Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 848
Filter
1.
J Clin Neurophysiol ; 41(3): 278-284, 2024 Mar 01.
Article in English | MEDLINE | ID: mdl-38436391

ABSTRACT

PURPOSE: Nerve conduction study (NCS) is essential for subclassifying Guillain-Barré syndrome (GBS). It is well known that the GBS subclassification can change through serial NCSs. However, the usefulness of serial NCSs is debatable, especially in patients with early stage GBS. METHODS: Follow-up NCS data within 3 weeks (early followed NCS, EFN) and within 3 to 10 weeks (late-followed NCS, LFN) were collected from 60 patients with GBS who underwent their first NCS (FN) within 10 days after symptom onset. Each NCS was classified into five subtypes (normal, demyelinating, axonal, inexcitable, and equivocal), according to Hadden's and Rajabally's criteria. We analyzed the frequency of significant changes in classification (SCCs) comprising electrodiagnostic aggravation and subtype shifts between demyelinating and axonal types according to follow-up timing. RESULTS: Between FN and EFN, 33.3% of patients with Hadden's criteria and 18.3% with Rajabally's criteria showed SCCs. Between FN and LFN, 23.3% of patients with Hadden's criteria and 21.7% with Rajabally's criteria showed SCCs, of which 71.4% (Hadden's criteria) and 46.2% (Rajabally's criteria) already showed SCCs from the EFN. The conditions of delayed SCCs between EFN and LFN were very early FN, mild symptoms at the FN, or persistent electrophysiological deterioration 3 weeks after symptom onset. CONCLUSIONS: A substantial proportion of patients with GBS showed significant changes in neurophysiological classification at the early stage. Serial NCS may be helpful for precise neurophysiological classification. This study suggests that follow-up NCSs should be performed within 3 weeks of symptom onset in patients with GBS in whom FN was performed within 10 days of symptom onset.


Subject(s)
Guillain-Barre Syndrome , Zinostatin , Humans , Guillain-Barre Syndrome/diagnosis , Nerve Conduction Studies , Neurophysiology
2.
Cells ; 12(22)2023 11 17.
Article in English | MEDLINE | ID: mdl-37998381

ABSTRACT

Cells respond to DNA damage by activating a complex array of signaling networks, which include the AMPK and mTOR pathways. After DNA double-strand breakage, ATM, a core component of the DNA repair system, activates the AMPK-TSC2 pathway, leading to the inhibition of the mTOR cascade. Recently, we showed that both AMPK and mTOR interact with SMYD3, a methyltransferase involved in DNA damage response. In this study, through extensive molecular characterization of gastrointestinal and breast cancer cells, we found that SMYD3 is part of a multiprotein complex that is involved in DNA damage response and also comprises AMPK and mTOR. In particular, upon exposure to the double-strand break-inducing agent neocarzinostatin, SMYD3 pharmacological inhibition suppressed AMPK cascade activation and thereby promoted the mTOR pathway, which reveals the central role played by SMYD3 in the modulation of AMPK-mTOR signaling balance during cancer cell response to DNA double-strand breaks. Moreover, we found that SMYD3 can methylate AMPK at the evolutionarily conserved residues Lys411 and Lys424. Overall, our data revealed that SMYD3 can act as a bridge between the AMPK and mTOR pathways upon neocarzinostatin-induced DNA damage in gastrointestinal and breast cancer cells.


Subject(s)
Breast Neoplasms , Zinostatin , Humans , Female , AMP-Activated Protein Kinases/metabolism , TOR Serine-Threonine Kinases/metabolism , DNA Damage , DNA , Histone-Lysine N-Methyltransferase/genetics
3.
Neuroimage ; 279: 120342, 2023 10 01.
Article in English | MEDLINE | ID: mdl-37619792

ABSTRACT

Early neurodevelopment is critically dependent on the structure and dynamics of spontaneous neuronal activity; however, the natural organization of newborn cortical networks is poorly understood. Recent adult studies suggest that spontaneous cortical activity exhibits discrete network states with physiological correlates. Here, we studied newborn cortical activity during sleep using hidden Markov modeling to determine the presence of such discrete neonatal cortical states (NCS) in 107 newborn infants, with 47 of them presenting with a perinatal brain injury. Our results show that neonatal cortical activity organizes into four discrete NCSs that are present in both cardinal sleep states of a newborn infant, active and quiet sleep, respectively. These NCSs exhibit state-specific spectral and functional network characteristics. The sleep states exhibit different NCS dynamics, with quiet sleep presenting higher fronto-temporal activity and a stronger brain-wide neuronal coupling. Brain injury was associated with prolonged lifetimes of the transient NCSs, suggesting lowered dynamics, or flexibility, in the cortical networks. Taken together, the findings suggest that spontaneously occurring transient network states are already present at birth, with significant physiological and pathological correlates; this NCS analysis framework can be fully automatized, and it holds promise for offering an objective, global level measure of early brain function for benchmarking neurodevelopmental or clinical research.


Subject(s)
Brain Injuries , Sleep, Slow-Wave , Zinostatin , Adult , Infant, Newborn , Infant , Female , Pregnancy , Humans , Brain Injuries/diagnostic imaging , Brain , Sleep , Benchmarking
4.
J Colloid Interface Sci ; 628(Pt A): 499-507, 2022 Dec 15.
Article in English | MEDLINE | ID: mdl-35933867

ABSTRACT

It is imperative to design and manufacture electrocatalysts towards oxygen reduction reaction (ORR) and oxygen evolution reaction (OER) for popularization of rechargeable Zn-air batteries. Herein, FeNi alloy confined in N-doped carbon nanosheets (FeNi@NCSs) was harvested via a facile complexation-pyrolysis strategy from the mixture of guanine and metal chlorides. After strictly exploring the pyrolysis temperature and metal types, the resulted FeNi@NCSs showed greatly improved performances on both the ORR (onset potential of 0.93 V and half-wave potential of 0.84 V) and OER (overpotential of 318 mV at 10 mA cm-2 and 379 mV at 100 mA cm-2). Further, the FeNi@NCSs based Zn-air battery exhibited a higher open circuit voltage (1.496 V), a larger power density (128.8 mW cm-2), and prominent durability (360 cycles, 120 h). This study provides an appealing approach to utilize biomass for synthesis of low-cost and high-efficiency electrocatalysts in energy associated systems.


Subject(s)
Carbon , Zinostatin , Alloys , Chlorides , Electrodes , Guanine , Oxygen , Zinc
5.
Mol Pharmacol ; 101(5): 322-333, 2022 05.
Article in English | MEDLINE | ID: mdl-35273080

ABSTRACT

Ceramide is a bioactive sphingolipid that mediates ionizing radiation- and chemotherapy-induced apoptosis. Neocarzinostatin (NCS) is a genotoxic anti-cancer drug that induces apoptosis in response to DNA double-strand breaks (DSBs) through ataxia telangiectasia mutated (ATM) activation. However, the involvement of ceramide in NCS-evoked nuclear events such as DSB-activated ATM has not been clarified. Here, we found that nuclear ceramide increased by NCS-mediated apoptosis through the enhanced assembly of ATM and the meiotic recombination 11/double-strand break repair/Nijmengen breakage syndrome 1 (MRN) complex proteins in human lymphoblastoid L-39 cells. NCS induced an increase of ceramide production through activation of neutral sphingomyelinase (nSMase) and suppression of sphingomyelin synthase (SMS) upstream of DSB-mediated ATM activation. In ATM-deficient lymphoblastoid AT-59 cells compared with L-39 cells, NCS treatment showed a decrease of apoptosis even though ceramide increase and DSBs were observed. Expression of wild-type ATM, but not the kinase-dead mutant ATM, in AT-59 cells increased NCS-induced apoptosis despite similar ceramide accumulation. Interestingly, NCS increased ceramide content in the nucleus through nSMase activation and SMS suppression and promoted colocalization of ceramide with phosphorylated ATM and foci of MRN complex. Inhibition of ceramide generation by the overexpression of SMS suppressed NCS-induced apoptosis through the inhibition of ATM activation and assembly of the MRN complex. In addition, inhibition of ceramide increased by the nSMase inhibitor GW4869 prevented NCS-mediated activation of the ATM. Therefore, our findings suggest the involvement of the nuclear ceramide with ATM activation in NCS-mediated apoptosis. SIGNIFICANCE STATEMENT: This study demonstrates that regulation of ceramide with neutral sphingomyelinase and sphingomyelin synthase in the nucleus in double-strand break-mimetic agent neocarzinostatin (NCS)-induced apoptosis. This study also showed that ceramide increase in the nucleus plays a role in NCS-induced apoptosis through activation of the ataxia telangiectasia mutated/meiotic recombination 11/double-strand break repair/Nijmengen breakage syndrome 1 complex in human lymphoblastoid cells.


Subject(s)
Ataxia Telangiectasia , Zinostatin , Apoptosis/genetics , Ataxia Telangiectasia/genetics , Ataxia Telangiectasia Mutated Proteins/genetics , Ataxia Telangiectasia Mutated Proteins/metabolism , Cell Cycle Proteins/metabolism , Ceramides/pharmacology , DNA Repair , DNA-Binding Proteins/metabolism , Humans , Protein Serine-Threonine Kinases , Sphingomyelin Phosphodiesterase/genetics , Sphingomyelin Phosphodiesterase/metabolism , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Zinostatin/pharmacology
6.
Nat Commun ; 12(1): 3686, 2021 06 17.
Article in English | MEDLINE | ID: mdl-34140498

ABSTRACT

Tumour hypoxia is associated with poor patient prognosis and therapy resistance. A unique transcriptional response is initiated by hypoxia which includes the rapid activation of numerous transcription factors in a background of reduced global transcription. Here, we show that the biological response to hypoxia includes the accumulation of R-loops and the induction of the RNA/DNA helicase SETX. In the absence of hypoxia-induced SETX, R-loop levels increase, DNA damage accumulates, and DNA replication rates decrease. Therefore, suggesting that, SETX plays a role in protecting cells from DNA damage induced during transcription in hypoxia. Importantly, we propose that the mechanism of SETX induction in hypoxia is reliant on the PERK/ATF4 arm of the unfolded protein response. These data not only highlight the unique cellular response to hypoxia, which includes both a replication stress-dependent DNA damage response and an unfolded protein response but uncover a novel link between these two distinct pathways.


Subject(s)
Cell Hypoxia , DNA Damage/genetics , DNA Helicases/metabolism , Gene Expression Regulation/genetics , Multifunctional Enzymes/metabolism , R-Loop Structures/genetics , RNA Helicases/metabolism , Unfolded Protein Response/genetics , Activating Transcription Factor 4/genetics , Activating Transcription Factor 4/metabolism , Cell Death/drug effects , Cell Death/genetics , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/genetics , Chromatin Immunoprecipitation , DNA Helicases/genetics , Gene Expression Regulation/drug effects , Humans , Multifunctional Enzymes/genetics , Nucleic Acid Synthesis Inhibitors/pharmacology , Oxygen/pharmacology , R-Loop Structures/drug effects , RNA Helicases/genetics , RNA-Seq , Unfolded Protein Response/drug effects , Up-Regulation , Zinostatin/pharmacology , eIF-2 Kinase/metabolism
7.
Cell Rep ; 33(2): 108240, 2020 10 13.
Article in English | MEDLINE | ID: mdl-33053351

ABSTRACT

DNA double strand breaks induce oscillatory expression of the transcription factor p53 that is dependent on ataxia telangiectasia mutated (ATM) activity and the rate of double strand break resolution. Although p53 dynamics are known to play a role in the regulation of cell fate determination, the consequences of the variability in dynamics associated with differences in repair rates and utilized repair pathways are unknown. Using single-cell time-lapse microscopy, we found that disruption of specific repair pathways has distinct impacts on p53 dynamics. The small-molecule rucaparib, an inhibitor of the alternative end-joining-associated protein poly (ADP-ribose) polymerase (PARP), increased p53 pulse duration, altering the temporal expression of multiple p53 target genes. As a result, combination treatments of the radiomimetic drug neocarzinostatin with rucaparib drove prolonged growth arrest beyond that of DNA damage alone. This study highlights how pharmacological manipulation of DNA repair pathways may be used to alter p53 dynamics to enhance therapeutic regimens.


Subject(s)
Cell Cycle Checkpoints , DNA Breaks, Double-Stranded , Indoles/pharmacology , Single-Cell Analysis , Tumor Suppressor Protein p53/metabolism , Cell Cycle Checkpoints/drug effects , Cell Proliferation/drug effects , DNA Breaks, Double-Stranded/drug effects , DNA Repair/drug effects , DNA-Activated Protein Kinase , Enzyme Activation/drug effects , Gene Expression Regulation/drug effects , HEK293 Cells , Humans , MCF-7 Cells , Zinostatin/pharmacology
8.
Cells ; 9(9)2020 09 19.
Article in English | MEDLINE | ID: mdl-32961751

ABSTRACT

Cells recovering from the G2/M DNA damage checkpoint rely more on Aurora A-PLK1 signaling than cells progressing through an unperturbed G2 phase, but the reason for this discrepancy is not known. Here, we devised a method based on a FRET reporter for PLK1 activity to sort cells in distinct populations within G2 phase. We employed mass spectroscopy to characterize changes in protein levels through an unperturbed G2 phase and validated that ATAD2 levels decrease in a proteasome-dependent manner. Comparing unperturbed cells with cells recovering from DNA damage, we note that at similar PLK1 activities, recovering cells contain higher levels of Cyclin B1 and increased phosphorylation of CDK1 targets. The increased Cyclin B1 levels are due to continuous Cyclin B1 production during a DNA damage response and are sustained until mitosis. Whereas partial inhibition of PLK1 suppresses mitotic entry more efficiently when cells recover from a checkpoint, partial inhibition of CDK1 suppresses mitotic entry more efficiently in unperturbed cells. Our findings provide a resource for proteome changes during G2 phase, show that the mitotic entry network is rewired during a DNA damage response, and suggest that the bottleneck for mitotic entry shifts from CDK1 to PLK1 after DNA damage.


Subject(s)
CDC2 Protein Kinase/genetics , Cell Cycle Proteins/genetics , Fibroblasts/metabolism , G2 Phase Cell Cycle Checkpoints/genetics , M Phase Cell Cycle Checkpoints/genetics , Mitosis/drug effects , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins/genetics , ATPases Associated with Diverse Cellular Activities/genetics , ATPases Associated with Diverse Cellular Activities/metabolism , Aurora Kinase A/genetics , Aurora Kinase A/metabolism , CDC2 Protein Kinase/metabolism , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cyclin B1/genetics , Cyclin B1/metabolism , DNA Damage , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Fibroblasts/cytology , Fibroblasts/drug effects , Flow Cytometry , Fluorescence Resonance Energy Transfer , G2 Phase Cell Cycle Checkpoints/drug effects , Gene Expression Regulation , Humans , M Phase Cell Cycle Checkpoints/drug effects , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Signal Transduction , Tumor Suppressor p53-Binding Protein 1/genetics , Tumor Suppressor p53-Binding Protein 1/metabolism , Zinostatin/pharmacology , Polo-Like Kinase 1
9.
Chemistry ; 26(63): 14351-14358, 2020 Nov 11.
Article in English | MEDLINE | ID: mdl-32533610

ABSTRACT

A 2-naphthol derivative 2 corresponding to the aromatic ring moiety of neocarzinostatin chromophore was found to degrade proteins under photo-irradiation with long-wavelength UV light without any additives under neutral conditions. Structure-activity relationship studies of the derivative revealed that methylation of the hydroxyl group at the C2 position of 2 significantly suppressed its photodegradation ability. Furthermore, a purpose-designed synthetic tumor-related biomarker, a H2 O2 -activatable photosensitizer 8 possessing a H2 O2 -responsive arylboronic ester moiety conjugated to the hydroxyl group at the C2 position of 2, showed significantly lower photodegradation ability compared to 2. However, release of the 2 from 8 by reaction with H2 O2 regenerated the photodegradation ability. Compound 8 exhibited selective photo-cytotoxicity against high H2 O2 -expressing cancer cells upon irradiation with long-wavelength UV light.


Subject(s)
Naphthols , Proteins , Zinostatin/analogs & derivatives , Animals , Cell Line, Tumor , Cell Survival/drug effects , Hydrogen Peroxide/chemistry , Mice , Naphthols/chemistry , Photosensitizing Agents/chemistry , Photosensitizing Agents/toxicity , Proteins/drug effects , Proteins/radiation effects , Zinostatin/chemistry , Zinostatin/toxicity
10.
J Cell Biol ; 218(4): 1282-1297, 2019 04 01.
Article in English | MEDLINE | ID: mdl-30745421

ABSTRACT

In response to DNA damage, the transcription factor p53 accumulates in a series of pulses. While p53 dynamics play a critical role in regulating stress responses, how p53 pulsing affects target protein expression is not well understood. Recently, we showed that p53 pulses generate diversity in target mRNA expression dynamics; however, given that mRNA and protein expression are not necessarily well correlated, it remains to be determined how p53 pulses impact target protein expression. Using computational and experimental approaches, we show that target protein decay rates filter p53 pulses: Distinct target protein expression dynamics are generated depending on the relationship between p53 pulse frequency and target mRNA and protein stability. Furthermore, by mutating the targets MDM2 and PUMA to alter their stabilities, we show that downstream pathways are sensitive to target protein decay rates. This study delineates the mechanisms by which p53 dynamics play a crucial role in orchestrating the timing of events in the DNA damage response network.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Breast Neoplasms/metabolism , DNA Breaks, Double-Stranded , Proto-Oncogene Proteins c-mdm2/metabolism , Proto-Oncogene Proteins/metabolism , Tumor Suppressor Protein p53/metabolism , Apoptosis Regulatory Proteins/genetics , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Female , Gene Expression Regulation, Neoplastic , Humans , Kinetics , MCF-7 Cells , Models, Biological , Mutation , Protein Stability , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-mdm2/genetics , RNA Stability , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction , Tumor Suppressor Protein p53/genetics , Zinostatin/pharmacology
11.
Nucleic Acids Res ; 46(17): 8926-8939, 2018 09 28.
Article in English | MEDLINE | ID: mdl-30113698

ABSTRACT

The Artemis nuclease and tyrosyl-DNA phosphodiesterase (TDP1) are each capable of resolving protruding 3'-phosphoglycolate (PG) termini of DNA double-strand breaks (DSBs). Consequently, both a knockout of Artemis and a knockout/knockdown of TDP1 rendered cells sensitive to the radiomimetic agent neocarzinostatin (NCS), which induces 3'-PG-terminated DSBs. Unexpectedly, however, a knockdown or knockout of TDP1 in Artemis-null cells did not confer any greater sensitivity than either deficiency alone, indicating a strict epistasis between TDP1 and Artemis. Moreover, a deficiency in Artemis, but not TDP1, resulted in a fraction of unrepaired DSBs, which were assessed as 53BP1 foci. Conversely, a deficiency in TDP1, but not Artemis, resulted in a dramatic increase in dicentric chromosomes following NCS treatment. An inhibitor of DNA-dependent protein kinase, a key regulator of the classical nonhomologous end joining (C-NHEJ) pathway sensitized cells to NCS, but eliminated the sensitizing effects of both TDP1 and Artemis deficiencies. These results suggest that TDP1 and Artemis perform different functions in the repair of terminally blocked DSBs by the C-NHEJ pathway, and that whereas an Artemis deficiency prevents end joining of some DSBs, a TDP1 deficiency tends to promote DSB mis-joining.


Subject(s)
DNA End-Joining Repair , DNA/genetics , Endonucleases/genetics , Epistasis, Genetic , Nuclear Proteins/genetics , Phosphoric Diester Hydrolases/genetics , Cell Survival/drug effects , Cytotoxins/pharmacology , DNA/chemistry , DNA/metabolism , DNA Breaks, Double-Stranded , DNA-Binding Proteins , Endonucleases/antagonists & inhibitors , Endonucleases/deficiency , HCT116 Cells , HEK293 Cells , Humans , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/deficiency , Nucleic Acid Synthesis Inhibitors/pharmacology , Phosphoric Diester Hydrolases/deficiency , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Tumor Suppressor p53-Binding Protein 1/genetics , Tumor Suppressor p53-Binding Protein 1/metabolism , Zinostatin/pharmacology
12.
DNA Repair (Amst) ; 68: 12-24, 2018 08.
Article in English | MEDLINE | ID: mdl-29807321

ABSTRACT

Polynucleotide kinase/phosphatase (PNKP) has been implicated in non-homologous end joining (NHEJ) of DNA double-strand breaks (DSBs). To assess the consequences of PNKP deficiency for NHEJ of 3'-phosphate-ended DSBs, PNKP-deficient derivatives of HCT116 and of HeLa cells were generated using CRISPR/CAS9. For both cell lines, PNKP deficiency conferred sensitivity to ionizing radiation as well as to neocarzinostatin (NCS), which specifically induces DSBs bearing protruding 3'-phosphate termini. Moreover, NCS-induced DSBs, detected as 53BP1 foci, were more persistent in PNKP -/- HCT116 cells compared to their wild-type (WT) counterparts. Surprisingly, PNKP-deficient whole-cell and nuclear extracts were biochemically competent in removing both protruding and recessed 3'-phosphates from synthetic DSB substrates, albeit much less efficiently than WT extracts, suggesting an alternative 3'-phosphatase. Measurements by ligation-mediated PCR showed that PNKP-deficient HeLa cells contained significantly more 3'-phosphate-terminated and fewer 3'-hydroxyl-terminated DSBs than parental cells 5-15 min after NCS treatment, but this difference disappeared by 1 h. These results suggest that, despite presence of an alternative 3'-phosphatase, loss of PNKP significantly sensitizes cells to 3'-phosphate-terminated DSBs, due to a 3'-dephosphorylation defect.


Subject(s)
DNA Breaks, Double-Stranded , DNA End-Joining Repair , DNA Repair Enzymes/genetics , Phosphotransferases (Alcohol Group Acceptor)/genetics , DNA/drug effects , DNA/metabolism , DNA/radiation effects , DNA Repair Enzymes/metabolism , Gene Knockdown Techniques , HCT116 Cells , HeLa Cells , Humans , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Radiation, Ionizing , Zinostatin/toxicity
13.
Sci Rep ; 8(1): 4153, 2018 03 07.
Article in English | MEDLINE | ID: mdl-29515139

ABSTRACT

Monocyte-derived macrophages (MDMs) are an important target for HIV-1 despite SAMHD1, a myeloid restriction factor for which HIV-1 lacks a counteracting accessory protein. The antiviral activity of SAMHD1 is modulated by phosphorylation of T592 by cyclin-dependent kinases (CDK). We show that treatment of MDMs with neocarzinostatin, a compound that introduces double strand breaks (DBS) in genomic DNA, results in the decrease of phosphorylated SAMHD1, activating its antiviral activity and blocking HIV-1 infection. The effect was specific for DSB as DNA damage induced by UV light irradiation did not affect SAMHD1 phosphorylation and did not block infection. The block to infection was at reverse transcription and was counteracted by Vpx, demonstrating that it was caused by SAMHD1. Neocarzinostatin treatment also activated an innate immune response that induced interferon-stimulated genes but this was not involved in the block to HIV-1 infection, as it was not relieved by an interferon-blocking antibody. In response to Neocarzinostatin-induced DNA damage, the level of the CDK inhibitor p21cip1 increased which could account for the decrease of phosphorylated SAMHD1. The results show that the susceptibility of MDMs to HIV-1 infection can be affected by stimuli that alter the phosphorylation state of SAMHD1, one of which is the DNA damage response.


Subject(s)
DNA Damage , HIV Infections/immunology , HIV-1/growth & development , Immunity, Innate , Macrophages/immunology , SAM Domain and HD Domain-Containing Protein 1/immunology , Female , HEK293 Cells , HIV Infections/pathology , Humans , Macrophages/pathology , Macrophages/virology , Male , Phosphorylation , SAM Domain and HD Domain-Containing Protein 1/genetics , Ultraviolet Rays/adverse effects , Zinostatin/pharmacology
14.
Anal Biochem ; 536: 78-89, 2017 11 01.
Article in English | MEDLINE | ID: mdl-28827125

ABSTRACT

We have developed a new method for estimating the localization of DNA damage such as apurinic/apyrimidinic sites (APs) on DNA using fluorescence anisotropy. This method is aimed at characterizing clustered DNA damage produced by DNA-damaging agents such as ionizing radiation and genotoxic chemicals. A fluorescent probe with an aminooxy group (AlexaFluor488) was used to label APs. We prepared a pUC19 plasmid with APs by heating under acidic conditions as a model for damaged DNA, and subsequently labeled the APs. We found that the observed fluorescence anisotropy (robs) decreases as averaged AP density (λAP: number of APs per base pair) increases due to homo-FRET, and that the APs were randomly distributed. We applied this method to three DNA-damaging agents, 60Co γ-rays, methyl methanesulfonate (MMS), and neocarzinostatin (NCS). We found that robs-λAP relationships differed significantly between MMS and NCS. At low AP density (λAP < 0.001), the APs induced by MMS seemed to not be closely distributed, whereas those induced by NCS were remarkably clustered. In contrast, the AP clustering induced by 60Co γ-rays was similar to, but potentially more likely to occur than, random distribution. This simple method can be used to estimate mutagenicity of ionizing radiation and genotoxic chemicals.


Subject(s)
DNA Damage , DNA/drug effects , Fluorescence Polarization/methods , Cobalt Radioisotopes/pharmacology , DNA/chemistry , Fluorescent Dyes/chemistry , Gamma Rays , Mesylates/pharmacology , Mutagens , Zinostatin/pharmacology
15.
Anticancer Res ; 37(7): 3615-3629, 2017 07.
Article in English | MEDLINE | ID: mdl-28668853

ABSTRACT

BACKGROUND/AIM: The aim of this study was to investigate the role of Neocarzinostatin (NCS) conjugated with epithelial cell adhesion molecule (EpCAM) aptamer in EpCAM-positive cancer cells. NCS is an antitumor antibiotic protein chromophore that has the ability to cleave double stranded DNA and can be used as a potential drug for the treatment of EpCAM-positive cancers. EpCAM aptamer is an oligonucleotide ligand that binds specifically to EpCAM, a protein overexpressed in tumor cells. MATERIALS AND METHODS: NCS was conjugated with EpCAM aptamer using Sulfo-Succinimidyl 6-(3-(2-pyridyldithio) - propionamide hexanoate) LC-(SPDP) cross-linker to deliver it to EpCAM-positive tumor cells. The conjugates were characterized using polyacrylamide gel electrophoresis (PAGE) and high-performance liquid chromatography (HPLC). Flow cytometry was used to study the binding efficiency of the aptamer and the conjugates in cancer cells. The effect of the conjugate on cancer cells was studied using propidium iodide (PI) to analyze the cell cycle phase changes. The apoptosis assay was performed using the IC50 concentration of NCS. Microarrays were performed to study the gene level changes in cancer cells upon treatment with NCS and the conjugate. RESULTS: Flow cytometry revealed significant binding of aptamer and conjugate in the MCF-7 and WERI-Rb1 cell lines. Briefly, 62% in MCF and 30% in WERI-Rb1 cells with conjugate treated cells (p<0.005). The cell-cycle analysis indicated G2 phase arrest in MCF-7 cells and S phase arrest in WERI-Rb1 cells (p<0.005). Microarray analysis showed differentially expressed genes involved in cell cycle, DNA damage, and apoptosis. The BrDU assay and the apoptosis assay showed that the expression of BrDU was reduced in conjugate-treated cells and the PARP levels were increased confirming the double stranded DNA breaks (p<0.005). In MCF-7 and WERI-Rb1 cells, most of the cells underwent necrosis (p<0.005). CONCLUSION: The EpCAM aptamer conjugated NCS showed specificity to EpCAM-positive cells. The effect of the conjugates on cancer cells were impressive as the conjugate arrested the cell cycle and promoted apoptosis and necrosis. The high levels of PARP expression confirmed the DNA breaks upon conjugate treatment. Our study demonstrates that the NCS conjugated with EpCAM can be targeted to cancer cells sparing normal cells.


Subject(s)
Epithelial Cell Adhesion Molecule/metabolism , Neoplasms/drug therapy , Zinostatin/pharmacology , Antibiotics, Antineoplastic/pharmacology , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Line, Tumor , DNA Breaks, Double-Stranded/drug effects , DNA Damage/drug effects , Humans , MCF-7 Cells , Neoplasms/metabolism , Oligonucleotides/metabolism
16.
Sci Signal ; 10(476)2017 Apr 25.
Article in English | MEDLINE | ID: mdl-28442631

ABSTRACT

Cellular systems show a wide range of signaling dynamics. Many of these dynamics are highly stereotyped, such as oscillations at a fixed frequency. However, most studies looking at the role of signaling dynamics focus on one or a few cell lines, leaving the diversity of dynamics across tissues or cell lines a largely unexplored question. We focused on the dynamics of the tumor suppressor protein p53, which regulates cell cycle arrest and apoptosis in response to DNA damage. We established live-cell reporters for 12 cancer cell lines expressing wild-type p53 and quantified p53 dynamics in response to double-strand break-inducing DNA damage. In many of the tested cell lines, we found that p53 abundance oscillated in response to ionizing radiation or the DNA-damaging chemotherapeutic neocarzinostatin and that the periodicity of the oscillations was fixed. In other cell lines, p53 abundance dynamically changed in different ways, such as a single broad pulse or a continuous induction. By combining single-cell assays of p53 signaling dynamics, small-molecule screening in live cells, and mathematical modeling, we identified molecules that perturbed p53 dynamics and determined that cell-specific variation in the efficiency of DNA repair and the activity of the kinase ATM (ataxia-telangiectasia mutated) controlled the signaling landscape of p53 dynamics. Because the dynamics of wild-type p53 varied substantially between cell lines, our study highlights the limitation of using one line as a model system and emphasizes the importance of studying the dynamics of other signaling pathways across different cell lines and genetic backgrounds.


Subject(s)
Ataxia Telangiectasia Mutated Proteins/metabolism , Gene Expression Regulation, Neoplastic , Protein Kinase Inhibitors/pharmacology , Tumor Suppressor Protein p53/metabolism , Antibiotics, Antineoplastic/pharmacology , Apoptosis/drug effects , Apoptosis/radiation effects , Ataxia Telangiectasia Mutated Proteins/genetics , Cell Line, Tumor , DNA Damage/drug effects , DNA Damage/radiation effects , DNA Repair/drug effects , DNA Repair/radiation effects , Humans , Mutation , Neoplasms/metabolism , Phosphorylation/drug effects , Phosphorylation/radiation effects , Protein Serine-Threonine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins c-mdm2/metabolism , Radiation, Ionizing , Signal Transduction/drug effects , Signal Transduction/radiation effects , Small Molecule Libraries/pharmacology , Tumor Suppressor Protein p53/genetics , Zinostatin/pharmacology
17.
J Gerontol A Biol Sci Med Sci ; 72(6): 763-770, 2017 Jun 01.
Article in English | MEDLINE | ID: mdl-27573809

ABSTRACT

In order to examine potential differences in genomic stability, we have challenged fibroblasts derived from five different mammalian species of variable longevity with the genotoxic agents, etoposide and neocarzinostatin. We report that cells from longer-lived species exhibit more tumor protein p53 binding protein 1 (53BP1) foci for a given degree of DNA damage relative to shorter-lived species. The presence of a greater number of 53BP1 foci was associated with decreased DNA fragmentation and a lower percentage of cells exhibiting micronuclei. These data suggest that cells from longer-lived species have an enhanced DNA damage response. We propose that the number of 53BP1 foci that form in response to damage reflects the intrinsic capacity of cells to detect and respond to DNA harms.


Subject(s)
DNA Damage , Fibroblasts/metabolism , Longevity , Tumor Suppressor p53-Binding Protein 1/metabolism , Animals , Cattle , Cell Cycle Checkpoints , Cell Line , Chiroptera , Cyclin A/metabolism , Cytotoxins/toxicity , DNA Fragmentation , Dogs , Etoposide/toxicity , Fibroblasts/drug effects , Genomic Instability , Histones/metabolism , Humans , Life Expectancy , Mice , Micronuclei, Chromosome-Defective , Micronucleus Tests , NIMA-Related Kinases/metabolism , Topoisomerase II Inhibitors/toxicity , Zinostatin/toxicity
18.
Biol Pharm Bull ; 39(10): 1623-1630, 2016 Oct 01.
Article in English | MEDLINE | ID: mdl-27431785

ABSTRACT

Neocarzinostatin (NCS) is a member of enediyne antibiotics with high anticancer potential. Our study was performed to explore the synergistic anti-glioma effects of NCS and paclitaxel (PTX) in vitro and in vivo. By 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay, the cytotoxicities of the drugs to human glioma cells U87MG and rat glioma cells C6 were evaluated. The results showed that the combinations of NCS and PTX can synergistically inhibit glioma cells survival. Cell apoptosis was detected by flow cytometry, and the results showed that the combinations of NCS and PTX synergistically enhanced apoptosis ratio of glioma cells. Western blot revealed that the cell signaling pathways of proliferation and apoptosis were synergistically regulated, in which Akt was synergistically inactivated, p53 was up-regulated with down-regulation of bcl-2. Meanwhile, with the subcutaneous model of U87MG cells and intracerebral implantation model of C6 cells, the combination strategy could synergistically delay the glioma growth and significantly prolong the survival of rats bearing orthotopic glioma. This study demonstrates that the combination of NCS and PTX can potentiate the effect on survival and apoptosis of glioma cells via suppression of Akt, bcl-2, and activations of p53; Meanwhile, the in vivo studies also confirmed that the combination of NCS and PTX synergistically inhibit the gliom growth. Our data about the combinational effects of NCS with PTX may provide an alternative strategy for glioma therapy.


Subject(s)
Antibiotics, Antineoplastic/therapeutic use , Antineoplastic Agents, Phytogenic/therapeutic use , Brain Neoplasms/drug therapy , Glioma/drug therapy , Paclitaxel/therapeutic use , Zinostatin/therapeutic use , Animals , Antibiotics, Antineoplastic/pharmacology , Antineoplastic Agents, Phytogenic/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Apoptosis/drug effects , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Synergism , Glioma/pathology , Humans , Male , Mice, Nude , Paclitaxel/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Rats, Wistar , Tumor Burden/drug effects , Tumor Suppressor Protein p53/metabolism , Zinostatin/pharmacology
19.
PLoS One ; 11(3): e0150372, 2016.
Article in English | MEDLINE | ID: mdl-26982737

ABSTRACT

Alterations in DNA damage response and repair have been observed in Huntington's disease (HD). We generated induced pluripotent stem cells (iPSC) from primary dermal fibroblasts of 5 patients with HD and 5 control subjects. A significant fraction of the HD iPSC lines had genomic abnormalities as assessed by karyotype analysis, while none of our control lines had detectable genomic abnormalities. We demonstrate a statistically significant increase in genomic instability in HD cells during reprogramming. We also report a significant association with repeat length and severity of this instability. Our karyotypically normal HD iPSCs also have elevated ATM-p53 signaling as shown by elevated levels of phosphorylated p53 and H2AX, indicating either elevated DNA damage or hypersensitive DNA damage signaling in HD iPSCs. Thus, increased DNA damage responses in the HD genotype is coincidental with the observed chromosomal aberrations. We conclude that the disease causing mutation in HD increases the propensity of chromosomal instability relative to control fibroblasts specifically during reprogramming to a pluripotent state by a commonly used episomal-based method that includes p53 knockdown.


Subject(s)
Gene Knockdown Techniques , Genomic Instability , Huntington Disease/pathology , Induced Pluripotent Stem Cells/pathology , Tumor Suppressor Protein p53/genetics , Adult , Aged , Cells, Cultured , DNA Damage , Humans , Huntington Disease/genetics , Karyotyping , Middle Aged , Nucleic Acid Synthesis Inhibitors/pharmacology , Signal Transduction/drug effects , Young Adult , Zinostatin/pharmacology
20.
Cell Cycle ; 15(4): 546-58, 2016.
Article in English | MEDLINE | ID: mdl-26745237

ABSTRACT

Because low levels of DNA double strand breaks (DSBs) appear not to activate the ATM-mediated prophase I checkpoint in full-grown oocytes, there may exist mechanisms to protect chromosome integrity during meiotic maturation. Using live imaging we demonstrate that low levels of DSBs induced by the radiomimetic drug Neocarzinostatin (NCS) increase the incidence of chromosome fragments and lagging chromosomes but do not lead to APC/C activation and anaphase onset delay. The number of DSBs, represented by γH2AX foci, significantly decreases between prophase I and metaphase II in both control and NCS-treated oocytes. Transient treatment with NCS increases >2-fold the number of DSBs in prophase I oocytes, but less than 30% of these oocytes enter anaphase with segregation errors. MRE11, but not ATM, is essential to detect DSBs in prophase I and is involved in H2AX phosphorylation during metaphase I. Inhibiting MRE11 by mirin during meiotic maturation results in anaphase bridges and also increases the number of γH2AX foci in metaphase II.  Compromised DNA integrity in mirin-treated oocytes indicates a role for MRE11 in chromosome integrity during meiotic maturation.


Subject(s)
DNA Damage/genetics , DNA Repair Enzymes/genetics , DNA-Binding Proteins/genetics , Meiosis/genetics , Oocytes/metabolism , Animals , Ataxia Telangiectasia Mutated Proteins/genetics , DNA Breaks, Double-Stranded/drug effects , DNA Damage/drug effects , Histones/genetics , MRE11 Homologue Protein , Metaphase/genetics , Mice , Oocytes/growth & development , Zinostatin/administration & dosage
SELECTION OF CITATIONS
SEARCH DETAIL
...