Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
Mol Biol Rep ; 48(2): 1651-1658, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33580460

ABSTRACT

SVCT2, Sodium-dependent Vitamin C Transporter 2, uniquely transports ascorbic acid (also known as vitamin C and ascorbate) into all types of cells. Vitamin C is an essential nutrient that must be obtained through the diet and plasma levels are tightly regulated by transporter activity. Vitamin C plays an important role in antioxidant defenses and is a cofactor for many enzymes that enable hormone synthesis, oxygen sensing, collagen synthesis and epigenetic pathways. Although SVCT2 has various functions, regulation of its expression/activity remains poorly understood. We found a p53-binding site, within the SVCT2 promoter, using a transcription factor binding-site prediction tool. In this study, we show that p53 can directly repress SVCT2 transcription by binding a proximal- (~-185 to -171 bp) and a distal- (~-1800 to -1787 bp) p53-responsive element (PRE), Chromatin immunoprecipitation assays showed that PRE-bound p53 interacts with the corepressor-histone deacetylase 3 (HDAC3), resulting in deacetylation of histones Ac-H4, at the proximal promoter, resulting in transcriptional silencing of SVCT2. Overall, our data suggests that p53 is a potent transcriptional repressor of SVCT2, a critical transporter of diet-derived ascorbic acid, across the plasma membranes of numerous essential tissue cell types.


Subject(s)
Antioxidants/metabolism , Histone Deacetylases/genetics , Sodium-Coupled Vitamin C Transporters/genetics , Tumor Suppressor Protein p53/genetics , Animals , Ascorbic Acid/genetics , Ascorbic Acid/metabolism , Binding Sites/genetics , Chromatin/genetics , Fibroblasts , Hep G2 Cells , Humans , Mice , Protein Binding , Repressor Proteins/genetics , Sodium-Coupled Vitamin C Transporters/antagonists & inhibitors
2.
Biochem Biophys Res Commun ; 527(1): 305-310, 2020 06 18.
Article in English | MEDLINE | ID: mdl-32446385

ABSTRACT

Non-small lung cancer (NSCLC) is the most common cancer in the world. The epidermal growth factor receptor (EGFR) gene is mutated in approximately 10% of lung cancer cases in the US and 50% of lung cancer in Asia. The representative target therapeutic agent, erlotinib (EGFR tyrosine kinase inhibitor; EGFR TKI), is effective in inactivating EGFR in lung cancer patients. However, approximately 50-60% of patients are resistant to EGFR TKI. These populations are associated with the EGFR mutation. To overcome resistance to EGFR TKI, we discovered a JAK1 inhibitor, CJ14939. We investigated the efficacy of CJ14939 in human NSCLC cell lines in vitro and in vivo. Our results showed that CJ14939 induced the inhibition of cell growth. Moreover, we demonstrated that combination treatment with erlotinib and CJ14939 induced cell death in vitro and inhibited tumor growth in vivo. In addition, we confirmed the suppression of phosphorylated EGFR, JAK1, and Stat3 expression in erlotinib and CJ14939-treated human NSCLC cell lines. Our results provide evidence that JAK inhibition overcomes resistance to EGFR TKI in human NSCLCs.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Non-Small-Cell Lung/drug therapy , Erlotinib Hydrochloride/pharmacology , Janus Kinase 1/antagonists & inhibitors , Lung Neoplasms/drug therapy , Protein Kinase Inhibitors/pharmacology , Animals , Antineoplastic Agents/chemistry , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Death/drug effects , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Resistance, Neoplasm/drug effects , Drug Screening Assays, Antitumor , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/genetics , ErbB Receptors/metabolism , Erlotinib Hydrochloride/chemistry , Female , Humans , Janus Kinase 1/metabolism , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Mice , Mice, Nude , Molecular Structure , Mutation , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Protein Kinase Inhibitors/chemistry , Structure-Activity Relationship , Tumor Cells, Cultured
3.
Invest New Drugs ; 38(6): 1696-1706, 2020 12.
Article in English | MEDLINE | ID: mdl-32462369

ABSTRACT

Inhibitor of apoptosis proteins (IAPs) are overexpressed in the majority of cancers and prevent apoptosis by inhibiting caspases. IAPs have therefore attracted considerable attention as potential targets for anticancer therapy. Here, we demonstrated that HM90822 (abbreviated HM822; a new synthetic IAP antagonist) induced apoptotic cell death via proteasome-dependent degradation of BIR2/3 domain-containing IAPs in human pancreatic cancer cells. HM822 inhibited the expression of XIAP and cIAP1/2 proteins in Panc-1 and BxPC-3 cells, which are sensitive to HM822. HM822 also induced IAP ubiquitination and promoted proteasome-dependent IAP degradation. However, cells expressing phospho-XIAP (Ser87) and AKT exhibited resistance to HM822. In other words, the overexpression of AKT-CA (constitutive active form for AKT) or AKT-WT induced resistance to HM822. In addition, in Panc-1 xenograft and orthotopic mouse models, we revealed that tumor growth was suppressed by the administration of HM822. Taken together, these results suggest that HM822 induces apoptosis through ubiquitin/proteasome-dependent degradation of BIR3 domain-containing IAPs. These findings suggest that phospho-XIAP and phospho-AKT may be used as biomarkers for predicting the efficacy of HM822 in pancreatic cancer patients.


Subject(s)
Antineoplastic Agents/therapeutic use , Inhibitor of Apoptosis Proteins/antagonists & inhibitors , Pancreatic Neoplasms/drug therapy , Animals , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Drug Resistance, Neoplasm , Female , Humans , Inhibitor of Apoptosis Proteins/genetics , Inhibitor of Apoptosis Proteins/metabolism , Mice, Inbred BALB C , Mice, Nude , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Tumor Burden/drug effects , Ubiquitination/drug effects
4.
Br J Cancer ; 120(9): 941-951, 2019 04.
Article in English | MEDLINE | ID: mdl-30944457

ABSTRACT

BACKGROUND: Mitogen-activated protein kinases (MEK 1/2) are central components of the RAS signalling pathway and are attractive targets for cancer therapy. These agents continue to be investigated in KRAS mutant colon cancer but are met with significant resistance. Clinical investigations have demonstrated that these strategies are not well tolerated by patients. METHODS: We investigated a biomarker of response for MEK inhibition in KRAS mutant colon cancers by LC-MS/MS analysis. We tested the MEK inhibitor in PIK3CA wild(wt) and mutant(mt) colon cancer cells. In addition, we tested the combinational effects of MEK and TNKS inhibitor in vitro and in vivo. RESULTS: We identified ß-catenin, a key mediator of the WNT pathway, in response to MEK inhibitor. MEK inhibition led to a decrease in ß-catenin in PIK3CA wt colon cancer cells but not in mt. Tumour regression was promoted by combination of MEK inhibition and NVP-TNS656, which targets the WNT pathway. Furthermore, inhibition of MEK promoted tumour regression in colon cancer patient-derived xenograft models expressing PIK3CA wt. CONCLUSIONS: We propose that inhibition of the WNT pathway, particularly ß-catenin, may bypass resistance to MEK inhibition in human PIK3CA mt colon cancer. Therefore, we suggest that ß-catenin is a potential predictive marker of MEK inhibitor resistance.


Subject(s)
Class I Phosphatidylinositol 3-Kinases/genetics , Colonic Neoplasms/drug therapy , Colonic Neoplasms/genetics , MAP Kinase Kinase 1/antagonists & inhibitors , MAP Kinase Kinase 3/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins p21(ras)/genetics , beta Catenin/metabolism , Acetamides/pharmacology , Animals , Biomarkers, Pharmacological/metabolism , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Class I Phosphatidylinositol 3-Kinases/antagonists & inhibitors , Class I Phosphatidylinositol 3-Kinases/metabolism , Colonic Neoplasms/metabolism , Drug Resistance, Viral , Humans , MAP Kinase Kinase 1/metabolism , MAP Kinase Kinase 3/metabolism , Mice , Mice, Inbred BALB C , Mice, Nude , Proto-Oncogene Proteins p21(ras)/antagonists & inhibitors , Proto-Oncogene Proteins p21(ras)/metabolism , Pyrimidinones/pharmacology , Xenograft Model Antitumor Assays , beta Catenin/antagonists & inhibitors
5.
Mol Cell ; 44(4): 545-58, 2011 Nov 18.
Article in English | MEDLINE | ID: mdl-22099303

ABSTRACT

Cellular antioxidant enzymes play crucial roles in aerobic organisms by eliminating detrimental oxidants and maintaining the intracellular redox homeostasis. Therefore, the function of antioxidant enzymes is inextricably linked to the redox-dependent activities of multiple proteins and signaling pathways. Here, we report that the VEGFR2 RTK has an oxidation-sensitive cysteine residue whose reduced state is preserved specifically by peroxiredoxin II (PrxII) in vascular endothelial cells. In the absence of PrxII, the cellular H(2)O(2) level is markedly increased and the VEGFR2 becomes inactive, no longer responding to VEGF stimulation. Such VEGFR2 inactivation is due to the formation of intramolecular disulfide linkage between Cys1199 and Cys1206 in the C-terminal tail. Interestingly, the PrxII-mediated VEGFR2 protection is achieved by association of two proteins in the caveolae. Furthermore, PrxII deficiency suppresses tumor angiogenesis in vivo. This study thus demonstrates a physiological function of PrxII as the residential antioxidant safeguard specific to the redox-sensitive VEGFR2.


Subject(s)
Antioxidants/metabolism , Aorta/enzymology , Endothelial Cells/enzymology , Endothelium, Vascular/enzymology , Neovascularization, Pathologic/enzymology , Peroxiredoxins , Vascular Endothelial Growth Factor Receptor-2 , Animals , Aorta/cytology , Carcinoma, Lewis Lung/enzymology , Carcinoma, Lewis Lung/pathology , Caveolae/enzymology , Cysteine/chemistry , Cysteine/metabolism , Disulfides/chemistry , Disulfides/metabolism , Endothelial Cells/cytology , Endothelium, Vascular/cytology , Gene Silencing , Humans , Hydrogen Peroxide/metabolism , Mice , Mice, Knockout , Mutagenesis, Site-Directed , Neoplasm Transplantation , Neovascularization, Pathologic/genetics , Oxidation-Reduction , Peroxiredoxins/antagonists & inhibitors , Peroxiredoxins/genetics , Peroxiredoxins/metabolism , Protein Binding , Protein Structure, Tertiary , RNA, Small Interfering , Reactive Oxygen Species/metabolism , Signal Transduction , Vascular Endothelial Growth Factor Receptor-2/genetics , Vascular Endothelial Growth Factor Receptor-2/metabolism
6.
Biochem Biophys Res Commun ; 473(1): 290-295, 2016 Apr 22.
Article in English | MEDLINE | ID: mdl-27012203

ABSTRACT

Hyaluronan has diverse biological activities depending on its molecular size. The hyaluronan fragments (50 kDa) can decrease adipogenic differentiation in vitro. However, in vivo anti-obesitic effects of hyaluronan fragments have not been elucidated. Therefore, we examined the anti-obesity effects of hyaluronan fragments on high-fat diet induced obesity in C57BL/6 mice. Oral administration of hyaluronan fragments (200 mg/kg for 8 weeks) decreased body weight, adipose tissues, serum lipid (low-density lipoprotein cholesterol, triglyceride), and leptin level. Hyaluronan fragments decreased the hypertrophy of adipose tissue and ameliorated liver steatosis. The mRNA expression of leptin was reduced in adipocyte by treatment with hyaluronan fragments. Additionally, hyaluronan fragments enhanced the mRNA expression of PPAR-α and its target genes UCP-2 and decreased mRNA expression of PPAR- γ and fatty acid synthase in liver. In conclusions, hyaluronan fragments had marked effects on inhibiting the development of obesity in obese mice fed the high-fat diet. It suggested that enhancing PPAR-α and suppressing PPAR-γ expression are two possible mechanisms for the anti-obesitic effect of hyaluronan fragments.


Subject(s)
Diet, High-Fat/adverse effects , Hyaluronic Acid/pharmacology , Obesity/therapy , Adipocytes/metabolism , Adiponectin/blood , Animals , Body Weight , Fatty Liver/pathology , Hyperlipidemias/metabolism , Leptin/blood , Leptin/metabolism , Lipid Metabolism , Lipids/blood , Lipoproteins, LDL/blood , Liver/metabolism , Mice , Mice, Inbred C57BL , Mice, Obese , Molecular Weight , Obesity/physiopathology , PPAR alpha/metabolism , Real-Time Polymerase Chain Reaction , Triglycerides/blood
7.
Biochem Biophys Res Commun ; 467(4): 623-8, 2015 Nov 27.
Article in English | MEDLINE | ID: mdl-26525853

ABSTRACT

Hyaluronan has diverse biological activities depending on its molecular size. High molecular weight hyaluronan (2000 kDa) is a major component of extracellular matrix, and has been used in wounding healing, extracellular matrix regeneration, and in the treatment of osteoarthritis. Hyaluronan fragments can stimulate inflammation or induce loss of extracellular matrix. Hyaluronan is expressed during adipocyte differentiation, and down regulation of hyaluronan synthesis can reduce adipogenic differentiation. However, the direct effects of hyaluronan fragments on adipocyte differentiation have not been elucidated. Therefore, we prepared hyaluronan fragments by enzymatic digestion, and examined the inhibitory effects of these hyaluronan fragments on the accumulation of lipid droplets and on adipogenic gene mRNA expression in differentiating 3T3-L1 pre-adipocytes. Medium sized hyaluronan fragments (50 kDa) decreased lipid droplet accumulation in a dose-dependent manner. However, high molecular weight hyaluronan did not inhibit lipid droplet accumulation when used at a concentration of 600 µg/ml. Two or 4 day treatments with medium molecular weight of hyaluronan resulted in similar inhibitory levels of lipid accumulation as did treatment for 8 days. Medium sized hyaluronan inhibited the differentiation of 3T3-L1 pre-adipocytes during the early stages of adipogenesis. When 3T3-L1 cells were treated with 180 µg/ml of medium sized hyaluronan, the mRNAs for the master adipogenic transcription factors PPAR-γ and C/EBP-α were inhibited. Additionally, medium molecular weight hyaluronan suppressed mRNA expression of PPAR-γ target genes, including aP2 and FAS. This study is the first to report that medium molecular weight hyaluronan fragments can inhibit adipocyte differentiation.


Subject(s)
Adipocytes/drug effects , Cell Differentiation/drug effects , Hyaluronic Acid/pharmacology , 3T3-L1 Cells , Adipocytes/cytology , Animals , Hyaluronic Acid/chemistry , Mice , Molecular Weight
8.
Bioorg Med Chem Lett ; 24(15): 3346-50, 2014 Aug 01.
Article in English | MEDLINE | ID: mdl-24961640

ABSTRACT

A newly designed curcumin mimic library (11a-11k) with 2-ethylamino groups in a chalcone structure and variously substituted triazole groups as side chains was synthesized using the Huisgen 1,3-cycloaddition reaction between various alkynes (a-k) and an intermediate (10), with CuSO4 and sodium ascorbate in a solution mixture of chloroform, ethanol, and water (5:3:1) at room temperature for 5h. In the lactate dehydrogenase (LDH) release assay involving co-treatment with tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) and/or synthetic curcumin derivatives using TRAIL-resistant human CRT-MG astroglioma cells, the novel curcumin mimic library was found to effectively stimulate the cytotoxicity of TRAIL, causing mild cytotoxicity when administered alone. In particular, 11a and 11j are promising candidates for TRAIL-sensitizers with potential use in combination chemotherapy for brain tumors.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Brain Neoplasms/drug therapy , Curcumin/chemistry , Diethylamines/chemistry , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Triazoles/chemistry , Antineoplastic Combined Chemotherapy Protocols/chemical synthesis , Antineoplastic Combined Chemotherapy Protocols/chemistry , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Molecular Structure , Structure-Activity Relationship , TNF-Related Apoptosis-Inducing Ligand/chemical synthesis , TNF-Related Apoptosis-Inducing Ligand/chemistry
9.
Heliyon ; 10(7): e28485, 2024 Apr 15.
Article in English | MEDLINE | ID: mdl-38596107

ABSTRACT

Background: Remimazolam has recently been introduced as a maintenance agent for general anesthesia. However, the effect of remimazolam on peripartum prognosis has not been reported. Therefore, this study aimed to compare the effects of remimazolam and propofol for uterotonic drugs following cesarean section. Methods: The electronic medical records of 51 adult women who underwent elective cesarean sections by single obstetrician under general anesthesia were collected. Participants were categorized into two groups: the propofol group and the remimazolam group. General anesthesia was maintained by continuous infusion of propofol or remimazolam after delivery. The number of uterotonic drugs administered during the cesarean section, the estimated blood loss (EBL), and length of hospital stay (LOS) after delivery were assessed. Results: Of the 51 patients included in the study, 35 were in the propofol group and 16 in the remimazolam group. In the remimazolam group, five patients (31.3%, 5/16) received more uterotonics than the standard regimen. Conversely, in the propofol group, 19 patients (54.3%, 19/35) were injected with more uterotonics than the standard regimen. Logistic regression analysis showed that abnormal positioning of the placenta (P = 0.079) and not using remimazolam (P = 0.100) were the most relevant factors associated with the increased use of uterotonics. There was no significant difference in EBL between the two groups. The use of remimazolam was clinically relevant with a shorter LOS (P = 0.059). Conclusions: The use of remimazolam as a maintenance agent did not result in significantly higher use of intrapartum uterotonics compared to the use of propofol. These results cannot exclude all adverse effects of remimazolam during cesarean delivery. Further randomized controlled trials must be conducted to obtain high-quality evidence.

10.
Cancer Immunol Res ; 12(4): 491-507, 2024 Apr 02.
Article in English | MEDLINE | ID: mdl-38289363

ABSTRACT

The development of first-generation immune-checkpoint inhibitors targeting PD-1/PD-L1 and CTLA-4 ushered in a new era in anticancer therapy. Although immune-checkpoint blockade therapies have shown clinical success, a substantial number of patients yet fail to benefit. Many studies are under way to discover next-generation immunotherapeutic targets. Immunoglobulin superfamily member 1 (IGSF1) is a membrane glycoprotein proposed to regulate thyroid function. Despite containing 12 immunoglobin domains, a possible role for IGSF1, in immune response, remains unknown. Here, our studies revealed that IGSF1 is predominantly expressed in tumors but not normal tissues, and increased expression is observed in PD-L1low non-small cell lung cancer (NSCLC) cells as compared with PD-L1high cells. Subsequently, we developed and characterized an IGSF1-specific human monoclonal antibody, WM-A1, that effectively promoted antitumor immunity and overcame the limitations of first-generation immune-checkpoint inhibitors, likely via a distinct mechanism of action. We further demonstrated high WM-A1 efficacy in humanized peripheral blood mononuclear cells (PBMC), and syngeneic mouse models, finding additive efficacy in combination with an anti-PD-1 (a well-characterized checkpoint inhibitor). These findings support IGSF1 as an immune target that might complement existing cancer immunotherapeutics.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Immunoglobulins , Lung Neoplasms , Membrane Proteins , Animals , Humans , Mice , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , B7-H1 Antigen , Immune Checkpoint Inhibitors/therapeutic use , Immunoglobulins/metabolism , Immunotherapy , Leukocytes, Mononuclear , Lung Neoplasms/drug therapy , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL