Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 171
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Invest New Drugs ; 39(6): 1559-1567, 2021 12.
Article in English | MEDLINE | ID: mdl-34117970

ABSTRACT

Background osteosarcoma is a rare, primary malignant bone tumour with limited available treatments for advanced or recurrent disease, resulting in a poor prognosis for patients. TAS-115 is a novel tyrosine kinase inhibitor under investigation in a phase I study in patients with solid tumours. We report data of osteosarcoma patients in the expansion cohort of this ongoing study. Patients and methods an analysis of this multicentre, open-label study was performed 6 months after the final patient was enrolled, and included patients aged ≥15 years, with unresectable or recurrent osteosarcoma, and who had refractory to standard therapy or for whom no standard therapy was available. TAS-115 650 mg/day was orally administered in a 5 days on/2 days off schedule. Results a total of 20 patients with osteosarcoma were enrolled. The most common adverse drug reactions (ADRs) were neutrophil count decreased (75%), aspartate aminotransferase increased (50%), and platelet count decreased (50%); 85% of patients had grade ≥ 3 ADRs. Long-term disease control (>1 year) with TAS-115 was achieved in three patients. The best overall response was stable disease (50%); no patient achieved a complete or partial response. Median progression-free survival was 3 months; 4-month and 12-month progression-free rates were 42% and 31%, respectively. Conclusion the safety and tolerability of TAS-115 and long-term disease stability for patients with unresectable or recurrent osteosarcoma were confirmed in this study, suggesting that TAS-115 is a promising novel therapy for advanced osteosarcoma patients. Trial registration number: JapicCTI-132333 (registered on November 8, 2013).


Subject(s)
Antineoplastic Agents/therapeutic use , Osteosarcoma/drug therapy , Protein Kinase Inhibitors/therapeutic use , Quinolines/therapeutic use , Thiourea/analogs & derivatives , Adolescent , Adult , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Biomarkers, Tumor , Dose-Response Relationship, Drug , Female , Humans , Male , Maximum Tolerated Dose , Middle Aged , Protein Kinase Inhibitors/adverse effects , Quinolines/administration & dosage , Quinolines/adverse effects , Survival Analysis , Thiourea/administration & dosage , Thiourea/adverse effects , Thiourea/therapeutic use , Young Adult
2.
Int J Med Sci ; 18(1): 137-149, 2021.
Article in English | MEDLINE | ID: mdl-33390782

ABSTRACT

The prognosis for patients with relapsed or refractory high-risk neuroblastoma remains dismal and novel therapeutic options are urgently needed. The RIST treatment protocol has a multimodal metronomic therapy design combining molecular-targeted drugs (Rapamycin and Dasatinib) with chemotherapy backbone (Irinotecan and Temozolomide), which is currently verified in a phase II clinical trial (NCT01467986). With the availability of novel and more potent ATP competitive mTOR inhibitors, we expect to improve the RIST combination therapy. By comparing the IC50 values of Torin-1, Torin-2, AZD3147 and PP242 we established that only Torin-2 inhibited cell viability of all three MycN-amplified neuroblastoma cell lines tested at nanomolar concentration. Single treatment of both mTOR inhibitors induced a significant G1 cell cycle arrest and combination treatment with Dasatinib reduced the expression of cell cycle regulator cyclin D1 or increased the expression of cell cycle inhibitor p21. The combinatorial index depicted for both mTOR inhibitors a synergistic effect with Dasatinib. Interestingly, compared to Rapamycin, the combination treatment with Torin-2 resulted in a broader mTOR pathway inhibition as indicated by reduced phosphorylation of AKT (Thr308, Ser473), 4E-BP (Ser65), and S6K (Thr389). Furthermore, substituting Rapamycin in the modified multimodal RIST protocol with Torin-2 reduced cell viability and induced apoptosis despite a significant lower Torin-2 drug concentration applied. The efficacy of nanomolar concentrations may significantly reduce unwanted immunosuppression associated with Rapamycin. However, at this point we cannot rule out that Torin-2 has increased toxicity due to its potency in more complex systems. Nonetheless, our results suggest that including Torin-2 as a substitute for Rapamycin in the RIST protocol may represent a valid option to be evaluated in prospective clinical trials for relapsed or treatment-refractory high-risk neuroblastoma.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Naphthyridines/administration & dosage , Neuroblastoma/drug therapy , Sirolimus/administration & dosage , TOR Serine-Threonine Kinases/antagonists & inhibitors , Administration, Metronomic , Cell Line, Tumor , Cell Survival/drug effects , Dasatinib/administration & dosage , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Drug Synergism , Humans , Indoles/administration & dosage , Inhibitory Concentration 50 , Irinotecan/administration & dosage , Neuroblastoma/pathology , Purines/administration & dosage , Pyrimidines/administration & dosage , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism , Temozolomide/administration & dosage , Thiourea/administration & dosage , Thiourea/analogs & derivatives
3.
Invest New Drugs ; 38(4): 1175-1185, 2020 08.
Article in English | MEDLINE | ID: mdl-31820255

ABSTRACT

TAS-115 is a novel MET, VEGFR, FMS and PDGFR inhibitor, developed to improve the continuity of drug administration with a relatively short half-life. We assessed its tolerability, safety, pharmacokinetics, efficacy, and pharmacodynamics in patients with solid tumors. This open-label, dose-escalation phase I study of TAS-115 consisted of three parts: part 1 (TAS-115 was administered orally once daily [SID]); part 2 and an expansion part (SID in a 5 days on/2 days off [5-on/2-off] schedule for 21 days per cycle). In part 1 (200-800 mg SID administered to 21 patients), systemic exposure after single administration increased almost dose-proportionally. Three dose-limiting toxicities (DLTs) were observed in three patients: grade 3 rash (650 mg), thrombocytopenia with bleeding, and rash (800 mg). The maximum tolerated dose (MTD) was determined as 650 mg SID. In part 2, the 5-on/2-off schedule was evaluated at the MTD to improve treatment exposure. No DLTs were observed and no patients required treatment interruption in cycle 1. During part 2 and the expansion part (N = 61), grade ≥3 treatment-related adverse events were reported in 47 patients, with neutropenia (24.6%), hypophosphatemia (21.3%), anemia, and thrombocytopenia (14.8% each), and leukocytopenia (11.5%) occurring in ≥10% of patients. The best overall response was stable disease in 31 of 82 patients (37.8%). An apparent reduction in fluorodesoxyglucose-uptake and bone scan index was observed in some patients. TAS-115 was generally well tolerated, with manageable toxicities and recommended phase II dose was estimated as 650 mg SID, 5-on/2-off. Furthermore, promising antitumor activity was observed.


Subject(s)
Antineoplastic Agents/administration & dosage , Neoplasms/drug therapy , Protein Kinase Inhibitors/administration & dosage , Quinolines/administration & dosage , Thiourea/analogs & derivatives , Adolescent , Adult , Aged , Aged, 80 and over , Antineoplastic Agents/adverse effects , Antineoplastic Agents/blood , Antineoplastic Agents/pharmacokinetics , Drug Administration Schedule , Female , Humans , Male , Maximum Tolerated Dose , Middle Aged , Neoplasms/blood , Neoplasms/metabolism , Protein Kinase Inhibitors/adverse effects , Protein Kinase Inhibitors/blood , Protein Kinase Inhibitors/pharmacokinetics , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Proto-Oncogene Proteins c-met/blood , Quinolines/adverse effects , Quinolines/blood , Quinolines/pharmacokinetics , Thiourea/administration & dosage , Thiourea/adverse effects , Thiourea/blood , Thiourea/pharmacokinetics , Vascular Endothelial Growth Factor A/blood , Vascular Endothelial Growth Factor Receptor-2/blood , Young Adult
4.
Bioorg Chem ; 88: 102941, 2019 07.
Article in English | MEDLINE | ID: mdl-31039470

ABSTRACT

Hydrogen sulfide (H2S) is a biologically important gaseous molecule that exhibits promising protective effects against a variety of pathological processes. For example, it was recognized as a blood pressure lowering agent. Aligned with the need for easily modifiable platforms for the H2S supply, we report here the preparation and the H2S release kinetics from a series of structurally diversified thioamides, thiolactams and thioureas. Three different thionation methods based on the usage of a phosphorus pentasulfide and Lawesson reagent were applied to prepare the target thioamides and thiolactams. Furthermore, obtained H2S donors were evaluated both in in vivo and in vitro studies. The kinetic parameters of the liberating H2S was determined and compared with NaHS and GYY4137 using two different detection technics i.e.; fluorescence labeling 7-azido-4-methyl-2H-chromen-2-one and 5,5'-dithiobis (2-nitrobenzoic acid), sulfhydryl probe, also known as the Ellman's reagent. We have proved that the amount of releasing H2S from these compounds is controllable through structural modifications. Finally, the present study shows a hypotensive response to an intravenous administration of the developed donors in the anesthetized rats.


Subject(s)
Blood Pressure/drug effects , Hydrogen Sulfide/analysis , Lactams/pharmacology , Thioamides/pharmacology , Thiourea/pharmacology , Administration, Intravenous , Animals , Kinetics , Lactams/administration & dosage , Lactams/chemistry , Male , Rats , Rats, Sprague-Dawley , Thioamides/administration & dosage , Thioamides/chemistry , Thiourea/administration & dosage , Thiourea/chemistry
5.
Water Sci Technol ; 77(11-12): 2876-2885, 2018 Jul.
Article in English | MEDLINE | ID: mdl-30065140

ABSTRACT

Allylthiourea is a known specific inhibitor for ammonium oxidiser to suppress its oxygen uptake, and is commonly used for various kinds of batch respirometric tests to detect heterotrophic respiration in activated sludge. However, when high heterotrophs were present in the sample, it appeared the inhibitor was noticeably degraded and reached below the inhibition threshold after a couple of days, which resulted in overestimation of the heterotrophic respiration. The biological decomposition of the inhibitor was expressed with a Monod-type rate expression having a half-saturation coefficient of 980 mg-COD/L and maximum specific growth rate of 1.0 d-1. The developed kinetic model, including the growth and decay of the heterotrophs and nitrifiers, indicated that the ATU with about 90 mg-ATU/L which was initially dosed to the system would reach below the inhibition threshold of 1.0 mg-ATU/L after 10 days when 750 mg-COD/L of heterotrophs were present. From the kinetic model, an empirical formula to calculate a safe minimum ATU dose for the batch respirometric test was elaborated. The model also provided a modified experimental procedure to accurately estimate the initial heterotrophic biomass concentration in the sample and its specific decay rate based on IWA Activated Sludge Models.


Subject(s)
Sewage/microbiology , Thiourea/analogs & derivatives , Biological Oxygen Demand Analysis , Biomass , Bioreactors , Heterotrophic Processes , Kinetics , Models, Theoretical , Nitrification , Oxygen/analysis , Thiourea/administration & dosage , Thiourea/chemistry , Thiourea/metabolism , Waste Disposal, Fluid/instrumentation , Waste Disposal, Fluid/methods
6.
Biochem Biophys Res Commun ; 493(4): 1371-1376, 2017 12 02.
Article in English | MEDLINE | ID: mdl-28988108

ABSTRACT

The role of UVB in skin photo damages has been widely reported. Overexposure to UVB will induce severe DNA damages in epidermal cells and cause most cytotoxic symptoms. In the present study, we tested the potential activity of salubrinal, a selective inhibitor of Eukaryotic Initiation Factor 2 (eIF2) -alpha phosphatase, against UV-induced skin cell damages. We first exposed human fibroblasts to UVB radiation and evaluated the cytosolic Ca2+ level as well as the induction of ER stress. We found that UVB radiation induced the depletion of ER Ca2+ and increased the expression of ER stress marker including phosphorylated PERK, CHOP, and phosphorylated IRE1α. We then determined the effects of salubrinal in skin cell death induced by UVB radiation. We observed that cells pre-treated with salubrinal had a higher survival rate compared to cells treated with UVB alone. Pre-treatment with salubrinal successfully re-established the ER function and Ca2+ homeostasis. Our results suggest that salubrinal can be a potential therapeutic agents used in preventing photoaging and photo damages.


Subject(s)
Cinnamates/pharmacology , Radiation-Protective Agents/pharmacology , Skin Aging/drug effects , Skin/drug effects , Skin/radiation effects , Thiourea/analogs & derivatives , Calcium/metabolism , Cell Death/drug effects , Cell Death/radiation effects , Cells, Cultured , Cinnamates/administration & dosage , Dose-Response Relationship, Drug , Endoplasmic Reticulum Stress/drug effects , Endoplasmic Reticulum Stress/radiation effects , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibroblasts/pathology , Homeostasis/drug effects , Humans , Radiation-Protective Agents/administration & dosage , Skin/metabolism , Skin Aging/pathology , Skin Aging/physiology , Sunscreening Agents/administration & dosage , Sunscreening Agents/pharmacology , Thiourea/administration & dosage , Thiourea/pharmacology , Ultraviolet Rays/adverse effects
7.
Bioorg Med Chem ; 25(14): 3736-3745, 2017 07 15.
Article in English | MEDLINE | ID: mdl-28533113

ABSTRACT

Serine racemase (SRR) is an enzyme that produces d-serine from l-serine. d-Serine acts as an endogenous coagonist of NMDA-type glutamate receptors (NMDARs), which regulate many physiological functions. Over-activation of NMDARs induces excitotoxicity, which is observed in many neurodegenerative disorders and epilepsy states. In our previous works on the generation of SRR gene knockout (Srr-KO) mice and its protective effects against NMDA- and Aß peptide-induced neurodegeneration, we hypothesized that the regulation of NMDARs' over-activation by inhibition of SRR activity is one such therapeutic strategy to combat these disease states. In the previous study, we performed in silico screening to identify four compounds with inhibitory activities against recombinant SRR. Here, we synthesized 21 derivatives of candidate 1, one of four hit compounds, and performed screening by in vitro evaluations. The derivative 13J showed a significantly lower IC50 value in vitro, and suppressed neuronal over-activation in vivo.


Subject(s)
Acrylamides/chemistry , Enzyme Inhibitors/chemistry , Protective Agents/chemistry , Racemases and Epimerases/antagonists & inhibitors , Thiourea/analogs & derivatives , Acrylamides/administration & dosage , Acrylamides/chemical synthesis , Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/metabolism , Animals , Binding Sites , Brain/drug effects , Brain/metabolism , Enzyme Inhibitors/metabolism , Enzyme Inhibitors/pharmacology , Humans , Hydrogen Bonding , Mice , Mice, Knockout , Mice, Transgenic , Molecular Docking Simulation , Optical Imaging , Protective Agents/chemical synthesis , Protective Agents/pharmacology , Protein Structure, Tertiary , Racemases and Epimerases/genetics , Racemases and Epimerases/metabolism , Receptors, N-Methyl-D-Aspartate/chemistry , Receptors, N-Methyl-D-Aspartate/metabolism , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Thiourea/administration & dosage , Thiourea/chemical synthesis , Thiourea/chemistry
8.
Carcinogenesis ; 37(8): 827-838, 2016 08.
Article in English | MEDLINE | ID: mdl-27267997

ABSTRACT

Androgen deprivation therapy in prostate cancer is extremely effective; however, due to the continuous expression and/or mutagenesis of androgen receptor (AR), the resistance to antihormonal therapy is a natural progression. Consequently, targeting the AR for degradation offers an alternate approach to overcome this resistance in prostate cancer. In this study, we demonstrate that carnosic acid, a benzenediol diterpene, binds the ligand-binding domain of the AR and degrades the AR via endoplasmic reticulum (ER) stress-mediated proteasomal degradative pathway. In vitro, carnosic acid treatment induced degradation of AR and decreased expression of prostate-specific antigen in human prostate cancer cell lines LNCaP and 22Rv1. Carnosic acid also promoted the expression of ER proteins including BiP and CHOP in a dose-dependent manner. Downregulation of CHOP by small interfering RNA somewhat restored expression of AR suggesting that AR degradation is dependent on ER stress pathway. Future studies will need to evaluate other aspects of the unfolded protein response pathway to characterize the regulation of AR degradation. Furthermore, cotreating cells individually with carnosic acid and proteasome inhibitor (MG-132) and carnosic acid and an ER stress modulator (salubrinal) restored protein levels of AR, suggesting that AR degradation is mediated by ER stress-dependent proteasomal degradation pathway. Degradation of AR and induction of CHOP protein were also evident in vivo along with a 53% reduction in growth of xenograft prostate cancer tumors. In addition, carnosic acid-induced ER stress in prostate cancer cells but not in normal prostate epithelial cells procured from patient biopsies. In conclusion, these data suggest that molecules such as carnosic acid could be further evaluated and optimized as a potential therapeutic alternative to target AR in prostate cancer.


Subject(s)
Abietanes/metabolism , Prostate-Specific Antigen/genetics , Prostatic Neoplasms/drug therapy , Receptors, Androgen/biosynthesis , Transcription Factor CHOP/biosynthesis , Abietanes/administration & dosage , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cinnamates/administration & dosage , Endoplasmic Reticulum Stress/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Leupeptins/administration & dosage , Male , Mice , Prostate-Specific Antigen/biosynthesis , Prostate-Specific Antigen/metabolism , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Proteolysis/drug effects , Thiourea/administration & dosage , Thiourea/analogs & derivatives , Transcription Factor CHOP/genetics , Unfolded Protein Response/drug effects , Xenograft Model Antitumor Assays
9.
Int J Cancer ; 138(11): 2665-77, 2016 Jun 01.
Article in English | MEDLINE | ID: mdl-26757413

ABSTRACT

It is estimated that approximately 90% of patients with advanced prostate cancer develop bone metastases; an occurrence that results in a substantial reduction in the quality of life and a drastic worsening of prognosis. The development of novel therapeutic strategies that impair the metastatic process and associated skeletal adversities is therefore critical to improving prostate cancer patient survival. Recognition of the importance of Cathepsin L (CTSL) to metastatic dissemination of cancer cells has led to the development of several CTSL inhibition strategies. The present investigation employed intra-cardiac injection of human PC-3ML prostate cancer cells into nude mice to examine tumor cell dissemination in a preclinical bone metastasis model. CTSL knockdown confirmed the validity of targeting this protease and subsequent intervention studies with the small molecule CTSL inhibitor KGP94 resulted in a significant reduction in metastatic tumor burden in the bone and an improvement in overall survival. CTSL inhibition by KGP94 also led to a significant impairment of tumor initiated angiogenesis. Furthermore, KGP94 treatment decreased osteoclast formation and bone resorptive function, thus, perturbing the reciprocal interactions between tumor cells and osteoclasts within the bone microenvironment which typically result in bone loss and aggressive growth of metastases. These functional effects were accompanied by a significant downregulation of NFκB signaling activity and expression of osteoclastogenesis related NFκB target genes. Collectively, these data indicate that the CTSL inhibitor KGP94 has the potential to alleviate metastatic disease progression and associated skeletal morbidities and hence may have utility in the treatment of advanced prostate cancer patients.


Subject(s)
Bone Neoplasms/genetics , Cathepsin L/genetics , Osteoclasts/metabolism , Prostatic Neoplasms/genetics , Animals , Bone Neoplasms/pathology , Bone Neoplasms/secondary , Bone Resorption/genetics , Bone Resorption/pathology , Cathepsin L/antagonists & inhibitors , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , Male , Mice , Neoplasm Metastasis , Osteoclasts/pathology , Prostatic Neoplasms/pathology , Thiosemicarbazones/administration & dosage , Thiourea/administration & dosage , Thiourea/analogs & derivatives , Tumor Burden/genetics , Xenograft Model Antitumor Assays
10.
Int J Mol Sci ; 17(11)2016 Oct 27.
Article in English | MEDLINE | ID: mdl-27801783

ABSTRACT

Natural compounds from soft corals have been increasingly used for their antitumor therapeutic properties. This study examined 11-epi-sinulariolide acetate (11-epi-SA), an active compound isolated from the cultured soft coral Sinularia flexibilis, to determine its potential antitumor effect on four hepatocellular carcinoma cell lines. Cell viability was investigated using 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay, and the results demonstrated that 11-epi-SA treatment showed more cytotoxic effect toward HA22T cells. Protein profiling of the 11-epi-SA-treated HA22T cells revealed substantial protein alterations associated with stress response and protein synthesis and folding, suggesting that the mitochondria and endoplasmic reticulum (ER) play roles in 11-epi-SA-initiated apoptosis. Moreover, 11-epi-SA activated caspase-dependent apoptotic cell death, suggesting that mitochondria-related apoptosis genes were involved in programmed cell death. The unfolded protein response signaling pathway-related proteins were also activated on 11-epi-SA treatment, and these changes were accompanied by the upregulated expression of growth arrest and DNA damage-inducible protein (GADD153) and CCAAT/enhancer binding protein (C/EBP) homologous protein (CHOP), the genes encoding transcription factors associated with growth arrest and apoptosis under prolonged ER stress. Two inhibitors, namely salubrinal (Sal) and SP600125, partially abrogated 11-epi-SA-related cell death, implying that the protein kinase R (PKR)-like endoplasmic reticulum kinase (PERK)-activating transcription factor (ATF) 6-CHOP or the inositol-requiring enzyme 1 alpha (IRE1α)-c-Jun N-terminal kinase (JNK)-cJun signal pathway was activated after 11-epi-SA treatment. In general, these results suggest that 11-epi-SA exerts cytotoxic effects on HA22T cells through mitochondrial dysfunction and ER stress cell death pathways.


Subject(s)
Carcinoma, Hepatocellular/drug therapy , Cell Proliferation/drug effects , Diterpenes/chemistry , Endoplasmic Reticulum Stress/drug effects , Liver Neoplasms/drug therapy , Animals , Anthozoa/chemistry , Anthracenes/administration & dosage , Apoptosis/drug effects , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Cell Survival/drug effects , Cinnamates/administration & dosage , Diterpenes/administration & dosage , Diterpenes/chemical synthesis , Gene Expression Regulation, Neoplastic/drug effects , Humans , JNK Mitogen-Activated Protein Kinases/biosynthesis , JNK Mitogen-Activated Protein Kinases/genetics , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Mitochondria/drug effects , Mitochondria/pathology , Signal Transduction/drug effects , Thiourea/administration & dosage , Thiourea/analogs & derivatives , Transcription Factor CHOP/biosynthesis , Transcription Factor CHOP/genetics , Unfolded Protein Response/drug effects
11.
Hum Mol Genet ; 22(18): 3680-9, 2013 Sep 15.
Article in English | MEDLINE | ID: mdl-23674520

ABSTRACT

Methylmalonic aciduria (MMA) cblB type is caused by mutations in the MMAB gene. This encodes the enzyme ATP:cob(I)alamin adenosyltransferase (ATR), which converts reduced cob(I)alamin to an active adenosylcobalamin cofactor. We recently reported the presence of destabilizing pathogenic mutations that retain some residual ATR activity. The aim of the present study was to seek pharmacological chaperones as a tailored therapy for stabilizing the ATR protein. High-throughput ligand screening of over 2000 compounds was performed; six were found to enhance the thermal stability of purified recombinant ATR. Further studies using a well-established bacterial system in which the recombinant ATR protein was expressed in the presence of these six compounds, showed them all to increase the stability of the wild-type ATR and the p.Ile96Thr mutant proteins. Compound V (N-{[(4-chlorophenyl)carbamothioyl]amino}-2-phenylacetamide) significantly increased this stability and did not act as an inhibitor of the purified protein. Importantly, compound V increased the activity of ATR in patient-derived fibroblasts harboring the destabilizing p.Ile96Thr mutation in a hemizygous state to within control range. When cobalamin was coadministrated with compound V, mutant ATR activity further improved. Oral administration of low doses of compound V to C57BL/6J mice for 12 days, led to increase in steady-state levels of ATR protein in liver and brain (disease-relevant organs). These results hold promise for the clinical use of pharmacological chaperones in MMA cblB type patients harboring chaperone-responsive mutations.


Subject(s)
Alkyl and Aryl Transferases/genetics , Amino Acid Metabolism, Inborn Errors/drug therapy , Benzeneacetamides/chemistry , Benzeneacetamides/pharmacology , Thiourea/analogs & derivatives , Alkyl and Aryl Transferases/chemistry , Alkyl and Aryl Transferases/metabolism , Amino Acid Metabolism, Inborn Errors/metabolism , Animals , Benzeneacetamides/administration & dosage , Binding Sites , Brain/drug effects , Brain/enzymology , Enzyme Stability , Female , High-Throughput Screening Assays , Humans , Liver/drug effects , Liver/enzymology , Mice , Mice, Inbred C57BL , Molecular Docking Simulation , Mutant Proteins/genetics , Mutant Proteins/metabolism , Thiourea/administration & dosage , Thiourea/chemistry , Thiourea/pharmacology , Vitamin B 12/administration & dosage , Vitamin B 12/pharmacology
12.
J Pharmacol Exp Ther ; 352(2): 338-45, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25503385

ABSTRACT

The mitochondrial outer membrane protein mitoNEET is a binding protein of the insulin sensitizer pioglitazone (5-[[4-[2-(5-ethylpyridin-2-yl)ethoxy]phenyl]methyl]-1,3-thiazolidine-2,4-dione) and is considered a novel target for the treatment of type II diabetes. Several small-molecule compounds have been identified as mitoNEET ligands using structure-based design or virtual docking studies. However, there are no reports about their therapeutic potential in animal models. Recently, we synthesized a novel small molecule, TT01001 [ethyl-4-(3-(3,5-dichlorophenyl)thioureido)piperidine-1-carboxylate], designed on the basis of pioglitazone structure. In this study, we assessed the pharmacological properties of TT01001 in both in vitro and in vivo studies. We found that TT01001 bound to mitoNEET without peroxisome proliferator-activated receptor-γ activation effect. In type II diabetes model db/db mice, TT01001 improved hyperglycemia, hyperlipidemia, and glucose intolerance, and its efficacy was equivalent to that of pioglitazone, without the pioglitazone-associated weight gain. Mitochondrial complex II + III activity of the skeletal muscle was significantly increased in db/db mice. We found that TT01001 significantly suppressed the elevated activity of the complex II + III. These results suggest that TT01001 improved type II diabetes without causing weight gain and ameliorated mitochondrial function of db/db mice. This is the first study that demonstrates the effects of a mitoNEET ligand on glucose metabolism and mitochondrial function in an animal disease model. These findings support targeting mitoNEET as a potential therapeutic approach for the treatment of type II diabetes.


Subject(s)
Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Type 2/drug therapy , Hypoglycemic Agents/therapeutic use , Iron-Binding Proteins/metabolism , Membrane Proteins/metabolism , Mitochondria, Muscle/drug effects , Mitochondrial Proteins/metabolism , Piperidines/therapeutic use , Thiourea/analogs & derivatives , Animals , Blood Glucose/analysis , DNA, Mitochondrial/metabolism , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/physiopathology , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/physiopathology , Escherichia coli/genetics , Fluorescence Resonance Energy Transfer , Humans , Hypoglycemic Agents/administration & dosage , Hypoglycemic Agents/pharmacology , Ligands , Male , Mice, Inbred Strains , Mitochondria, Muscle/enzymology , Mitochondria, Muscle/physiology , Mitochondrial Proteins/genetics , PPAR gamma/metabolism , Piperidines/administration & dosage , Piperidines/pharmacology , Surface Plasmon Resonance , Thiourea/administration & dosage , Thiourea/pharmacology , Thiourea/therapeutic use
13.
Int J Neuropsychopharmacol ; 18(9)2015 Mar 11.
Article in English | MEDLINE | ID: mdl-25762718

ABSTRACT

BACKGROUND: Histamine is a modulatory neurotransmitter regulating neuronal activity. Antidepressant drugs target modulatory neurotransmitters, thus ultimately regulating glutamatergic transmission and plasticity. Histamine H3 receptor (H3R) antagonists have both pro-cognitive and antidepressant effects; however, the mechanism by which they modulate glutamate transmission is not clear. We measured the effects of the H3R antagonist clobenpropit in the Flinders Sensitive Line (FSL), a rat model of depression with impaired memory and altered glutamatergic transmission. METHODS: Behavioral tests included the forced swim test, memory tasks (passive avoidance, novel object recognition tests), and anxiety-related paradigms (novelty suppressed feeding, social interaction, light/dark box tests). Hippocampal protein levels were detected by Western blot. Hippocampal plasticity was studied by in slice field recording of CA3-CA1 long-term synaptic potentiation (LTP), and glutamatergic transmission by whole-cell patch clamp recording of excitatory postsynaptic currents (EPSCs) in CA1 pyramidal neurons. RESULTS: Clobenpropit, administered systemically or directly into the hippocampus, decreased immobility during the forced swim test; systemic injections reversed memory deficits and increased hippocampal GluN2A protein levels. FSL rats displayed anxiety-related behaviors not affected by clobenpropit treatment. Clobenpropit enhanced hippocampal plasticity, but did not affect EPSCs. H1R and H2R antagonists prevented the clobenpropit-induced increase in LTP and, injected locally into the hippocampus, blocked clobenpropit's effect in the forced swim test. CONCLUSIONS: Clobenpropit's antidepressant effects and the enhanced synaptic plasticity require hippocampal H1R and H2R activation, suggesting that clobenpropit acts through disinhibition of histamine release. Clobenpropit reverses memory deficits and increases hippocampal GluN2A expression without modifying anxiety-related phenotypes or EPSCs in CA1 pyramidal neurons.


Subject(s)
Antidepressive Agents/pharmacology , Depression/drug therapy , Excitatory Postsynaptic Potentials/drug effects , Glutamic Acid/metabolism , Hippocampus/drug effects , Histamine H3 Antagonists/pharmacology , Imidazoles/pharmacology , Long-Term Potentiation/drug effects , Thiourea/analogs & derivatives , Animals , Antidepressive Agents/administration & dosage , Anxiety/drug therapy , Behavior, Animal/drug effects , Disease Models, Animal , Histamine H3 Antagonists/administration & dosage , Imidazoles/administration & dosage , Male , Memory Disorders/drug therapy , Patch-Clamp Techniques , Pyramidal Cells/drug effects , Rats , Rats, Sprague-Dawley , Thiourea/administration & dosage , Thiourea/pharmacology
14.
Biometals ; 28(5): 891-902, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26174742

ABSTRACT

Zinc (Zn(2+)) is considered to be one of the factors aggravating brain damage after cerebral ischemia. Since Zn(2+) activates microglia, immune cells in the brain, this metal is proposed to modulate neuroinflammatory responses in the post-ischemic brain. Interleukin (IL)-23 is a heterodimeric cytokine composed of the p19 subunit unique to IL-23 and the p40 subunit common to IL-12. IL-23 has been shown to play a critical role in the progression of ischemic brain injury. However, whether Zn(2+) participates in the expression of IL-23 in microglia remains unknown. In this study, we examined the effect of Zn(2+) on IL-23 p19 mRNA expression using rat immortalized microglia HAPI cells. Exposure to Zn(2+) dose- and time-dependently induced the expression of IL-23 p19 mRNA in HAPI cells. Inhibitors of MAPK and NF-κB pathways failed to suppress this induction. Interestingly, we found that Zn(2+) stimulated the phosphorylation of eIF2α and promoted the nuclear accumulation of activating transcription factor 4 (ATF4). Treatment with salubrinal, an eIF2α dephosphorylation inhibitor, enhanced Zn(2+)-induced ATF4 accumulation and IL-23 p19 mRNA expression. In addition, reporter assay using the IL-23 p19 promoter region revealed that ATF4 directly transactivated IL-23 p19 promoter and that dominant-negative ATF4 suppressed Zn(2+)-induced activation of IL-23 p19 promoter. Taken together, these findings suggest that Zn(2+) up-regulates expression of the IL-23 p19 gene via the eIF2α/ATF4 axis in HAPI cells.


Subject(s)
Activating Transcription Factor 4/biosynthesis , Brain Ischemia/drug therapy , Eukaryotic Initiation Factor-2/biosynthesis , Interleukin-23 Subunit p19/biosynthesis , Zinc/administration & dosage , Activating Transcription Factor 4/genetics , Animals , Brain Ischemia/genetics , Brain Ischemia/pathology , Cinnamates/administration & dosage , Eukaryotic Initiation Factor-2/genetics , Gene Expression Regulation/drug effects , Humans , Interleukin-23 Subunit p19/antagonists & inhibitors , Interleukin-23 Subunit p19/genetics , Microglia/drug effects , Microglia/pathology , Neuroimmunomodulation/drug effects , Promoter Regions, Genetic , RNA, Messenger/biosynthesis , Rats , Thiourea/administration & dosage , Thiourea/analogs & derivatives , Zinc/chemistry
15.
Int J Mol Sci ; 16(8): 17637-54, 2015 Jul 31.
Article in English | MEDLINE | ID: mdl-26263980

ABSTRACT

The increase in antibiotic resistant bacteria demands the development of new antibiotics against preferably new targets. The common approach is to test compounds for their ability to kill bacteria or to design molecules that inhibit essential protein activities in vitro. In the first case, the mode of action of the drug is unknown and in the second case, it is not known whether the compound will pass the impermeable barrier of the bacterial envelope. We developed an assay that detects the target of a compound, as well as its ability to pass the membrane(s) simultaneously. The Escherichia coli cytoskeletal protein MreB recruits protein complexes (elongasomes) that are essential for cell envelope growth. An in cell Förster Resonance Energy Transfer (FRET) assay was developed to detect the interaction between MreB molecules and between MreB and the elongasome proteins RodZ, RodA and PBP2. Inhibition of the polymerization of MreB by S-(3,4-dichlorobenzyl) isothiourea (A22) or of the activity of PBP2 by mecilinam resulted in loss or reduction of all measured interactions. This suggests that the interactions between the elongasome proteins are governed by a combination of weak affinities and substrate availability. This validated in cell FRET assay can be used to screen for cell envelope growth inhibitors.


Subject(s)
Escherichia coli Proteins/genetics , Escherichia coli/genetics , Fluorescence Resonance Energy Transfer , Gene Expression Regulation, Bacterial/drug effects , Cytoskeletal Proteins/biosynthesis , Cytoskeletal Proteins/chemistry , Escherichia coli/drug effects , Escherichia coli/growth & development , Escherichia coli Proteins/biosynthesis , Escherichia coli Proteins/chemistry , Growth Inhibitors/administration & dosage , Growth Inhibitors/chemistry , Membrane Proteins/biosynthesis , Membrane Proteins/chemistry , Penicillin-Binding Proteins/biosynthesis , Penicillin-Binding Proteins/chemistry , Substrate Specificity , Thiourea/administration & dosage , Thiourea/analogs & derivatives
16.
Radiats Biol Radioecol ; 55(3): 250-9, 2015.
Article in Russian | MEDLINE | ID: mdl-26310017

ABSTRACT

The study of radioprotective activity of NO-synthase inhibitor, N-S-isothiourea derivative T1023 showed that this compound has a significant therapeutic range of radioprotective activity (5.5-6.0) and its optimal radioprotective dose is 1/4 LD16. The value of its Radiation Dose-Reduction Factor totaled 1.4-1.8. We have demonstrated a pronounced pharmacodynamic interaction of T1023 with some known radioprotectors. The character of the interaction was determined by its vasoactive properties. Combined use of T1023 and cystamine, which causes a decrease in vascular tone, was accompanied by a statistically significant weakening of the radioprotective effect. But, the combined use of T1023 with serotonergic and adrenergic radioprotectors having a pressor action caused a statistically significant increase in the radioprotective effect. Moreover, T1023 combined with such radioprotectors caused the synergistic radioprotective effect even when used at small doses that do not have any radioprotective effect alone. The findings suggest that NOS inhibitors can be effective radioprotectors and are able to create new opportunities for the development of safer radioprotective agents. The very same compound T1023, according to current criteria of pharmacological screening, is certainly promising for further investigations.


Subject(s)
Enzyme Inhibitors/administration & dosage , Radiation Protection , Radiation-Protective Agents/administration & dosage , Thiourea/analogs & derivatives , Animals , Cystamine/administration & dosage , Enzyme Inhibitors/chemical synthesis , Gamma Rays , Humans , Mice , Nitric Oxide Synthase/antagonists & inhibitors , Radiation Dosage , Radiation Injuries, Experimental , Radiation-Protective Agents/chemical synthesis , Thiourea/administration & dosage
17.
J Cardiovasc Pharmacol ; 64(4): 360-7, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25286361

ABSTRACT

: PP1-12, a new protein phosphatase-1 inhibitor, is designed and synthesized to modulate the endoplasmic reticulum (ER) stress apoptotic pathway, which is involved in various cardiovascular diseases. In this study, we examined the effect of PP1-12 on ventricular remodeling and heart function after myocardial infarction. Rats that survived within 24 hours after coronary ligation were randomly divided into 6 groups and treated with normal saline, vehicle, PP1-12 at 1, 3, and 10 mg·kg·d and perindopril at 2 mg·kg·d for 4 weeks, respectively. At the end of the follow-up point, we evaluated echocardiographic and hemodynamic parameters, myocardial pathomorphology, apoptosis, and interstitial fibrosis, as well as the expression levels of important proteins involved in ER stress and apoptosis. Left ventricular geometry and function were ameliorated by PP1-12. PP1-12 inhibited interstitial fibrosis and reduced apoptosis of cardiomyocytes in a dose-dependent manner. PP1-12 decreased GRP78 and caspase-12 expression and increased p-eIF2α and Bcl-2/Bax expression. These results suggest that PP1-12 efficiently inhibits left ventricular remodeling and improves heart function. The mechanism involved may be associated with the ability of PP1-12 to depress myocardial apoptosis induced by ER stress.


Subject(s)
Acrylamides/therapeutic use , Endoplasmic Reticulum Stress/drug effects , Enzyme Inhibitors/therapeutic use , Heart Ventricles/drug effects , Myocardial Infarction/drug therapy , Protein Phosphatase 1/antagonists & inhibitors , Thiourea/analogs & derivatives , Ventricular Remodeling/drug effects , Acrylamides/administration & dosage , Acrylamides/pharmacology , Animals , Apoptosis/drug effects , Blotting, Western , Dose-Response Relationship, Drug , Echocardiography , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/pharmacology , Heart Ventricles/pathology , Heart Ventricles/physiopathology , Hemodynamics/drug effects , In Situ Nick-End Labeling , Male , Myocardial Infarction/enzymology , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Rats, Sprague-Dawley , Thiourea/administration & dosage , Thiourea/pharmacology , Thiourea/therapeutic use , Ventricular Function, Left
18.
Brain ; 136(Pt 4): 1161-76, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23518711

ABSTRACT

Activating transcription factor 5 (ATF5) is a basic-leucine-zipper transcription factor of the ATF/CREB family. The Atf5 gene generates two transcripts, Atf5α and Atf5ß, of which Atf5α is known to be selectively translated upon endoplasmic reticulum stress response in non-neuronal cells. ATF5 is highly expressed in the developing brain where it modulates proliferation of neural progenitor cells. These cells show a high level of ATF5 that has to decrease to allow them to differentiate into mature neurons or glial cells. This has led to the extended notion that differentiated neural cells do not express ATF5 unless they undergo tumourigenic transformation. However, no systematic analysis of the distribution of ATF5 in adult brain or of its potential role in neuronal endoplasmic reticulum stress response has been reported. By immunostaining here we confirm highest ATF5 levels in neuroprogenitor cells of the embryonic and adult subventricular zone but also found ATF5 in a large variety of neurons in adult mouse brain. By combining Atf5 in situ hybridization and immunohistochemistry for the neuronal marker NeuN we further confirmed Atf5 messenger RNA in adult mouse neurons. Quantitative reverse transcriptase polymerase chain reaction demonstrated that Atf5α is the most abundant transcript in adult mouse encephalon and injection of the endoplasmic reticulum stress inducer tunicamycin into adult mouse brain increased neuronal ATF5 levels. Accordingly, ATF5 levels increased in hippocampal neurons of a mouse model of status epilepticus triggered by intra-amygdala injection of kainic acid, which leads to abnormal hippocampal neuronal activity and endoplasmic reticulum stress. Interestingly, ATF5 upregulation occurred mainly in hippocampal neuronal fields that do not undergo apoptosis in this status epilepticus model such as CA1 and dentate gyrus, thus suggesting a neuroprotective role. This was confirmed in a primary neuronal culture model in which ATF5 overexpression resulted in decreased endoplasmic reticulum stress-induced apoptosis and the opposite result was achieved by Atf5 RNA interference. Furthermore, in vivo administration of the eIF2α phosphatase inhibitor salubrinal resulted in increased ATF5 hippocampal levels and attenuated status epilepticus-induced neuronal death in the vulnerable CA3 subfield. In good agreement with the neuroprotective effect of increased ATF5, we found that apoptosis-resistant epileptogenic foci from patients with temporal lobe epilepsy also showed increased levels of ATF5. Thus, our results demonstrate that adult neurons express ATF5 and that they increase its levels upon endoplasmic reticulum stress as a pro-survival mechanism, thus opening a new field for neuroprotective strategies focused on ATF5 modulation.


Subject(s)
Activating Transcription Factors/biosynthesis , Endoplasmic Reticulum Stress/physiology , Neurons/metabolism , Neuroprotective Agents/metabolism , Status Epilepticus/metabolism , Status Epilepticus/pathology , Animals , Apoptosis/drug effects , Apoptosis/physiology , Cinnamates/administration & dosage , Cinnamates/pharmacology , Disease Models, Animal , Endoplasmic Reticulum Stress/drug effects , Humans , Mice , Mice, Inbred C57BL , Neurons/drug effects , Neurons/pathology , Status Epilepticus/drug therapy , Thiourea/administration & dosage , Thiourea/analogs & derivatives , Thiourea/pharmacology
19.
Digestion ; 90(3): 167-78, 2014.
Article in English | MEDLINE | ID: mdl-25339182

ABSTRACT

BACKGROUND/AIMS: Endoplasmic reticulum (ER) stress in the intestine is closely associated with the development of inflammatory bowel disease (IBD). However, the role of the protein kinase RNA-like ER kinase in this disease is not fully known. We studied whether an inhibitor of the dephosphorylation of eukaryotic initiation factor 2α, salubrinal, improves murine experimental colitis through the amelioration of ER stress. METHODS: Colitis was induced by the administration of 3% dextran sulfate sodium (DSS) for 5 days. Mice were injected salubrinal intraperitoneally from the commencement of DSS treatment and were sacrificed on day 10. The severity of colitis was evaluated histologically using a scoring system.Myeloperoxidase activity and the expression of proinflammatory cytokine genes in the colon were analyzed. The expression levels of ER stress-related proteins were evaluated by Western blotting. RESULTS: The administration of salubrinal significantly attenuated body weight loss and improved colitis, as assessed histologically. The elevation of myeloperoxidase activity and the expression of proinflammatory cytokine genes were suppressed in salubrinal-treated mice. The expression of glucose-regulated protein 78, activating translation factor 4, and heat-shock protein 70 was elevated in mice treated with salubrinal. CONCLUSION: The amelioration of ER stress may be a therapeutic target for the treatment of IBD.


Subject(s)
Cinnamates/administration & dosage , Colitis/drug therapy , DNA-Binding Proteins/metabolism , Endoplasmic Reticulum Stress/drug effects , Eukaryotic Initiation Factor-2/drug effects , Thiourea/analogs & derivatives , Transcription Factors/metabolism , eIF-2 Kinase/metabolism , Animals , Colitis/chemically induced , Colitis/metabolism , Colitis/pathology , Colon/drug effects , Colon/metabolism , Colon/pathology , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/drug effects , DNA-Binding Proteins/genetics , Dextran Sulfate , Disease Models, Animal , Endoplasmic Reticulum Chaperone BiP , HSP70 Heat-Shock Proteins/metabolism , Heat-Shock Proteins/metabolism , Injections, Intraperitoneal , Interleukins/genetics , Interleukins/metabolism , Male , Mice , Mice, Inbred C57BL , Peroxidase/metabolism , RNA, Messenger/metabolism , Regulatory Factor X Transcription Factors , Thiourea/administration & dosage , Transcription Factors/antagonists & inhibitors , Transcription Factors/drug effects , Transcription Factors/genetics , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , Weight Loss/drug effects , eIF-2 Kinase/drug effects , eIF-2 Kinase/genetics
20.
Article in English | MEDLINE | ID: mdl-24845199

ABSTRACT

KB-R7943 (2-[2-[4-(4-nitrobenzyloxy)phenyl]ethyl]isothiourea) was developed as a specific inhibitor of the sarcolemmal sodium-calcium exchanger (NCX) with potential experimental and therapeutic use. However, in cardiomyocytes KB-R7943 also effectively blocks several K(+) currents including the delayed rectifier, IKr, and background inward rectifier, IK1. In the present study we analyze the effects of KB-R7943 on the ATP-dependent potassium current (IKATP) recorded by whole-cell patch-clamp in ventricular cardiomyocytes from a mammal (mouse) and a fish (crucian carp). IKATP was induced by external application of a mitochondrial uncoupler CCCP (3×10(-7) M) and internal perfusion of the cell with ATP-free pipette solution. A weakly inwardly rectifying current with a large outward component, recorded in the presence of CCCP, was blocked with 10(-5) M glibenclamide by 56.1±4.6% and 56.9±3.6% in crucian carp and mouse ventricular myocytes, respectively. In fish cardiomyocytes IKATP was blocked by KB-R7943 with an IC50 value of 3.14×10(-7) M, while in mammalian cells IC50 was 2.8×10(-6) M (P<0.05). 10(-5) M KB-R7943 inhibited CCCP-induced IKATP by 99.9±0.13% and 97.5±1.2% in crucian carp and mouse ventricular myocytes, respectively. In crucian carp the IKATP is about an order of magnitude more sensitive to KB-R7943 than the background IK1, but in mammals IKATP and IK1 are almost equally sensitive to KB-R7943. Therefore, the ability of KB-R7943 to block IKATP should be taken into account together with INCX inhibition when investigating possible cardioprotective effects of this compound.


Subject(s)
Myocytes, Cardiac/drug effects , Potassium/metabolism , Thiourea/analogs & derivatives , Adenosine Triphosphate/metabolism , Animals , Heart/drug effects , Heart Ventricles/drug effects , Heart Ventricles/metabolism , Ion Transport/genetics , Mice , Myocytes, Cardiac/metabolism , Patch-Clamp Techniques , Sodium-Calcium Exchanger/metabolism , Thiourea/administration & dosage
SELECTION OF CITATIONS
SEARCH DETAIL