Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 244
Filter
1.
Expert Opin Investig Drugs ; 33(1): 9-17, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38265202

ABSTRACT

INTRODUCTION: Chronic hepatitis C virus (HCV) persists as a public health concern worldwide. Consequently, optimizing HCV therapy remains an important objective. While current therapies are generally highly effective, advanced antiviral agents are needed to maximize cure rates with potentially shorter treatment durations in a broader patient population, particularly those patients with advanced diseases who remain difficult to treat. AREAS COVERED: This review summarizes the in vitro anti-HCV activity, preclinical pharmacological properties of bemnifosbuvir (BEM, AT-527), a novel prodrug that is metabolically converted to AT-9010, the active guanosine triphosphate analogue that potently and selectively inhibits several viral RNA polymerases, including the HCV NS5B polymerase. Results from clinical proof-of-concept and phase 2 combination studies are also discussed. EXPERT OPINION: BEM exhibits potent pan-genotype activity against HCV, and has favorable safety, and drug interaction profiles. BEM is approximately 10-fold more potent than sofosbuvir against HCV genotypes (GT) tested in vitro. When combined with a potent NS5A inhibitor, BEM is expected to be a promising once-daily oral antiviral for chronic HCV infection of all genotypes and fibrosis stages with potentially short treatment durations.


Subject(s)
Guanosine Monophosphate/analogs & derivatives , Hepatitis C, Chronic , Hepatitis C , Phosphoramides , Humans , Hepacivirus , Hepatitis C, Chronic/drug therapy , Antiviral Agents/adverse effects , Sofosbuvir/pharmacology , Sofosbuvir/therapeutic use , Hepatitis C/drug therapy , Genotype , Drug Therapy, Combination , Viral Nonstructural Proteins
2.
ACS Nano ; 16(9): 15141-15154, 2022 09 27.
Article in English | MEDLINE | ID: mdl-35977379

ABSTRACT

Nanomedicine-based and unmodified drug interventions to address COVID-19 have evolved over the course of the pandemic as more information is gleaned and virus variants continue to emerge. For example, some early therapies (e.g., antibodies) have experienced markedly decreased efficacy. Due to a growing concern of future drug resistant variants, current drug development strategies are seeking to find effective drug combinations. In this study, we used IDentif.AI, an artificial intelligence-derived platform, to investigate the drug-drug and drug-dose interaction space of six promising experimental or currently deployed therapies at various concentrations: EIDD-1931, YH-53, nirmatrelvir, AT-511, favipiravir, and auranofin. The drugs were tested in vitro against a live B.1.1.529 (Omicron) virus first in monotherapy and then in 50 strategic combinations designed to interrogate the interaction space of 729 possible combinations. Key findings and interactions were then further explored and validated in an additional experimental round using an expanded concentration range. Overall, we found that few of the tested drugs showed moderate efficacy as monotherapies in the actionable concentration range, but combinatorial drug testing revealed significant dose-dependent drug-drug interactions, specifically between EIDD-1931 and YH-53, as well as nirmatrelvir and YH-53. Checkerboard validation analysis confirmed these synergistic interactions and also identified an interaction between EIDD-1931 and favipiravir in an expanded range. Based on the platform nature of IDentif.AI, these findings may support further explorations of the dose-dependent drug interactions between different drug classes in further pre-clinical and clinical trials as possible combinatorial therapies consisting of unmodified and nanomedicine-enabled drugs, to combat current and future COVID-19 strains and other emerging pathogens.


Subject(s)
COVID-19 Drug Treatment , SARS-CoV-2 , Amides , Artificial Intelligence , Auranofin , Guanosine Monophosphate/analogs & derivatives , Humans , Phosphoramides , Pyrazines
3.
Ned Tijdschr Geneeskd ; 1662022 07 21.
Article in Dutch | MEDLINE | ID: mdl-36036702

ABSTRACT

The anti-inflammatory agents dexamethasone (corticosteroid), and tocilizumab and sarilumab (IL6-inhibitors) are effective in the treatment of late COVID-19. Other anti-inflammatory agents, like anakinra (IL1-inhibitor), baricitinib and tofacitinib (JAK-inhibitors) and lenzilumab (GM-CSF-inhibitor) have also shown positive results in late COVID-19. For the treatment of early COVID-19, the inhalation corticosteroid budesonide is regarded as an off-label treatment option. Virus-inhibitors, like remdesivir, molnupiravir and nirmatrelvir/ritonavir decrease the risk of hospitalization and the development of severe COVID-19 by patients with early symptoms. Monoclonal antibodies have shown limited or no efficacy against the omicron-variant of SARS-CoV-2. Fluvoxamine, l-arginine, AT-527 and ensovibep are considered as potential promising new therapies for the treatment of early COVID-19.


Subject(s)
COVID-19 Drug Treatment , Adrenal Cortex Hormones , Anti-Inflammatory Agents/therapeutic use , Antiviral Agents/therapeutic use , Guanosine Monophosphate/analogs & derivatives , Humans , Phosphoramides , Recombinant Fusion Proteins , SARS-CoV-2
4.
Nat Commun ; 13(1): 621, 2022 02 02.
Article in English | MEDLINE | ID: mdl-35110538

ABSTRACT

The guanosine analog AT-527 represents a promising candidate against Severe Acute Respiratory Syndrome coronavirus type 2 (SARS-CoV-2). AT-527 recently entered phase III clinical trials for the treatment of COVID-19. Once in cells, AT-527 is converted into its triphosphate form, AT-9010, that presumably targets the viral RNA-dependent RNA polymerase (RdRp, nsp12), for incorporation into viral RNA. Here we report a 2.98 Å cryo-EM structure of the SARS-CoV-2 nsp12-nsp7-nsp82-RNA complex, showing AT-9010 bound at three sites of nsp12. In the RdRp active-site, one AT-9010 is incorporated at the 3' end of the RNA product strand. Its modified ribose group (2'-fluoro, 2'-methyl) prevents correct alignment of the incoming NTP, in this case a second AT-9010, causing immediate termination of RNA synthesis. The third AT-9010 is bound to the N-terminal domain of nsp12 - known as the NiRAN. In contrast to native NTPs, AT-9010 is in a flipped orientation in the active-site, with its guanine base unexpectedly occupying a previously unnoticed cavity. AT-9010 outcompetes all native nucleotides for NiRAN binding, inhibiting its nucleotidyltransferase activity. The dual mechanism of action of AT-527 at both RdRp and NiRAN active sites represents a promising research avenue against COVID-19.


Subject(s)
Antiviral Agents/chemistry , Antiviral Agents/pharmacology , Guanosine Monophosphate/analogs & derivatives , Phosphoramides/chemistry , Phosphoramides/pharmacology , RNA-Dependent RNA Polymerase/antagonists & inhibitors , SARS-CoV-2/enzymology , Viral Proteins/antagonists & inhibitors , Viral Proteins/metabolism , COVID-19/virology , Cryoelectron Microscopy , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Guanosine Monophosphate/chemistry , Guanosine Monophosphate/pharmacology , Humans , RNA-Dependent RNA Polymerase/chemistry , RNA-Dependent RNA Polymerase/genetics , RNA-Dependent RNA Polymerase/metabolism , SARS-CoV-2/chemistry , SARS-CoV-2/drug effects , SARS-CoV-2/genetics , Viral Proteins/genetics
5.
Photosynth Res ; 152(1): 13-22, 2022 Apr.
Article in English | MEDLINE | ID: mdl-34988868

ABSTRACT

Chlorophyll f is a new type of chlorophyll isolated from cyanobacteria. The absorption and fluorescence characteristics of chlorophyll f permit these oxygenic-photosynthetic organisms to thrive in environments where white light is scarce but far-red light is abundant. To explore the ligand properties of chlorophyll f and its energy transfer profiles we established two different in vitro reconstitution systems. The reconstituted peridinin-chlorophyll f protein complex (chlorophyll f-PCP) showed a stoichiometry ratio of 4:1 between peridinin and chlorophyll f, consistent with the peridinin:chlorophyll a ratio from native PCP complexes. Using emission wavelength at 712 nm, the excitation fluorescence featured a broad peak at 453 nm and a shoulder at 511 nm confirming energy transfer from peridinin to chlorophyll f. In addition, by using a synthetic peptide mimicking the first transmembrane helix of light-harvesting chlorophyll proteins of plants, we report that chlorophyll f, similarly to chlorophyll b, did not interact with the peptide contrarily to chlorophyll a, confirming the accessory role of chlorophyll f in photosystems. The binding of chlorophyll f, even in the presence of chlorophylls a and b, by PCP complexes shows the flexibility of chlorophyll-protein complexes and provides an opportunity for the introduction of new chlorophyll species to extend the photosynthetic spectral range.


Subject(s)
Dinoflagellida , Chlorophyll/analogs & derivatives , Chlorophyll/metabolism , Chlorophyll A/metabolism , Dinoflagellida/metabolism , Guanosine Monophosphate/analogs & derivatives , Phosphoramides
6.
Environ Sci Pollut Res Int ; 29(4): 5923-5930, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34435288

ABSTRACT

There exists a visible evidence of linkage between pollutant exposure and nutritional deficiency in many organisms. The aim of the present study was to analyze the proximate composition of juvenile African catfish (Clarias gariepinus) exposed to polydimethylsiloxane (antifoam) for 56 days using a renewal bioassay. The moisture, crude protein, ash, and fat content of the fish samples were analyzed using standard methods. Antifoam concentrations at 0.0mg/L, 63.96mg/L, 127.9mg/L, 255.82mg/L, and 511.64mg/L were used for the sublethal exposure after which the proximate composition was analyzed. The water quality variables including dissolved oxygen, conductivity, and total dissolved solids varied as the concentration increased. The moisture and lipid contents were highest at 511.64mg/L concentration while the control(0.0mg/L) had the highest percentage composition of ash, protein, carbohydrate, and fiber contents which were normal values. The differences in proximate values were slightly significant at P>0.05 among the different concentrations. The findings in this study may be an indication that antifoam can significantly affect some water quality variables and proximate composition, while also portraying the risk associated with the consumption of such exposed fish.


Subject(s)
Catfishes , Water Pollutants, Chemical , Animals , Dimethylpolysiloxanes , Guanosine Monophosphate/analogs & derivatives , Phosphoramides
7.
J Toxicol Sci ; 46(8): 359-369, 2021.
Article in English | MEDLINE | ID: mdl-34334557

ABSTRACT

Predicting drug-induced side effects in the cardiovascular system is very important because it can lead to the discontinuation of new drugs/candidates or the withdrawal of marketed drugs. Although chronic assessment of cardiac contractility is an important issue in safety pharmacology, an in vitro evaluation system has not been fully developed. We previously developed an imaging-based contractility assay system to detect acute cardiotoxicity using human iPS cell-derived cardiomyocytes (hiPSC-CMs). To extend the system to chronic toxicity assessment, we examined the effects of the anti-hepatitis C virus (HCV) drug candidate BMS-986094, a guanosine nucleotide analogue, which was withdrawn from phase 2 clinical trials because of unexpected contractility toxicities. Additionally, we examined sofosbuvir, another nucleotide analogue inhibitor of HCV that has been approved as an anti-HCV drug. Motion imaging analysis revealed the difference in cardiotoxicity between the cardiotoxic BMS-986094 and the less toxic sofosbuvir in hiPSC-CMs, with a minimum of 4 days of treatment. In addition, we found that BMS-986094-induced contractility impairment was mediated by a decrease in calcium transient. These data suggest that chronic treatment improves the predictive power for the cardiotoxicity of anti-HCV drugs. Thus, hiPSC-CMs can be a useful tool to assess drug-induced chronic cardiotoxicity in non-clinical settings.


Subject(s)
Antiviral Agents/toxicity , Cardiotoxicity/etiology , Guanosine Monophosphate/analogs & derivatives , Guanosine/analogs & derivatives , Guanosine/toxicity , Induced Pluripotent Stem Cells/cytology , Myocytes, Cardiac/drug effects , Antiviral Agents/adverse effects , Calcium/metabolism , Cells, Cultured , Chronic Disease , Guanosine/adverse effects , Guanosine Monophosphate/adverse effects , Guanosine Monophosphate/toxicity , Humans , Molecular Imaging , Myocardial Contraction/drug effects , Myocytes, Cardiac/metabolism , Safety
8.
Article in English | MEDLINE | ID: mdl-33558299

ABSTRACT

The impact of severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), the causative agent of COVID-19, is global and unprecedented. Although remdesivir has recently been approved by the FDA to treat SARS-CoV-2 infection, no oral antiviral is available for outpatient treatment. AT-527, an orally administered double prodrug of a guanosine nucleotide analog, was previously shown to be highly efficacious and well tolerated in hepatitis C virus (HCV)-infected subjects. Here, we report the potent in vitro activity of AT-511, the free base of AT-527, against several coronaviruses, including SARS-CoV-2. In normal human airway epithelial cells, the concentration of AT-511 required to inhibit replication of SARS-CoV-2 by 90% (EC90) was 0.47 µM, very similar to its EC90 against human coronavirus (HCoV)-229E, HCoV-OC43, and SARS-CoV in Huh-7 cells. Little to no cytotoxicity was observed for AT-511 at concentrations up to 100 µM. Substantial levels of the active triphosphate metabolite AT-9010 were formed in normal human bronchial and nasal epithelial cells incubated with 10 µM AT-511 (698 ± 15 and 236 ± 14 µM, respectively), with a half-life of at least 38 h. Results from steady-state pharmacokinetic and tissue distribution studies of nonhuman primates administered oral doses of AT-527, as well as pharmacokinetic data from subjects given daily oral doses of AT-527, predict that twice daily oral doses of 550 mg AT-527 will produce AT-9010 trough concentrations in human lung that exceed the EC90 observed for the prodrug against SARS-CoV-2 replication. This suggests that AT-527 may be an effective treatment option for COVID-19.


Subject(s)
Antiviral Agents/pharmacology , COVID-19 Drug Treatment , Guanosine Monophosphate/analogs & derivatives , Guanosine/pharmacology , Phosphoramides/pharmacology , Prodrugs/pharmacology , SARS-CoV-2/drug effects , Administration, Oral , Animals , COVID-19/virology , Cell Line , Cell Line, Tumor , Chlorocebus aethiops , Coronavirus 229E, Human/metabolism , Coronavirus OC43, Human/metabolism , Cricetinae , Epithelial Cells/virology , Guanosine Monophosphate/pharmacology , Humans , Lung/virology , SARS-CoV-2/metabolism , Vero Cells , Virus Replication/drug effects
9.
Life Sci ; 248: 117477, 2020 May 01.
Article in English | MEDLINE | ID: mdl-32119961

ABSTRACT

AIMS: A newly emerged Human Coronavirus (HCoV) is reported two months ago in Wuhan, China (COVID-19). Until today >2700 deaths from the 80,000 confirmed cases reported mainly in China and 40 other countries. Human to human transmission is confirmed for COVID-19 by China a month ago. Based on the World Health Organization (WHO) reports, SARS HCoV is responsible for >8000 cases with confirmed 774 deaths. Additionally, MERS HCoV is responsible for 858 deaths out of about 2500 reported cases. The current study aims to test anti-HCV drugs against COVID-19 RNA dependent RNA polymerase (RdRp). MATERIALS AND METHODS: In this study, sequence analysis, modeling, and docking are used to build a model for Wuhan COVID-19 RdRp. Additionally, the newly emerged Wuhan HCoV RdRp model is targeted by anti-polymerase drugs, including the approved drugs Sofosbuvir and Ribavirin. KEY FINDINGS: The results suggest the effectiveness of Sofosbuvir, IDX-184, Ribavirin, and Remidisvir as potent drugs against the newly emerged HCoV disease. SIGNIFICANCE: The present study presents a perfect model for COVID-19 RdRp enabling its testing in silico against anti-polymerase drugs. Besides, the study presents some drugs that previously proved its efficiency against the newly emerged viral infection.


Subject(s)
Adenosine Monophosphate/analogs & derivatives , Alanine/analogs & derivatives , Antiviral Agents/chemistry , Betacoronavirus/enzymology , Coronavirus Infections/drug therapy , Guanosine Monophosphate/analogs & derivatives , Pneumonia, Viral/drug therapy , RNA-Dependent RNA Polymerase/antagonists & inhibitors , Ribavirin/chemistry , Sofosbuvir/chemistry , Viral Proteins/antagonists & inhibitors , Adenosine Monophosphate/chemistry , Adenosine Monophosphate/metabolism , Alanine/chemistry , Alanine/metabolism , Alphacoronavirus/enzymology , Alphacoronavirus/genetics , Amino Acid Sequence , Antiviral Agents/metabolism , Betacoronavirus/genetics , COVID-19 , Catalytic Domain , Computational Biology/methods , Coronavirus Infections/virology , Drug Repositioning/methods , Guanosine Monophosphate/chemistry , Guanosine Monophosphate/metabolism , Guanosine Triphosphate/chemistry , Guanosine Triphosphate/metabolism , Humans , Molecular Docking Simulation , Pneumonia, Viral/virology , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , RNA-Dependent RNA Polymerase/chemistry , RNA-Dependent RNA Polymerase/metabolism , Ribavirin/metabolism , SARS-CoV-2 , Sequence Alignment , Sequence Homology, Amino Acid , Sofosbuvir/metabolism , Thermodynamics , Uridine Triphosphate/chemistry , Uridine Triphosphate/metabolism , Viral Proteins/chemistry , Viral Proteins/metabolism , COVID-19 Drug Treatment
10.
Biochem Pharmacol ; 163: 250-259, 2019 05.
Article in English | MEDLINE | ID: mdl-30772266

ABSTRACT

Phosphorothioate oligonucleotides (PS-oligos) containing sulfur atom attached in a nonbridging position to the phosphorus atom at one or more internucleotide bond(s) are often used in medicinal applications. Their hydrolysis in cellular media proceeds mainly from the 3'-end, resulting in the appearance of nucleoside 5'-O-phosphorothioates ((d)NMPS), whose further metabolism is poorly understood. We hypothesize that the enzyme responsible for (d)NMPS catabolism could be Hint1, an enzyme that belongs to the histidine triad (HIT) superfamily and is present in all organisms. We previously found that (d)NMPS were desulfurated in vitro to yield (d)NMP and H2S in a Hint1-assisted reaction. Here, we demonstrate that AMPS/GMPS/dGMPS introduced into HeLa/A549 cells are intracellularly converted into AMP/GMP/dGMP and H2S. The level of the released H2S was relative to the concentration of the compounds used and the reaction time. Using RNAi technology, we have shown decreased levels of AMPS/GMPS desulfuration in HeLa/A549 cells with reduced Hint1 levels. Finally, after transfection of a short Rp-d(APSAPSA) oligomer into HeLa cells, the release of H2S was observed. These results suggest that the metabolic pathway of PS-oligos includes hydrolysis into (d)NMPS (by cellular nucleases) followed by Hint1-promoted conversion of the resulting (d)NMPS into (d)NMP accompanied by H2S elimination. Our observations may be also important for possible medicinal applications of (d)NMPS because H2S is a gasotransmitter involved in many physiological and pathological processes.


Subject(s)
Hydrogen Sulfide/metabolism , Nerve Tissue Proteins/metabolism , Phosphorothioate Oligonucleotides/metabolism , A549 Cells , Adenosine Monophosphate/metabolism , Guanosine Monophosphate/analogs & derivatives , Guanosine Monophosphate/metabolism , HeLa Cells , Humans , Lysine/analogs & derivatives , Lysine/metabolism , Nerve Tissue Proteins/genetics , RNA Interference
11.
J Gastroenterol ; 54(5): 449-458, 2019 May.
Article in English | MEDLINE | ID: mdl-30684016

ABSTRACT

BACKGROUND: The emergence of a deletion mutant at hepatitis C virus (HCV) NS5A-P32 (P32del) has recently been reported in a subset of chronic hepatitis C patients who experience virologic failure after direct-acting antiviral drug (DAA) treatment. This mutation confers extremely high resistance to NS5A inhibitors. No effective treatment has been established for cases with this mutation. METHODS: We used a JFH1-based recombinant virus with NS5A from a genotype 1b strain to introduce a P32del mutation. We inoculated human hepatocyte chimeric mice with sera from a patient with ledipasvir/sofosbuvir therapy failure carrying a genotype 1b HCV with NS5A L31M and P32del or from a DAA-naïve patient carrying wild-type virus. RESULTS: JFH1-based chimeric viruses with P32del showed sufficient levels of replication for in vitro assay despite the suppression of viral growth and infectious virus production. Variants with P32del exhibited severe resistance to all tested NS5A inhibitors, including daclatasvir, ledipasvir, elbasvir and velpatasvir, but were as susceptible to NS3/4A inhibitors, NS5B inhibitors, interferon alfa-2b, and ribavirin as wild-type viruses in the in vitro assay. The P32del mutant virus caused persistent infection in all inoculated chimeric mice with high viral titer and frequency. The virus was resistant to the ledipasvir/GS-558093 (a nucleotide analog inhibitor of NS5B polymerase) regimen but susceptible to either simeprevir plus GS-558093 or peg-interferon alfa-2b, compared to the wild-type virus. CONCLUSION: Therapies combining at least two drugs among NS3/4A inhibitors, NS5B inhibitors and non-selective antiviral agents may be effective for HCV-infected patients with NS5A-P32del.


Subject(s)
Antiviral Agents/therapeutic use , Hepacivirus/genetics , Hepatitis C/drug therapy , Viral Nonstructural Proteins/genetics , Aged , Animals , Antiviral Agents/pharmacology , Benzimidazoles/pharmacology , Benzimidazoles/therapeutic use , Benzofurans/pharmacology , Carbamates/pharmacology , Cell Line , Chimera , Drug Resistance, Viral/genetics , Drug Therapy, Combination , Fluorenes/pharmacology , Fluorenes/therapeutic use , Guanosine Monophosphate/analogs & derivatives , Guanosine Monophosphate/pharmacology , Guanosine Monophosphate/therapeutic use , Hepacivirus/drug effects , Hepatocytes , Heterocyclic Compounds, 4 or More Rings/pharmacology , Humans , Imidazoles/pharmacology , Interferon alpha-2/pharmacology , Interferon alpha-2/therapeutic use , Interferon-alpha/pharmacology , Interferon-alpha/therapeutic use , Male , Mice , Middle Aged , Polyethylene Glycols/pharmacology , Polyethylene Glycols/therapeutic use , Pyrrolidines , Recombinant Proteins/pharmacology , Recombinant Proteins/therapeutic use , Ribavirin/pharmacology , Sequence Deletion , Serine Proteases , Simeprevir/pharmacology , Simeprevir/therapeutic use , Valine/analogs & derivatives , Viral Nonstructural Proteins/antagonists & inhibitors
12.
Nat Commun ; 9(1): 4213, 2018 10 11.
Article in English | MEDLINE | ID: mdl-30310068

ABSTRACT

Incorporation of mismatched nucleotides during DNA replication or repair leads to transition or transversion mutations and is considered as a predominant source of base substitution mutagenesis in cancer cells. Watson-Crick like dG:dT base pairing is considered to be an important source of genome instability. Here we show that DNA polymerase (pol) µ insertion of 7,8-dihydro-8'-oxo-dGTP (8-oxodGTP) or deoxyguanosine triphosphate (dGTP) into a model double-strand break DNA repair substrate with template base T results in efficient ligation by DNA ligase. These results indicate that pol µ-mediated dGTP mismatch insertion opposite template base T coupled with ligation could be a feature of mutation prone nonhomologous end joining during double-strand break repair.


Subject(s)
DNA Repair , DNA-Directed DNA Polymerase/metabolism , Deoxyguanine Nucleotides/metabolism , Mutagenesis/genetics , Thymine/metabolism , Base Pair Mismatch , DNA/metabolism , Guanosine Monophosphate/analogs & derivatives , Guanosine Monophosphate/metabolism , Humans
13.
Environ Sci Pollut Res Int ; 25(22): 21879-21886, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29796890

ABSTRACT

To understand whether flavor enhancers pose potential risks to the environment, it is important to assess its effects on insects. Therefore, the objective of this study was to evaluate the toxicity of flavor enhancers on the survival and behaviors of the red imported fire ant, Solenopsis invicta. In this study, we found that the mortality of S. invicta workers that were fed glutamic acid monosodium salt hydrate, glycine, L-alanine, succinic acid, succinic acid disodium, inosinate 5'-monophosphate disodium salt hydrate, and GMP were significantly higher than the mortality rates of workers fed sucrose. Moreover, glycine and GMP exhibited the strongest toxicities and caused 100% mortality in workers after 84 h. LC50 values were 0.004 g/ml and 0.02 g/ml for GMP and glycine, respectively. Additionally, at sublethal doses, both GMP and glycine solutions decreased foraging and digging behaviors. Our results suggest that flavor enhancers are toxic to insects and also likely to have a negative impact at sublethal concentrations.


Subject(s)
Ants/drug effects , Ants/physiology , Flavoring Agents/toxicity , Animals , Behavior, Animal/drug effects , Ecotoxicology/methods , Flavoring Agents/administration & dosage , Glycine/administration & dosage , Glycine/toxicity , Guanosine Monophosphate/administration & dosage , Guanosine Monophosphate/analogs & derivatives , Guanosine Monophosphate/toxicity , Insecticides/toxicity , Larva/drug effects , Lethal Dose 50 , Lysine/administration & dosage , Lysine/analogs & derivatives , Lysine/toxicity , Mortality
14.
J Biol Inorg Chem ; 22(8): 1211-1222, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29038915

ABSTRACT

Extensive research has linked the amyloid-beta (Aß) peptide to neurological dysfunction in Alzheimer's disease (AD). Insoluble Aß plaques in the AD patient brain contain high concentrations of advanced glycation end-products (AGEs) as well as transition metal ions. This research elucidated the roles of Aß, sugars, and Cu2+ in the oxidative stress mechanism of AD at the molecular level. Mass spectral (MS) analysis of the reactions of Aß with two representative sugars, ribose-5-phosphate (R5P) and methylglyoxal (MG), revealed Lys-16 and Arg-5 as the primary glycation sites. Quantitative analysis of superoxide [Formula: see text] production by a cyt c assay showed that Lys-16 generated four times as much [Formula: see text] as Arg-5. Lys-16 and Arg-5 in Aß1-40 are both adjacent to histidine residues, which are suggested to catalyze glycation. Additionally, Lys-16 is close to the central hydrophobic core (Leu-17-Ala-21) and to His-13, both of which are known to lower the pKa of the residue, leading to increased deprotonation of the amine and an enhanced glycation reactivity compared to Arg-5. Gel electrophoresis results indicated that all three components of AD plaques-Aß1-40, sugars, and Cu2+-are necessary for DNA damage. It is concluded that the glycation of Aß1-40 with sugars generates significant amounts of [Formula: see text], owing to the rapid glycation of Lys-16 and Arg-5. In the presence of Cu2+, [Formula: see text] converts to hydroxyl radical (HO·), the source of oxidative stress in AD.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/metabolism , Arginine/metabolism , Copper/pharmacology , Lysine/metabolism , Oxidative Stress/drug effects , Amino Acid Sequence , Cytochromes c/metabolism , DNA Damage , Deoxyguanine Nucleotides/metabolism , Glycosylation/drug effects , Guanosine Monophosphate/analogs & derivatives , Guanosine Monophosphate/metabolism , Models, Molecular , Oxidation-Reduction/drug effects , Protein Conformation
15.
Antiviral Res ; 143: 151-161, 2017 07.
Article in English | MEDLINE | ID: mdl-28412183

ABSTRACT

Recent cases of severe toxicity during clinical trials have been associated with antiviral ribonucleoside analogs (e.g. INX-08189 and balapiravir). Some have hypothesized that the active metabolites of toxic ribonucleoside analogs, the triphosphate forms, inadvertently target human mitochondrial RNA polymerase (POLRMT), thus inhibiting mitochondrial RNA transcription and protein synthesis. Others have proposed that the prodrug moiety released from the ribonucleoside analogs might instead cause toxicity. Here, we report the mitochondrial effects of several clinically relevant and structurally diverse ribonucleoside analogs including NITD-008, T-705 (favipiravir), R1479 (parent nucleoside of balapiravir), PSI-7851 (sofosbuvir), and INX-08189 (BMS-986094). We found that efficient substrates and chain terminators of POLRMT, such as the nucleoside triphosphate forms of R1479, NITD-008, and INX-08189, are likely to cause mitochondrial toxicity in cells, while weaker chain terminators and inhibitors of POLRMT such as T-705 ribonucleoside triphosphate do not elicit strong in vitro mitochondrial effects. Within a fixed 3'-deoxy or 2'-C-methyl ribose scaffold, changing the base moiety of nucleotides did not strongly affect their inhibition constant (Ki) against POLRMT. By swapping the nucleoside and prodrug moieties of PSI-7851 and INX-08189, we demonstrated that the cell-based toxicity of INX-08189 is mainly caused by the nucleoside component of the molecule. Taken together, these results show that diverse 2' or 4' mono-substituted ribonucleoside scaffolds cause mitochondrial toxicity. Given the unpredictable structure-activity relationship of this ribonucleoside liability, we propose a rapid and systematic in vitro screen combining cell-based and biochemical assays to identify the early potential for mitochondrial toxicity.


Subject(s)
Antiviral Agents/toxicity , Mitochondria/drug effects , Ribonucleosides/chemistry , Ribonucleosides/toxicity , Adenosine/analogs & derivatives , Amides/toxicity , Cell Line/drug effects , Cytidine/analogs & derivatives , Cytidine/toxicity , DNA-Directed RNA Polymerases/drug effects , Guanosine Monophosphate/analogs & derivatives , Guanosine Monophosphate/toxicity , Humans , Inhibitory Concentration 50 , Mitochondrial Proteins/metabolism , Nucleosides/toxicity , Prodrugs/pharmacology , Protein Biosynthesis/drug effects , Pyrazines/toxicity , RNA/metabolism , RNA, Mitochondrial , Sofosbuvir/toxicity , Structure-Activity Relationship , Transcription Initiation Site/drug effects , Transcription, Genetic/drug effects
16.
Toxicol Sci ; 155(2): 348-362, 2017 02.
Article in English | MEDLINE | ID: mdl-27864544

ABSTRACT

BMS-986094, a 2'-C-methylguanosine prodrug that was in development for treatment of chronic hepatitis C infection was withdrawn from Phase 2 clinical trials because of unexpected cardiac and renal adverse events. Investigative nonclinical studies were conducted to extend the understanding of these findings using more comprehensive endpoints. BMS-986094 was given orally to female CD-1 mice (25 and 150 mg/kg/d) for 2 weeks (53/group) and to cynomolgus monkeys (15 and 30 mg/kg/d) for up to 6 weeks (2-3/sex/group for cardiovascular safety, and 5/sex/group for toxicology). Endpoints included toxicokinetics; echocardiography, telemetric hemodynamics and electrocardiography, and tissue injury biomarkers (monkey); and light and ultrastructural pathology of heart, kidney, and skeletal muscle (mouse/monkey). Dose-related and time-dependent findings included: severe toxicity in mice at 150 mg/kg/d and monkeys at 30 mg/kg/d; decreased left ventricular (LV) ejection fraction, fractional shortening, stroke volume, and dP/dt; LV dilatation, increased QTc interval, and T-wave flattening/inversion (monkeys at ≥ 15 mg/kg/d); cardiomyocyte degeneration (mice at 150 mg/kg/d and monkeys at ≥ 15 mg/kg/d) with myofilament lysis/myofbril disassembly; time-dependent proteinuria and increased urine ß-2 microglobulin, calbindin, clusterin; kidney pallor macroscopically; and tubular dilatation (monkeys); tubular regeneration (mice 150 mg/kg/d); and acute proximal tubule degeneration ultrastructurally (mice/monkeys); and skeletal muscle degeneration with increased urine myoglobin and serum sTnI. These studies identified changes not described previously in studies of BMS-986094 including premonitory cardiovascular functional changes as well as additional biomarkers for muscle and renal toxicities. Although the mechanism of potential toxicities observed in BMS-986094 studies was not established, there was no evidence for direct mitochondrial toxicity.


Subject(s)
Guanosine Monophosphate/analogs & derivatives , Heart/drug effects , RNA-Dependent RNA Polymerase/antagonists & inhibitors , Viral Nonstructural Proteins/antagonists & inhibitors , Animals , Female , Guanosine Monophosphate/therapeutic use , Guanosine Monophosphate/toxicity , Heart/physiology , Hepatitis C, Chronic/drug therapy , Kidney/drug effects , Macaca fascicularis , Male , Mice , Muscle, Skeletal/drug effects , Toxicokinetics
18.
J Med Virol ; 88(12): 2044-2051, 2016 12.
Article in English | MEDLINE | ID: mdl-27604059

ABSTRACT

In the last few months, a new Zika virus (ZIKV) outbreak evolved in America. In accordance, World Health Organization (WHO) in February 2016 declared it as Public Health Emergency of International Concern (PHEIC). ZIKV infection was reported in more than 60 countries and the disease was spreading since 2007 but with little momentum. Many antiviral drugs are available in market or in laboratories under clinical trials, could affect ZIKV infection. In silico docking study were performed on the ZIKV polymerase to test some of Hepatitis C Virus (HCV) drugs (approved and in clinical trials). The results show potency of almost all of the studied compounds on ZIKV polymerase and hence inhibiting the propagation of the disease. In addition, the study suggested two nucleotide inhibitors (IDX-184 and MK0608) that may be tested as drugs against ZIKV infection. J. Med. Virol. 88:2044-2051, 2016. © 2016 Wiley Periodicals, Inc.


Subject(s)
Antiviral Agents/pharmacology , RNA-Dependent RNA Polymerase/antagonists & inhibitors , Zika Virus Infection/drug therapy , Zika Virus/drug effects , Zika Virus/enzymology , Clinical Trials as Topic , Computer Simulation , Drug Discovery , Enzyme Inhibitors/pharmacology , Guanosine Monophosphate/analogs & derivatives , Guanosine Monophosphate/pharmacology , Guanosine Monophosphate/therapeutic use , Hepacivirus/drug effects , Hepatitis C/drug therapy , Hepatitis C/virology , Humans , Molecular Docking Simulation , Tubercidin/analogs & derivatives , Tubercidin/pharmacology , Tubercidin/therapeutic use , Zika Virus Infection/virology
19.
Toxicol Sci ; 153(2): 396-408, 2016 10.
Article in English | MEDLINE | ID: mdl-27466212

ABSTRACT

BMS-986094, the prodrug of a guanosine nucleotide analogue (2'-C-methylguanosine), was withdrawn from clinical trials due to serious safety issues. Nonclinical investigative studies were conducted as a follow up to evaluate the potential for BMS-986094-related mitochondrial-toxicity. In vitro, BMS-986094 was applied to human hepatoma cells (HepG2 and Huh-7) or cardiomyocytes (hiPSCM) up to 19 days to assess mitochondrial DNA content and specific gene expression. There were no mitochondrial DNA changes at concentrations ≤10 µM. Transcriptional effects, such as reductions in Huh-7 MT-ND1 and MT-ND5 mRNA content and hiPSCM MT-ND1, MT-COXII, and POLRMT protein expression levels, occurred only at cytotoxic concentrations (≥10 µM) suggesting these transcriptional effects were a consequence of the observed toxicity. Additionally, BMS-986094 has a selective weak affinity for inhibition of RNA polymerases as opposed to DNA polymerases. In vivo, BMS-986094 was given orally to cynomolgus monkeys for 3 weeks or 1 month at doses of 15 or 30 mg/kg/day. Samples of heart and kidney were collected for assessment of mitochondrial respiration, mitochondrial DNA content, and levels of high energy substrates. Although pronounced cardiac and renal toxicities were observed in some monkeys at 30 mg/kg/day treated for 3-4 weeks, there were no changes in mitochondrial DNA content or ATP/GTP levels. Collectively, these data suggest that BMS-986094 is not a direct mitochondrial toxicant.


Subject(s)
DNA, Mitochondrial/drug effects , Guanosine Monophosphate/analogs & derivatives , Adenosine Triphosphate/metabolism , Animals , Cell Line , DNA, Mitochondrial/biosynthesis , DNA, Mitochondrial/physiology , Dose-Response Relationship, Drug , Female , Guanosine Monophosphate/metabolism , Guanosine Monophosphate/toxicity , Guanosine Triphosphate/metabolism , Heart/drug effects , Heart Function Tests , Humans , Inosine Monophosphate/metabolism , Kidney/drug effects , Kidney/metabolism , Kidney Function Tests , Macaca fascicularis , Male
20.
DNA Res ; 23(4): 395-402, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27260513

ABSTRACT

The next-generation sequencing studies of breast cancer have reported that the tumour suppressor P53 (TP53) gene is mutated in more than 40% of the tumours. We studied the levels of oxidative lesions, including 8-oxo-7,8-dihydro-2'-deoxyguanosine (8-oxodG), along the coding strand of the exon 5 in breast cancer patients as well as in a reactive oxygen species (ROS)-attacked breast cancer cell line using the ligation-mediated polymerase chain reaction technique. We detected a significant 'in vitro' generation of 8-oxodG between the codons 163 and 175, corresponding to a TP53 region with high mutation prevalence, after treatment with xanthine plus xanthine oxidase, a ROS-generating system. Then, we evaluated the occurrence of oxidative lesions in the DNA-binding domain of the TP53 in the core needle biopsies of 113 of women undergoing breast investigation for diagnostic purpose. An increment of oxidative damage at the -G- residues into the codons 163 and 175 was found in the cancer cases as compared to the controls. We found significant associations with the pathological stage and the histological grade of tumours. As the major news of this study, this largest analysis of genomic footprinting of oxidative lesions at the TP53 sequence level to date provided a first roadmap describing the signatures of oxidative lesions in human breast cancer. Our results provide evidence that the generation of oxidative lesions at single nucleotide resolution is not an event highly stochastic, but causes a characteristic pattern of DNA lesions at the site of mutations in the TP53, suggesting causal relationship between oxidative DNA adducts and breast cancer.


Subject(s)
Breast Neoplasms/genetics , DNA, Neoplasm/chemistry , Exons , Guanosine Monophosphate/analogs & derivatives , Tumor Suppressor Protein p53/genetics , Adult , Aged , Breast Neoplasms/pathology , Case-Control Studies , Cell Line, Tumor , Codon , DNA Adducts/chemistry , DNA Adducts/genetics , DNA, Neoplasm/genetics , Female , Guanosine Monophosphate/chemistry , Guanosine Monophosphate/genetics , Humans , Middle Aged , Oxidative Stress , Point Mutation
SELECTION OF CITATIONS
SEARCH DETAIL
...