Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
1.
Int J Biol Macromol ; 265(Pt 1): 130921, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38492688

ABSTRACT

The design of small molecule inhibitors that target the programmed death ligand-1 (PD-L1) is a forefront issue in immune checkpoint blocking therapy. Small-molecule inhibitors have been shown to exert therapeutic effects by inducing dimerization of the PD-L1 protein, however, the specific mechanisms underlying this dimerization process remain largely unexplored. Furthermore, there is a notable lack of comparative studies examining the binding modes of structurally diverse inhibitors. In view of the research gaps, this work employed molecular dynamics simulations to meticulously examine the interactions between two distinct types of inhibitors and PD-L1 in both monomeric and dimeric forms, and predicted the dimerization mechanism. The results revealed that inhibitors initially bind to a PD-L1 monomer, subsequently attracting another monomer to form a dimer. Notably, symmetric inhibitors observed superior binding efficiency compared to other inhibitors. Key residues, including Ile54, Tyr56, Met115 and Tyr123 played a leading role in binding. Structurally, symmetric inhibitors were capable of thoroughly engaging the binding pocket, promoting a more symmetrical formation of PD-L1 dimers. Furthermore, symmetric inhibitors formed more extensive hydrophobic interactions with protein residues. The insights garnered from this research are expected to significantly contribute to the rational design and optimization of small molecule inhibitors targeting PD-L1.


Subject(s)
B7-H1 Antigen , Programmed Cell Death 1 Receptor , Dimerization , B7-H1 Antigen/metabolism , Programmed Cell Death 1 Receptor/metabolism , Small Molecule Libraries/chemistry , Molecular Dynamics Simulation
2.
Cell Mol Life Sci ; 80(11): 313, 2023 Oct 05.
Article in English | MEDLINE | ID: mdl-37796323

ABSTRACT

Papain-like protease (PLpro), a non-structural protein encoded by SARS-CoV-2, is an important therapeutic target. Regions 1 and 5 of an existing drug, GRL0617, can be optimized to produce cooperativity with PLpro binding, resulting in stronger binding affinity. This work investigated the origin of the cooperativity using molecular dynamics simulations combined with the interaction entropy (IE) method. The regions' improvement exhibits cooperativity by calculating the binding free energies between the complex of PLpro-inhibitor. The thermodynamic integration method further verified the cooperativity generated in the drug improvement. To further determine the specific source of cooperativity, enthalpy and entropy in the complexes were calculated using molecular mechanics/generalized Born surface area and IE. The results show that the entropic change is an important contributor to the cooperativity. Our study also identified residues P248, Q269, and T301 that play a significant role in cooperativity. The optimization of the inhibitor stabilizes these residues and minimizes the entropic loss, and the cooperativity observed in the binding free energy can be attributed to the change in the entropic contribution of these residues. Based on our research, the application of cooperativity can facilitate drug optimization, and provide theoretical ideas for drug development that leverage cooperativity by reducing the contribution of entropy through multi-locus binding.


Subject(s)
COVID-19 , SARS-CoV-2 , Humans , Entropy , Molecular Dynamics Simulation
3.
Environ Int ; 180: 108237, 2023 10.
Article in English | MEDLINE | ID: mdl-37802009

ABSTRACT

Environmental benzo(a)pyrene (BaP) and its ultimate metabolite BPDE (benzo(a)pyrene-7,8-dihydrodiol-9,10-epoxide) are universal and inevitable persistent organic pollutants and endocrine disrupting chemicals. Angiogenesis in placental decidua plays a pivotal role in healthy pregnancy. Ferroptosis is a newly identified and iron-dependent cell death mode. However, till now, BaP/BPDE exposure, ferroptosis, defective angiogenesis, and miscarriage have never been correlated; and their regulatory mechanisms have been rarely explored. In this study, we used assays with BPDE-exposed HUVECs (human umbilical vein endothelial cells), decidual tissues and serum samples collected from unexplained recurrent miscarriage and their matched healthy control groups, and placental tissues of BaP-exposed mouse miscarriage model. We found that BaP/BPDE exposure caused ferroptosis and then directly suppressed angiogenesis and eventually induced miscarriage. In mechanism, BaP/BPDE exposure up-regulated free Fe2+ level and promoted lipid peroxidation and also up-regulated MARCHF1 (a novel E3 ligase of GPX4) level to promote the ubiquitination degradation of GPX4, both of which resulted in HUVEC ferroptosis. Furthermore, we also found that GPX4 protein down-regulated the protein levels of VEGFA and ANG-1, two key proteins function for angiogenesis, and thus suppressed HUVEC angiogenesis. In turn, supplement with GPX4 could suppress ferroptosis, recover angiogenesis, and alleviate miscarriage. Moreover, the levels of free Fe2+ and VEGFA in serum might predict the risk of miscarriage. Overall, this study uncovered the crosstalk among BaP/BPDE exposure, ferroptosis, angiogenesis, and miscarriage, discovering novel toxicological effects of BaP/BPDE on human reproductive health. This study also warned the public to avoid exposure to polycyclic aromatic hydrocarbons during pregnancy to effectively prevent adverse pregnancy outcomes.


Subject(s)
Abortion, Spontaneous , Ferroptosis , Mice , Animals , Pregnancy , Humans , Female , 7,8-Dihydro-7,8-dihydroxybenzo(a)pyrene 9,10-oxide/metabolism , 7,8-Dihydro-7,8-dihydroxybenzo(a)pyrene 9,10-oxide/pharmacology , Benzo(a)pyrene , Endothelial Cells/metabolism , Placenta/metabolism , Proteins
4.
Phys Chem Chem Phys ; 25(34): 22941-22951, 2023 Aug 30.
Article in English | MEDLINE | ID: mdl-37593785

ABSTRACT

Recent studies have shown that DNA methylation is an important epigenetic marker. Two prominent forms are methylation of the C5 position of cytosine and methylation of the C6 position of adenine. Given the vital significance of DNA methylation, investigating the mechanisms that influence protein binding remains a compelling pursuit. This study used molecular dynamics simulations to investigate the binding patterns of R2R3 protein and four differentially methylated DNAs. The alanine scanning combined with interaction entropy method was used to identify key residues that respond to different methylation patterns. The order of protein binding ability to DNA is as follows: unmethylated DNA > A11 methylation (5'-A6mAC-3') (6m2A system) > A10 methylation (5'-6mAAC-3') (6m1A system) > both A10 and A11 methylation (5'-6mA6mAC-3') (6mAA system) > C12 methylation (5'-AA5mC-3') (5mC system). All methylation systems lead to the sixth α helix (H6) (residues D105 to L116) moving away from the binding interface, and in the 5mC and 6m1A systems, the third α helix (H3) (residues G54 to L65) exhibits a similar trend. When the positively charged amino acids in H3 and H6 move away from the binding interface, their electrostatic and van der Waals interactions with the negatively charged DNA are weakened. Structural changes induced by methylation contributed to the destabilization of the hydrogen bond network near the original binding site, except for the 6m2A system. Moreover, there is a positive correlation between the number of methylated sites and the probability of distorting the DNA structure. Our study explores how different methylation patterns affect binding and structural adaptability, and have implications for drug discovery and understanding diseases related to abnormal methylation.


Subject(s)
5-Methylcytosine , DNA , Kinetics , Adenine
5.
Int J Biol Macromol ; 247: 125690, 2023 Aug 30.
Article in English | MEDLINE | ID: mdl-37423448

ABSTRACT

DNA methylation as an important epigenetic marker, has gained attention for the significance of three oxidative modifications (hydroxymethyl-C (hmC), formyl-C (fC), and carboxyl-C (caC)). Mutations occurring in the methyl-CpG-binding domain (MBD) of MeCP2 result in Rett. However, uncertainties persist regarding DNA modification and MBD mutation-induced interaction changes. Here, molecular dynamics simulations were used to investigate the underlying mechanisms behind changes due to different modifications of DNA and MBD mutations. Alanine scanning combined with the interaction entropy method was employed to accurately evaluate the binding free energy. The results show that MBD has the strongest binding ability for mCDNA, followed by caC, hmC, and fCDNA, with the weakest binding ability observed for CDNA. Further analysis revealed that mC modification induces DNA bending, causing residues R91 and R162 closer to the DNA. This proximity enhances van der Waals and electrostatic interactions. Conversely, the caC/hmC and fC modifications lead to two loop regions (near K112 and K130) closer to DNA, respectively. Furthermore, DNA modifications promote the formation of stable hydrogen bond networks, however mutations in the MBD significantly reduce the binding free energy. This study provides detailed insight into the effects of DNA modifications and MBD mutations on binding ability. It emphasizes the necessity for research and development of targeted Rett compounds that induce conformational compatibility between MBD and DNA, enhancing the stability and strength of their interactions.


Subject(s)
Rett Syndrome , Humans , Rett Syndrome/genetics , Rett Syndrome/metabolism , Methyl-CpG-Binding Protein 2/chemistry , DNA/chemistry , Mutation , DNA Methylation , Protein Binding
6.
J Pharm Pharmacol ; 75(8): 1021-1033, 2023 Aug 01.
Article in English | MEDLINE | ID: mdl-37343148

ABSTRACT

OBJECTIVES: This study was to explore the potential effects and mechanism of naringenin against vascular senescence in atherosclerosis focusing on the SIRT1-mediated signalling pathway. METHODS: Aged apoE-/- mice were administrated with naringenin continuously for three months. Lipid parameters in serum and pathological changes and associated protein expression in aorta were examined. In vitro, endothelial cells were treated with H2O2 to induce senescence. KEY FINDINGS: Dyslipidemia, atherosclerotic lesion formation and vascular senescence were found in ApoE-/- mice, which were significantly ameliorated by naringenin treatment. Naringenin decreased reactive oxygen species overproduction and enhanced the activities of antioxidant enzymes in aorta. It also decreased mitoROS production and increased protein expressions of mitochondrial biogenesis-related genes in aorta. Moreover, naringenin treatment enhanced aortic protein expression and activity of SIRT1. Meanwhile, naringenin increased deacetylation and protein expression of SIRT1's target genes FOXO3a and PGC1α. In vitro study, the benefits of naringenin on endothelial senescence, oxidative stress and mitochondrial injury as well as protein expressions and acetylated levels of FOXO3a and PGC1α were diminished in cells transfected with SIRT1 siRNA. CONCLUSIONS: Naringenin could ameliorate vascular senescence and atherosclerosis and the activation of SIRT1, with subsequent deacetylation and regulation of FOXO3a and PGC1α, is involved in this process.


Subject(s)
Atherosclerosis , Endothelial Cells , Mice , Animals , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Endothelial Cells/metabolism , Sirtuin 1/metabolism , Hydrogen Peroxide , Mice, Knockout, ApoE , Atherosclerosis/metabolism , Oxidative Stress , Apolipoproteins E/metabolism , Cellular Senescence
7.
J Hazard Mater ; 455: 131543, 2023 08 05.
Article in English | MEDLINE | ID: mdl-37167865

ABSTRACT

Environmental Benzo(a)pyrene (BaP) and its ultimate metabolite BPDE (benzo(a)pyrene-7,8-dihydrodiol-9,10-epoxide) are typical persistent organic pollutants and endocrine disrupting chemicals. BaP/BPDE exposure might cause human trophoblast cell dysfunctions and induce miscarriage. However, the underlying mechanisms remain largely elusive. In this study, we found that BPDE exposure induced human trophoblast cell pyroptosis by up-regulating NLRP3/Caspase1/GSDMD pathway. We also identified that lnc-HZ14 was highly expressed in BPDE-exposed trophoblast cells and in recurrent miscarriage (RM) vs healthy control (HC) villous tissues. Lnc-HZ14 promoted trophoblast cell pyroptosis by promoting IRF1-mediated ZBP1 transcription, increasing METTL3-mediated m6A methylation on NLRP3 mRNA and its stability, and also enhancing ZBP1/NLRP3 protein interactions. Knockdown of lnc-HZ14/ZBP1/NLRP3 axis could efficiently alleviate BPDE-induced trophoblast cell pyroptosis. Higher level of pyroptosis, as indicated by the up-regulation of lnc-HZ14/ZBP1/NLRP3 axis, was found in RM vs HC villous tissues. In BaP-exposed mouse model, BaP exposure induced placental tissue pyroptosis and miscarriage by up-regulating murine Zbp1/Nlrp3 axis, and knockdown of Nlrp3 could efficiently reduce placenta pyroptosis and alleviate BaP-induced mouse miscarriage. Serum IL-1ß protein level might act as a promising indicator to predict the risk of miscarriage. These findings provided new insights into BaP/BPDE-induced trophoblast cell pyroptosis and miscarriage and might be helpful for further assessment of the toxicological effects of BaP/BPDE on the female reproduction.


Subject(s)
7,8-Dihydro-7,8-dihydroxybenzo(a)pyrene 9,10-oxide , Abortion, Spontaneous , Pregnancy , Humans , Female , Mice , Animals , Trophoblasts , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Pyroptosis , Abortion, Spontaneous/chemically induced , Abortion, Spontaneous/metabolism , Benzo(a)pyrene/metabolism , Placenta/metabolism , Methyltransferases/metabolism , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/pharmacology
8.
Int J Mol Sci ; 23(10)2022 May 21.
Article in English | MEDLINE | ID: mdl-35628577

ABSTRACT

A novel function of retinoid X receptor beta (RXRß) in endothelial cells has been reported by us during the formation of atherosclerosis. Here, we extended the study to explore the cellular mechanisms of RXRß protein stability regulation. In this study, we discovered that murine double minute-2 (MDM2) acts as an E3 ubiquitin ligase to target RXRß for degradation. The result showed that MDM2 directly interacted with and regulated RXRß protein stability. MDM2 promoted RXRß poly-ubiquitination and degradation by proteasomes. Moreover, mutated MDM2 RING domain (C464A) or treatment with an MDM2 inhibitor targeting the RING domain of MDM2 lost the ability of MDM2 to regulate RXRß protein expression and ubiquitination. Furthermore, treatment with MDM2 inhibitor alleviated oxidized low-density lipoprotein-induced mitochondrial damage, activation of TLR9/NF-κB and NLRP3/caspase-1 pathway and production of pro-inflammatory cytokines in endothelial cells. However, all these beneficial effects were reduced by the transfection of RXRß siRNA. Moreover, pharmacological inhibition of MDM2 attenuated the development of atherosclerosis and reversed mitochondrial damage and related inflammation in the atherosclerotic process in LDLr-/- mice, along with the increased RXRß protein expression in the aorta. Therefore, our study uncovers a previously unknown ubiquitination pathway and suggests MDM2-mediated RXRß ubiquitination as a new therapeutic target in atherosclerosis.


Subject(s)
Atherosclerosis , Proto-Oncogene Proteins c-mdm2 , Animals , Atherosclerosis/genetics , Endothelial Cells/metabolism , Inflammation/genetics , Mice , Proto-Oncogene Proteins c-mdm2/genetics , Proto-Oncogene Proteins c-mdm2/metabolism , Tumor Suppressor Protein p53/metabolism , Ubiquitination
9.
Ecotoxicol Environ Saf ; 237: 113564, 2022 Jun 01.
Article in English | MEDLINE | ID: mdl-35483139

ABSTRACT

Human trophoblast cell apoptosis may induce miscarriage. Trophoblast cells are sensitive to environmental BaP-7,8-dihydrodiol-9,10-epoxide (BPDE). However, how BPDE induces human trophoblast cell apoptosis is still largely elusive. In this work, we used BPDE-treated human trophoblast cells and villous tissues collected from recurrent miscarriage and health control groups to explore the underlying mechanism of BPDE-induced human trophoblast cell apoptosis. Continued with our recent work, we found that lncRNA HZ01 (lnc-HZ01) could induce human trophoblast cell apoptosis. In mechanism, lnc-HZ01 up-regulated p53 expression level by suppressing its MDM2-mediated proteasomal degradation. Meanwhile, we found that p53 acted as lnc-HZ01 transcription factor and promoted lnc-HZ01 transcription. Thus, lnc-HZ01 and p53 composed a positive feedback loop in human trophoblast cells. In normal trophoblast cells, relatively low levels of lnc-HZ01 and p53 suppressed p53/caspase-3 apoptosis pathway, giving normal pregnancy. Upon BPDE exposure, BPDE up-regulated the expression levels of lnc-HZ01 and p53, triggered this positive feedback loop, activated the p53/caspase-3 apoptosis pathway, and then induced miscarriage. Collectively, we discovered new mechanism by which lnc-HZ01 regulated BPDE-induced human trophoblast cell apoptosis, providing scientific basis for the diagnosis and treatment of unexplained recurrent miscarriage.


Subject(s)
Abortion, Habitual , RNA, Long Noncoding , 7,8-Dihydro-7,8-dihydroxybenzo(a)pyrene 9,10-oxide/metabolism , 7,8-Dihydro-7,8-dihydroxybenzo(a)pyrene 9,10-oxide/toxicity , Abortion, Habitual/chemically induced , Abortion, Habitual/metabolism , Apoptosis , Caspase 3/metabolism , Feedback , Female , Humans , Pregnancy , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Trophoblasts/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
10.
Life (Basel) ; 12(3)2022 Mar 11.
Article in English | MEDLINE | ID: mdl-35330157

ABSTRACT

Fucosylation is one of the most important glycan terminal modifications that affects multiple biological activities of proteins. 2-Fluorofucose (2FF), its specific inhibitor, has recently been reported to reveal numerous biological effects by blocking fucosylation both in vitro and in vivo. The current study aimed to evaluate the effect of 2FF on hydrogen peroxide (H2O2)-induced oxidative damage in vitro. In our study, treatment with H2O2 increased the level of fucosylation, and 2FF improved the cell viability in H2O2-treated HepG2 cells. Our study also showed that 2FF significantly decreased the overproduction of reactive oxygen species (ROS) induced by H2O2 and the activities of catalase, glutathione and Mn-superoxide dismutase were remarkably increased by 2FF pretreatment. Furthermore, 2FF attenuated H2O2-induced early mitochondria dysfunction. The second part of the study revealed that 2FF enhanced antioxidant capacity by affecting Nrf2/keap1 and NF-κB signaling pathways in HepG2 cells. Being pretreated with 2FF significantly increased the nuclear translocation of Nrf2 and simultaneously promoted the expression of downstream proteins, such as HO-1 and NQO1. Moreover, 2FF remarkably suppressed the expression of inflammation-associated proteins. Taken together, these data suggest that 2FF might have a potential therapeutic effect for oxidative stress.

11.
Int J Mol Sci ; 20(7)2019 Apr 01.
Article in English | MEDLINE | ID: mdl-30939745

ABSTRACT

The compound, 2,3,5,4'-tetrahydroxystilbene-2-O-ß-d-glucoside (TSG), a primary bioactive polyphenolic component of Polygonum multiflorum exerts numerous pharmacological activities. However, its protective effect against non-alcoholic steatohepatitis (NASH), in the context of metabolic syndrome, remains poorly understood. The aim of the present study is to evaluate the effects of TSG treatment on middle-aged (12-mo-old) male LDLr-/- mice, which were fed a high fat diet for 12 weeks to induce metabolic syndrome and NASH. At the end of the experiment, the blood samples of mice were collected for determination of metabolic parameters. Liver and aorta tissues were collected for analysis, such as histology, immunofluorescence, hepatic lipid content, real-time PCR, and western blot. Our data show that TSG treatment improved the different aspects of NASH (steatosis, inflammation, and fibrosis) and atherosclerosis, as well as some of the metabolic basal characteristics. These modulatory effects of TSG are mediated, at least in part, through regulating key regulators of lipid metabolism (SREBP1c, PPARα and their target genes, ABCG5 and CYP7A1), inflammation (CD68, TNF-α, IL-6 and ICAM), fibrosis (α-SMA and TNFß) and oxidative stress (NADPH-oxidase 2/4, CYP2E1 and antioxidant enzymes). These results suggest that TSG may be a promising candidate for preventing and treating the progression of NASH.


Subject(s)
Aging/pathology , Atherosclerosis/drug therapy , Glucosides/therapeutic use , Non-alcoholic Fatty Liver Disease/drug therapy , Stilbenes/therapeutic use , Animals , Aorta/drug effects , Aorta/metabolism , Atherosclerosis/etiology , Diet, High-Fat/adverse effects , Glucosides/pharmacology , Lipid Metabolism , Liver/drug effects , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease/etiology , Receptors, LDL/genetics , Stilbenes/pharmacology
12.
ACS Appl Mater Interfaces ; 11(1): 1587-1594, 2019 Jan 09.
Article in English | MEDLINE | ID: mdl-30540166

ABSTRACT

Few-layer black phosphorus (BP) has recently emerged as a promising two-dimensional (2D) material for electronic and optoelectronic devices because of its high mobility and tunable band gap. However, BP is known to quickly degrade and oxidize in ambient conditions by breaking of the P-P bonds. As a result, there is a growing need to encapsulate BP that avoids oxygen and water while retaining the high electric performance of the devices. Here, we demonstrate a hydrophobic polymer encapsulation technique with improved thermal conductivity for high current density, which preserves the electrical properties of BP back-gate transistors compared to the commonly used Al2O3 encapsulation with improved mobility and minimal traps. The on-off ratio increases by more than an order of magnitude at room temperature and more than 4 orders of magnitude at cryogenic temperatures. High field transport shows the first systematic study on unprecedented breakdown characteristics up to -5.5 V for the 0.16 µm transistors with a high current of 1.2 mA/µm at 20 K. These discoveries open up a new way to achieve high-performance 2D semiconductors with significantly improved breakdown voltage, on-off ratios, and stability under ambient conditions for practical applications in electronic and optoelectronic devices.

13.
Nat Commun ; 9(1): 4778, 2018 11 14.
Article in English | MEDLINE | ID: mdl-30429471

ABSTRACT

Atomically-thin layered molybdenum disulfide (MoS2) has attracted tremendous research attention for their potential applications in high performance DC and radio frequency electronics, especially for flexible electronics. Bilayer MoS2 is expected to have higher electron mobility and higher density of states with higher performance compared with single layer MoS2. Here, we systematically investigate the synthesis of high quality bilayer MoS2 by chemical vapor deposition on molten glass with increasing domain sizes up to 200 µm. High performance transistors with optimized high-κ dielectrics deliver ON-current of 427 µA µm-1 at 300 K and a record high ON-current of 1.52 mA µm-1 at 4.3 K. Moreover, radio frequency transistors are demonstrated with an extrinsic high cut-off frequency of 7.2 GHz and record high extrinsic maximum frequency of oscillation of 23 GHz, together with gigahertz MoS2 mixers on flexible polyimide substrate, showing the great potential for future high performance DC and high-frequency electronics.

14.
ACS Appl Mater Interfaces ; 10(45): 38700-38714, 2018 Nov 14.
Article in English | MEDLINE | ID: mdl-30360090

ABSTRACT

Photothermal therapy is a promising approach for antitumor application although regrettably restricted by available photothermal agents. Physical entrapment of organic near-infrared dyes into nanosystems was extensively studied to reverse the dilemma. However, problems still remained, such as drug bursting and leakage. We developed here an amphiphilic prodrug conjugate by chemically modifying indocyanine green derivative (ICG-COOH) and paclitaxel (PTX) to hyaluronic acid (HA) backbone for integration of photothermal-chemotherapy and specific tumor imaging. The prepared ICG-HA-PTX conjugates could self-assemble into nanomicelles to improve the stability and reduce systemic toxicity of the therapeutic agents. The high local concentration of ICG-COOH in nanomicelles resulted in fluorescence self-quenching, leading to no fluorescence signal being detected in circulation. When the nanomicelles reached the tumor site via electron paramagnetic resonance effect and HA-mediated active targeting, the overexpressed esterase in tumor cells ruptured the ester linkage between drugs and HA, achieving tumor-targeted therapy and specific imaging. A series of in vitro and in vivo experiments demonstrated that the easily prepared ICG- HA-PTX nanomicelles with high stability, smart release behavio r, and excellent tumor targeting ability showed formidable synergy in tumor inhibition, which provided new thoughts in developing an organic near-infrared-dye-based multifunctional delivery system for tumor theranostics.


Subject(s)
Breast Neoplasms/diagnostic imaging , Breast Neoplasms/therapy , Hyperthermia, Induced/methods , Indocyanine Green/administration & dosage , Nanostructures/administration & dosage , Paclitaxel/administration & dosage , Phototherapy/methods , Prodrugs/administration & dosage , Animals , Breast Neoplasms/metabolism , Female , Humans , Indocyanine Green/chemistry , Indocyanine Green/pharmacokinetics , MCF-7 Cells , Mice , Mice, Inbred BALB C , Mice, Nude , Micelles , NIH 3T3 Cells , Nanostructures/chemistry , Optical Imaging/methods , Paclitaxel/chemistry , Paclitaxel/pharmacokinetics , Prodrugs/chemistry , Prodrugs/pharmacokinetics , Xenograft Model Antitumor Assays
15.
Eur J Pharmacol ; 830: 76-86, 2018 Jul 05.
Article in English | MEDLINE | ID: mdl-29704496

ABSTRACT

Salusin-α is an endogenous bioactive peptide and likely to prevent atherosclerosis. But its protective effect against atherosclerosis in vivo remains poorly understood. The aim of the present study was to determine the potential effects of salusin-α on atherosclerosis and its associated metabolic disorders in high fat diet (HFD)-fed low density lipoprotein receptor deficient (LDLr-/-) mice, and also explore the possible underlying mechanisms involved. Our data showed that after 12 weeks treatment, salusin-α ameliorated HFD-induced weight gain, hyperlipidemia, and serum levels of interleukin-6 (IL-6) and tumor necrosis factor-α (TNF-α). Salusin-α suppressed HFD-induced hepatic steatosis and regulated gene expression of fatty acid synthase, acetyl coenzyme A carboxylase-α, peroxisome proliferator-activated receptor-α, camitine palmitoyltransferase-1α and CYP7A1 in liver. Salusin-α reduced atherosclerotic plaque area and macrophage foam cell formation. Salusin-α prevented hepatic and aortic inflammation as evidenced by the reduced macrophage recruitment and mRNA expression of IL-6 and TNF-α in both liver and aorta. Salusin-α also reduced hepatic and aortic oxidative stress by normalizing activities of antioxidant enzymes in liver and suppressing reactive oxygen species generation and protein expressions of NADPH-oxidase (NOX) 2 and NOX4 in both liver and aorta. Our present data suggest that salusin-α could reduce hepatic steatosis and atherosclerosis via its pleiotropic effects, including amelioration of lipid profiles, regulation of some key molecules involved in lipid metabolism in liver, anti-oxidative effect and anti-inflammatory action.


Subject(s)
Atherosclerosis/drug therapy , Fatty Liver/drug therapy , Intercellular Signaling Peptides and Proteins/therapeutic use , Acetyl Coenzyme A/genetics , Animals , Atherosclerosis/blood , Atherosclerosis/genetics , Carnitine O-Palmitoyltransferase/genetics , Cholesterol 7-alpha-Hydroxylase/genetics , Diet, High-Fat , Fatty Acid Synthases/genetics , Fatty Liver/blood , Fatty Liver/genetics , Hyperlipidemias/blood , Hyperlipidemias/drug therapy , Intercellular Signaling Peptides and Proteins/pharmacology , Interleukin-6/blood , Lipids/blood , Male , Mice, Knockout , PPAR alpha/genetics , Receptors, LDL/genetics , Tumor Necrosis Factor-alpha/blood , Weight Gain/drug effects
16.
J Physiol ; 596(5): 827-855, 2018 03 01.
Article in English | MEDLINE | ID: mdl-29313986

ABSTRACT

KEY POINTS: Abnormal mitochondrial morphology and function in cardiomyocytes are frequently observed under persistent Gq protein-coupled receptor (Gq PCR) stimulation. Cardiac signalling mechanisms for regulating mitochondrial morphology and function under pathophysiological conditions in the heart are still poorly understood. We demonstrate that a downstream kinase of Gq PCR, protein kinase D (PKD) induces mitochondrial fragmentation via phosphorylation of dynamin-like protein 1 (DLP1), a mitochondrial fission protein. The fragmented mitochondria enhance reactive oxygen species generation and permeability transition pore opening in mitochondria, which initiate apoptotic signalling activation. This study identifies a novel PKD-specific substrate in cardiac mitochondria and uncovers the role of PKD on cardiac mitochondria, with special emphasis on the molecular mechanism(s) underlying mitochondrial injury with abnormal mitochondrial morphology under persistent Gq PCR stimulation. These findings provide new insights into the molecular basis of cardiac mitochondrial physiology and pathophysiology, linking Gq PCR signalling with the regulation of mitochondrial morphology and function. ABSTRACT: Regulation of mitochondrial morphology is crucial for the maintenance of physiological functions in many cell types including cardiomyocytes. Small and fragmented mitochondria are frequently observed in pathological conditions, but it is still unclear which cardiac signalling pathway is responsible for regulating the abnormal mitochondrial morphology in cardiomyocytes. Here we demonstrate that a downstream kinase of Gq protein-coupled receptor (Gq PCR) signalling, protein kinase D (PKD), mediates pathophysiological modifications in mitochondrial morphology and function, which consequently contribute to the activation of apoptotic signalling. We show that Gq PCR stimulation induced by α1 -adrenergic stimulation mediates mitochondrial fragmentation in a fission- and PKD-dependent manner in H9c2 cardiac myoblasts and rat neonatal cardiomyocytes. Upon Gq PCR stimulation, PKD translocates from the cytoplasm to the outer mitochondrial membrane (OMM) and phosphorylates a mitochondrial fission protein, dynamin-like protein 1 (DLP1), at S637. PKD-dependent phosphorylation of DLP1 initiates DLP1 association with the OMM, which then enhances mitochondrial fragmentation, mitochondrial superoxide generation, mitochondrial permeability transition pore opening and apoptotic signalling. Finally, we demonstrate that DLP1 phosphorylation at S637 by PKD occurs in vivo using ventricular tissues from transgenic mice with cardiac-specific overexpression of constitutively active Gαq protein. In conclusion, Gq PCR-PKD signalling induces mitochondrial fragmentation and dysfunction via PKD-dependent DLP1 phosphorylation in cardiomyocytes. This study is the first to identify a novel PKD-specific substrate, DLP1 in mitochondria, as well as the functional role of PKD in cardiac mitochondria. Elucidation of these molecular mechanisms by which PKD-dependent enhanced fission mediates cardiac mitochondrial injury will provide novel insight into the relationship among mitochondrial form, function and Gq PCR signalling.


Subject(s)
Dynamins/metabolism , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Mitochondria/pathology , Mitochondrial Dynamics , Myocytes, Cardiac/pathology , Protein Kinase C/metabolism , Animals , Mice , Mice, Transgenic , Mitochondria/metabolism , Mitochondrial Membrane Transport Proteins , Mitochondrial Permeability Transition Pore , Myocytes, Cardiac/metabolism , Phosphorylation , Rats , Rats, Sprague-Dawley , Signal Transduction
17.
Can J Physiol Pharmacol ; 96(1): 8-17, 2018 Jan.
Article in English | MEDLINE | ID: mdl-28863273

ABSTRACT

The aim of this study was to evaluate the potential effects of 2,3,4',5-tetrahydroxystilbene-2-O-ß-d-glucoside (TSG) on the development of atherosclerotic plaque in ApoE-/- mice, and explore the mechanisms involved. Our data showed that after 8 weeks of treatment, TSG ameliorated serum levels of total cholesterol, triglyceride, and low density lipoprotein cholesterol, and increased serum levels of high density lipoprotein cholesterol in ApoE-/- mice. TSG suppressed hepatic steatosis, the formation of atherosclerotic lesions, and the formation of macrophage foam cells in ApoE-/- mice. Moreover, TSG improved the expressions of hepatic SR-BI, ABCG5, and CYP7A1, and up-regulated the protein expressions of aortic ABCA1 and ABCG1. An in-vitro study showed that TSG promoted macrophage cholesterol efflux and increased the protein expressions of ABCA1 and ABCG1. Our findings provide evidence for a positive role of TSG in preventing atherosclerosis by promoting reverse cholesterol transport. These effects may be achieved by stimulating cholesterol efflux through ABCA1 and ABCG1, promoting SR-BI-mediated cholesterol uptake in the liver, increasing secretion of cholesterol into bile by ABCG5, and improving cholesterol metabolism by the CYP7A1 pathway. In addition, antioxidative and anti-inflammatory effects of TSG may also contribute to its inhibitory effects on atherosclerosis. Further study is needed to investigate whether other potential mechanisms are involved in TSG-mediated atheroprotection.


Subject(s)
Apolipoproteins E/deficiency , Atherosclerosis/drug therapy , Atherosclerosis/metabolism , Cholesterol/metabolism , Glucosides/therapeutic use , Stilbenes/therapeutic use , Animals , Atherosclerosis/complications , Atherosclerosis/pathology , Biological Transport/drug effects , Fatty Liver/complications , Fatty Liver/drug therapy , Foam Cells/drug effects , Foam Cells/metabolism , Glucosides/pharmacology , Hyperlipidemias/complications , Hyperlipidemias/drug therapy , Hyperlipidemias/metabolism , Inflammation/complications , Inflammation/pathology , Lipid Metabolism/drug effects , Macrophages/drug effects , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Oxidative Stress/drug effects , RAW 264.7 Cells , Stilbenes/pharmacology
18.
Cell ; 171(3): 557-572.e24, 2017 Oct 19.
Article in English | MEDLINE | ID: mdl-29053968

ABSTRACT

Chromosome conformation capture technologies have revealed important insights into genome folding. Yet, how spatial genome architecture is related to gene expression and cell fate remains unclear. We comprehensively mapped 3D chromatin organization during mouse neural differentiation in vitro and in vivo, generating the highest-resolution Hi-C maps available to date. We found that transcription is correlated with chromatin insulation and long-range interactions, but dCas9-mediated activation is insufficient for creating TAD boundaries de novo. Additionally, we discovered long-range contacts between gene bodies of exon-rich, active genes in all cell types. During neural differentiation, contacts between active TADs become less pronounced while inactive TADs interact more strongly. An extensive Polycomb network in stem cells is disrupted, while dynamic interactions between neural transcription factors appear in vivo. Finally, cell type-specific enhancer-promoter contacts are established concomitant to gene expression. This work shows that multiple factors influence the dynamics of chromatin interactions in development.


Subject(s)
Chromatin/metabolism , Genome , Neurogenesis , Animals , CCCTC-Binding Factor , Embryonic Stem Cells/metabolism , Enhancer Elements, Genetic , Exons , Gene Expression , Gene Regulatory Networks , Mice , Promoter Regions, Genetic , Repressor Proteins/metabolism , Transcription Factors/metabolism
19.
Toxicology ; 389: 74-84, 2017 08 15.
Article in English | MEDLINE | ID: mdl-28743511

ABSTRACT

Both RhoA/ROCK and Raf-1/CK2 pathway play essential roles in cell proliferation, apoptosis, differentiation, and multiple other common cellular functions. We previously reported that vimentin is responsible for TNF-α-induced cell apoptosis. Herein, we investigated the regulation of RhoA/ROCK and Raf-1/CK2 signaling on vimentin filaments and endothelial apoptosis mediated by TNF-α. Treatment with TNF-α significantly induced the activation of RhoA and ROCK, and the expression of ROCK1. RhoA deficiency could obviously inhibit ROCK activation and ROCK1 expression induced by TNF-α. Both RhoA deficiency and ROCK activity inhibition (Y-27632) greatly inhibited endothelial apoptosis and preserved cell viability in TNF-α-induced human umbilical vein endothelial cells (HUVECs). Also vimentin phosphorylation and the remodeling of vimentin or phospho-vimentin induced by TNF-α were obviously attenuated by RhoA suppression and ROCK inhibition. TNF-α-mediated vimentin cleavage was significantly inhibited by RhoA suppression and ROCK inhibition through decreasing the activation of caspase3 and 8. Furthermore, TNF-α treatment greatly enhanced the activation of Raf-1. Suppression of Raf-1 or CK2 by its inhibitor (GW5074 or TBB) blocked vimentin phosphorylation, remodeling and endothelial apoptosis, and preserved cell viability in TNF-α-induced HUVECs. However, Raf-1 inhibition showed no significant effect on TNF-α-induced ROCK expression and activation, suggesting that the regulation of Raf-1/CK2 signaling on vimentin was independent of ROCK. Taken together, these results indicate that both RhoA/ROCK and Raf-1/CK2 pathway are responsible for TNF-α-mediated endothelial cytotoxicity via regulating vimentin cytoskeleton.


Subject(s)
Apoptosis/drug effects , Casein Kinase II/metabolism , Cytoskeleton/drug effects , Human Umbilical Vein Endothelial Cells/drug effects , Proto-Oncogene Proteins c-raf/metabolism , Tumor Necrosis Factor-alpha/toxicity , Vimentin/metabolism , rho-Associated Kinases/metabolism , rhoA GTP-Binding Protein/metabolism , Casein Kinase II/antagonists & inhibitors , Cell Survival/drug effects , Cells, Cultured , Cytoskeleton/enzymology , Cytoskeleton/pathology , Human Umbilical Vein Endothelial Cells/enzymology , Human Umbilical Vein Endothelial Cells/pathology , Humans , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-raf/antagonists & inhibitors , RNA Interference , Signal Transduction/drug effects , Transfection , cdc42 GTP-Binding Protein/genetics , cdc42 GTP-Binding Protein/metabolism , rho-Associated Kinases/antagonists & inhibitors , rhoA GTP-Binding Protein/genetics
20.
Front Cell Neurosci ; 11: 154, 2017.
Article in English | MEDLINE | ID: mdl-28603491

ABSTRACT

NOSH-NBP, a novel nitric oxide (NO) and hydrogen sulfide (H2S)-releasing hybrid, protects brain from ischemic stroke. This study mainly aimed to investigate the therapeutic effect of NOSH-NBP on ischemic stroke and the underlying mechanisms. In vivo, transient middle cerebral artery occlusion (tMCAO) was performed in C57BL/6 mice, with NO-NBP and H2S-NBP as controls. NO and H2S scavengers, carboxy-PTIO and BSS, respectively, were used to quench NO and H2S of NOSH-NBP. In vitro, BV2 microglia/BMDM were induced to the M1/2 phenotype, and conditioned medium (CM) experiments in BV2 microglia, neurons and b.End3 cerebral microvascular endothelial cells (ECs) were performed. Microglial/macrophage activation/polarization was assessed by flow cytometry, Western blot, RT-qPCR, and ELISA. Neuronal and EC survival was measured by TUNEL, flow cytometry, MTT and LDH assays. Transmission electron microscopy, EB extravasation, brain water content, TEER measurement and Western blot were used to detect blood-brain barrier (BBB) integrity and function. Interestingly, NOSH-NBP significantly reduced cerebral infarct volume and ameliorated neurological deficit, with superior effects compared with NO-NBP and/or H2S-NBP in mice after tMCAO. Both NO and H2S-releasing groups contributed to protection by NOSH-NBP. Additionally, NOSH-NBP decreased neuronal death and attenuated BBB dysfunction in tMCAO-treated mice. Furthermore, NOSH-NBP promoted microglia/macrophage switch from an inflammatory M1 phenotype to the protective M2 phenotype in vivo and in vitro. Moreover, the TLR4/MyD88/NF-κB pathway and NLRP3 inflammasome were involved in the inhibitory effects of NOSH-NBP on M1 polarization, while peroxisome proliferator activated receptor gamma signaling contributed to NOSH-NBP induced M2 polarization. These findings indicated that NOSH-NBP is a potential therapeutic agent that preferentially promotes microglial/macrophage M1-M2 switch in ischemic stroke.

SELECTION OF CITATIONS
SEARCH DETAIL
...