Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
Pharmaceutics ; 16(6)2024 Jun 13.
Article in English | MEDLINE | ID: mdl-38931920

ABSTRACT

Zastaprazan (JP-1366), a novel potassium-competitive acid blocker, is a new drug for the treatment of erosive esophagitis. JP-1366 is highly metabolized in human, mouse, and dog hepatocytes but moderately metabolized in rat and monkey hepatocytes when estimated from the metabolic stability of this compound in hepatocyte suspension and when 18 phase I metabolites and 5 phase II metabolites [i.e., N-dearylation (M6), hydroxylation (M1, M19, M21), dihydroxylation (M7, M8, M14, M22), trihydroxylation (M13, M18), hydroxylation and reduction (M20), dihydroxylation and reduction (M9, M16), hydrolysis (M23), hydroxylation and glucuronidation (M11, M15), hydroxylation and sulfation (M17), dihydroxylation and sulfation (M10, M12), N-dearylation and hydroxylation (M3, M4), N-dearylation and dihydroxylation (M5), and N-dearylation and trihydroxylation (M2)] were identified from JP-1366 incubation with the hepatocytes from humans, mice, rats, dogs, and monkeys. Based on the cytochrome P450 (CYP) screening test and immune-inhibition analysis with CYP antibodies, CYP3A4 and CYP3A5 played major roles in the metabolism of JP-1366 to M1, M3, M4, M6, M8, M9, M13, M14, M16, M18, M19, M21, and M22. CYP1A2, 2C8, 2C9, 2C19, and 2D6 played minor roles in the metabolism of JP-1366. UDP-glucuronosyltransferase (UGT) 2B7 and UGT2B17 were responsible for the glucuronidation of M1 to M15. However, JP-1366 and active metabolite M1 were not substrates for drug transporters such as organic cation transporter (OCT) 1/2, organic anion transporter (OAT) 1/3, organic anion transporting polypeptide (OATP)1B1/1B3, multidrug and toxic compound extrusion (MATE)1/2K, P-glycoprotein (P-gp), and breast cancer-resistant protein (BCRP). Only M1 showed substrate specificity for P-gp. The findings indicated that drug-metabolizing enzymes, particularly CYP3A4/3A5, may have a significant role in determining the pharmacokinetics of zastaprazan while drug transporters may only have a small impact on the absorption, distribution, and excretion of this compound.

2.
Pharmacol Res Perspect ; 11(3): e01090, 2023 06.
Article in English | MEDLINE | ID: mdl-37147903

ABSTRACT

The global prevalence of GERD is substantially increasing each year, and GERD is a chronic disease that reduces the quality of life of patients. The efficacy of conventional drugs is diverse, and most require long-term or lifetime administration; thus, the development of more effective therapeutic agents is needed. Herein, a more effective treatment for GERD was tested. We investigated whether JP-1366 affected gastric H+/K+-ATPase activity and used the Na+/K+-ATPase assay to confirm the selectivity of H+/K+-ATPase inhibition. To clarify the mechanism of enzyme inhibition, JP-1366 and TAK-438 were analyzed by Lineweaver-Burk. Also, we investigated the effects of JP-1366 in various models involving reflux esophagitis. We found that JP-1366 mediates strong, selective, and dose-dependent inhibition of H+/K+-ATPase. We found that JP-1366 significantly suppressed gastric acid secretion in histamine-treated pylorus-ligated rats in a dose-dependent manner. Additionally, we confirmed that JP-1366 inhibited histamine-stimulated gastric acid secretion in the HPD model. JP-1366 exhibited a more than 2-fold higher inhibitory effect on esophageal injury than TAK-438 in GERD lesions and had a more potent inhibitory effect in indomethacin- or aspirin-induced gastric ulcer rat models than TAK-438. Additionally, JP-1366 inhibited gastric ulcers. These results support the possibility that JP-1366 is a good candidate drug for treating acid-related diseases.


Subject(s)
Gastroesophageal Reflux , Proton Pump Inhibitors , Rats , Animals , Proton Pump Inhibitors/pharmacology , Proton Pump Inhibitors/therapeutic use , Histamine , Potassium/therapeutic use , Quality of Life , Gastric Acid , Gastroesophageal Reflux/drug therapy , Adenosine Triphosphatases
3.
Aliment Pharmacol Ther ; 57(7): 763-772, 2023 04.
Article in English | MEDLINE | ID: mdl-36732884

ABSTRACT

BACKGROUND: Zastaprazan (JP-1366) is a novel potassium-competitive acid blocker with favourable preclinical safety and efficacy profile being developed for the treatment of acid-related diseases. AIMS: To investigate the safety, tolerability, pharmacodynamics and pharmacokinetics of zastaprazan. METHODS: A randomised, open-label, placebo- and active-controlled, single and multiple ascending dose clinical trial was conducted in healthy Korean male subjects. Intragatric pH and serum gastrin were measured to assess the pharmacodynamics, while serial blood and urine samples were collected to assess the pharmacokinetics. Pharmacogenomic evaluation was conducted to explore genetic variants, which can affect the pharmacodynamics and pharmacokinetics. Safety and tolerability including hepatotoxicity were evaluated. RESULTS: Suppression of gastric acid secretion increased as the dose of zastaprazan increased. The percentage of time that gastric pH was over 4 (%Time pH >4) with zastaprazan 20 mg (85.19%) and 40 mg (91.84%) were similar to or greater than that with esomeprazole 40 mg (72.06%). Zastaprazan was rapidly absorbed within 2 h and eliminated with a half-life of 6-10 h. Pharmacogenomic analysis found no genetic variant of drug metabolising enzymes including CYP2C19 or drug transporters associated with the exposure of zastaprazan. Zastaprazan was well tolerated with no clinically significant changes in safety and tolerability assessments. CONCLUSIONS: Zastaprazan was safe and well tolerated after a single oral dose up to 60 mg and multiple oral doses up to 40 mg. It also showed rapid, potent suppression of gastric acid secretion. Pharmacodynamic and pharmacokinetic profile of zastaprazan was suitable for treatment of patients with acid-related diseases.


Subject(s)
Esomeprazole , Potassium , Humans , Male , Healthy Volunteers , Double-Blind Method , Gastrins , Dose-Response Relationship, Drug , Administration, Oral
4.
J Cancer ; 11(14): 4059-4072, 2020.
Article in English | MEDLINE | ID: mdl-32368288

ABSTRACT

Histone deacetylase inhibitors (HDACis) are well-known epigenetic regulators with therapeutic potential in various diseases. Recent studies have shown that HDACis are involved in immune-mediated anti-cancer effects and may modulate the activity of immunotherapy agents. CG-745, a histone deacetylase inhibitor, has shown anti-cancer effects in pancreatic cancer, colorectal cancer, and non-small cell lung cancer. However, the exact role of CG-745 within the immune system is largely unknown. In this study, we have shown that CG-745 induces microenvironment changes promoting anti-cancer effect of anti-PD-1 antibody in syngeneic mouse models. Specifically, CG-745 induces or extends IL-2 and IFN-γ expression with or without additional stimulation, and increases proliferation of cytotoxic T cells and NK cells, while inhibiting proliferation of regulatory T cells. The analysis of immune cell distribution in the tumor microenvironment and spleen reveals that CG-745 suppresses M2 macrophage polarization and decreases the myeloid-derived suppressor cells. Recent advances in immunotherapy highlight the anti-cancer effects of immune checkpoint inhibitor despite a relatively limited clinical benefit in the subset of patients. Our results indicate that CG-745 enables the synergistic effects of the immune checkpoint inhibitor combination therapy in various cancers by suppressing tumor microenvironment.

5.
Int J Mol Sci ; 21(4)2020 Feb 21.
Article in English | MEDLINE | ID: mdl-32098220

ABSTRACT

Histone deacetylases have been a target of therapy for organ fibrosis. Here, we report the protective effect of CG200745 (CG), a novel histone deacetylase inhibitor, on tubulointerstitial fibrosis in Col4a3-/- mice, a murine model of Alport syndrome. Morphological analyses revealed CG treatment markedly alleviated kidney fibrosis in Col4a3-/- mice at the age of 7 weeks. CG prevented the activation of transforming growth factor ß (TGFß) and its downstream SMAD signaling in the kidney of Col4a3-/- mice. As critical upstream regulators of TGFß signaling, immunoblotting of whole kidney lysate of Col4a3-/- mice reveled that intra-renal renin-angiotensin system (RAS) was activated with concurrent upregulation of inflammation and apoptosis, which were effectively suppressed by CG treatment. CG suppressed both activation of RAS and up-regulation of TGFß signals in angiotensin II-stimulated HK-2 cells, a human kidney proximal tubular epithelial cell line. CG inhibited activation of TGFß-driven signals and fibrosis in NRK-49F cells, a rat kidney fibroblast cell line, under angiotensin II-rich conditions. Collectively, CG was found to be effective both in proximal tubular epithelial cells by inhibiting local RAS and TGFß signaling activation, as well as in fibroblasts by blocking their transition to myofibroblasts, attenuating renal fibrosis in a murine model of Alport syndrome.


Subject(s)
Histone Deacetylase Inhibitors/pharmacology , Hydroxamic Acids/pharmacology , Kidney Tubules, Proximal/metabolism , Naphthalenes/pharmacology , Nephritis, Hereditary , Signal Transduction , Animals , Autoantigens/metabolism , Cell Line , Collagen Type IV/deficiency , Collagen Type IV/metabolism , Disease Models, Animal , Fibrosis , Humans , Kidney Tubules, Proximal/pathology , Male , Mice , Mice, Knockout , Nephritis, Hereditary/drug therapy , Nephritis, Hereditary/genetics , Nephritis, Hereditary/metabolism , Nephritis, Hereditary/pathology , Rats , Transforming Growth Factor beta/metabolism
6.
Molecules ; 24(15)2019 Jul 31.
Article in English | MEDLINE | ID: mdl-31370295

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is a fatal lung disease with poor prognosis and progression to lung fibrosis related to genetic factors as well as environmental factors. In fact, it was discovered that in South Korea many people who used humidifier disinfectants containing polyhexamethylene guanidine (PHMG), died of lung fibrosis. Currently two anti-fibrotic drugs, pirfenidone and nintedanib, have been approved by the FDA, but unfortunately, do not cure the disease. Since the histone deacetylase (HDAC) activity is associated with progression to chronic diseases and with fibrotic phenomena in the kidney, heart and lung tissues, we investigated the anti-fibrotic effects of CG-745, an HDAC inhibitor. After lung fibrosis was induced in two animal models by bleomycin and PHMG instillation, the regulation of fibrosis and epithelial mesenchymal transition (EMT)-related markers was assessed. CG-745 exhibited potent prevention of collagen production, inflammatory cell accumulation, and cytokines release in both models. Additionally, N-cadherin and vimentin expression were lowered significantly by the treatment of CG-745. The anti-fibrotic effects of CG-745 proven by the EMT regulation may suggest a potential therapeutic effect of CG-745 on lung fibrosis.


Subject(s)
Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/drug effects , Idiopathic Pulmonary Fibrosis/drug therapy , Lung/drug effects , Animals , Biguanides/toxicity , Bleomycin/toxicity , Disease Models, Animal , Epithelial-Mesenchymal Transition/drug effects , Histone Deacetylase Inhibitors/chemistry , Histone Deacetylases/chemistry , Humans , Idiopathic Pulmonary Fibrosis/chemically induced , Idiopathic Pulmonary Fibrosis/pathology , Indoles/chemistry , Indoles/therapeutic use , Lung/pathology , Mice , Pyridones/chemistry , Pyridones/therapeutic use , Republic of Korea/epidemiology
7.
Int J Mol Sci ; 20(3)2019 Jan 25.
Article in English | MEDLINE | ID: mdl-30691015

ABSTRACT

The novel histone deacetylase inhibitor CG200745 was initially developed to treat various hematological and solid cancers. We investigated the molecular mechanisms associated with the renoprotective effects of CG200745 using deoxycorticosterone acetate (DOCA)-salt hypertensive (DSH) rats. DOCA strips (200 mg/kg) were implanted into rats one week after unilateral nephrectomy. Two weeks after DOCA implantation, DSH rats were randomly divided into two groups that received either physiological saline or CG200745 (5 mg/kg/day) for another two weeks. The extent of glomerulosclerosis and tubulointerstitial fibrosis was determined by Masson's trichrome staining. The renal expression of fibrosis and inflammatory markers was detected by semiquantitative immunoblotting, a polymerase chain reaction, and immunohistochemistry. Pathological signs such as glomerulosclerosis, tubulointerstitial fibrosis, increased systolic blood pressure, decreased creatinine clearance, and increased albumin-to-creatinine ratios in DSH rats were alleviated by CG200745 treatment compared to those manifestations in positive control animals. Furthermore, this treatment counteracted the increased expression of αSMA, TGF-ß1, and Bax, and the decreased expression of Bcl-2 in the kidneys of DSH rats. It also attenuated the increase in the number of apoptotic cells in DSH rats. Thus, CG200745 can effectively prevent the progression of renal injury in DSH rats by exerting anti-inflammatory, anti-fibrotic, and anti-apoptotic effects.


Subject(s)
Desoxycorticosterone Acetate/adverse effects , Histone Deacetylase Inhibitors/administration & dosage , Hydroxamic Acids/administration & dosage , Hypertension/drug therapy , Kidney Diseases/prevention & control , Naphthalenes/administration & dosage , Actins/metabolism , Albumins/metabolism , Animals , Apoptosis/drug effects , Biomarkers/metabolism , Creatinine/metabolism , Disease Models, Animal , Histone Deacetylase Inhibitors/pharmacology , Hydroxamic Acids/pharmacology , Hypertension/chemically induced , Hypertension/complications , Kidney Diseases/metabolism , Male , Naphthalenes/pharmacology , Random Allocation , Rats , Rats, Sprague-Dawley , Transforming Growth Factor beta1/metabolism
8.
FASEB J ; 33(3): 4300-4313, 2019 03.
Article in English | MEDLINE | ID: mdl-30540497

ABSTRACT

SRC-family kinases (SFKs) have been implicated in Alzheimer's disease (AD), but their mode of action was scarcely understood. Here, we show that LYN plays an essential role in amyloid ß (Aß)-triggered neurotoxicity and tau hyperphosphorylation by phosphorylating Fcγ receptor IIb2 (FcγRIIb2). We found that enzyme activity of LYN was increased in the brain of AD patients and was promoted in neuronal cells exposed to Aß 1-42 (Aß1-42). Knockdown of LYN expression inhibited Aß1-42-induced neuronal cell death. Of note, LYN interacted with FcγRIIb2 upon exposure to Aß1-42 and phosphorylated FcγRIIb2 at Tyr273 within immunoreceptor tyrosine-based inhibitory motif in neuronal cells. With the use of the structure-based drug design, we isolated KICG2576, an ATP-competitive inhibitor of LYN. Determination of cocrystal structure illustrated that KICG2576 bound to the cleft in the LYN kinase domain and inhibited LYN with a half-maximal inhibitory concentration value of 0.15 µM. KICG2576 inhibited Aß- or FcγRIIb2-induced cell death, and this effect was better than pyrazolopyrimidine 1, a widely used inhibitor of SFK. Upon exposure to Aß, KICG2576 blocked the phosphorylation of FcγRIIb2 and translocation of phosphatidylinositol 3,4,5-trisphosphate 5-phosphatase 2, a binding protein to the phosphorylated FcγRIIb2, to the plasma membrane, resulting in the inhibition of tau hyperphosphorylation, the downstream event of Aß1-42-FcγRIIb2 binding. Furthermore, intracerebroventricular injection of KICG2576 into mice ameliorated Aß-induced memory impairment. These results suggest that LYN plays a crucial role in Aß1-42-mediated neurotoxicity and tau pathology, providing a therapeutic potential of LYN in AD.-Gwon, Y., Kim, S.-H., Kim, H. T., Kam, T.-I., Park, J., Lim, B., Cha, H., Chang, H.-J., Hong, Y. R., Jung, Y.-K. Amelioration of amyloid ß-FcγRIIb neurotoxicity and tau pathologies by targeting LYN.


Subject(s)
Amyloid beta-Peptides/metabolism , Neurons/metabolism , Receptors, IgG/metabolism , tau Proteins/metabolism , Alzheimer Disease/metabolism , Animals , Cell Line , Cell Membrane/metabolism , Hippocampus/metabolism , Humans , Memory Disorders/metabolism , Mice , Peptide Fragments/metabolism , Phosphatidylinositols/metabolism , Phosphorylation/physiology , Rats , src-Family Kinases/metabolism
9.
Sci Rep ; 8(1): 11546, 2018 08 01.
Article in English | MEDLINE | ID: mdl-30068917

ABSTRACT

Tubulointerstitial fibrosis is a common feature of kidney disease. Histone deacetylase (HDAC) inhibitors have been reported to attenuate renal fibrosis progression. Here, we investigated the effect of CG200745, a novel HDAC inhibitor, on renal fibrosis development in a mouse model of unilateral ureteral obstruction (UUO). To examine the effects of CG200745 on renal fibrosis in UUO, C57BL/6 J male mice were divided into three groups: control, UUO, and CG200745 (30 mg/kg/day)-treated UUO groups. CG 200745 was administered through drinking water for 1 week. Human proximal tubular epithelial (HK-2) cells were also treated with CG200745 (10 µM) with or without TGF-ß (2 ng/mL). Seven days after UUO, plasma creatinine did not differ among the groups. However, plasma neutrophil gelatinase-associated lipocalin (NGAL) levels were markedly increased in the UUO group, which were attenuated by CG200745 treatment. UUO kidneys developed marked fibrosis as indicated by collagen deposition and increased α-smooth muscle actin (SMA) and fibronectin expression. CG200745 treatment attenuated these fibrotic responses and suppressed UUO-induced production of transforming growth factor-beta1 (TGF-ß) and phosphorylation of Smad-2/3. CG200745 treatment also attenuated UUO-induced inflammation as indicated by the expression of inflammatory markers. Furthermore, CG200745 attenuated phosphorylation of p38 mitogen-activated protein kinase in UUO kidneys. In HK-2 cells, TGF-ß induced the expression of α-SMA and fibronectin, which were attenuated by CG200745 cotreatment. These results demonstrate that CG200745, a novel HDAC inhibitor, has a renoprotective effect by suppressing renal fibrosis and inflammation in a UUO mouse model.


Subject(s)
Fibrosis/drug therapy , Histone Deacetylase Inhibitors/administration & dosage , Hydroxamic Acids/administration & dosage , Naphthalenes/administration & dosage , Ureteral Obstruction/complications , Animals , Cell Line , Disease Models, Animal , Epithelial Cells/drug effects , Fibrosis/pathology , Humans , Kidney/pathology , Lipocalins/blood , Mice , Mice, Inbred C57BL , Phosphorylation , Protein Processing, Post-Translational , Smad2 Protein/metabolism , Smad3 Protein/metabolism , Transforming Growth Factor beta/analysis , Treatment Outcome
10.
Cell Chem Biol ; 25(4): 426-438.e4, 2018 04 19.
Article in English | MEDLINE | ID: mdl-29429898

ABSTRACT

Kynurenine 3-monooxygenase (KMO) inhibitors have been developed for the treatment of neurodegenerative disorders. The mechanisms of flavin reduction and hydrogen peroxide production by KMO inhibitors are unknown. Herein, we report the structure of human KMO and crystal structures of Saccharomyces cerevisiae (sc) and Pseudomonas fluorescens (pf) KMO with Ro 61-8048. Proton transfer in the hydrogen bond network triggers flavin reduction in p-hydroxybenzoate hydroxylase, but the mechanism triggering flavin reduction in KMO is different. Conformational changes via π-π interactions between the loop above the flavin and substrate or non-substrate effectors lead to disorder of the C-terminal α helix in scKMO and shifts of domain III in pfKMO, stimulating flavin reduction. Interestingly, Ro 61-8048 has two different binding modes. It acts as a competitive inhibitor in scKMO and as a non-substrate effector in pfKMO. These findings provide understanding of the catalytic cycle of KMO and insight for structure-based drug design of KMO inhibitors.


Subject(s)
Enzyme Inhibitors/pharmacology , Hydrogen Peroxide/metabolism , Kynurenine 3-Monooxygenase/antagonists & inhibitors , Kynurenine 3-Monooxygenase/metabolism , Pseudomonas fluorescens/enzymology , Saccharomyces cerevisiae/enzymology , Sulfonamides/pharmacology , Thiazoles/pharmacology , Amino Acid Sequence , Animals , Flavins/metabolism , Humans , Kynurenine 3-Monooxygenase/chemistry , Molecular Docking Simulation , Oxidation-Reduction/drug effects , Protein Conformation/drug effects , Pseudomonas fluorescens/chemistry , Saccharomyces cerevisiae/chemistry , Sequence Alignment
11.
Sci Rep ; 7: 41615, 2017 01 30.
Article in English | MEDLINE | ID: mdl-28134290

ABSTRACT

Pancreatic cancer is predominantly lethal, and is primarily treated using gemcitabine, with increasing resistance. Therefore, novel agents that increase tumor sensitivity to gemcitabine are needed. Histone deacetylase (HDAC) inhibitors are emerging therapeutic agents, since HDAC plays an important role in cancer initiation and progression. We evaluated the antitumor effect of a novel HDAC inhibitor, CG200745, combined with gemcitabine/erlotinib on pancreatic cancer cells and gemcitabine-resistant pancreatic cancer cells. Three pancreatic cancer-cell lines were used to evaluate the antitumor effect of CG200745 combined with gemcitabine/erlotinib. CG200745 induced the expression of apoptotic proteins (PARP and caspase-3) and increased the levels of acetylated histone H3. CG200745 with gemcitabine/erlotinib showed significant growth inhibition and synergistic antitumor effects in vitro. In vivo, gemcitabine/erlotinib and CG200745 reduced tumor size up to 50%. CG200745 enhanced the sensitivity of gemcitabine-resistant pancreatic cancer cells to gemcitabine, and decreased the level of ATP-binding cassette-transporter genes, especially multidrug resistance protein 3 (MRP3) and MRP4. The novel HDAC inhibitor, CG200745, with gemcitabine/erlotinib had a synergistic anti-tumor effect on pancreatic cancer cells. CG200745 significantly improved pancreatic cancer sensitivity to gemcitabine, with a prominent antitumor effect on gemcitabine-resistant pancreatic cancer cells. Therefore, improved clinical outcome is expected in the future.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Deoxycytidine/analogs & derivatives , Drug Resistance, Neoplasm/drug effects , Histone Deacetylase Inhibitors/pharmacology , Hydroxamic Acids/pharmacology , Naphthalenes/pharmacology , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Deoxycytidine/pharmacology , Disease Models, Animal , Humans , Mice , Pancreatic Neoplasms/metabolism , Xenograft Model Antitumor Assays , Gemcitabine
12.
Biochem Biophys Res Commun ; 478(1): 1-6, 2016 09 09.
Article in English | MEDLINE | ID: mdl-27475498

ABSTRACT

Polmacoxib is not only a selective COX-2 inhibitor but also a potent inhibitor of carbonic anhydrases (CAs). Both CA I and CA II are highly expressed in the GI tract and kidneys, organs that are also thought to be the sites at which selective COX-2 inhibitors show their side effects. By inhibition assays, we show that both CA I and CA II are strongly inhibited by polmacoxib, while CA II also demonstrates direct competition with COX-2. To understand, at the molecular level, how polmacoxib interacts with CA I and II, we solved the first crystal structures of CA I and CA II in complex with polmacoxib, at 2.0 Å and 1.8 Å, respectively. Interestingly, three polmacoxib molecules bind to the active site of CA I, whereas only one molecule binds CA II. In the active site, the three molecules of polmacoxib organize itself along hydrophobic interaction as "stack-on-formation", and fully occupy a cone-shaped active pocket in CA I. The binding mode of polmacoxib to CA II was found different than its binding to celecoxib and valdecoxib. Our results provide structural insight into inhibition of CA I and CA II by polmacoxib, to assess its potential clinical efficacy.


Subject(s)
Carbonic Anhydrase II/antagonists & inhibitors , Carbonic Anhydrase II/chemistry , Carbonic Anhydrase I/antagonists & inhibitors , Carbonic Anhydrase I/chemistry , Carbonic Anhydrase Inhibitors/pharmacology , Cyclooxygenase 2 Inhibitors/pharmacology , Furans/pharmacokinetics , Sulfonamides/pharmacokinetics , Carbonic Anhydrase I/metabolism , Carbonic Anhydrase II/metabolism , Catalytic Domain/drug effects , Crystallography, X-Ray , Humans , Models, Molecular , Protein Conformation/drug effects , Protein Multimerization/drug effects
13.
Invest New Drugs ; 33(5): 1048-57, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26076682

ABSTRACT

PURPOSE: The aim of the present study was to assess the safety, maximum tolerated dose (MTD), pharmacokinetics, pharmacodynamics, and efficacy of single and multiple doses of intravenous CG200745, a novel histone deacetylase (HDAC) inhibitor, in patients with advanced solid malignancies. EXPERIMENTAL DESIGN: Two to six patients received intravenous CG200745 according to the 2 + 4 dose-escalating method. This first-in-human trial was comprised of two parts: Part 1 was a single ascending dose, and Part 2 was multiple ascending doses weekly for 3 weeks, and then 1 week off. For the first cycle, pharmacokinetic sampling for CG200745 and pharmacodynamic sampling for acetylated histone H4 in peripheral blood mononuclear cells (PBMCs) were performed on day 1 for Part 1 and on days 1 and 15 for Part 2. Examination of acetylated histone H4 in pre- and post-biopsy samples was performed in accessible patients. RESULTS: In all, 28 patients were treated at 13 dose levels (1.8-250 mg/m(2)) and received a total of 71 cycles of CG200745. Hematologic toxicities included grade 3/4 neutropenia (22.2 %) that did not last a week and non-hematologic toxicities included fatigue (22.2 %) and anorexia (16.7 %) that did not exceed grade 2. No dose-limiting toxic effects were noted. Dose proportionality was observed for both the maximum concentration and area under the curve. The elimination half-life was 5.67 ± 2.69 h (mean ± standard deviation). An increase in PBMC acetylated histone H4 was observed at dose levels up to 51 mg/m(2), which plateaued at higher dose levels. At 24 h, 75 % of patients (6/8) showed higher relative acetylation in tumor tissue compared to PBMCs. Although there was no partial or complete response, 57.1 % of patients (16/28) had stable disease that lasted at least 6 weeks. CONCLUSIONS: CG200745 can be safely administered at effective dose levels that inhibit HDAC in PBMCs and tumor tissue. Although MTD was not reached, further escalation was not performed because acetylated histone H4 plateaued at dose levels higher than 51 mg/m(2). Additional phase II trials are recommended at 250 mg/m(2).


Subject(s)
Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Hydroxamic Acids/pharmacology , Hydroxamic Acids/therapeutic use , Naphthalenes/pharmacology , Naphthalenes/therapeutic use , Neoplasms/drug therapy , Adolescent , Adult , Aged , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacokinetics , Child , Child, Preschool , Dose-Response Relationship, Drug , Drug Administration Schedule , Female , Half-Life , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylase Inhibitors/therapeutic use , Histones/metabolism , Humans , Hydroxamic Acids/adverse effects , Hydroxamic Acids/pharmacokinetics , Leukocytes, Mononuclear/metabolism , Male , Maximum Tolerated Dose , Middle Aged , Naphthalenes/adverse effects , Naphthalenes/pharmacokinetics , Young Adult
14.
Biochem Biophys Res Commun ; 451(4): 541-7, 2014 Sep 05.
Article in English | MEDLINE | ID: mdl-25117441

ABSTRACT

To understand the role of His and Glu in the catalytic activity of Bacillus licheniformis α-amylase (BLA), His235 was replaced with Glu. The mutant enzyme, H235E, was characterized in terms of its mode of action using labeled and unlabeled maltooctaose (Glc8). H235E predominantly produced maltotridecaose (Glc13) from Glc8, exhibiting high substrate transglycosylation activity, with Km=0.38mM and kcat/Km=20.58mM(-1)s(-1) for hydrolysis, and Km2=18.38mM and kcat2/Km2=2.57mM(-1)s(-1) for transglycosylation, while the wild-type BLA exhibited high hydrolysis activity exclusively. Glu235-located on a wide open groove near subsite +1-is likely involved in transglycosylation via formation of an α-1,4-glycosidic linkage and may recognize and stabilize the non-reducing end glucose of the acceptor molecule.


Subject(s)
alpha-Amylases/genetics , alpha-Amylases/metabolism , Amino Acid Sequence , Apraxia, Ideomotor , Bacillus/enzymology , Binding Sites , Glutamic Acid/metabolism , Glycosylation , Histidine/metabolism , Hydrolysis , Models, Molecular , Oligosaccharides/metabolism
15.
J Biol Chem ; 283(42): 28641-8, 2008 Oct 17.
Article in English | MEDLINE | ID: mdl-18703518

ABSTRACT

TreX is an archaeal glycogen-debranching enzyme that exists in two oligomeric states in solution, as a dimer and tetramer. Unlike its homologs, TreX from Sulfolobus solfataricus shows dual activities for alpha-1,4-transferase and alpha-1,6-glucosidase. To understand this bifunctional mechanism, we determined the crystal structure of TreX in complex with an acarbose ligand. The acarbose intermediate was covalently bound to Asp363, occupying subsites -1 to -3. Although generally similar to the monomeric structure of isoamylase, TreX exhibits two different active-site configurations depending on its oligomeric state. The N terminus of one subunit is located at the active site of the other molecule, resulting in a reshaping of the active site in the tetramer. This is accompanied by a large shift in the "flexible loop" (amino acids 399-416), creating connected holes inside the tetramer. Mutations in the N-terminal region result in a sharp increase in alpha-1,4-transferase activity and a reduced level of alpha-1,6-glucosidase activity. On the basis of geometrical analysis of the active site and mutational study, we suggest that the structural lid (acids 99-97) at the active site generated by the tetramerization is closely associated with the bifunctionality and in particular with the alpha-1,4-transferase activity. These results provide a structural basis for the modulation of activities upon TreX oligomerization that may represent a common mode of action for other glycogen-debranching enzymes in higher organisms.


Subject(s)
Exodeoxyribonucleases/chemistry , Glycogen Debranching Enzyme System/chemistry , Glycogen/chemistry , Phosphoproteins/chemistry , Sulfolobus solfataricus/metabolism , Acarbose/chemistry , Amino Acid Sequence , Aspartic Acid/chemistry , Catalytic Domain , DNA Mutational Analysis , Hydrogen-Ion Concentration , Molecular Conformation , Molecular Sequence Data , Mutation , Protein Conformation , Protein Structure, Tertiary , Sequence Homology, Amino Acid
16.
Biochem Biophys Res Commun ; 366(1): 98-103, 2008 Feb 01.
Article in English | MEDLINE | ID: mdl-18060856

ABSTRACT

Di-O-alpha-maltosyl-beta-cyclodextrin ((G2)(2)-beta-CD) was synthesized from 6-O-alpha-maltosyl-beta-cyclodextrin (G2-beta-CD) via a transglycosylation reaction catalyzed by TreX, a debranching enzyme from Sulfolobus solfataricus P2. TreX showed no activity toward glucosyl-beta-CD, but a transfer product (1) was detected when the enzyme was incubated with maltosyl-beta-CD, indicating specificity for a branched glucosyl chain bigger than DP2. Analysis of the structure of the transfer product (1) using MALDI-TOF/MS and isoamylase or glucoamylase treatment revealed it to be dimaltosyl-beta-CD, suggesting that TreX transferred the maltosyl residue of a G2-beta-CD to another molecule of G2-beta-CD by forming an alpha-1,6-glucosidic linkage. When [(14)C]-maltose and maltosyl-beta-CD were reacted with the enzyme, the radiogram showed no labeled dimaltosyl-beta-CD; no condensation product between the two substrates was detected, indicating that the synthesis of dimaltosyl-beta-CD occurred exclusively via transglycosylation of an alpha-1,6-glucosidic linkage. Based on the HPLC elution profile, the transfer product (1) was identified to be isomers of 6(1),6(3)- and 6(1),6(4)-dimaltosyl-beta-CD. Inhibition studies with beta-CD on the transglycosylation activity revealed that beta-CD was a mixed-type inhibitor, with a K(i) value of 55.6 micromol/mL. Thus, dimaltosyl-beta-CD can be more efficiently synthesized by a transglycosylation reaction with TreX in the absence of beta-CD. Our findings suggest that the high yield of (G2)(2)-beta-CD from G2-beta-CD was based on both the transglycosylation action mode and elimination of the inhibitory effect of beta-CD.


Subject(s)
Cyclodextrins/chemical synthesis , Glucosyltransferases/chemistry , Maltose/chemistry , Catalysis , Enzyme Activation , Enzyme Stability , Glycosylation , Isomerism
17.
Biosci Biotechnol Biochem ; 71(6): 1564-7, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17587692

ABSTRACT

To elucidate the relationship between the substrate size and geometric shape of the catalytic site of Thermus maltogenic amylase, Gly50, Asp109, and Val431, located at the interface of the dimer, were replaced with bulky amino acids. The k(cat)/K(m) value of the mutant for amylose increased significantly, whereas that for amylopectin decreased as compared to that of the wild-type enzyme. Thus, the substituted bulky amino acid residues modified the shape of the catalytic site, such that the ability of the enzyme to distinguish between small and large molecules like amylose and amylopectin was enhanced.


Subject(s)
Amylases/metabolism , Mutation , Substrate Specificity/genetics , Thermus/enzymology , Amino Acids , Amylases/genetics , Amylopectin/metabolism , Amylose/metabolism , Binding Sites/genetics , Dimerization , Kinetics
18.
J Agric Food Chem ; 55(12): 4735-40, 2007 Jun 13.
Article in English | MEDLINE | ID: mdl-17488117

ABSTRACT

A bread-baking process was developed using a potential novel enzyme, cyclodextrin glucanotransferase[3-18] (CGTase[3-18]), that had previously been engineered to have enhanced hydrolyzing activity with little cyclodextrin (CD) formation activity toward starch. CGTase[3-18] was primarily manipulated to be displayed on the cell surface of Saccharomyces cerevisiae. S. cerevisiae carrying pdeltaCGT integrated into the chromosome exhibited starch-hydrolyzing activity at the same optimal pH and temperature as the free enzyme. Volumes of the bread loaves and rice cakes prepared using S. cerevisiae/pdeltaCGT increased by 20% and 45%, respectively, with no detectable CD. Retrogradation rates of the bread and rice cakes decreased significantly during storage. In comparison to the wild type, S. cerevisiae/pdeltaCGT showed improved viability during four freeze-thaw cycles. The results indicated that CGTase[3-18] displayed on the surface of yeast hydrolyzed starch to glucose and maltose that can be used more efficiently for yeast fermentation. Therefore, display of an antistaling enzyme on the cell surface of yeast has potential for enhancing the baking process.


Subject(s)
Bread , Glucosyltransferases/genetics , Yeasts/genetics , Cell Membrane/enzymology , Cloning, Molecular , Cooking , Enzyme Stability , Gene Transfer Techniques , Genetic Engineering , Glucosyltransferases/metabolism , Oryza , Plasmids , Polymerase Chain Reaction , Recombinant Proteins/metabolism
19.
Biosci Biotechnol Biochem ; 71(5): 1348-52, 2007 May.
Article in English | MEDLINE | ID: mdl-17485831

ABSTRACT

A treX in the trehalose biosynthesis gene cluster of Sulfolobus solfataricus ATCC 35092 has been reported to produce TreX, which hydrolyzes the alpha-1,6-branch portion of amylopectin and glycogen. TreX exhibited 4-alpha-D-glucan transferase activity, catalyzing the transfer of alpha-1,4-glucan oligosaccharides from one molecule to another in the case of linear maltooligosaccharides (G3-G7), and it produced cyclic glucans from amylopectin and amylose like 4-alpha-glucanotransferase. These results suggest that TreX is a novel isoamylase possessing the properties of 4-alpha-glucanotransferase.


Subject(s)
Glycogen Debranching Enzyme System/genetics , Isoamylase/metabolism , Sulfolobus solfataricus/genetics , Trehalose/biosynthesis , Genes, Bacterial , Glucans/biosynthesis , Glucans/chemistry , Glycogen Debranching Enzyme System/metabolism , Operon , Sulfolobus solfataricus/enzymology
20.
Biochim Biophys Acta ; 1764(10): 1633-8, 2006 Oct.
Article in English | MEDLINE | ID: mdl-17035108

ABSTRACT

A W229H mutant of 4-alpha-glucanotransferase (4-alpha-GTase) from Pyrococcus furiosus was constructed and its catalytic properties were studied to investigate the role of W229 in the catalytic specificities of the enzyme. Various activities and kinetic parameters were determined for the wild-type and W229H mutant enzymes. The transglycosylation factor and transglycosylation activity of the mutant enzyme markedly decreased, but its hydrolysis activity was scarcely affected. It was discovered that the k(cat)/K(m) value of transglycosylation activity significantly decreased to about 15% of that of the wild type, while k(cat)/K(m) value of hydrolysis activity changed little for the mutant enzyme. The hydrophobicity of W229 was thought to be critical to the transglycosylation activity of the enzyme based on the enzyme's modeled tertiary structures.


Subject(s)
Glycogen Debranching Enzyme System/chemistry , Pyrococcus furiosus/enzymology , Tryptophan/chemistry , Amino Acid Sequence , Catalysis , Glycogen Debranching Enzyme System/genetics , Glycosylation , Hydrophobic and Hydrophilic Interactions , Kinetics , Models, Chemical , Molecular Sequence Data , Mutagenesis, Site-Directed , Mutation , Protein Structure, Tertiary , Tryptophan/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...