic flows to coordinate morphogenesis across the large expanse of the embryo. We also outline how, in the late cleavage divisions, the cell cycle is inter-regulated with the activation of gene expression to ensure a reliable maternal-to-zygotic transition. Finally, we discuss how precise transcriptional regulation of the timing of mitosis ensures that tissue morphogenesis and cell proliferation are tightly controlled during gastrulation.


Subject(s)
Cell Cycle Checkpoints/physiology , Drosophila/embryology , Embryonic Development/physiology , Animals , CDC2 Protein Kinase , Cell Cycle/genetics , Drosophila Proteins , Embryo, Mammalian , Embryo, Nonmammalian/metabolism , Embryonic Development/genetics , Gene Expression Regulation, Developmental , Mitosis , Morphogenesis , Xenopus/embryology , Zygote/metabolism
20.
Med Sci Monit ; 27: e930083, 2021 Jun 14.
Article in English | MEDLINE | ID: mdl-34125740

ABSTRACT

BACKGROUND Helenalin is a pseudoguaianolide natural product with anti-cancer activities. This study investigated the underlying mechanism of the anti-prostate cancer effects of helenalin in vitro. MATERIAL AND METHODS CCK-8 assay was performed to detect the optimal concentrations of helenalin in DU145 and PC-3 cells. After exposure to helenalin and/or reactive oxygen species (ROS) inhibitor, ROS production was assessed by DCFH-DA staining. Thioredoxin reductase-1 (TrxR1) expression was detected by RT-qPCR and western blot. Moreover, apoptosis and cell cycle were evaluated by flow cytometry. Following TrxR1 knockdown or overexpression, TrxR1 expression, ROS generation, apoptosis, cell cycle, migration, and invasion were examined in cells co-treated with helenalin. RESULTS Helenalin distinctly repressed the viability of prostate cancer cells in a concentration-dependent manner. We chose 8 µM and 4 µM as the optimal concentrations of helenalin for DU145 and PC-3 cells, respectively. Helenalin treatment markedly triggered ROS production and lowered TrxR1 expression, which was ameliorated by ROS inhibitor. Exposure to helenalin facilitated apoptosis as well as G0/G1 cell cycle arrest, which was reversed by ROS inhibitor. Helenalin relieved the inhibitory effect of TrxR1 on ROS production. Furthermore, helenalin ameliorated the decrease in apoptosis rate and the shortening of G0/G1 phase as well as the increase in migration and invasion induced by TrxR1 overexpression. CONCLUSIONS Our findings revealed that helenalin accelerated ROS-mediated apoptosis and cell cycle arrest via targeting TrxR1 in human prostate cancer cells.


Subject(s)
Prostatic Neoplasms/metabolism , Sesquiterpenes, Guaiane/pharmacology , Thioredoxin Reductase 1/metabolism , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Cycle Checkpoints/drug effects , Cell Cycle Checkpoints/physiology , Cell Line, Tumor , Cell Survival/drug effects , G1 Phase Cell Cycle Checkpoints , Humans , Male , Reactive Oxygen Species , Sesquiterpenes, Guaiane/metabolism , Thioredoxin Reductase 1/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...