Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 406
1.
Int J Mol Sci ; 24(17)2023 Aug 28.
Article En | MEDLINE | ID: mdl-37686160

The hepatitis B virus (HBV) is constantly exposed to significant oxidative stress characterized by elevated levels of reactive oxygen species (ROS), such as H2O2, during infection in hepatocytes of patients. In this study, we demonstrated that H2O2 inhibits HBV replication in a p53-dependent fashion in human hepatoma cell lines expressing sodium taurocholate cotransporting polypeptide. Interestingly, H2O2 failed to inhibit the replication of an HBV X protein (HBx)-null HBV mutant, but this defect was successfully complemented by ectopic expression of HBx. Additionally, H2O2 upregulated p53 levels, leading to increased expression of seven in absentia homolog 1 (Siah-1) levels. Siah-1, an E3 ligase, induced the ubiquitination-dependent proteasomal degradation of HBx. The inhibitory effect of H2O2 was nearly abolished not only by treatment with a representative antioxidant, N-acetyl-L-cysteine but also by knockdown of either p53 or Siah-1 using specific short hairpin RNA, confirming the role of p53 and Siah-1 in the inhibition of HBV replication by H2O2. The present study provides insights into the mechanism that regulates HBV replication under conditions of oxidative stress in patients.


Carcinoma, Hepatocellular , Hepatitis B virus , Hepatitis B , Liver Neoplasms , Humans , Carcinoma, Hepatocellular/genetics , Hepatitis B virus/drug effects , Hydrogen Peroxide/pharmacology , Liver Neoplasms/genetics , Tumor Suppressor Protein p53/genetics , Virus Replication , Viral Regulatory and Accessory Proteins/drug effects , Trans-Activators/drug effects
2.
J Hepatol ; 76(1): 53-62, 2022 01.
Article En | MEDLINE | ID: mdl-34478763

BACKGROUND & AIMS: HBV causes hepatocellular carcinoma (HCC). While it was recently shown that the ability of HBV X protein (HBx) to impair the Smc5/6 (structural maintenance of chromosome 5/6) complex is important for viral transcription, HBx is also a potent driver of HCC. However, the mechanism by which HBx expression induces hepatocarcinogenesis is unclear. METHODS: Degradation of the Smc5/6 complex and accumulation of DNA damage were observed in both in vivo and in vitro HBV infection models. Rescue experiments were performed using nitazoxanide (NTZ), which inhibits degradation of the Smc5/6 complex by HBx. RESULTS: HBx-triggered degradation of the Smc5/6 complex causes impaired homologous recombination (HR) repair of DNA double-strand breaks (DSBs), leading to cellular transformation. We found that DNA damage accumulated in the liver tissue of HBV-infected humanized chimeric mice, HBx-transgenic mice, and human tissues. HBx suppressed the HR repair of DSBs, including that induced by the CRISPR-Cas9 system, in an Smc5/6-dependent manner, which was rescued by restoring the Smc5/6 complex. NTZ restored HR repair in, and colony formation by, HBx-expressing cells. CONCLUSIONS: Degradation of the Smc5/6 complex by HBx increases viral transcription and promotes cellular transformation by impairing HR repair of DSBs. LAY SUMMARY: The hepatitis B virus expresses a regulatory protein called HBV X protein (or HBx). This protein degrades the Smc5/6 complex in human hepatocytes, which is essential for viral replication. We found that this process also plays a key role in the accumulation of DNA damage, which contributes to HBx-mediated tumorigenesis.


Cell Cycle Proteins/adverse effects , Chromosomal Proteins, Non-Histone/adverse effects , Recombinational DNA Repair/drug effects , Trans-Activators/drug effects , Viral Regulatory and Accessory Proteins/drug effects , Animals , Carcinoma, Hepatocellular/complications , Carcinoma, Hepatocellular/pathology , Disease Models, Animal , Liver/drug effects , Liver/pathology , Liver Neoplasms/complications , Liver Neoplasms/pathology , Mice , Recombinational DNA Repair/immunology , Statistics, Nonparametric
3.
Diabetes ; 70(12): 2850-2859, 2021 12.
Article En | MEDLINE | ID: mdl-34610983

Controllable genetic manipulation is an indispensable tool in research, greatly advancing our understanding of cell biology and physiology. However in ß-cells, transgene silencing, low inducibility, ectopic expression, and off-targets effects are persistent challenges. In this study, we investigated whether an inducible Tetracycline (Tet)-Off system with ß-cell-specific mouse insulin promoter (MIP)-itTA-driven expression of tetracycline operon (TetO)-CreJaw/J could circumvent previous issues of specificity and efficacy. Following assessment of tissue-specific gene recombination, ß-cell architecture, in vitro and in vivo glucose-stimulated insulin secretion, and whole-body glucose homeostasis, we discovered that expression of any tetracycline-controlled transactivator (e.g., improved itTA, reverse rtTA, or tTA) in ß-cells significantly reduced Insulin gene expression and decreased insulin content. This translated into lower pancreatic insulin levels and reduced insulin secretion in mice carrying any tTA transgene, independent of Cre recombinase expression or doxycycline exposure. Our study echoes ongoing challenges faced by fundamental researchers working with ß-cells and highlights the need for consistent and comprehensive controls when using the tetracycline-controlled transactivator systems (Tet-On or Tet-Off) for genome editing.


Insulin-Secreting Cells/metabolism , Insulin/genetics , Insulin/metabolism , Animals , Cells, Cultured , Female , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Integrases/genetics , Integrases/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Promoter Regions, Genetic/drug effects , Promoter Regions, Genetic/genetics , Tetracycline/pharmacology , Trans-Activators/drug effects , Trans-Activators/genetics , Transgenes/drug effects
4.
Plant Sci ; 310: 110983, 2021 Sep.
Article En | MEDLINE | ID: mdl-34315599

The Arabidopsis thaliana R2R3-MYB transcription factor AtMYB32 and its homologs AtMYB4 and AtMYB7 play crucial roles in the regulation of phenylpropanoid metabolism. In addition, AtMYB4 and AtMYB7 are involved in the response to abiotic stress. However, the function of AtMYB32 remains unclear. In this study, we found that AtMYB32 is induced by abscisic acid (ABA) and repressed by drought stress. AtMYB32 positively regulates ABA-mediated seed germination and early seedling development. The expression of ABSCISIC ACID-INSENSITIVE 3 (ABI3), ABI4 and ABI5, which encode key positive regulators of ABA signaling, was upregulated in response to ABA in AtMYB32-overexpressing plants and downregulated in the atmyb32-1 mutant. In addition, we found that the atmyb32-1 mutant was drought resistant. Consistent with the drought-resistant phenotype, the transcript levels of C-repeat binding factor 4 (CBF4) were higher in the atmyb32-1 mutant in response to drought stress. Electrophoretic mobility shift assays (EMSAs) and chromatin immunoprecipitation (ChIP) assays revealed that AtMYB32 binds directly to the ABI3, ABI4, ABI5 and CBF4 promoters both in vitro and in vivo. Genetically, ABI4 was found to be epistatic to AtMYB32 for ABA-induced inhibition of seed germination and early seedling development. Taken together, our findings revealed that AtMYB32 regulates the ABA response by directly promoting ABI3, ABI4 and ABI5 expression and that the drought stress response likely occurs because of repression of CBF4 expression.


Abscisic Acid/pharmacology , Arabidopsis Proteins/metabolism , Arabidopsis/drug effects , Arabidopsis/metabolism , Basic-Leucine Zipper Transcription Factors/metabolism , Trans-Activators/metabolism , Transcription Factors/metabolism , Arabidopsis Proteins/drug effects , Arabidopsis Proteins/genetics , Basic-Leucine Zipper Transcription Factors/drug effects , Droughts , Gene Expression Regulation, Plant/drug effects , Gene Expression Regulation, Plant/genetics , Trans-Activators/drug effects , Transcription Factors/drug effects
5.
Mol Cancer ; 20(1): 78, 2021 05 26.
Article En | MEDLINE | ID: mdl-34039363

BACKGROUND: Distant metastasis is the major cause of death in patients with colorectal cancer (CRC). Previously, we identified KITENIN as a metastasis-enhancing gene and suggested that the oncogenic KITENIN complex is involved in metastatic dissemination of KITENIN-overexpressing CRC cells. Here, we attempted to find substances targeting the KITENIN complex and test their ability to suppress distant metastasis of CRC. METHODS: We screened a small-molecule compound library to find candidate substances suppressing the KITENIN complex in CRC cells. We selected a candidate compound and examined its effects on the KITENIN complex and distant metastasis through in vitro assays, a molecular docking model, and in vivo tumor models. RESULTS: Among several compounds, we identified DKC1125 (Disintegrator of KITENIN Complex #1125) as the best candidate. DKC1125 specifically suppressed KITENIN gain of function. After binding KH-type splicing regulatory protein (KSRP), DKC1125 degraded KITENIN and Dvl2 by recruiting RACK1 and miRNA-124, leading to the disintegration of the functional KITENIN-KSRP-RACK1-Dvl2 complex. A computer docking model suggested that DKC1125 specifically interacted with the binding pocket of the fourth KH-domain of KSRP. KITENIN-overexpressing CRC cells deregulated certain microRNAs and were resistant to 5-fluorouracil, oxaliplatin, and cetuximab. DKC1125 restored sensitivity to these drugs by normalizing expression of the deregulated microRNAs, including miRNA-124. DKC1125 effectively suppressed colorectal liver metastasis in a mouse model. Interestingly, the combination of DKC1125 with 5-fluorouracil suppressed metastasis more effectively than either drug alone. CONCLUSION: DKC1125 targets the KITENIN complex and could therefore be used as a novel therapeutic to suppress liver metastasis in CRC expressing high levels of KITENIN.


Antineoplastic Agents/pharmacology , Carrier Proteins/drug effects , Colorectal Neoplasms/pathology , Membrane Proteins/drug effects , RNA-Binding Proteins/drug effects , RNA-Binding Proteins/metabolism , Trans-Activators/drug effects , Trans-Activators/metabolism , Animals , Antineoplastic Agents/chemistry , Drug Discovery , Humans , Mice , Molecular Docking Simulation , Neoplasm Metastasis/pathology , RNA-Binding Proteins/antagonists & inhibitors , Trans-Activators/antagonists & inhibitors
6.
Cell ; 181(3): 702-715.e20, 2020 04 30.
Article En | MEDLINE | ID: mdl-32315619

Protein phosphatase 2A (PP2A) enzymes can suppress tumors, but they are often inactivated in human cancers overexpressing inhibitory proteins. Here, we identify a class of small-molecule iHAPs (improved heterocyclic activators of PP2A) that kill leukemia cells by allosterically assembling a specific heterotrimeric PP2A holoenzyme consisting of PPP2R1A (scaffold), PPP2R5E (B56ε, regulatory), and PPP2CA (catalytic) subunits. One compound, iHAP1, activates this complex but does not inhibit dopamine receptor D2, a mediator of neurologic toxicity induced by perphenazine and related neuroleptics. The PP2A complex activated by iHAP1 dephosphorylates the MYBL2 transcription factor on Ser241, causing irreversible arrest of leukemia and other cancer cells in prometaphase. In contrast, SMAPs, a separate class of compounds, activate PP2A holoenzymes containing a different regulatory subunit, do not dephosphorylate MYBL2, and arrest tumor cells in G1 phase. Our findings demonstrate that small molecules can serve as allosteric switches to activate distinct PP2A complexes with unique substrates.


Protein Phosphatase 2/metabolism , Apoptosis , Cell Cycle Proteins/drug effects , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Enzyme Activators/metabolism , G1 Phase , Humans , Multiprotein Complexes/metabolism , Multiprotein Complexes/physiology , Phenothiazines/pharmacology , Phosphorylation , Protein Phosphatase 2/physiology , Protein Subunits/metabolism , Trans-Activators/drug effects , Trans-Activators/metabolism , Transcription Factors/metabolism
7.
BMC Res Notes ; 13(1): 168, 2020 Mar 19.
Article En | MEDLINE | ID: mdl-32192538

OBJECTIVE: The present study was carried out to investigate the transcriptional response of marA (Multiple antibiotic resistance A gene), soxS (Superoxide S gene) and rob (Right-origin-binding gene) under carbapenem stress. RESULTS: 12 isolates were found over-expressing AcrAB-TolC efflux pump system and showed reduced expression of OmpF (Outer membrane porin) gene were selected for further study. Among them, over expression of marA and rob was observed in 7 isolates. Increasing pattern of expression of marA and rob against meropenem was observed. The clones of marA and rob showed reduced susceptibility towards carbapenems.


Carbapenems/pharmacology , Cross Infection/microbiology , DNA-Binding Proteins/drug effects , Escherichia coli Infections/microbiology , Escherichia coli Proteins/drug effects , Escherichia coli , Regulon/drug effects , Trans-Activators/drug effects , Escherichia coli/drug effects , Escherichia coli/genetics , Escherichia coli/isolation & purification , Humans , India
8.
Cells ; 9(3)2020 03 04.
Article En | MEDLINE | ID: mdl-32143420

Regenerative medicine is a rapidly expanding area in research and clinical applications. Therapies involving the use of small molecule chemicals aim to simplify the creation of specific drugs for clinical applications. Adult mesenchymal stem cells have recently shown the capacity to differentiate into several cell types applicable for regenerative medicine (specifically neural cells, using chemicals). Valproic acid was an ideal candidate due to its clinical stability. It has been implicated in the induction of neural differentiation; however, the mechanism and the downstream events were not known. In this study, we showed that using valproic acid on adult mesenchymal stem cells induced neural differentiation within 24 h by upregulating the expression of suppressor of cytokine signaling 5 (SOCS5) and Fibroblast growth factor 21 (FGF21), without increasing the potential death rate of the cells. Through this, the Janus Kinase/Signal Transducer and Activator of Transcription (JAK/STAT) pathway is downregulated, and the mitogen-activated protein kinase (MAPK) cascade is activated. The bioinformatics analyses revealed the expression of several neuro-specific proteins as well as a range of functional and structural proteins involved in the formation and development of the neural cells.


Cell Differentiation/drug effects , DNA-Binding Proteins/drug effects , Mesenchymal Stem Cells/drug effects , Valproic Acid/pharmacology , DNA-Binding Proteins/metabolism , Humans , MAP Kinase Signaling System/drug effects , Mesenchymal Stem Cells/metabolism , Protein-Tyrosine Kinases/drug effects , Protein-Tyrosine Kinases/metabolism , Trans-Activators/drug effects , Trans-Activators/metabolism
9.
J Cell Physiol ; 235(11): 7889-7899, 2020 11.
Article En | MEDLINE | ID: mdl-31912905

The irreversible ERBB1/2/4 inhibitor neratinib causes plasma membrane-associated K-RAS to mislocalize into intracellular vesicles liminal to the plasma membrane; this effect is enhanced by HDAC inhibitors and is now a Phase I trial (NCT03919292). The combination of neratinib and HDAC inhibitors killed pancreatic cancer and lymphoma T cells. Neratinib plus HDAC inhibitor exposure was as efficacious as (paclitaxel+gemcitabine) at killing pancreatic cancer cells. Neratinib reduced the phosphorylation of PAK1, Merlin, LATS1/2, AKT, mTOR, p70 S6K, and ERK1/2 which required expression of Rubicon, Beclin1, and Merlin. Neratinib altered pancreatic tumor cell morphology which was associated with MST4 degradation reduced Ezrin phosphorylation and enhanced phosphorylation of MAP4K4 and LATS1/2. Knockdown of the MAP4K4 activator and sensor of membrane rigidity RAP2A reduced basal LATS1/2 and YAP phosphorylation but did not prevent neratinib from stimulating LATS1/2 or YAP phosphorylation. Beclin1 knockdown prevented MST4 degradation, Ezrin dephosphorylation and neratinib-induced alterations in tumor cell morphology. Our findings demonstrate that neratinib enhances LATS1/2 phosphorylation independently of RAP2A/MAP4K4 and that MST4 degradation and Ezrin dephosphorylation may represent a universal trigger for the biological actions of neratinib.


Antineoplastic Agents/pharmacology , Pancreatic Neoplasms/drug therapy , Quinolines/pharmacology , Signal Transduction/drug effects , Adaptor Proteins, Signal Transducing/drug effects , Adaptor Proteins, Signal Transducing/metabolism , Autophagy/drug effects , Cell Line, Tumor , Cell Membrane/drug effects , Humans , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/physiology , Pancreatic Neoplasms/metabolism , Phosphatidylinositol 3-Kinases/drug effects , Phosphatidylinositol 3-Kinases/metabolism , Protein Serine-Threonine Kinases/drug effects , Protein Serine-Threonine Kinases/metabolism , Trans-Activators/drug effects , Trans-Activators/metabolism , Transcription Factors/drug effects , Transcription Factors/metabolism , Transcriptional Coactivator with PDZ-Binding Motif Proteins , YAP-Signaling Proteins
10.
Int Microbiol ; 23(2): 215-224, 2020 May.
Article En | MEDLINE | ID: mdl-31342213

The increasing incidence of antimicrobial-resistant bacterial pathogens has focused researchers on quorum sensing inhibition strategies instead of those conventional approaches to fight bacterial infections. Anti-quorum sensing (QS) activity of aqueous extract from Forsythia suspense (FSE) was assessed, and its potential QS inhibition mechanisms were also analyzed. The minimal inhibitory concentration (MIC) of FSE to Chromobacterium violaceum 12472 is 0.5 mg mL-1. Inhibition of QS-regulated violacein production and biofilm formation in C. violaceum 12472 by FSE occurred in a concentration-dependent manner at sub-MIC, with > 70.12 and > 85.31% inhibition at 0.25 mg mL-1, respectively. N-Acyl homoserine lactones (AHLs) extracted from cultures of C. violaceum 31532 grown in the presence of FSE could not change the violacein production in C. violaceum 026, which indicated that FSE did not inhibit AHL synthesis. We also found that FSE cannot degrade AHLs. Finally, in silico molecular docking was conducted. The computed binding energy data suggested that components of F. suspense have a tendency to inhibit CviR with varying binding affinities and the energy score of Pinoresinol (- 26.02 kcal/mol) is higher than that of C6-HSL (- 16.09 kcal mol-1). We concluded that FSE acts as an antagonist of bacterial quorum sensing by competing with AHL receptor binding site.


Chromobacterium/drug effects , Forsythia , Plant Extracts/pharmacology , Quorum Sensing/drug effects , 4-Butyrolactone/analogs & derivatives , 4-Butyrolactone/metabolism , Anti-Bacterial Agents/pharmacology , Biofilms/drug effects , Indoles/metabolism , Microbial Sensitivity Tests , Molecular Docking Simulation/methods , Plant Extracts/chemistry , Repressor Proteins/drug effects , Repressor Proteins/metabolism , Trans-Activators/drug effects , Trans-Activators/metabolism
11.
Exp Clin Endocrinol Diabetes ; 128(2): 125-132, 2020 Feb.
Article En | MEDLINE | ID: mdl-30257265

BACKGROUND: Small for gestational age (SGA) due to intrauterine malnourishment is closely related to metabolic syndrome and type 2 diabetes mellitus. Growth Hormone (GH) treatment has been demonstrated to influence metabolic parameters and islet function of SGA individuals. The present study demonstrates the effects of early GH treatment on glucose tolerance and expression of pancreatic duodenal homeobox 1 (Pdx1) of SGA rats during adulthood. METHODS: SGA rat model was induced by restricting food intake during pregnancy. GH or normal saline was administered during postnatal days 21-35 of SGA rats and appropriate for gestational age (AGA) rats, respectively. RESULTS: In early adulthood (postnatal day 70), as compared to AGA rats, SGA rats showed: (1) decreased body weight; (2) increased postprandial blood glucose; and (3) down-regulated Pdx1 with increased histone deacetylase (HDAC) and down-regulated histone H3-lysine 4 methyltransferase SET7/9. Exogenous GH administration led to a restoration of body weight and normalized glucose tolerance due to an enhanced Pdx1 expression, accompanied by decreased HDAC and up-regulated SET7/9 in SGA rats in early adulthood. CONCLUSION: Our results demonstrate positive effects on glucose metabolism by an early and short GH treatment in SGA adulthood.


Body Weight/drug effects , Glucose Intolerance/drug therapy , Glucose Intolerance/metabolism , Growth Hormone/pharmacology , Homeodomain Proteins/drug effects , Homeodomain Proteins/metabolism , Infant, Small for Gestational Age/metabolism , Trans-Activators/drug effects , Trans-Activators/metabolism , Animals , Disease Models, Animal , Female , Male , Pregnancy , Rats , Rats, Sprague-Dawley
12.
Sci Rep ; 9(1): 13744, 2019 09 24.
Article En | MEDLINE | ID: mdl-31551455

Methicillin resistant Staphylococcus aureus (MRSA) is a predominant human pathogen with high morbidity that is listed in the WHO high priority pathogen list. Being a primary cause of persistent human infections, biofilm forming ability of S. aureus plays a pivotal role in the development of antibiotic resistance. Hence, targeting biofilm is an alternative strategy to fight bacterial infections. The present study for the first time demonstrates the non-antibacterial biofilm inhibitory efficacy of 5-Dodecanolide (DD) against ATCC strain and clinical isolates of S. aureus. In addition, DD is able to inhibit adherence of MRSA on human plasma coated Titanium surface. Further, treatment with DD significantly reduced the eDNA synthesis, autoaggregation, staphyloxanthin biosynthesis and ring biofilm formation. Reduction in staphyloxanthin in turn increased the susceptibility of MRSA to healthy human blood and H2O2 exposure. Quantitative PCR analysis revealed the induced expression of agrA and agrC upon DD treatment. This resulted down regulation of genes involved in biofilm formation such as fnbA and fnbB and up regulation of RNAIII, hld, psmα and genes involved in biofilm matrix degradation such as aur and nuc. Inefficacy of DD on the biofilm formation of agr mutant further validated the agr mediated antibiofilm potential of DD. Notably, DD was efficient in reducing the in vivo colonization of MRSA in Caenorhabditis elegans. Results of gene expression studies and physiological assays unveiled the agr mediated antibiofilm efficacy of DD.


Anti-Bacterial Agents/pharmacology , Bacterial Proteins/genetics , Biofilms/drug effects , Methicillin-Resistant Staphylococcus aureus/drug effects , Trans-Activators/drug effects , Up-Regulation/drug effects , Virulence/drug effects , Bacterial Proteins/drug effects , Gene Expression/drug effects , Humans , Hydrogen Peroxide/pharmacology , Microbial Sensitivity Tests/methods , Staphylococcal Infections/drug therapy , Staphylococcal Infections/microbiology , Virulence Factors
13.
Microb Pathog ; 132: 343-354, 2019 Jul.
Article En | MEDLINE | ID: mdl-31100406

In the present study, an attempt has been made to explore the antibiofilm activity of bioactive compound 1-hydroxy-1-norresistomycin (HNM) derived from coral mucus associated actinomycete Streptomyces variabilis. Initially, different concentration of HNM inhibited the biofilm formation of human clinical pathogens Escherichia coli, Vibrio cholerae and Staphylococcus aureus was determined using crystal-violet staining assay. The light microscopic analysis showed that HNM reduced the biofilm formation and adherence of bacterial cells on the surface of coverslip. HNM also damages the 3D architecture with reduced thickness as well as cell aggregation of biofilm forming bacteria analysed by confocal laser scanning microscopy (CLSM). In addition, HNM also demonstrated the efficiency in inhibiting theoretical adhesion by altering the surface hydrophobicity that can potentially hamper cellular adhesion and prevent biofilm formation. Furthermore, the molecular docking showed the significant interaction between HNM and key biofilm forming proteins proved an excellent antibiofilm activity of HNM. Together, these results suggest that the HNM can serve as potential antibiofilm agent in controlling the infections of E. coli, V. cholerae and S. aureus.


Biofilms/drug effects , Escherichia coli/drug effects , Pyrenes/pharmacology , Staphylococcus aureus/drug effects , Vibrio cholerae/drug effects , Actinobacteria/metabolism , Animals , Anthozoa/microbiology , AraC Transcription Factor/drug effects , Bacterial Adhesion/drug effects , Bacterial Proteins/drug effects , Computer Simulation , Cysteine Endopeptidases/drug effects , Escherichia coli Proteins/drug effects , Humans , Hydrogen Bonding , Molecular Docking Simulation , Pyrenes/chemistry , Streptomyces/metabolism , Trans-Activators/drug effects , Zebrafish
14.
Clin Transl Oncol ; 21(12): 1624-1633, 2019 Dec.
Article En | MEDLINE | ID: mdl-30963468

BACKGROUND: Synovial sarcoma (SS) is an aggressive soft-tissue sarcoma with a poor prognosis owing to its resistance to radiation and chemotherapy. Thus, novel therapeutic strategies for SS are urgently required. Anlotinib, a new oral tyrosine kinase inhibitor, is designed to primarily inhibit multi-targets in vasculogenesis and angiogenesis. This study was designed to characterize its antitumor efficacy and possible mechanism in patients with advanced refractory synovial sarcoma. METHODS: Anlotinib's antitumor effect was evaluated in vivo and vitro. Downstream targets of anlotinib in treating synovial sarcoma were analyzed through microarray assay. Cell proliferation and apoptosis analyses were performed to evaluate the impact of candidate downstream gene depletion in synovial sarcoma cells. Microarray assay were carried out to investigate potential signal network related with candidate downstream gene. RESULTS: Anlotinib significantly suppresses synovial sarcoma proliferation in PDTX model and cell lines. Additionally, GINS1 (also named as PSF1, Partner of SLD Five 1), rather than other conventional gene target, was demonstrated to be a vital target of anlotinib's antitumor effect in synovial sarcoma through microarray assay. Expression of GINS1 was remarkably higher in synovial sarcoma tumor samples and related with poor outcome. Knockdown of GINS1 expression could remarkably inhibit proliferation and promote apoptosis in vitro. Meanwhile, through microarray assay, CITED2, EGR1, SGK1 and SPP1 were identified and further validated by qPCR/WB as downstream targets of GINS1. CONCLUSION: Anlotinib might suppress proliferation of SS through a novel downstream GINS1-regulated network which plays a vital function in SS proliferation and also demonstrated that targeting the GINS1-regulated signal pathway could be a potential strategy for management of SS.


Bone Neoplasms/drug therapy , DNA-Binding Proteins/drug effects , Indoles/therapeutic use , Neoplasm Proteins/drug effects , Protein Kinase Inhibitors/therapeutic use , Quinolines/therapeutic use , Sarcoma, Synovial/drug therapy , Apoptosis/drug effects , Bone Neoplasms/genetics , Cell Proliferation/drug effects , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Disease Progression , Early Growth Response Protein 1/drug effects , Early Growth Response Protein 1/genetics , Early Growth Response Protein 1/metabolism , Gene Knockdown Techniques , Humans , Immediate-Early Proteins/drug effects , Immediate-Early Proteins/genetics , Immediate-Early Proteins/metabolism , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Osteopontin/drug effects , Osteopontin/genetics , Osteopontin/metabolism , Protein Array Analysis , Protein Serine-Threonine Kinases/drug effects , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , RNA, Messenger/analysis , Repressor Proteins/drug effects , Repressor Proteins/genetics , Repressor Proteins/metabolism , Sarcoma, Synovial/genetics , Trans-Activators/drug effects , Trans-Activators/genetics , Trans-Activators/metabolism
15.
Life Sci ; 220: 1-7, 2019 Mar 01.
Article En | MEDLINE | ID: mdl-30690083

AIMS: To investigate the role and underlying mechanism of 4E-BP1 and S6K1 in regulating autophagy and hepatitis B virus (HBV) replication. MAIN METHODS: The mRNA relative expression of HBx and its DNA level were detected by real-time PCR. The relative levels of hepatitis B surface antigen (HBsAg) were measured by enzyme-linked immunosorbent assay (ELISA). HBx DNA level of HepG2 cells transfected with pcDNA3.1(+)-HBV1.3 plasmids were detected by Southern blot. Moreover, we determined autophagy through electron microscopy, confocal microscopy and Western blot. KEY FINDINGS: Rapamycin promoted autophagy and the X protein synthesis concomitantly with elevation in Akt phosphorylation and Beclin1 expression. Either Beclin1 or Akt depletion suppresses the Rapa-enhanced HBV replication, whereas mTOR silencing inhibited HBV replication concurring with a decreased in both S6K1 and 4E-BP1 phosphorylation. Unexpectedly, Akt inhibitor suppressed Rapa-dependent autophagic flux and increased the level of p62/SQSTM1. While S6K1 ablation impaired autophagy and decreased X protein expression, 4E-BP1 silencing slightly influenced autophagy and increased X protein level. SIGNIFICANCE: The underlying mechanism of 4E-BP1 and S6K1, two main downstream effectors of mTOR, in mediating HBV replication and HBV-induced autophagy remains largely unknown. Here, we propose that Akt is required for both HBV replication and Rapa-induced autophagy, and 4E-BP1 and S6K1 play a distinct role in the virus replication and autophagic process.


Adaptor Proteins, Signal Transducing/physiology , Phosphoproteins/physiology , Ribosomal Protein S6 Kinases, 70-kDa/physiology , Virus Replication/drug effects , Adaptor Proteins, Signal Transducing/metabolism , Autophagy/drug effects , Cell Cycle Proteins , DNA Replication/drug effects , Hep G2 Cells , Hepatitis B Surface Antigens/metabolism , Hepatitis B virus/metabolism , Hepatitis B virus/physiology , Humans , Phosphoproteins/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Signal Transduction/drug effects , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/physiology , Trans-Activators/drug effects , Viral Regulatory and Accessory Proteins
16.
J Cell Physiol ; 234(7): 11923-11941, 2019 07.
Article En | MEDLINE | ID: mdl-30523639

Pyrroloquinoline quinone (PQQ) acts as a powerful modulator of PGC-1α activation and therefore regulates multiple pathways involved in cellular energy homeostasis. In the present study, we assessed the effects of L6 myotubes incubation with 0.5, 1, and 3 µM PQQ solution for 2 and 24 hr with respect to the cells' lipid metabolism. We demonstrated that PQQ significantly elevates PGC-1α content in a dose- and time-dependent manner with the highest efficiency for 0.5 and 1 µM. The level of free fatty acids was diminished (24 hr: -66%), while an increase in triacylglycerol (TAG) amount was most pronounced after 0.5 µM (2 hr: +93%, 24 hr: +139%) treatment. Ceramide (CER) content was elevated after 2 hr incubation with 0.5 µM and after prolonged exposure to all PQQ concentrations. The cells treated with PQQ for 2 hr exhibited decreased sphinganine (SFA) and sphinganine-1-phosphate (SFA1P) level, while 24 hr incubation resulted in an elevated sphingosine (SFO) amount. In summary, PGC-1α activation promotes TAG and CER synthesis.


Lipid Metabolism/physiology , Mitochondria/drug effects , Muscle Fibers, Skeletal/metabolism , PQQ Cofactor/pharmacology , Animals , Ceramides/metabolism , Lipid Metabolism/drug effects , Mitochondria/metabolism , Muscle Fibers, Skeletal/drug effects , Trans-Activators/drug effects , Trans-Activators/metabolism , Transcription Factors/drug effects , Transcription Factors/metabolism , Triglycerides/metabolism
17.
Skelet Muscle ; 8(1): 33, 2018 10 27.
Article En | MEDLINE | ID: mdl-30368256

BACKGROUND: The tetracycline-responsive system (Tet-ON/OFF) has proven to be a valuable tool for manipulating gene expression in an inducible, temporal, and tissue-specific manner. The purpose of this study was to create and characterize a new transgenic mouse strain utilizing the human skeletal muscle α-actin (HSA) promoter to drive skeletal muscle-specific expression of the reverse tetracycline transactivator (rtTA) gene which we have designated as the HSA-rtTA mouse. METHODS: To confirm the HSA-rtTA mouse was capable of driving skeletal muscle-specific expression, we crossed the HSA-rtTA mouse with the tetracycline-responsive histone H2B-green fluorescent protein (H2B-GFP) transgenic mouse in order to label myonuclei. RESULTS: Reverse transcription-PCR confirmed skeletal muscle-specific expression of rtTA mRNA, while single-fiber analysis showed highly effective GFP labeling of myonuclei in both fast- and slow-twitch skeletal muscles. Pax7 immunohistochemistry of skeletal muscle cross-sections revealed no appreciable GFP expression in satellite cells. CONCLUSIONS: The HSA-rtTA transgenic mouse allows for robust, specific, and inducible gene expression across muscles of different fiber types. The HSA-rtTA mouse provides a powerful tool to manipulate gene expression in skeletal muscle.


Gene Targeting/methods , Green Fluorescent Proteins/genetics , Muscle, Skeletal/metabolism , Tetracycline/pharmacology , Transgenes , Animals , Green Fluorescent Proteins/metabolism , Humans , Mice , Mice, Inbred C57BL , Muscle, Skeletal/pathology , PAX7 Transcription Factor/genetics , PAX7 Transcription Factor/metabolism , Trans-Activators/drug effects
18.
Endocrinology ; 159(11): 3643-3654, 2018 11 01.
Article En | MEDLINE | ID: mdl-30215691

Prolonged elevation of glucose can adversely affect ß-cell function. Oxidative stress, which has been implicated in glucose-induced ß-cell dysfunction, can activate c-jun N-terminal kinase (JNK). However, whether JNK is causal in glucose-induced ß-cell dysfunction in vivo is unclear. Therefore, we aimed at investigating the causal role of JNK activation in in vivo models of glucose-induced ß-cell dysfunction. Glucose-induced ß-cell dysfunction was investigated in the presence or absence of JNK inhibition. JNK inhibition was achieved using either (i) the JNK-specific inhibitor SP600125 or (ii) JNK-1-null mice. (i) Rats or mice were infused intravenously with saline or glucose with or without SP600125. (ii) JNK-1 null mice and their littermate wild-type controls were infused intravenously with saline or glucose. Following the glucose infusion periods in rats and mice, ß-cell function was assessed in isolated islets or in vivo using hyperglycemic clamps. Forty-eight-hour hyperglycemia at ~20 mM in rats or 96-hour hyperglycemia at ~13 mM in mice impaired ß-cell function in isolated islets and in vivo. Inhibition of JNK using either SP600125 or JNK-1-null mice prevented glucose-induced ß-cell dysfunction in isolated islets and in vivo. Islets of JNK-1-null mice exposed to hyperglycemia in vivo showed an increase in Pdx-1 and insulin 2 mRNA, whereas islets of wild-type mice did not. Together, these data show that JNK pathway is involved in glucose-induced ß-cell dysfunction in vivo and is thus a potential therapeutic target for type 2 diabetes.


Anthracenes/pharmacology , Glucose/pharmacology , Insulin-Secreting Cells/drug effects , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinase 8/genetics , Protein Kinase Inhibitors/pharmacology , Animals , Diabetes Mellitus, Type 2/metabolism , Glucose Clamp Technique , Homeodomain Proteins/drug effects , Homeodomain Proteins/genetics , Hyperglycemia/metabolism , In Vitro Techniques , Insulin/genetics , Insulin-Secreting Cells/metabolism , Mice , Mice, Knockout , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Rats , Signal Transduction , Trans-Activators/drug effects , Trans-Activators/genetics
19.
Endocrinology ; 159(9): 3143-3157, 2018 09 01.
Article En | MEDLINE | ID: mdl-29514186

Pharmacologic expansion of endogenous ß cells is a promising therapeutic strategy for diabetes. To elucidate the molecular pathways that control ß-cell growth we screened ∼2400 bioactive compounds for rat ß-cell replication-modulating activity. Numerous hit compounds impaired or promoted rat ß-cell replication, including CC-401, an advanced clinical candidate previously characterized as a c-Jun N-terminal kinase inhibitor. Surprisingly, CC-401 induced rodent (in vitro and in vivo) and human (in vitro) ß-cell replication via dual-specificity tyrosine phosphorylation-regulated kinase (DYRK) 1A and 1B inhibition. In contrast to rat ß cells, which were broadly growth responsive to compound treatment, human ß-cell replication was only consistently induced by DYRK1A/B inhibitors. This effect was enhanced by simultaneous glycogen synthase kinase-3ß (GSK-3ß) or activin A receptor type II-like kinase/transforming growth factor-ß (ALK5/TGF-ß) inhibition. Prior work emphasized DYRK1A/B inhibition-dependent activation of nuclear factor of activated T cells (NFAT) as the primary mechanism of human ß-cell-replication induction. However, inhibition of NFAT activity had limited effect on CC-401-induced ß-cell replication. Consequently, we investigated additional effects of CC-401-dependent DYRK1A/B inhibition. Indeed, CC-401 inhibited DYRK1A-dependent phosphorylation/stabilization of the ß-cell-replication inhibitor p27Kip1. Additionally, CC-401 increased expression of numerous replication-promoting genes normally suppressed by the dimerization partner, RB-like, E2F and multivulval class B (DREAM) complex, which depends upon DYRK1A/B activity for integrity, including MYBL2 and FOXM1. In summary, we present a compendium of compounds as a valuable resource for manipulating the signaling pathways that control ß-cell replication and leverage a DYRK1A/B inhibitor (CC-401) to expand our understanding of the molecular pathways that control ß-cell growth.


Cell Proliferation/drug effects , Glycogen Synthase Kinase 3 beta/antagonists & inhibitors , Insulin-Secreting Cells/drug effects , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/antagonists & inhibitors , Pyrazolones/pharmacology , Receptor, Transforming Growth Factor-beta Type I/antagonists & inhibitors , Adult , Animals , Cell Cycle Proteins/drug effects , Cell Cycle Proteins/metabolism , Cyclin-Dependent Kinase Inhibitor p27/drug effects , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Female , Forkhead Box Protein M1/drug effects , Forkhead Box Protein M1/metabolism , Humans , In Vitro Techniques , Kv Channel-Interacting Proteins/drug effects , Kv Channel-Interacting Proteins/metabolism , Male , Middle Aged , NFATC Transcription Factors/drug effects , NFATC Transcription Factors/metabolism , Rats , Repressor Proteins/drug effects , Repressor Proteins/metabolism , Trans-Activators/drug effects , Trans-Activators/metabolism , Transcription Factors/drug effects , Transcription Factors/metabolism , Dyrk Kinases
20.
Pathol Oncol Res ; 24(2): 407-417, 2018 Apr.
Article En | MEDLINE | ID: mdl-28577130

Simultaneous use of cisplatin (CIS) and gemcitabine (GEN) for treating bladder cancer has increased because of their complementary effects. However, the molecular mechanisms underlying the activities of these two antineoplastic drugs are not fully known. Here, molecular biology techniques and microscopy were used to investigate transcriptomic and morphological changes in low and high-grade urinary bladder transitional carcinoma cell lines [RT4 - wild type TP53; 5637 - two TP53 mutations, one in codon 72 (Arg-Pro) and other in codon 280 (Arg-Thr) and T24 - in-frame deletion of tyrosine 126 in the TP53 allele] simultaneously treated with CIS/GEN. Gene expression profile was evaluated by PCR arrays; cell morphology by scanning and transmission electron microscopy, and apoptosis was analyzed using fluorescent dye. Results showed concomitantly upregulation of CDKN2B (G1/S transition), GADD45A (DNA repair and apoptosis) and SERTAD1 (regulation of transcription) gene, increased number of nuclear chamfers and apoptotic cells, and reduced number of microfilaments, organelles and in the size of the nucleus in 5637 and T24 cells after simultaneous treatment with CIS/GEN. In conclusion, independently of the TP53 mutation status and tumor grade, CIS/GEN induced gene modulation accompanied by changes in cell morphologies, which confirm the antiproliferative activity of the treatment protocol. These findings help to understand the pathways modulated by these antineoplastic agents and may provide insights for anti-cancer chemotherapy.


Antineoplastic Combined Chemotherapy Protocols/pharmacology , Carcinoma, Transitional Cell/pathology , Gene Expression Regulation, Neoplastic/drug effects , Urinary Bladder Neoplasms/pathology , Apoptosis/drug effects , Carcinoma, Transitional Cell/metabolism , Cell Cycle/drug effects , Cell Cycle Proteins/biosynthesis , Cell Cycle Proteins/drug effects , Cell Line, Tumor , Cisplatin/pharmacology , Cyclin-Dependent Kinase Inhibitor p15/biosynthesis , Cyclin-Dependent Kinase Inhibitor p15/drug effects , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Humans , Mutation , Nuclear Proteins/biosynthesis , Nuclear Proteins/drug effects , Trans-Activators/biosynthesis , Trans-Activators/drug effects , Transcription Factors , Tumor Suppressor Protein p53/genetics , Up-Regulation , Urinary Bladder Neoplasms/metabolism , Gemcitabine
...