Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Small ; 20(6): e2304743, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37803930

ABSTRACT

Converting solar energy into hydrogen energy using conjugated polymers (CP) is a promising solution to the energy crisis. Improving water solubility plays one of the critical factors in enhancing the hydrogen evolution rate (HER) of CP photocatalysts. In this study, a novel concept of incorporating hydrophilic side chains to connect the backbones of CPs to improve their HER is proposed. This concept is realized through the polymerization of carbazole units bridged with octane, ethylene glycol, and penta-(ethylene glycol) to form three new side-chain-braided (SCB) CPs: PCz2S-OCt, PCz2S-EG, and PCz2S-PEG. Verified through transient absorption spectra, the enhanced capability of PCz2S-PEG for ultrafast electron transfer and reduced recombination effects has been demonstrated. Small- and wide-angle X-ray scattering (SAXS/WAXS) analyses reveal that these three SCB-CPs form cross-linking networks with different mass fractal dimensions (f) in aqueous solution. With the lowest f value of 2.64 and improved water/polymer interfaces, PCz2S-PEG demonstrates the best HER, reaching up to 126.9 µmol h-1 in pure water-based photocatalytic solution. Moreover, PCz2S-PEG exhibits comparable performance in seawater-based photocatalytic solution under natural sunlight. In situ SAXS analysis further reveals nucleation-dominated generation of hydrogen nanoclusters with a size of ≈1.5 nm in the HER of PCz2S-PEG under light illumination.

2.
Cell Mol Gastroenterol Hepatol ; 17(2): 292-308.e1, 2024.
Article in English | MEDLINE | ID: mdl-37820788

ABSTRACT

BACKGROUND & AIMS: Metabolic reprogramming is essential for the activation and functions of macrophages, including bacterial killing and cytokine production. Bromodomain-containing protein 4 (BRD4) has emerged as a critical regulator of innate immune response. However, the potential role of BRD4 in the metabolic reprogramming of macrophage activation upon Helicobacter pylori infection remains unclear. METHODS: Bone marrow-derived macrophages (BMDMs) from wild-type (WT) and Brd4-myeloid deletion conditional knockout (Brd4-CKO) mice were infected with H pylori. RNA sequencing was performed to evaluate the differential gene expression between WT and Brd4-deficient BMDMs upon infection. An in vivo model of H pylori infection using WT and Brd4-CKO mice was used to confirm the role of BRD4 in innate immune response to infection. RESULTS: Depletion of Brd4 in BMDMs showed impaired H pylori-induced glycolysis. In addition, H pylori-induced expression of glycolytic genes, including Slc2a1 and Hk2, was decreased in Brd4-deficient BMDMs. BRD4 was recruited to the promoters of Slc2a1 and Hk2 via hypoxia-inducible factor-1α, facilitating their expression. BRD4-mediated glycolysis stabilized H pylori-induced nitric oxide synthase (Nos2) messenger RNA to produce nitric oxide. The NO-mediated killing of H pylori decreased in Brd4-deficient BMDMs, which was rescued by pyruvate. Furthermore, Brd4-CKO mice infected with H pylori showed reduced gastric inflammation and increased H pylori colonization with reduced inducible NO synthase expression in gastric macrophages. CONCLUSIONS: Our study identified BRD4 as a key regulator of hypoxia-inducible factor-1α-dependent glycolysis and macrophage activation. Furthermore, we show a novel regulatory role of BRD4 in innate immunity through glycolysis to stabilize Nos2 messenger RNA for NO production to eliminate H pylori infection.


Subject(s)
Helicobacter Infections , Helicobacter pylori , Animals , Mice , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Helicobacter Infections/microbiology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Helicobacter pylori/metabolism , Macrophages/metabolism , Nitric Oxide Synthase/metabolism , RNA, Messenger/metabolism , Glycolysis , Nitric Oxide Synthase Type II/metabolism
3.
Cogn Neurodyn ; 16(3): 733-744, 2022 Jun.
Article in English | MEDLINE | ID: mdl-35603055

ABSTRACT

This paper studies the dynamical characteristics of discrete analogue of impulsive recurrent neural networks with both discrete and finite distributed asynchronous time-varying delays. Firstly, the discrete impulsive system of the corresponding continuous-time model is reformed by impulsive maps and semi-discrete method. Secondly, by employing a famous delay impulsive differential inequality, several novel sufficient conditions are derived to ensure the uniqueness of equilibrium point and its global exponential stability in Lagrange sense for the discussed discrete-time impulsive system. Meanwhile, it is illustrated that the discrete-time analogue retains the uniqueness of equilibrium point of the corresponding continuous-time model, and some corollaries follow. Finally, one example is given to demonstrate the validity of our obtained results.

4.
Nucleic Acids Res ; 50(2): 750-762, 2022 01 25.
Article in English | MEDLINE | ID: mdl-34935961

ABSTRACT

P-TEFb modulates RNA polymerase II elongation through alternative interaction with negative and positive regulation factors. While inactive P-TEFbs are mainly sequestered in the 7SK snRNP complex in a chromatin-free state, most of its active forms are in complex with its recruitment factors, Brd4 and SEC, in a chromatin-associated state. Thus, switching from inactive 7SK snRNP to active P-TEFb (Brd4/P-TEFb or SEC/P-TEFb) is essential for global gene expression. Although it has been shown that cellular signaling stimulates the disruption of 7SK snRNP, releasing dephosphorylated and catalytically inactive P-TEFb, little is known about how the inactive released P-TEFb is reactivated. Here, we show that the Cdk9/CycT1 heterodimer released from 7SK snRNP is completely dissociated into monomers in response to stress. Brd4 or SEC then recruits monomerized Cdk9 and CycT1 to reassemble the core P-TEFb. Meanwhile, the binding of monomeric dephosphorylated Cdk9 to either Brd4 or SEC induces the autophosphorylation of T186 of Cdk9. Finally, the same mechanism is employed during nocodazole released entry into early G1 phase of cell cycle. Therefore, our studies demonstrate a novel mechanism by which Cdk9 and CycT1 monomers are reassembled on chromatin to form active P-TEFb by its interaction with Brd4 or SEC to regulate transcription.


Subject(s)
Cell Cycle Proteins/metabolism , Cyclin T/metabolism , Cyclin-Dependent Kinase 9/metabolism , DNA-Binding Proteins/metabolism , Positive Transcriptional Elongation Factor B/metabolism , Ribonucleoproteins, Small Nuclear/metabolism , Transcription Factors/metabolism , Transcriptional Elongation Factors/metabolism , Cell Cycle , Cell Line , Cyclin T/chemistry , Cyclin-Dependent Kinase 9/chemistry , Enzyme Activation , Humans , Models, Biological , Phosphorylation , Protein Binding , Protein Multimerization , Recombinant Proteins , Ribonucleoproteins, Small Nuclear/chemistry , Stress, Physiological
5.
JCI Insight ; 6(7)2021 04 08.
Article in English | MEDLINE | ID: mdl-33830083

ABSTRACT

Macrophage-mediated inflammatory response has been implicated in the pathogenesis of obesity and insulin resistance. Brd4 has emerged as a key regulator in the innate immune response. However, the role of Brd4 in obesity-associated inflammation and insulin resistance remains uncharacterized. Here, we demonstrated that myeloid lineage-specific Brd4 knockout (Brd4-CKO) mice were protected from high-fat diet-induced (HFD-induced) obesity with less fat accumulation, higher energy expenditure, and increased lipolysis in adipose tissue. Brd4-CKO mice fed a HFD also displayed reduced local and systemic inflammation with improved insulin sensitivity. RNA-Seq of adipose tissue macrophages (ATMs) from HFD-fed WT and Brd4-CKO mice revealed that expression of antilipolytic factor Gdf3 was significantly decreased in ATMs of Brd4-CKO mice. We also found that Brd4 bound to the promoter and enhancers of Gdf3 to facilitate PPARγ-dependent Gdf3 expression in macrophages. Furthermore, Brd4-mediated expression of Gdf3 acted as a paracrine signal targeting adipocytes to suppress the expression of lipases and the associated lipolysis in cultured cells and mice. Controlling the expression of Gdf3 in ATMs could be one of the mechanisms by which Brd4 modulates lipid metabolism and diet-induced obesity. This study suggests that Brd4 could be a potential therapeutic target for obesity and insulin resistance.


Subject(s)
Adipose Tissue/cytology , Growth Differentiation Factor 3/genetics , Macrophages/metabolism , Nuclear Proteins/metabolism , Obesity/etiology , Transcription Factors/metabolism , Adipocytes/metabolism , Adipose Tissue/metabolism , Animals , Diet, High-Fat/adverse effects , Energy Metabolism/genetics , Gene Expression Regulation , Growth Differentiation Factor 3/metabolism , Insulin Resistance/genetics , Lipase/genetics , Lipase/metabolism , Lipid Metabolism/physiology , Lipolysis/genetics , Male , Mice, Knockout , Nuclear Proteins/genetics , PPAR gamma/metabolism , Promoter Regions, Genetic , Transcription Factors/genetics
6.
Cell Death Dis ; 11(8): 667, 2020 08 21.
Article in English | MEDLINE | ID: mdl-32820150

ABSTRACT

H. pylori infection is one of the leading causes of gastric cancer and the pathogenicity of H. pylori infection is associated with its ability to induce chronic inflammation and apoptosis resistance. While H. pylori infection-induced expression of pro-inflammatory cytokines for chronic inflammation is well studied, the molecular mechanism underlying the apoptosis resistance in infected cells is not well understood. In this study, we demonstrated that H. pylori infection-induced apoptosis resistance in gastric epithelial cells triggered by Raptinal, a drug that directly activates caspase-3. This resistance resulted from the induction of cIAP2 (encoded by BIRC3) since depletion of BIRC3 by siRNA or inhibition of cIAP2 via BV6 reversed H. pylori-suppressed caspase-3 activation. The induction of cIAP2 was regulated by H. pylori-induced BIRC3 eRNA synthesis. Depletion of BIRC3 eRNA decreased H. pylori-induced cIAP2 and reversed H. pylori-suppressed caspase-3 activation. Mechanistically, H. pylori stimulated the recruitment of bromodomain-containing factor Brd4 to the enhancer of BIRC3 and promoted BIRC3 eRNA and mRNA synthesis. Inhibition of Brd4 diminished the expression of BIRC3 eRNA and the anti-apoptotic response to H. pylori infection. Importantly, H. pylori isogenic cagA-deficient mutant failed to activate the synthesis of BIRC3 eRNA and the associated apoptosis resistance. Finally, in primary human gastric epithelial cells, H. pylori also induced resistance to Raptinal-triggered caspase-3 activation by activating the Brd4-dependent BIRC3 eRNA synthesis in a CagA-dependent manner. These results identify a novel function of Brd4 in H. pylori-mediated apoptosis resistance via activating BIRC3 eRNA synthesis, suggesting that Brd4 could be a potential therapeutic target for H. pylori-induced gastric cancer.


Subject(s)
Apoptosis/physiology , Baculoviral IAP Repeat-Containing 3 Protein/metabolism , Enhancer Elements, Genetic/genetics , Antigens, Bacterial/metabolism , Bacterial Proteins/metabolism , Baculoviral IAP Repeat-Containing 3 Protein/physiology , Caspase 3/metabolism , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Epithelial Cells/metabolism , Gastric Mucosa/metabolism , Helicobacter Infections/metabolism , Helicobacter pylori/pathogenicity , Humans , Nuclear Proteins/metabolism , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Stomach/pathology , Stomach Neoplasms/metabolism , Transcription Factors/metabolism
7.
Biomed Res Int ; 2016: 5173205, 2016.
Article in English | MEDLINE | ID: mdl-27529070

ABSTRACT

In the past decade, much emphasis has been put on the transcriptional activation of HIV-1, which is proposed as a promised strategy for eradicating latent HIV-1 provirus. Two drugs, prostratin and hexamethylene bisacetamide (HMBA), have shown potent effects as inducers for releasing HIV-1 latency when used alone or in combination, although their cellular target(s) are currently not well understood, especially under drug combination. Here, we have shown that HMBA and prostratin synergistically release HIV-1 latency via different mechanisms. While prostratin strongly stimulates HMBA-induced HIV-1 transcription via improved P-TEFb activation, HMBA is capable of boosting NF-κB-dependent transcription initiation by suppressing prostratin-induced expression of the deubiquitinase A20, a negative feedback regulator in the NF-κB signaling pathway. In addition, HMBA was able to increase prostratin-induced phosphorylation and degradation of NF-κB inhibitor IκBα, thereby enhancing and prolonging prostratin-induced nuclear translocation of NF-κB, a prerequisite for stimulation of transcription initiation. Thus, by blocking the negative feedback circuit, HMBA functions as a signaling enhancer of the NF-κB signaling pathway.


Subject(s)
Acetamides/administration & dosage , HIV Infections/drug therapy , I-kappa B Proteins/genetics , Phorbol Esters/administration & dosage , Tumor Necrosis Factor alpha-Induced Protein 3/biosynthesis , Drug Synergism , Feedback, Physiological/drug effects , Gene Expression Regulation/drug effects , HEK293 Cells , HIV Infections/genetics , HIV Infections/virology , HIV-1/drug effects , HIV-1/genetics , HeLa Cells , Humans , NF-kappa B/genetics , Phosphorylation/drug effects , Positive Transcriptional Elongation Factor B/biosynthesis , Proviruses/drug effects , Proviruses/genetics , Signal Transduction/drug effects , Transcriptional Activation/drug effects , Tumor Necrosis Factor alpha-Induced Protein 3/genetics
8.
Nucleic Acids Res ; 44(14): 6853-67, 2016 08 19.
Article in English | MEDLINE | ID: mdl-27353326

ABSTRACT

The association of DSIF and NELF with initiated RNA Polymerase II (Pol II) is the general mechanism for inducing promoter-proximal pausing of Pol II. However, it remains largely unclear how the paused Pol II is released in response to stimulation. Here, we show that the release of the paused Pol II is cooperatively regulated by multiple P-TEFbs which are recruited by bromodomain-containing protein Brd4 and super elongation complex (SEC) via different recruitment mechanisms. Upon stimulation, Brd4 recruits P-TEFb to Spt5/DSIF via a recruitment pathway consisting of Med1, Med23 and Tat-SF1, whereas SEC recruits P-TEFb to NELF-A and NELF-E via Paf1c and Med26, respectively. P-TEFb-mediated phosphorylation of Spt5, NELF-A and NELF-E results in the dissociation of NELF from Pol II, thereby transiting transcription from pausing to elongation. Additionally, we demonstrate that P-TEFb-mediated Ser2 phosphorylation of Pol II is dispensable for pause release. Therefore, our studies reveal a co-regulatory mechanism of Brd4 and SEC in modulating the transcriptional pause release by recruiting multiple P-TEFbs via a Mediator- and Paf1c-coordinated recruitment network.


Subject(s)
Positive Transcriptional Elongation Factor B/metabolism , Promoter Regions, Genetic , RNA Polymerase II/metabolism , Acetamides/pharmacology , Cell Cycle Proteins , HCT116 Cells , HeLa Cells , Humans , Models, Biological , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Phosphorylation/drug effects , RNA, Small Interfering/metabolism , Transcription Elongation, Genetic/drug effects , Transcription Factors/genetics , Transcription Factors/metabolism , Transcriptional Elongation Factors/metabolism
9.
J Biol Chem ; 291(27): 14363-14372, 2016 Jul 01.
Article in English | MEDLINE | ID: mdl-27143356

ABSTRACT

The adenovirus early region 1A (E1A) oncoprotein hijacks host cells via direct interactions with many key cellular proteins, such as KAT2B, also known as PCAF (p300/CBP associated factor). E1A binds the histone acetyltransferase (HAT) domain of KAT2B to repress its transcriptional activation. However, the molecular mechanism by which E1A inhibits the HAT activity is not known. Here we demonstrate that a short and relatively conserved N-terminal motif (cNM) in the intrinsically disordered E1A protein is crucial for KAT2B interaction, and inhibits its HAT activity through a direct competition with acetyl-CoA, but not its substrate histone H3. Molecular modeling together with a series of mutagenesis experiments suggests that the major helix of E1A cNM binds to a surface of the acetyl-CoA pocket of the KAT2B HAT domain. Moreover, transient expression of the cNM peptide is sufficient to inhibit KAT2B-specific H3 acetylation H3K14ac in vivo Together, our data define an essential motif cNM in N-terminal E1A as an acetyl-CoA entry blocker that directly associates with the entrance of acetyl-CoA binding pocket to block the HAT domain access to its cofactor.


Subject(s)
Adenovirus E1A Proteins/physiology , Lysine Acetyltransferases/antagonists & inhibitors , Acetylation , Adenovirus E1A Proteins/chemistry , Amino Acid Motifs , Amino Acid Sequence , Humans , Hydrophobic and Hydrophilic Interactions , Kinetics , Models, Molecular , Sequence Homology, Amino Acid
10.
Biomed Res Int ; 2014: 968027, 2014.
Article in English | MEDLINE | ID: mdl-25136641

ABSTRACT

Prostratin has been proposed as a promising reagent for eradicating the latent HIV-1 provirus by inducing HIV-1 transcription activation. The molecular mechanism of this activation, however, is far from clear. Here, we show that the protein kinase D3 (PKD3) is essential for prostratin-induced transcription activation of latent HIV-1 provirus. First, silencing PKD3, but not the other members of PKD family, blocked prostratin-induced transcription of HIV-1. Second, overexpressing the constitutively active form of PKD3, but not the wild-type or kinase-dead form of PKD3, augmented the expression of HIV-1. Consistent with this observation, we found that prostratin could trigger PKD3 activation by inducing the phosphorylation of its activation loop. In addition, we identified PKCε of the novel PKC subfamily as the upstream kinase for this phosphorylation. Finally, the activation effect of PKD3 on HIV-1 transcription was shown to depend on the presence of κB element and the prostratin-induced activation of NF-κB, as indicated by the fact that silencing PKD3 blocked prostratin-induced NF-κB activation and NF-κB-dependent HIV-1 transcription. Therefore, for the first time, PKD3 is implicated in the transcription activation of latent HIV-1 provirus, and our results revealed a molecular mechanism of prostratin-induced HIV-1 transcription via PKCε/PKD3/NF-κB signaling pathway.


Subject(s)
HIV-1/genetics , Protein Kinase C/genetics , Proviruses/genetics , Virus Integration/genetics , Gene Expression Regulation, Viral/drug effects , HEK293 Cells , HIV-1/pathogenicity , Humans , Indoles/pharmacology , Maleimides/pharmacology , NF-kappa B/genetics , NF-kappa B/metabolism , Phorbol Esters/pharmacology , Promoter Regions, Genetic , Protein Kinase C/metabolism , Protein Kinase C-epsilon/genetics , Protein Kinase C-epsilon/metabolism , Proviruses/drug effects , Proviruses/pathogenicity , Signal Transduction/drug effects , Signal Transduction/genetics , Transcriptional Activation/drug effects , Virus Integration/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...