Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 96
Filter
1.
Cell ; 161(3): 595-609, 2015 Apr 23.
Article in English | MEDLINE | ID: mdl-25892225

ABSTRACT

Organisms must be able to respond to low oxygen in a number of homeostatic and pathological contexts. Regulation of hypoxic responses via the hypoxia-inducible factor (HIF) is well established, but evidence indicates that other, HIF-independent mechanisms are also involved. Here, we report a hypoxic response that depends on the accumulation of lactate, a metabolite whose production increases in hypoxic conditions. We find that the NDRG3 protein is degraded in a PHD2/VHL-dependent manner in normoxia but is protected from destruction by binding to lactate that accumulates under hypoxia. The stabilized NDRG3 protein binds c-Raf to mediate hypoxia-induced activation of Raf-ERK pathway, promoting angiogenesis and cell growth. Inhibiting cellular lactate production abolishes the NDRG3-mediated hypoxia responses. Our study, therefore, elucidates the molecular basis for lactate-induced hypoxia signaling, which can be exploited for the development of therapies targeting hypoxia-induced diseases.


Subject(s)
Hypoxia/metabolism , Lactic Acid/metabolism , Cell Hypoxia , Cell Line , Gene Expression Regulation , Humans , Hypoxia-Inducible Factor-Proline Dioxygenases/metabolism , Intracellular Signaling Peptides and Proteins , MAP Kinase Signaling System , Neovascularization, Pathologic/metabolism , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/metabolism , Oxygen/metabolism , Protein Binding , raf Kinases/metabolism
2.
Luminescence ; 38(7): 1330-1338, 2023 Jul.
Article in English | MEDLINE | ID: mdl-36519806

ABSTRACT

Uric acid (2,6,8-trihydroxypurine) is a metabolic product of purine, which is one of the important markers of human health. The development of a rapid, facile, highly sensitive, and selective method for uric acid detection is critical for the diagnosis of related diseases and is still a strategic challenge. In this study, we developed a highly sensitive and selective colorimetric assay for the detection of uric acid using biogenic palladium nanoparticles (Pd NPs). The synthesized nanoparticles were shown to acquire peroxidase mimetic activity that oxidized 3,3',5,5'-tetramethylbenzidine and produced a blue colour in an assay. The developed colorimetric assay is instrument-free detection of uric acid with a limit of detection of 0.05 µM and a 1.11 µM limit of quantification (LOQ). This is the first report determining the LOQ for a colorimetric assay that gives the lowest quantity of analyte that can be evaluated with more precision under the specified conditions of the analysis. The developed assay had a linear response at low uric acid concentrations of 0.05 to 1 µM and a 0.99841 linear regression correlation coefficient. This colorimetric detection provides a rapid, cost-effective, and easy-to-use platform for the clinical diagnosis of uric acid biomarkers.


Subject(s)
Metal Nanoparticles , Uric Acid , Humans , Peroxidase/metabolism , Colorimetry/methods , Palladium , Hydrogen Peroxide/analysis
3.
Chem Biodivers ; 19(1): e202100600, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34725898

ABSTRACT

Protein tyrosine phosphatases (PTPs) are essential modulators of signal transduction pathways and has been implicated in many human diseases such as cancer, diabetes, obesity, autoimmune disorders, and neurological diseases, indicating that PTPs are next-generation drug targets. Since PTPN1, PTPN2, and PTPN11 have been reported to be negative regulators of insulin action, the identification of PTP inhibitors may be an effective strategy to develop therapeutic agents for the treatment of type 2 diabetes. In this study, we observed for the first time that nepetin inhibits the catalytic activity of PTPN1, PTPN2, and PTPN11 in vitro, indicating that nepetin acts as a multi-targeting inhibitor of PTPN1, PTPN2, and PTPN11. Furthermore, treatment of mature 3T3-L1 adipocytes with 20 µM nepetin stimulates glucose uptake through AMPK activation. Taken together, our findings provide evidence that nepetin, a multi-targeting inhibitor of PTPN1, PTPN2, and PTPN11, could be a promising therapeutic candidate for the treatment of type 2 diabetes.


Subject(s)
Enzyme Inhibitors/chemistry , Flavones/chemistry , Protein Tyrosine Phosphatases/antagonists & inhibitors , 3T3-L1 Cells , AMP-Activated Protein Kinases/metabolism , Animals , Biocatalysis , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/pathology , Enzyme Inhibitors/metabolism , Enzyme Inhibitors/therapeutic use , Flavones/metabolism , Flavones/therapeutic use , Glucose/metabolism , Humans , Insulin Resistance , Mice , Phosphorylation/drug effects , Protein Tyrosine Phosphatase, Non-Receptor Type 1/antagonists & inhibitors , Protein Tyrosine Phosphatase, Non-Receptor Type 1/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 11/antagonists & inhibitors , Protein Tyrosine Phosphatase, Non-Receptor Type 11/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 2/antagonists & inhibitors , Protein Tyrosine Phosphatase, Non-Receptor Type 2/metabolism , Protein Tyrosine Phosphatases/metabolism
4.
Int J Mol Sci ; 23(2)2022 Jan 13.
Article in English | MEDLINE | ID: mdl-35055051

ABSTRACT

Depletion of protein phosphatase-1 catalytic subunit beta (PPP1CB), a serine/threonine protein phosphatase and potent adipogenic activator, suppresses the differentiation of 3T3-L1 preadipocytes into mature adipocytes. Therefore, PPP1CB is considered as a potential therapeutic target for obesity. We screened 1033 natural products for PPP1CB inhibitors and identified chebulinic acid, which is abundantly present in the seeds of Euphoria longana and fruits of Terminalia chebula. Chebulinic acid strongly inhibited the hydrolysis of 6,8-difluoro-4-methylumbelliferyl phosphate by PPP1CB (IC50 = 300 nM) and demonstrated potent antiadipogenic effects in 3T3-L1 preadipocytes in a concentration-dependent manner. Additional studies have demonstrated that chebulinic acid suppresses early differentiation by downregulating key transcription factors that control adipogenesis in 3T3-L1 cells. These results suggested that chebulinic acid may be a potential therapeutic agent for treating obesity by inhibiting PPP1CB activity.


Subject(s)
Adipocytes/drug effects , Adipocytes/metabolism , Adipogenesis/drug effects , Hydrolyzable Tannins/pharmacology , Protein Phosphatase 1/antagonists & inhibitors , 3T3-L1 Cells , Adipocytes/cytology , Adipogenesis/genetics , Adipokines/genetics , Adipokines/metabolism , Animals , Anti-Obesity Agents/pharmacology , Biological Products/pharmacology , Cell Survival/drug effects , Enzyme Activation/drug effects , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Gene Expression Regulation/drug effects , Hydrolyzable Tannins/chemistry , Mice , Molecular Structure , Protein Phosphatase 1/metabolism , Recombinant Proteins , Transcription Factors/genetics , Transcription Factors/metabolism
5.
Int J Mol Sci ; 23(7)2022 Apr 01.
Article in English | MEDLINE | ID: mdl-35409287

ABSTRACT

Ginkgolic acid (C13:0) (GA), isolated from Ginkgo biloba, is a potential therapeutic agent for type 2 diabetes. A series of GA analogs were designed and synthesized for the evaluation of their structure-activity relationship with respect to their antidiabetic effects. Unlike GA, the synthetic analog 1e exhibited improved inhibitory activity against PTPN9 and significantly stimulated glucose uptake via AMPK phosphorylation in differentiated 3T3-L1 adipocytes and C2C12 myotubes; it also induced insulin-dependent AKT activation in C2C12 myotubes in a concentration-dependent manner. Docking simulation results showed that 1e had a better binding affinity through a unique hydrophobic interaction with a PTPN9 hydrophobic groove. Moreover, 1e ameliorated palmitate-induced insulin resistance in C2C12 cells. This study showed that 1e increases glucose uptake and suppresses palmitate-induced insulin resistance in C2C12 myotubes via PTPN9 inhibition; thus, it is a promising therapeutic candidate for treating type 2 diabetes.


Subject(s)
Diabetes Mellitus, Type 2 , Insulin Resistance , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/metabolism , Glucose/metabolism , Humans , Insulin/metabolism , Muscle Fibers, Skeletal/metabolism , Palmitates/metabolism , Salicylates , Signal Transduction , Structure-Activity Relationship
6.
Int J Mol Sci ; 23(9)2022 Apr 30.
Article in English | MEDLINE | ID: mdl-35563411

ABSTRACT

The emergence of the high correlation between type 2 diabetes and obesity with complicated conditions has led to the coinage of the term "diabesity". AMP-activated protein kinase (AMPK) activators and peroxisome proliferator-activated receptor (PPARγ) antagonists have shown therapeutic activity for diabesity, respectively. Hence, the discovery of compounds that activate AMPK as well as antagonize PPARγ may lead to the discovery of novel therapeutic agents for diabesity. In this study, the knockdown of PTPN6 activated AMPK and suppressed adipogenesis in 3T3-L1 cells. By screening a library of 1033 natural products against PTPN6, we found ethyl gallate to be the most selective inhibitor of PTPN6 (Ki = 3.4 µM). Subsequent assay identified ethyl gallate as the best PPARγ antagonist (IC50 = 5.4 µM) among the hit compounds inhibiting PTPN6. Ethyl gallate upregulated glucose uptake and downregulated adipogenesis in 3T3-L1 cells as anticipated. These results strongly suggest that ethyl gallate, which targets both PTPN6 and PPARγ, is a potent therapeutic candidate to combat diabesity.


Subject(s)
Diabetes Mellitus, Type 2 , Gallic Acid , PPAR gamma , Protein Tyrosine Phosphatase, Non-Receptor Type 6 , 3T3-L1 Cells , AMP-Activated Protein Kinases/metabolism , Adipocytes/metabolism , Adipogenesis , Animals , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/metabolism , Gallic Acid/analogs & derivatives , Gallic Acid/pharmacology , Mice , Obesity/drug therapy , Obesity/metabolism , PPAR gamma/drug effects , PPAR gamma/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 6/drug effects , Protein Tyrosine Phosphatase, Non-Receptor Type 6/metabolism
7.
Chembiochem ; 22(5): 830-833, 2021 03 02.
Article in English | MEDLINE | ID: mdl-33103305

ABSTRACT

The protein p73 acts as a transcription factor, resulting in tumour suppression. MDM2, an oncogenic protein, can negatively influence p73-mediated apoptosis by binding to p73 transactivation domains (TAD). Inhibition of the protein-protein interaction between p73 and oncogenic proteins is an attractive strategy for promoting p73-mediated apoptosis. Herein, we describe the use of a modified p73-TAD peptide for the FRET-based assay of the binding of p73-TAD to MDM2. The FRET probe, equipped with 1-naphthylamine (λex =330 nm, λem =445 nm), serves as a FRET acceptor, and the tryptophan of the protein acts as FRET donor (λex =280 nm, λem =340 nm). Sensitized emission from the FRET probe was observed upon excitation of the protein-FRET-probe complex at the excitation wavelength of Trp. Furthermore, addition of the MDM2 inhibitor Nutiln-3 drastically reduced the FRET signal, thus indicating that the FRET probe competes with Nutiln-3 for MDM2 binding. The developed FRET binding assay might be applicable in high-throughput screening of novel drugs that inhibit interactions between p73 and MDM2.


Subject(s)
Antineoplastic Agents/pharmacology , Fluorescence Resonance Energy Transfer/methods , Fluorescent Dyes/chemistry , Protein Interaction Domains and Motifs/drug effects , Proto-Oncogene Proteins c-mdm2/antagonists & inhibitors , Tumor Protein p73/antagonists & inhibitors , Humans , Protein Binding , Proto-Oncogene Proteins c-mdm2/metabolism , Tumor Protein p73/metabolism
8.
Molecules ; 26(4)2021 Feb 05.
Article in English | MEDLINE | ID: mdl-33562835

ABSTRACT

Natural products have continued to offer tremendous opportunities for drug development, as they have long been used in traditional medicinal systems. SHP2 has served as an anticancer target. To identify novel SHP2 inhibitors with potential anticancer activity, we screened a library containing 658 natural products. Polyphyllin D was found to selectively inhibit SHP2 over SHP1, whereas two other identified compounds (echinocystic acid and oleanolic acid) demonstrated dual SHP1 and SHP2 inhibition. In a cell-based assay, polyphyllin D exhibited cytotoxicity in Jurkat cells, an acute lymphoma leukemia cell line, whereas the other two compounds were ineffective. Polyphyllin D also decreased the level of phosphorylated extracellular signal-regulated kinase (p-ERK), a proliferation marker in Jurkat cells. Furthermore, knockdown of protein tyrosine phosphatase (PTP)N6 (SHP1) or PTPN11 (SHP2) decreased p-ERK levels. However, concurrent knockdown of PTPN6 and PTPN11 in Jurkat cells recovered p-ERK levels. These results demonstrated that polyphyllin D has potential anticancer activity, which can be attributed to its selective inhibition of SHP2 over SHP1.


Subject(s)
Antineoplastic Agents/pharmacology , Diosgenin/analogs & derivatives , Enzyme Inhibitors/pharmacology , Protein Tyrosine Phosphatase, Non-Receptor Type 11/antagonists & inhibitors , Saponins/pharmacology , Cell Proliferation/drug effects , Diosgenin/pharmacology , Humans , Jurkat Cells
9.
Molecules ; 26(6)2021 Mar 14.
Article in English | MEDLINE | ID: mdl-33799458

ABSTRACT

Inhibition of the megakaryocyte protein tyrosine phosphatase 2 (PTP-MEG2, also named PTPN9) activity has been shown to be a potential therapeutic strategy for the treatment of type 2 diabetes. Previously, we reported that PTP-MEG2 knockdown enhances adenosine monophosphate activated protein kinase (AMPK) phosphorylation, suggesting that PTP-MEG2 may be a potential antidiabetic target. In this study, we found that phloridzin, isolated from Ulmus davidiana var. japonica, inhibits the catalytic activity of PTP-MEG2 (half-inhibitory concentration, IC50 = 32 ± 1.06 µM) in vitro, indicating that it could be a potential antidiabetic drug candidate. Importantly, phloridzin stimulated glucose uptake by differentiated 3T3-L1 adipocytes and C2C12 muscle cells compared to that by the control cells. Moreover, phloridzin led to the enhanced phosphorylation of AMPK and Akt relevant to increased insulin sensitivity. Importantly, phloridzin attenuated palmitate-induced insulin resistance in C2C12 muscle cells. We also found that phloridzin did not accelerate adipocyte differentiation, suggesting that phloridzin improves insulin sensitivity without significant lipid accumulation. Taken together, our results demonstrate that phloridzin, an inhibitor of PTP-MEG2, stimulates glucose uptake through the activation of both AMPK and Akt signaling pathways. These results strongly suggest that phloridzin could be used as a potential therapeutic candidate for the treatment of type 2 diabetes.


Subject(s)
Insulin Resistance/physiology , Phlorhizin/pharmacology , Protein Tyrosine Phosphatases, Non-Receptor/antagonists & inhibitors , Protein Tyrosine Phosphatases/antagonists & inhibitors , 3T3 Cells , Adipocytes/drug effects , Adipocytes/metabolism , Animals , Cell Differentiation/drug effects , Cell Line , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/metabolism , Glucose/metabolism , Hypoglycemic Agents/pharmacology , Insulin/metabolism , Mice , Palmitates/pharmacology , Phosphorylation/drug effects , Signal Transduction/drug effects
10.
FASEB J ; 33(11): 12288-12298, 2019 11.
Article in English | MEDLINE | ID: mdl-31431058

ABSTRACT

The major side effect of tacrolimus (Tac) is nephrotoxicity. We studied whether supplementation of coenzyme Q10, (CoQ10) a potent antioxidant, can reduce Tac-induced nephrotoxicity via improving mitochondrial function. In an in vitro study, CoQ10 reduced the production of Tac-induced mitochondrial reactive oxygen species and abolished the loss of mitochondrial membrane potential in proximal tubular cell line. Assessment of mitochondrial function revealed that CoQ10 decreased oxygen consumption and mitochondrial respiration rate increased by Tac, suggesting improvement of mitochondrial function to synthesize ATP with CoQ10 treatment. The effect of the CoQ10in vitro study was observed in an experimental model of chronic Tac-induced nephropathy. CoQ10 attenuated Tac-induced oxidative stress and was accompanied by function and histologic improvement. On electron microscopy, addition of CoQ10 increased not only the number but also the volume of mitochondria compared with Tac treatment only. Our data indicate that CoQ10 improves Tac-induced mitochondrial dysfunction in kidney. Supplementary CoQ10 treatment may be a promising approach to reduce Tac-induced nephrotoxicity.-Yu, J. H., Lim, S. W., Luo, K., Cui, S., Quan, Y., Shin, Y. J., Lee, K. E., Kim, H. L., Ko, E. J., Chung, B. H., Kim, J. H., Chung, S. J., Yang, C. W. Coenzyme Q10 alleviates tacrolimus-induced mitochondrial dysfunction in kidney.


Subject(s)
Kidney/drug effects , Mitochondria/drug effects , Tacrolimus/toxicity , Ubiquinone/analogs & derivatives , Apoptosis/drug effects , Cells, Cultured , Humans , Kidney/metabolism , Kidney Tubules, Proximal/drug effects , Membrane Potential, Mitochondrial/drug effects , Mitochondria/physiology , Reactive Oxygen Species/metabolism , Ubiquinone/pharmacology
11.
Chem Biodivers ; 17(2): e1900414, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31797547

ABSTRACT

Protein tyrosine phosphatases are involved in diverse human diseases, including cancer, diabetes and inflammatory disorders. Loss of Vaccinia-H1 related phosphatase (VHR) has been shown to arrest at the G1-S and G2-M transitions of the cell cycle, and to increases cell death of prostate cancer cells through JNK activation, suggesting that VHR can be considered as an anticancer target. In this study, 658 natural products were screened through in vitro enzyme assay to identify VHR inhibitor. Among the VHR-inhibitory compounds, 1,2,3,4,6-O-pentagalloylglucose (PGG) was selected for further study as it has been reported to show antitumor effects against tumor model mice, but its direct target has not been identified. PGG inhibited the catalytic activity of VHR (Ki =53 nm) in vitro. Furthermore, the incubation of HeLa cervical cancer cells with PGG dramatically decreased cell viability and markedly increased the protein levels of the cleaved PARP, a hallmark of apoptosis. In addition, treatment of HeLa cells with PGG significantly reduced the protein levels of cyclin D1, Bcl-2 and STAT3 phosphorylation. Taken together, these results suggest that PGG could be a potential therapeutic candidate for the treatment of cervical cancer through VHR inhibition.


Subject(s)
Antineoplastic Agents/chemistry , Dual Specificity Phosphatase 3/antagonists & inhibitors , Hydrolyzable Tannins/chemistry , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cyclin D1/genetics , Cyclin D1/metabolism , Down-Regulation/drug effects , Dual Specificity Phosphatase 3/genetics , Dual Specificity Phosphatase 3/metabolism , HeLa Cells , Humans , Hydrolyzable Tannins/metabolism , Hydrolyzable Tannins/pharmacology , Kinetics , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism
12.
Molecules ; 25(15)2020 Jul 23.
Article in English | MEDLINE | ID: mdl-32717976

ABSTRACT

Hydrogen peroxide is a low-reactivity reactive oxygen species (ROS); however, it can easily penetrate cell membranes and produce highly reactive hydroxyl radical species through Fenton's reaction. Its presence in abnormal amounts can lead to serious diseases in humans. Although the development of a simple, ultrasensitive, and selective method for H2O2 detection is crucial, this remains a strategic challenge. The peroxidase mimetic activity of palladium nanoclusters (PdNCs) has not previously been evaluated. In this study, we developed an ultrasensitive and selective colorimetric detection method for H2O2 using PdNCs. An unprecedented eco-friendly, cost-effective, and facile biological method was developed for the synthesis of PdNCs. This is the first report of the biosynthesis of PdNCs. The synthesized nanoclusters had a significantly narrow size distribution profile and high stability. The nanoclusters were demonstrated to possess a peroxidase mimetic activity that could oxidize peroxidase substrate 3,3',5,5'-tetramethylbenzidine (TMB). Various interfering substances in serum (100 µM phenylalanine, cysteine, tryptophan, arginine, glucose, urea, Na+, Fe2+, PO43-, Mn+2, Ca2+, Mg2+, Zn2+, NH4+, and K+) were included to evaluate the selectivity of the assay, and oxidation of TMB occurred only in the presence of H2O2. Therefore, PdNCs show an efficient nanozyme for the peroxidase mimetic activity. The assay produced a sufficient signal at the ultralow concentration of 0.0625 µM H2O2. This colorimetric assay provides a real-time, rapid, and easy-to-use platform for the detection of H2O2 for clinical purposes.


Subject(s)
Erigeron/growth & development , Palladium/pharmacology , Reactive Oxygen Species/analysis , Benzidines/chemistry , Calorimetry , Erigeron/metabolism , Hydrogen Peroxide/analysis , Molecular Mimicry , Nanocomposites , Palladium/chemistry , Photosynthesis
13.
Molecules ; 25(11)2020 Jun 02.
Article in English | MEDLINE | ID: mdl-32498444

ABSTRACT

Recent developments in the area of nanotechnology have focused on the development of nanomaterials with catalytic activities. The enzyme mimics, nanozymes, work efficiently in extreme pH and temperature conditions, and exhibit resistance to protease digestion, in contrast to enzymes. We developed an environment-friendly, cost-effective, and facile biological method for the synthesis of ZnO-Pd nanosheets. This is the first biosynthesis of ZnO-Pd nanosheets. The synthesized nanosheets were characterized by UV-visible spectroscopy, X-ray diffraction (XRD), scanning electron microscopy, transmission electron microscopy, and energy-dispersive X-ray. The d-spacing (inter-atomic spacing) of the palladium nanoparticles in the ZnO sheets was found to be 0.22 nm, which corresponds to the (111) plane. The XRD pattern revealed that the 2θ values of 21.8°, 33.3°, 47.7°, and 56.2° corresponded with the crystal planes of (100), (002), (112), and (201), respectively. The nanosheets were validated to possess peroxidase mimetic activity, which oxidized the 3,3',5,5'-tetramethylbenzidine (TMB) substrate in the presence of H2O2. After 20 min of incubation time, the colorless TMB substrate oxidized into a dark-blue-colored one and a strong peak was observed at 650 nm. The initial velocities of Pd-ZnO-catalyzed TMB oxidation by H2O2 were analyzed by Michaelis-Menten and Lineweaver-Burk plots, resulting in 64 × 10-6 M, 8.72 × 10-9 Msec-1, and 8.72 × 10-4 sec-1 of KM, Vmax, and kcat, respectively.


Subject(s)
Biomimetic Materials/chemical synthesis , Palladium/chemistry , Zinc Oxide/chemistry , Biomimetic Materials/chemistry , Green Chemistry Technology , Metal Nanoparticles/chemistry , Microscopy, Electron, Transmission , Particle Size , Peroxidase/metabolism , X-Ray Diffraction
14.
Pharmacol Res ; 139: 325-336, 2019 01.
Article in English | MEDLINE | ID: mdl-30472463

ABSTRACT

Adipogenesis involved in hypertrophy and hyperplasia of adipocytes is responsible for expanding the mass of adipose tissues in obese individuals. Peroxisome proliferator-activated receptor γ (PPARγ) and CCAAT/enhancer-binding protein α (C/EBPα) are two principal transcription factors induced by delicate signaling pathways, including signal transducer and activator of transcription 5 (STAT5), in adipogenesis. Here, we demonstrated a novel role of ginkgetin, a biflavone from Ginkgo biloba leaves, as a STAT5 inhibitor that blocks the differentiation of preadipocytes into adipocytes. During the differentiation of 3T3-L1 cells, ginkgetin treatment during the first 2 days markedly inhibited the formation of lipid-bearing adipocytes. PPARγ and C/EBPα expression was decreased in 3T3-L1 cells during adipogenesis following ginkgetin treatment, whereas no change was observed in C/EBPß or C/EBPδ expression. Inhibition of PPARγ and C/EBPα expression by ginkgetin occurred through the prevention of STAT5 activation during the initiation phase of adipogenesis. In addition, ginkgetin-mediated the inhibition of adipogenesis was recapitulated in the differentiation of primary preadipocytes. Lastly, we confirmed the inhibitory effects of ginkgetin on the hypertrophy of white adipose tissues from high-fat diet-fed mice. These results indicate that ginkgetin is a potential anti-adipogenesis and anti-obesity drug.


Subject(s)
Adipogenesis/drug effects , Anti-Obesity Agents/pharmacology , Anti-Obesity Agents/therapeutic use , Biflavonoids/pharmacology , Biflavonoids/therapeutic use , 3T3-L1 Cells , Animals , CCAAT-Enhancer-Binding Protein-alpha/genetics , CCAAT-Enhancer-Binding Protein-alpha/metabolism , Diet, High-Fat , Ginkgo biloba , Male , Mice , Mice, Inbred C57BL , PPAR gamma/genetics , PPAR gamma/metabolism , Plant Leaves , Signal Transduction/drug effects
15.
Bioorg Chem ; 90: 103087, 2019 09.
Article in English | MEDLINE | ID: mdl-31284101

ABSTRACT

Natural products as antidiabetic agents have been shown to stimulate insulin signaling via the inhibition of the protein tyrosine phosphatases relevant to insulin resistance. Previously, we have identified PTPN9 and DUSP9 as potential antidiabetic targets and a multi-targeting natural product thereof. In this study, knockdown of PTPN11 increased AMPK phosphorylation in differentiated C2C12 muscle cells by 3.8 fold, indicating that PTPN11 could be an antidiabetic target. Screening of a library of 658 natural products against PTPN9, DUSP9, or PTPN11 identified chebulinic acid (CA) as a strong allosteric inhibitor with a slow cooperative binding to PTPN9 (IC50 = 34 nM) and PTPN11 (IC50 = 37 nM), suggesting that it would be a potential antidiabetic candidate. Furthermore, CA stimulated glucose uptake and resulted in increased AMP-activated protein kinase (AMPK) phosphorylation. Taken together, we demonstrated that CA increased glucose uptake as a dual inhibitor of PTPN9 and PTPN11 through activation of the AMPK signaling pathway. These results strongly suggest that CA could be used as a potential therapeutic candidate for the treatment of type 2 diabetes.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Hydrolyzable Tannins/pharmacology , Hypoglycemic Agents/pharmacology , Protein Tyrosine Phosphatase, Non-Receptor Type 11/antagonists & inhibitors , Protein Tyrosine Phosphatases, Non-Receptor/antagonists & inhibitors , Glucose/metabolism , Humans , Insulin/metabolism , Insulin Resistance , Phosphorylation , Signal Transduction
16.
Bioorg Chem ; 91: 103145, 2019 10.
Article in English | MEDLINE | ID: mdl-31357073

ABSTRACT

The root bark of Ulmus davidiana var. japonica (Ulmaceae), commonly known as yugeunpi, has been used as a traditional Korean medicine for the treatment of gastroenteric and inflammatory disorders. As part of continuing projects to discover bioactive natural products from traditional medicinal plants with pharmacological potential, phytochemical investigation of the root bark of this plant was carried out. This led to the successful isolation of a new chromane derivative (1) and 22 known compounds: catechin derivatives (2-5), megastigmane glycoside (6), dihydrochalcone glycosides (7 and 8), flavanone glycosides (9 and 10), coumarins (11 and 12), lignan derivatives (13-17), and phenolic compounds (18-23). The structure of the new compound (1) was determined with 1D and 2D NMR spectroscopy and HR-ESIMS, and its absolute configurations were achieved by chemical reactions and the gauge-including atomic orbital (GIAO) NMR chemical shifts calculations. All the isolated compounds were evaluated for their potential biological activities including neuro-protective, anti-neuroinflammatory, and anti-Helicobacter pylori activities. Among the isolates, compounds 1, 8, and 20 displayed stronger potency by causing a greater increase in the production and the activity of nerve growth factor (NGF) in C6 glioma cells (147.04 ±â€¯4.87, 206.27 ±â€¯6.70, and 143.70 ±â€¯0.88%, respectively), whereas compounds 11, 14, and 19 inhibited nitric oxide (NO) production in lipopolysaccharide (LPS)-stimulated murine microglial cells (IC50 of 18.72, 12.31, and, 21.40 µM, respectively). In addition, compounds 1, 11, 18, and 20 showed anti-H. pylori activity with MIC values of 25 or 50 µM against two strains of H. pylori 51 and 43504. These findings provide scientific evidence that supports the traditional usage of U. davidiana var. japonica root bark in the treatment of gastroenteric and inflammatory disorders.


Subject(s)
Anti-Bacterial Agents/pharmacology , Anti-Inflammatory Agents/pharmacology , Neuroprotective Agents/pharmacology , Plant Bark/chemistry , Plant Extracts/pharmacology , Ulmus/chemistry , Animals , Cells, Cultured , Glioma/drug therapy , Glioma/metabolism , Glioma/pathology , Helicobacter Infections/drug therapy , Helicobacter Infections/microbiology , Helicobacter pylori/drug effects , Inflammation/drug therapy , Inflammation/metabolism , Inflammation/pathology , Mice , Microglia/drug effects , Microglia/pathology , Nerve Growth Factor/metabolism , Nitric Oxide/metabolism , Plant Roots/chemistry , Rats
17.
Chembiochem ; 19(23): 2495-2501, 2018 12 04.
Article in English | MEDLINE | ID: mdl-30238680

ABSTRACT

A highly selective detection method of native protein tyrosine phosphatase 1B (PTP1B) is described using a target specific probe equipped with 1-naphthylamine (λex =330 nm, λem =445 nm). Irradiation of a mixture of PTP1B and Probe 1 with ultraviolet light of 280 nm (corresponding to PTP1B excitation maximum) resulted in significant fluorescence increase at 445 nm, following FRET characteristics. This phenomenon does not occur with other closely related phosphatases or cellular abundant alkaline phosphatase (APP). Probe 1, the most potent and selective probe, was found to competitively inhibit PTP1B (Ki ≈42 nm), whereas APP inhibition was found to be in the low micromolar range. Furthermore, Probe 1 discriminates between PTP1B and several other phosphatases. Here, we report real-time label-free FRET detection of pure PTP1B as well as induced human PTP1B in Escherichia coli cell lysate. In contrast to 6,8-difluoro-4-methylumbelliferyl phosphate (DiFMUP), a representative fluorescence turn-on PTP substrate, our FRET probe successfully differentiated human cervical carcinoma cell lysate, SiHa, which has a high expression level of PTP1B, from PTP1B-knockdown SiHa cell lysate (that is, siRNA was used for PTP1B knockdown).


Subject(s)
1-Naphthylamine/analogs & derivatives , Fluorescent Dyes/chemistry , Protein Tyrosine Phosphatase, Non-Receptor Type 1/analysis , 1-Naphthylamine/chemical synthesis , 1-Naphthylamine/toxicity , Animals , Cattle , Cell Line, Tumor , Enzyme Assays/methods , Fluorescence , Fluorescence Resonance Energy Transfer/methods , Fluorescent Dyes/chemical synthesis , Fluorescent Dyes/toxicity , Humans , Oligopeptides/chemical synthesis , Oligopeptides/chemistry , Oligopeptides/toxicity
18.
Bioorg Chem ; 81: 264-269, 2018 12.
Article in English | MEDLINE | ID: mdl-30153591

ABSTRACT

Several protein tyrosine phosphatases (PTPs) that disrupt the insulin-signaling pathway were investigated by siRNAs to identify potential antidiabetic targets. Individual knockdown of PTPN9 and DUSP9 in 3T3-L1 preadipocytes increased AMPK phosphorylation, respectively, and furthermore, concurrent knockdown of both PTPN9 and DUSP9 synergistically increased AMPK phosphorylation. Next, 658 natural products were screened to identify dual inhibitors of both PTPN9 and DUSP9. Based on the selectivity and inhibition potency of the compounds, ginkgolic acid (GA) was selected for further study as a potential antidiabetic drug candidate. GA inhibited the enzymatic activity of PTPN9 (Ki = 53 µM) and DUSP9 (Ki = 2.5 µM) in vitro and resulted in a significant increase of glucose-uptake in differentiated C2C12 muscle cells and 3T3-L1 adipocytes. In addition, GA increased phosphorylation of AMPK in 3T3L1 adipocytes. In this study, GA as a dual targeting inhibitor of PTPN9 and DUSP9 increased glucose uptake in 3T3L1 and C2C12 cells by activating the AMPK signaling pathway. These results strongly suggest GA could be used as a therapeutic candidate for type 2 diabetes.


Subject(s)
Dual-Specificity Phosphatases/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Hypoglycemic Agents/pharmacology , Mitogen-Activated Protein Kinase Phosphatases/antagonists & inhibitors , Protein Tyrosine Phosphatases, Non-Receptor/antagonists & inhibitors , Salicylates/pharmacology , AMP-Activated Protein Kinases/metabolism , Animals , Cell Line , Dual-Specificity Phosphatases/genetics , Gene Knockdown Techniques , Glucose/metabolism , Mice , Mitogen-Activated Protein Kinase Phosphatases/genetics , Phosphorylation/drug effects , Phosphorylation/genetics , Protein Tyrosine Phosphatases, Non-Receptor/genetics
19.
Pharmacol Res ; 119: 422-430, 2017 05.
Article in English | MEDLINE | ID: mdl-28274853

ABSTRACT

Phospho-cofilin (p-cofilin), which has a phosphate group on Ser-3, is involved in actin polymerization. Its dephosphorylated form promotes filopodia formation and cell migration by enhancing actin depolymerization. Protein phosphatase slingshot homologs (SSHs), known as dual-specificity phosphatases, catalyze hydrolytic removal of the Ser-3 phosphate group from phospho-cofilin. Aberrant SSH activity results in cancer metastasis, implicating SSHs as potential therapeutic targets for cancer metastasis. In this study, we screened 658 natural products purified from traditional oriental medicinal plants to identify three potent SSH inhibitors with submicromolar or single-digit micromolar Ki values: gossypol, hypericin, and sennoside A. The three compounds were purified from cottonseed, Saint John's wort, and rhubarb, respectively. Sennoside A markedly increased cofilin phosphorylation in pancreatic cancer cells, leading to impaired actin dynamics in pancreatic cancer cells with or without EGF stimulation and reduced motility and invasiveness in vitro and in vivo. Collaboratively, these results demonstrate that sennoside A is a novel inhibitor of SSHs and suggest that it may be valuable in the development of pharmaceutical drugs for treating cancer metastasis.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Movement/drug effects , Neoplasm Invasiveness/prevention & control , Pancreatic Neoplasms/drug therapy , Phosphoprotein Phosphatases/antagonists & inhibitors , Phosphoprotein Phosphatases/metabolism , Senna Extract/pharmacology , Actin Depolymerizing Factors/metabolism , Actins/metabolism , Cell Line, Tumor , Human Umbilical Vein Endothelial Cells , Humans , Neoplasm Invasiveness/pathology , Pancreas/drug effects , Pancreas/metabolism , Pancreas/pathology , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Phosphorylation/drug effects , Sennosides
20.
Anal Chem ; 88(14): 7178-82, 2016 07 19.
Article in English | MEDLINE | ID: mdl-27367584

ABSTRACT

We report a chromogenic and fluorescence turn-on probe based on crotonoyl ester-functionalized oxazolidinoindole for the selective detection of cysteine in neutral buffer. The probe rapidly formed indocyanophenolate through the Michael addition and a subsequent cyclization reaction of cysteine, inducing both a dramatic bathochromic shift (>130 nm) and a large fluorescence turn-on response (F/F0 12) in the UV-vis and fluorescence spectra and affording a micromolar limit of detection (LOD = 5.0 µM) of cysteine in HEPES buffer. When cysteine was added, the probe exhibited a dual optical change with strong green fluorescence and dramatic red color by the oxazolidinoindole-to-hydroxyethylindolium transformation. Further cellular application of the probe was successfully performed for the mitochondrial imaging of HeLa cells.


Subject(s)
Cysteine/analysis , Fluorescent Dyes/chemistry , Indoles/chemistry , Mitochondria/metabolism , Oxazoles/chemistry , Crotonates/chemistry , Fluorescence , Fluorescent Dyes/chemical synthesis , HeLa Cells , Humans , Indoles/chemical synthesis , Limit of Detection , Microscopy, Confocal/methods , Microscopy, Fluorescence/methods , Models, Chemical , Oxazoles/chemical synthesis
SELECTION OF CITATIONS
SEARCH DETAIL