Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 68
Filter
1.
New Phytol ; 242(2): 507-523, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38362849

ABSTRACT

Polyploidization is a major event driving plant evolution and domestication. However, how reshaped epigenetic modifications coordinate gene transcription to generate phenotypic variations during wheat polyploidization is currently elusive. Here, we profiled transcriptomes and DNA methylomes of two diploid wheat accessions (SlSl and AA) and their synthetic allotetraploid wheat line (SlSlAA), which displayed elongated root hair and improved root capability for nitrate uptake and assimilation after tetraploidization. Globally decreased DNA methylation levels with a reduced difference between subgenomes were observed in the roots of SlSlAA. DNA methylation changes in first exon showed strong connections with altered transcription during tetraploidization. Homoeolog-specific transcription was associated with biased DNA methylation as shaped by homoeologous sequence variation. The hypomethylated promoters showed significantly enriched binding sites for MYB, which may affect gene transcription in response to root hair growth. Two master regulators in root hair elongation pathway, AlCPC and TuRSL4, exhibited upregulated transcription levels accompanied by hypomethylation in promoter, which may contribute to the elongated root hair. The upregulated nitrate transporter genes, including NPFs and NRTs, also are significantly associated with hypomethylation, indicating an epigenetic-incorporated regulation manner in improving nitrogen use efficiency. Collectively, these results provided new insights into epigenetic changes in response to crop polyploidization and underscored the importance of epigenetic regulation in improving crop traits.


Subject(s)
DNA Methylation , Tetraploidy , DNA Methylation/genetics , Triticum/genetics , Epigenesis, Genetic , Transcriptome , Gene Expression Regulation, Plant
2.
Langmuir ; 40(2): 1266-1276, 2024 01 16.
Article in English | MEDLINE | ID: mdl-38157426

ABSTRACT

Liquid-liquid phase separation (LLPS) of fused in sarcoma (FUS) has emerged as a fundamental principle underpinning cellular function and malfunction. However, we know little about the FUS phase transition process from individual molecules to nanoscale condensates, which plays important roles in neurodegenerative diseases. Here, we propose the fluorescence correlation spectroscopy (FCS) method to quantitatively study the phase separation process of FUS protein with the fluorescent tag-enhanced green fluorescent protein (EGFP), from individual molecules to nanoscale condensates. The characteristic diffusion time (τD) of the protein condensates can be obtained from the FCS curve, which increases with the growth of the protein hydration radius. The bigger the τD value of the protein condensates, the larger the condensates formed by the phase separation of FUS. By this method, we discovered that the critical concentration for FUS to phase separation was 20 nM. We then plotted FUS phase diagrams based on τD under different concentrations of NaCl and found that both low-salt and high-salt concentrations tended to promote FUS-EGFP phase separation. Our results showed that ATP has a good inhibitory effect on FUS phase separation, and its inhibition constant IC50 was 3.2 mM. Finally, we evaluated the inhibition efficiency of single-stranded DNA sequences (ssDNA) on FUS phase separation and demonstrated that ssDNA containing three copies of TCCCCGT had relatively strong inhibition efficiency. In summary, our work provides detailed insight into the FUS phase transition process from individual molecules to nanoscale condensates at nanomolar concentrations and can be exploited for drug screening of neurodegenerative diseases.


Subject(s)
RNA-Binding Protein FUS , Humans , Neurodegenerative Diseases/metabolism , Phase Separation , Spectrum Analysis , RNA-Binding Protein FUS/chemistry
3.
Analyst ; 149(9): 2719-2727, 2024 Apr 29.
Article in English | MEDLINE | ID: mdl-38525957

ABSTRACT

Protein phase separation plays a very important role in many biological processes and is closely related to the occurrence and development of some serious diseases. So far, the fluorescence imaging method and fluorescence correlation spectroscopy (FCS) have been frequently used to study the phase separation behavior of proteins. Due to the wide size distribution of protein condensates in phase separation from nano-scale to micro-scale in solution and living cells, it is difficult for the fluorescence imaging method and conventional FCS to fully reflect the real state of protein phase separation in the solution due to the low spatio-temporal resolution of the conventional fluorescence imaging method and the limited detection area of FCS. Here, we proposed a novel method for studying the protein phase separation process by objective scanning-based fluorescence cross-correlation spectroscopy (Scan-FCCS). In this study, CRDBP proteins were used as a model and respectively fused with fluorescent proteins (EGFP and mCherry). We first compared conventional FCS and Scan-FCS methods for characterizing the CRDBP protein phase separation behaviors and found that the reproducibility of Scan-FCS is significantly improved by the scanning mode. We studied the self-fusion process of mCherry-CRDBP and EGFP-CRDBP and observed that the phase change concentration of CRDBP was 25 nM and the fusion of mCherry-CRDBP and EGFP-CRDBP at 500 nM was completed within 70 min. We studied the effects of salt concentration and molecular crowding agents on the phase separation of CRDBP and found that salt can prevent the self-fusion of CRDBP and molecular crowding agents can improve the self-fusion of CRDBP. Furthermore, we found the recruitment behavior of CRDBP to ß-catenin proteins and studied their recruitment dynamics. Compared to conventional FCS, Scan-FCCS can significantly improve the reproducibility of measurements due to the dramatic increase of detection zone, and more importantly, this method can provide information about self-fusion and recruitment dynamics in protein phase separation.


Subject(s)
Green Fluorescent Proteins , Spectrometry, Fluorescence , Spectrometry, Fluorescence/methods , Green Fluorescent Proteins/chemistry , Luminescent Proteins/chemistry , Recombinant Fusion Proteins/chemistry , Red Fluorescent Protein , Phase Separation
4.
Electrophoresis ; 43(4): 548-558, 2022 02.
Article in English | MEDLINE | ID: mdl-34783369

ABSTRACT

Protein phosphorylation is one of the most basic mechanisms for regulating and controlling protein biological activity and function, and it is also a very important posttranslational modification process. Protein phosphorylation participates in and regulates many life activities such as signal transduction, gene expression, cell cycle, and so on. In this paper, we propose a method for the determination of the protein phosphorylation combining capillary electrophoresis (CE) with ATP analog labeling technique. We synthesized two new ATP analogs (ATP-NB and ATP-TATD-NB) functionalized by norbornene. Using Abl kinase as a model, we established a method for the determination of the kinase activity in solution and lysate by CE with laser-induced fluorescence detection (CE-LIF). This method was used to evaluate the efficiencies of kinase inhibitors. The IC50 values obtained are basically consistent with the reports. By D-A reaction (inverse electron demand Diels-Alder reaction) to label TZ-BODIPY fluorescence, we also realized the phosphorylation fluorescence detection of substrate peptide. Then, we used fluorescence confocal microscopy imaging technology to study the phosphorylation of proteins in vivo by the D-A reaction of ATP-NB and TZ-BODIPY. Our preliminary results documented that the combination of CE-LIF with analog ATP-NB labeling technique is an effective strategy for the determination of the protein phosphorylation and the kinase activity and for screening of kinase inhibitors. The D-A reaction of ATP-NB and TZ-BODIPY also laid the foundation for the subsequent in situ study of protein phosphorylation.


Subject(s)
Electrophoresis, Capillary , Proteins , Adenosine Triphosphate/metabolism , Cycloaddition Reaction , Electrophoresis, Capillary/methods , Phosphorylation , Proteins/metabolism
5.
Analyst ; 147(7): 1357-1366, 2022 Mar 28.
Article in English | MEDLINE | ID: mdl-35253833

ABSTRACT

The identification of molecular targets for achieving beneficial effects from small-molecule drugs is a crucial and currently unsolved challenge, which leads to high costs and long development cycles. Therefore, it is urgent to develop methods for easily and quickly acquiring information about protein-drug interaction at a molecular level. In this study, we propose a novel method for the study of protein-drug interaction by fluorescence correlation spectroscopy (FCS) based on organic solvent-induced protein aggregation. We used ß-secretase (BACE-1) and dihydrofolate reductase (DHFR) as model proteins. Fluorescence-labelled proteins aggregated in aqueous solutions containing organic solvents. In the presence of drugs, the aggregation of proteins was inhibited greatly, and FCS was used to characterize protein aggregates. The decrease in the characteristic diffusion time (τD) of protein aggregates demonstrated a strong interaction between proteins and drug molecules. We presented a new parameter IC50 to assess the inhibitory effects of drugs on the basis of the changes in the τD of fluorescence-labelled proteins under different concentrations of the drugs in the presence of organic solvents. We acquired a remarkable difference in the IC50 values for different drugs and in terms of the trend, our results were consistent with those reported by other methods. Compared with current methods, our approach is simple, low-cost, and time-saving, and has the potential to become a promising and universal tool for drug screening at the molecular level.


Subject(s)
Protein Aggregates , Proteins , Drug Interactions , Solvents/chemistry , Spectrometry, Fluorescence/methods
6.
Biochemistry ; 60(19): 1498-1505, 2021 05 18.
Article in English | MEDLINE | ID: mdl-33870698

ABSTRACT

Protein oligomerization plays a very important role in many physiological processes. p53 acts as a key tumor suppressor by regulating cell cycle arrest, DNA repair, and apoptosis, and its antitumor activity is regulated by the hetero- and homo-oligomerization of MDMX and MDM2 proteins. So far, some traditional methods have been utilized to study the oligomerization of MDMX and MDM2 in vitro, but they have not clarified some controversial issues or whether the extracellular results can represent the intracellular results. Here, we put forward an in situ method for studying protein homo- and hetero-oligomerization in single living cells by using fluorescence correlation spectroscopy. In this study, MDMX and MDM2 were labeled with fluorescent proteins using lentiviral transfection. Autocorrelation spectroscopy and cross-correlation spectroscopy methods were used to study the oligomerization of MDMX and MDM2 in situ and the effect of regulation of MDMX oligomerization on p53-MDMX interactions in single living cells. We observed the homo- and hetero-oligomerization of MDMX and MDM2 in living cells. Meanwhile, the levels of the homo-oligomers of MDMX and MDM2 were increased due to the lack of hetero-oligomerization. Finally, the binding affinity of MDMX for p53 was improved with an increase in the level of MDMX hetero-oligomerization.


Subject(s)
Cell Cycle Proteins/metabolism , Proto-Oncogene Proteins c-mdm2/metabolism , Proto-Oncogene Proteins/metabolism , Apoptosis , Cell Cycle Proteins/physiology , Fluorescence , Humans , In Situ Hybridization, Fluorescence/methods , Nuclear Proteins/metabolism , Protein Binding , Proto-Oncogene Proteins/physiology , Proto-Oncogene Proteins c-mdm2/physiology , Single-Cell Analysis , Spectrometry, Fluorescence/methods , Tumor Suppressor Protein p53/metabolism
7.
Anal Chem ; 93(5): 3042-3051, 2021 02 09.
Article in English | MEDLINE | ID: mdl-33502862

ABSTRACT

Chemiluminescence (CL) resonance energy transfer (CRET) has received great attention due to its fascinating applications in in vivo imaging and photodynamic therapy. Here, we report a highly efficient CRET polymer dot (CRET-Pdots)-based system using catalytic CL reagents as energy donors and fluorescent polymers and dyes as energy acceptors. CRET-Pdots consist of Fe(III) deuteroporphyrin IX (CL catalyst), fluorescent polymers, and dyes. The CL intensity and duration are markedly enhanced by using ultrasensitive catalytic CL reaction of luminol analogue-H2O2, and the CL emission wavelength can be adjusted by one-step/two-step energy-transfer strategies. CRET-Pdots show intensive multicolor CL (about 3000× enhanced), an adjustable emission wavelength (470-720 nm), long CL duration (over 8 h), and a high CRET efficiency (50%). CRET-Pdots possess excellent biocompatibility, sensitive response to reactive oxygen species (ROS), and ultrahigh catalytic activity. They are successfully used for high-contrast real-time ROS imaging and in vivo tumor-targeted imaging with an excellent signal-to-noise ratio (over 90).


Subject(s)
Ferric Compounds , Hydrogen Peroxide , Energy Transfer , Luminescence , Luminescent Measurements , Luminol
8.
Anal Chem ; 93(2): 886-894, 2021 01 19.
Article in English | MEDLINE | ID: mdl-33393764

ABSTRACT

Protein oligomerization and protein-protein interaction are crucial to regulate protein functions and biological processes. p73 protein is a very important transcriptional factor and can promote apoptosis and cell cycle arrest, and its transcriptional activity is regulated by p73 oligomerization and p73-MDM2 interaction. Although extracellular studies on p73 oligomerization and p73-MDM2 interaction have been carried out, it is unclear how p73 oligomerization and p73-MDM2 interaction occur in living cells. In our study, we described an in situ method for studying p73 oligomerization and p73-MDM2 interaction in living cells by combining fluorescence cross-correlation spectroscopy with a fluorescent protein labeling technique. Lentiviral transfection was used to transfect cells with a plasmid for either p73 or MDM2, each fused to a different fluorescent protein. p73 oligomerization was evaluated using brightness per particle, and the p73-MDM2 interaction was quantified using the cross-correlation value. We constructed a series of p73 mutants in three domains (transactivation domain, DNA binding domain, and oligomerization domain) and MDM2 mutants. We systematically studied p73 oligomerization and the effects of p73 oligomerization and the p73 and MDM2 structures on the p73-MDM2 interaction in single living cells. We have found that the p73 protein can form oligomers and that the p73 structure changes in the oligomerization domain significantly influence its oligomerization. p73 oligomerization and the structure changes significantly affect the p73-MDM2 interaction. Furthermore, the effects of inhibitors on p73 oligomerization and p73-MDM2 interaction were studied.


Subject(s)
Proto-Oncogene Proteins c-mdm2/chemistry , Single Molecule Imaging , Tumor Protein p73/analysis , Humans , Tumor Cells, Cultured
9.
Analyst ; 146(14): 4506-4514, 2021 Jul 21.
Article in English | MEDLINE | ID: mdl-34190230

ABSTRACT

Protein phosphorylation is a very important mechanism for regulating and controlling the activity and function of proteins, and is closely associated with signal transduction, gene expression, cell cycle and other life activities in organisms. In this paper, we proposed a new strategy for studying protein phosphorylation in living cells by combining fluorescence resonance energy transfer (FRET) with a small molecule adenosine 5'-triphosphate (ATP) analogue. We synthesized a new ATP analogue functionalized by norbornene (ATP-NB), and a tetrazine modified fluorescent probe Cyanine3 (TZ-Cy3). Based on the inverse electron demand Diels-Alder (D-A) reaction, ATP-NB phosphorylated proteins in solution and in living cells were in situ labelled with TZ-Cy3. By combining FRET with fluorescence correlation spectroscopy (FRET-FCS) and imaging technology, we established an efficient method for studying the phosphorylation of proteins in solution and in living cells using an ATP analogue instead of natural ATP. We studied the effects of phosphatase inhibitors on the phosphorylation of proteins in living cells. Our results documented that ATP-NB is a small molecule ATP analogue with hydrophobicity, which can penetrate cells and efficiently phosphorylate proteins in living cells. This strategy is well suitable for in situ study of protein phosphorylation in living cells.


Subject(s)
Adenosine Triphosphate , Fluorescence Resonance Energy Transfer , Adenosine Triphosphate/metabolism , Fluorescent Dyes , Phosphorylation , Spectrometry, Fluorescence
10.
Analyst ; 146(8): 2581-2590, 2021 Apr 26.
Article in English | MEDLINE | ID: mdl-33899064

ABSTRACT

The mitogen-activated protein kinase (MAPK) pathway is a major module for cellular signal transduction. The dysregulation of the MAPK pathway has been involved in the pathogenesis of multiple diseases ranging from cancers to chronic inflammations. So far, we have not fully understood the influences of external factors and signaling networks on the MAPK pathway due to the lack of in situ methods for simultaneous detection of multiple kinases in the pathway in living cells. Herein, we present a new strategy for in situ and simultaneously monitoring MAPK pathway kinases in single living cells combining multi-channel fluorescence correlation spectroscopy (FCS) with affinity fluorescent probes. We chose rapidly growing fibrosarcoma kinase (RAF), mitogen-activated protein kinase (MEK), and extracellular signal-regulated kinase (ERK) as representative members in the MAPK pathway. We designed and synthesized three fluorescent affinity probes and experimental results demonstrated that the three probes specifically targeted endogenous BRAF, MEK1/2, and ERK1/2 in living cells. Based on the multi-channel FCS system, we studied the influences of biological substances, drugs and oxidative stress on the activities of endogenous MAPK kinases and the cross-talk between the MAPK and PI3K-mTOR pathways. We have found that serum, sorafenib, and hydrogen peroxide can regulate multiple MAPK kinases and the effects of external stimuli can transmit to the MAPK pathway; furthermore, we have observed that the MAPK pathway can be activated by modulating the PI3K-mTOR pathway. Our results illustrated the complexity of a cellular signal network and the necessity of in situ and simultaneous determination of biomolecules in living cells.


Subject(s)
Extracellular Signal-Regulated MAP Kinases , Mitogen-Activated Protein Kinase Kinases , Spectrum Analysis , Extracellular Signal-Regulated MAP Kinases/metabolism , Mitogen-Activated Protein Kinase Kinases/metabolism , Signal Transduction
11.
Analyst ; 146(2): 478-486, 2021 Jan 21.
Article in English | MEDLINE | ID: mdl-33174562

ABSTRACT

Protein expression is closely related to many biological processes including cell growth, differentiation and signaling. It is a challenge to selectively monitor newly synthesized proteins under both physiological and pathological conditions due to shortage of efficient analytical methods. Here, we proposed a new strategy to selectively monitor newly synthesized proteins in cells by combining fluorescence correlation spectroscopy (FCS) with bioorthogonal noncanonical amino acid tagging (BONCAT) technique. Firstly, homopropargylglycine (HPG), an alkyne surrogate of methionine, was metabolically incorporated into newly synthesized proteins in living cells, and the proteins containing the alkyne functional group were subsequently labeled with chemoselective fluorescence reporters using the Cu(i)-catalyzed azide-alkyne cycloaddition (CuAAC) reaction. Then, FCS was used to analyze the newly synthesized proteins based on the difference in the characteristic diffusion times of labeled proteins and free fluorescent dyes. We optimized the conditions of HPG metabolic incorporation and the CuAAC click reaction and applied this new method to study autophagic protein degradation and in situ monitor secreted proteins in cells. Compared to current methods, our method is simple, fast, and without separation, and it may become a promising approach for in situ studying protein expression in living cells.


Subject(s)
Amino Acids/chemistry , Protein Biosynthesis , Proteins/chemistry , Alkynes/chemistry , Autophagy , Cell Line, Tumor , Glycine/analogs & derivatives , Glycine/chemistry , Humans , Proteins/metabolism , Proteolysis , Spectrometry, Fluorescence
12.
Analyst ; 146(11): 3623-3632, 2021 Jun 07.
Article in English | MEDLINE | ID: mdl-33929479

ABSTRACT

DNA methylation is a critical part of epigenetics and plays a vital role in maintaining normal cell function, genetic imprinting, and human tumorigenesis. Thus, it is important to develop a sensitive method for the determination of DNA methyltransferase (MTase) activity. Here, we present a simple and sensitive method based on single molecule fluorescence correlation spectroscopy (FCS) and polystyrene polymer dots (PS Pdots) for the quantitative detection of DNA adenine methylation (Dam) MTase activity and its inhibitor screening in homogeneous solution without separation. Its principle is based on the measurement of the characteristic diffusion time (τD) of unmethylated and methylated DNA-fluorescent probes by FCS. A hairpin DNA probe including the 5'-GATC-3' sequence is used by doubly labelling fluorophore Alexa Fluor 488 (Alexa 488) and biotin at the 5'- and 3'-terminus, respectively. Dam MTase catalyzed the methylation of the sequence of 5'-GATC-3', and DpnI cleaved the sequence of 5'-G-Am-TC-3'. Streptavidin conjugated PS Pdots were used to react with DNA probes without methylation to further increase the difference in τD values between methylated and unmethylated DNA-Alexa 488 probes. We used the FCS method to measure the τD values of DNA-Alexa 488 probes and further obtained the activity of Dam MTase. It is found that the τD value of the methylated DNA probe is negatively correlated with the logarithm of Dam MTase concentration in the range from 0.025 U mL-1 to 3 U mL-1. The detection limit is as low as 0.025 U mL-1. Furthermore, we evaluated the inhibition effect of drug-related DNA methylation and the half-maximal inhibitory concentration (IC50) value is consistent with a previous study. The results demonstrated that our proposed method will become a promising platform for the determination of Dam MTase activity and inhibitor screening.


Subject(s)
Biosensing Techniques , Site-Specific DNA-Methyltransferase (Adenine-Specific) , DNA/genetics , DNA Methylation , Humans , Polymers , Polystyrenes , Site-Specific DNA-Methyltransferase (Adenine-Specific)/genetics , Site-Specific DNA-Methyltransferase (Adenine-Specific)/metabolism
13.
Anal Chem ; 92(10): 7020-7027, 2020 05 19.
Article in English | MEDLINE | ID: mdl-32340449

ABSTRACT

The drug-target protein interaction is the basis of drug screening and precise therapy in modern clinical medicine. How to acquire the information about the drug-target protein interaction in single living cell is a great challenge due to the shortage of efficient methods. Here we propose a new strategy for in situ studying the drug-target protein interaction in single living cells based on the competition of candidate drugs to the fluorescent probe-target complexes and fluorescence correlation spectroscopy (FCS) with a microfluidic chip. In this study, we used ABL kinase (target) as a model and synthesized a fluorescent probe (Cy3-dasatinib) with an affinity to the target using ABL inhibitor dasatinib as a precursor. We systematically investigated the association of the probe with targets and the dissociation of the drug-target complexes in the presence of candidate drug. We presented a new parameter IC50 (τD) to assess the inhibitory effect of drugs on the basis of the changes in the characteristic diffusion time (τD) and the binding ratio (y) of fluorescent probes during the drug competition process in living cells. We found a remarkable difference of IC50 (τD) values in living cells and in solutions, suggesting it is quite necessary to evaluate the drug-target interactions in living cells. Compared with current methods, our approach can be used to in situ and real-time study the drug-target interaction in living cells, and it may become a promising and universal tool for in situ drug research at molecular level.


Subject(s)
Antineoplastic Agents/chemistry , Carbocyanines/chemistry , Dasatinib/chemistry , Fluorescent Dyes/chemistry , Protein Kinase Inhibitors/chemistry , Proto-Oncogene Proteins c-abl/antagonists & inhibitors , Antineoplastic Agents/pharmacology , Dasatinib/pharmacology , Humans , Molecular Structure , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-abl/metabolism , Spectrometry, Fluorescence
14.
Anal Chem ; 92(4): 2988-2996, 2020 02 18.
Article in English | MEDLINE | ID: mdl-32003968

ABSTRACT

mRNA-protein interactions play key roles in facilitating various biological functions in gene expression regulations and even the progression of diseases. However, it is still a challenge to directly monitor mRNA-protein interactions in a single living cell at present. Herein, we propose a new strategy for real-time studying of mRNA-protein interactions in a single living cell using fluorescence cross-correlation spectroscopy (FCCS) and molecular beacon (MB) labeling techniques. The c-myc mRNA and coding region determinant binding protein (CRDBP) were used as models. We first evaluated the performances of unmodified (2'-deoxy) and modified (2'-O-methyl) MBs and found that the 2'-O-methyl loop MB (2'-O-methyl loop domain, 2'-deoxy stem region) has high affinity to target mRNA and good nuclease resistance. Then we constructed stable cell line expressing mCherry-CRDBP using lentivirus infection, and on the basis of FCCS, we established an efficient method for quantifying the interaction of c-myc mRNA with CRDBP in a single living cell. The RNA binding domains of CRDBP cover two RNA recognition motifs (RRM) and four K homologies (KH). Furthermore, we constructed the truncated variants and point mutants on RNA binding domains of CRDBP, systematically studied the effects of RNA binding domains of CRDBP on c-myc mRNA-CRDBP interaction in living cells, and found that KH3-4 is indispensable for c-myc mRNA binding, KH1-2 plays a supplementary role, and RRM1-2 shows no binding ability to c-myc mRNA. Our work reveals the mechanisms of c-myc mRNA-CRDBP interactions and provides a general strategy for quantifying the interactions of endogenous mRNA with protein in a single living cell.


Subject(s)
Proto-Oncogene Proteins c-myc/chemistry , RNA, Messenger/chemistry , RNA-Binding Proteins/chemistry , Humans , Open Reading Frames/genetics , Proto-Oncogene Proteins c-myc/genetics , RNA, Messenger/genetics , RNA-Binding Proteins/genetics , Spectrometry, Fluorescence , Tumor Cells, Cultured
15.
Zhongguo Dang Dai Er Ke Za Zhi ; 22(8): 909-915, 2020 Aug.
Article in Zh | MEDLINE | ID: mdl-32800041

ABSTRACT

OBJECTIVE: To study the regulatory mechanism of MS275, a histone deacetylase inhibitor, on the p38 MAPK signaling pathway in rats with convulsion in the developmental stage. METHODS: Thirty-two male rats were randomly divided into four groups: control, pentylenetetrazol (PTZ), PTZ+3 mg/kg MS275, and PTZ+6 mg/kg MS275 (n=8 each). A rat model of convulsion in the developmental stage was prepared by an intraperitoneal injection of PTZ. The rats in the control group were given an injection of normal saline alone. MS275 was given by an intraperitoneal injection at 2 hours before PTZ injection. At 24 hours after successful modeling, 6 rats were taken from each group. Western blot and qRT-PCR were used to measure the protein and mRNA expression of p38, MK2, cAMP response element-binding protein (CREB), and interleukin-6 (IL-6) in the hippocampus. Hematoxylin-eosin (HE) staining was used to observe brain pathological changes. Western blot was used to measure the expression of CD11b as a marker for the activation of microglial cells. RESULTS: Compared with the control group, the PTZ group had significant increases in the mRNA and protein expression of p38, MK2, CREB, and IL-6 (P<0.05). MS275 significantly inhibited the mRNA and protein expression of the above markers in the rats with convulsion in the developmental stage (P<0.05), and 6 mg/kg MS275 had a significantly better inhibitory effect on the mRNA and protein expression of IL-6 and CREB than 3 mg/kg MS275 (P<0.05). HE staining showed that the PTZ group had marked neuron apoptosis, cellular edema, and inflammatory cell infiltration, while MS275 intervention alleviated neuron apoptosis and cellular edema and reduced inflammatory cell infiltration in the rats with convulsion. The PTZ group had a significant increase in the activation of microglial cells, while MS275 significantly inhibited the activation of microglial cells in the rats with convulsion (P<0.05); 6 mg/kg MS275 had a significantly better inhibitory effect than 3 mg/kg MS275 (P<0.05). CONCLUSIONS: In rats with convulsion in the developmental stage, the histone deacetylase inhibitor MS275 can inhibit the p38 MAPK signaling pathway, the apoptosis of hippocampal neurons, and the activation of microglial cells and thus reduce inflammatory response and convulsion-induced brain injury in a dose-dependent manner.


Subject(s)
Seizures , Animals , Male , Pentylenetetrazole , Rats , Rats, Sprague-Dawley , Signal Transduction , p38 Mitogen-Activated Protein Kinases
16.
J Biol Inorg Chem ; 24(2): 179-189, 2019 03.
Article in English | MEDLINE | ID: mdl-30701360

ABSTRACT

The abnormal aggregation and deposition of human islet amyloid polypeptide (hIAPP) are implicated in the pathogeny of type 2 diabetes mellitus (T2DM). Many aromatic ring-containing Ru complexes inhibit the aggregation of hIAPP. A new Ru complex Ru(bipy)(met)2·3H2O (1), where bipy is 2,2'-bipyridine and met is methionine, was synthesized and employed to resist the fibril formation of hIAPP and to promote the biocompatibility of metal complexes. Two polypyridyl Ru complexes, namely [Ru(bipy)3]Cl2(2) and Ru(bipy)2Cl2(3), were used for comparison. Results reveal that the three Ru complexes can inhibit hIAPP aggregation and depolymerize mature hIAPP fibrils. Interaction studies show that Ru complexes bind to hIAPP through metal coordination, hydrophobic interaction, and other intermolecular forces. The binding of the three compounds is spontaneous and exothermic. The compounds also rescue peptide-induced cytotoxicity to some extent. Similar to 3, the novel methionine-Ru complex 1 exhibits an enhanced inhibitory effect and binding affinity to hIAPP possibly because of the smaller steric hindrance and more profitable molecular configuration of 1 than those of 2. The newly designed amino acid-Ru complex may provide new insights into the treatment of T2DM and related amyloidosis diseases. Methionine-Ru complex effectively impedes the fibril formation of human islet amyloid polypeptide.


Subject(s)
Coordination Complexes/pharmacology , Islet Amyloid Polypeptide/antagonists & inhibitors , Methionine/pharmacology , Ruthenium/pharmacology , Animals , Cells, Cultured , Coordination Complexes/chemical synthesis , Coordination Complexes/chemistry , Humans , Islet Amyloid Polypeptide/metabolism , Methionine/chemistry , Molecular Structure , Particle Size , Rats , Ruthenium/chemistry , Surface Properties
17.
Anal Chem ; 90(6): 3871-3877, 2018 03 20.
Article in English | MEDLINE | ID: mdl-29468867

ABSTRACT

Protein tyrosine kinases play a pivotal role in intracellular signal transduction pathways and oncogenic transformation. It is necessary to develop a simple, cost-effective, and sensitive kinase assay for study of protein kinases and discovery of kinase-target drugs. In this paper, we present a simple and sensitive method for homogeneous detection of protein kinase activity and screening of inhibitor by measuring surface charge change on the peptide-modified gold nanoparticles (GNPs) as kinase substrates. In this assay, Abl (Abelson murine leukemia viral oncogene) kinase was used as a model. In the presence of Abl kinase and ATP, the surface negative charge on GNPs significantly increases due to phosphorylation of the peptide-modified GNPs. The surface charge on the peptide-modified GNPs was measured by zeta potential analyzer. Under the optimum conditions, the zeta potential on the peptide-modified GNPs was linearly dependent on Abl kinase concentration, the linear range was from 1 to 40 nM and the detection limit was 1 nM. This method was used to evaluate the inhibition efficiency of inhibitors, and the obtained IC50 values were well in agreement with the results reported in the references. Furthermore, this method was successfully applied to determine Abl kinase activity in the cell lysates. Compared to current methods, this new method shows simplicity, short analysis time, high sensitivity, and will become a promising platform for kinase-related fundamental research and inhibitor screening.


Subject(s)
Enzyme Assays/methods , Gold/chemistry , Metal Nanoparticles/chemistry , Peptides/metabolism , Proto-Oncogene Proteins c-abl/metabolism , Biosensing Techniques/methods , Dasatinib/pharmacology , Drug Evaluation, Preclinical/methods , Humans , K562 Cells , Limit of Detection , Peptides/chemistry , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-abl/analysis , Proto-Oncogene Proteins c-abl/antagonists & inhibitors , Static Electricity , Surface Properties
18.
Anal Chem ; 90(1): 1006-1013, 2018 01 02.
Article in English | MEDLINE | ID: mdl-29211436

ABSTRACT

Telomerase is a key enzyme for maintaining the telomere length and is regarded as a versatile cancer biomarker and a potential drug target due to its important role in cancer and aging. It is necessary to develop a sensitive and reliable method for detection of telomerase activity due to its very low level in cells. In this Article, we propose an ultrasensitive and robust method for quantitative determination of telomerase activity by combining single molecule fluorescence correlation spectroscopy (FCS) with telomerase repeat amplification protocol (TRAP). The principle of this new method (FCS-TRAP) is based on measurement of the change in characteristic diffusion time and molecule number of TRAP products by FCS. The characteristic diffusion time is related to the length of TRAP products, and the molecule number represents the concentration of TRAP products. We optimized the conditions of TRAP procedure and FCS measurements. We observed that the telomerase activities are positively correlated to characteristic diffusion time and molecule number of TRAP products at optimal conditions. This method was successfully used for determination of telomerase activity of different cells, and detection of a single cell was realized. Meanwhile, this method was used to evaluate the inhibition efficiency of inhibitors, and the IC50 values obtained were in good agreement with the references. Compared to current TRAP methods, this method shows reliable quantification, ultrahigh sensitivity, and short detection time and is without separation. We believe that the FCS-TRAP method has a potential application in clinical diagnosis and screening of telomerase inhibitors.


Subject(s)
Polymerase Chain Reaction/methods , Spectrometry, Fluorescence/methods , Telomerase/analysis , Base Sequence , Benzoxazoles/chemistry , DNA/chemistry , Fluorescent Dyes/chemistry , HeLa Cells , Humans , Intercalating Agents/chemistry , Quinolinium Compounds/chemistry
19.
Anal Chem ; 90(11): 6929-6935, 2018 06 05.
Article in English | MEDLINE | ID: mdl-29732881

ABSTRACT

Chemiluminescence (CL) is a promising bioimaging method due to no interferences of light source and autofluorescence. However, compared to fluorescent emission, most CL reactions show short emission time and wavelength and weak emission intensity, which limit their applications in in vivo imaging. Here, we report mimic-enzyme catalytic CL polymer dots (hemin-Pdots) consisting of hemin and fluorescent conjugated polymer based on chemiluminescence resonance energy transfer. Hemin-Pdots show about 700× enhanced CL and over 10 h light emission in the presence of CL substrates and H2O2. These properties are mainly due to high-catalytic activity of hemin-Pdots and slow-diffusion-controlled heterogeneous reaction. Hemin-Pdots also possess excellent biocompatibility, good stability, emission wavelength redshift, and ultrasensitive response to reactive oxygen species (ROS), and they were successfully used for real-time imaging ROS levels in the peritoneal cavity and normal and tumor tissues of mice. Hemin-Pdots as new CL probes have wide applications in bioassays, bioimaging, and photodynamic therapy.


Subject(s)
Luminescence , Optical Imaging , Polymers/chemistry , Quantum Dots/chemistry , Reactive Oxygen Species/analysis , Animals , Catalysis , Cell Line, Tumor , Female , HEK293 Cells , Humans , Mice , Mice, Nude , Neoplasms, Experimental/diagnostic imaging , Neoplasms, Experimental/metabolism , Reactive Oxygen Species/metabolism
20.
Anal Chem ; 90(10): 6144-6151, 2018 05 15.
Article in English | MEDLINE | ID: mdl-29671327

ABSTRACT

Protein-protein interactions play a central role in signal transduction, transcription regulations, enzymatic activity, and protein synthesis. The p53 protein is a key transcription factor, and its activity is precisely regulated by the p53-MDM2 interaction. Although the p53-MDM2 interaction has been studied, it is still not clear how p53 structures and external factors influence the p53-MDM2 interaction in living cells. Here, we developed a direct method for monitoring the p53-MDM2 interaction in single living cells using single-molecule fluorescence cross-correlation spectroscopy with a microfluidic chip. First, we labeled p53 and MDM2 proteins with enhanced green fluorescent protein (EGFP) and mCherry, respectively, using lentivirus infection. We then designed various mutants covering the three main domains of p53 (tetramerization, transactivation, and DNA-binding domains) and systematically studied effects of p53 protein primary, secondary, and quaternary structures on p53-MDM2 binding affinity in single living cells. We found that p53 dimers and tetramers can bind to MDM2, that the binding affinity of p53 tetramers is higher than that of p53 dimers, and that the affinity is closely correlated to the helicity of the p53 transactivation domain. The hot-spot mutation R175H in the DNA-binding domain reduced the binding of p53 to MDM2. Finally, we studied effects of inhibitors on p53-MDM2 interactions and dissociation dynamics of p53-MDM2 complexes in single living cells. We found that inhibitors Nutlin 3α and MI773 efficiently inhibited the p53-MDM2 interaction, but RITA did not work in living cells. This study provides a direct way for quantifying the relationship between protein structure and protein-protein interactions and evaluation of inhibitors in living cells.


Subject(s)
Proto-Oncogene Proteins c-mdm2/chemistry , Single Molecule Imaging , Single-Cell Analysis , Tumor Suppressor Protein p53/chemistry , Cell Survival , Humans , Protein Binding , Spectrometry, Fluorescence , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL