Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.344
Filter
1.
Antiviral Res ; 209: 105453, 2023 01.
Article in English | MEDLINE | ID: mdl-36379378

ABSTRACT

The unprecedented magnitude of the 2013-2016 Ebola virus (EBOV) epidemic in West Africa resulted in over 11 000 deaths and spurred an international public health emergency. A second outbreak in 2018-2020 in DRC resulted in an additional >3400 cases and nearly 2300 deaths (WHO, 2020). These large outbreaks across geographically diverse regions highlight the need for the development of effective oral therapeutic agents that can be easily distributed for self-administration to populations with active disease or at risk of infection. Herein, we report the in vivo efficacy of N4-hydroxycytidine (EIDD-1931), a broadly active ribonucleoside analog and the active metabolite of the prodrug EIDD-2801 (molnupiravir), in murine models of lethal EBOV infection. Twice daily oral dosing with EIDD-1931 at 200 mg/kg for 7 days, initiated either with a prophylactic dose 2 h before infection, or as therapeutic treatment starting 6 h post-infection, resulted in 92-100% survival of mice challenged with lethal doses of EBOV, reduced clinical signs of Ebola virus disease (EVD), reduced serum virus titers, and facilitated weight loss recovery. These results support further investigation of molnupiravir as a potential therapeutic or prophylactic treatment for EVD.


Subject(s)
Ebolavirus , Hemorrhagic Fever, Ebola , Ribonucleosides , Animals , Mice , Hemorrhagic Fever, Ebola/drug therapy , Hemorrhagic Fever, Ebola/prevention & control , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Ribonucleosides/pharmacology
2.
Int J Mol Sci ; 23(19)2022 Sep 20.
Article in English | MEDLINE | ID: mdl-36232311

ABSTRACT

The novel coronavirus, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), was identified as the pathogenic cause of coronavirus disease 2019 (COVID-19). The RNA-dependent RNA polymerase (RdRp) of SARS-CoV-2 is a potential target for the treatment of COVID-19. An RdRp complex:dsRNA structure suitable for docking simulations was prepared using a cryo-electron microscopy (cryo-EM) structure (PDB ID: 7AAP; resolution, 2.60 Å) that was reported recently. Structural refinement was performed using energy calculations. Structure-based virtual screening was performed using the ChEMBL database. Through 1,838,257 screenings, 249 drugs (37 approved, 93 clinical, and 119 preclinical drugs) were predicted to exhibit a high binding affinity for the RdRp complex:dsRNA. Nine nucleoside triphosphate analogs with anti-viral activity were included among these hit drugs, and among them, remdesivir-ribonucleoside triphosphate and favipiravir-ribonucleoside triphosphate adopted a similar docking mode as that observed in the cryo-EM structure. Additional docking simulations for the predicted compounds with high binding affinity for the RdRp complex:dsRNA suggested that 184 bioactive compounds could be anti-SARS-CoV-2 drug candidates. The hit bioactive compounds mainly consisted of a typical noncovalent major groove binder for dsRNA. Three-layer ONIOM (MP2/6-31G:AM1:AMBER) geometry optimization calculations and frequency analyses (MP2/6-31G:AMBER) were performed to estimate the binding free energy of a representative bioactive compound obtained from the docking simulation, and the fragment molecular orbital calculation at the MP2/6-31G level of theory was subsequently performed for analyzing the detailed interactions. The procedure used in this study represents a possible strategy for discovering anti-SARS-CoV-2 drugs from drug libraries that could significantly shorten the clinical development period for drug repositioning.


Subject(s)
COVID-19 Drug Treatment , Ribonucleosides , Adenosine Monophosphate/analogs & derivatives , Alanine/analogs & derivatives , Amides , Antiviral Agents/chemistry , Cryoelectron Microscopy , Humans , Molecular Docking Simulation , Nucleosides , Polyphosphates , Pyrazines , RNA, Viral , RNA-Dependent RNA Polymerase , Reproduction , Ribonucleosides/pharmacology , SARS-CoV-2
3.
Int J Mol Sci ; 23(10)2022 May 21.
Article in English | MEDLINE | ID: mdl-35628582

ABSTRACT

The accumulation of specific metabolic intermediates is known to promote cancer progression. We analyzed the role of 4-pyridone-3-carboxamide-1-ß-D-ribonucleoside (4PYR), a nucleotide metabolite that accumulates in the blood of cancer patients, using the 4T1 murine in vivo breast cancer model, and cultured cancer (4T1) and endothelial cells (ECs) for in vitro studies. In vivo studies demonstrated that 4PYR facilitated lung metastasis without affecting primary tumor growth. In vitro studies demonstrated that 4PYR affected extracellular adenine nucleotide metabolism and the intracellular energy status in ECs, shifting catabolite patterns toward the accumulation of extracellular inosine, and leading to the increased permeability of lung ECs. These changes prevailed over the direct effect of 4PYR on 4T1 cells that reduced their invasive potential through 4PYR-induced modulation of the CD73-adenosine axis. We conclude that 4PYR is an oncometabolite that affects later stages of the metastatic cascade by acting specifically through the regulation of EC permeability and metabolic controls of inflammation.


Subject(s)
Breast Neoplasms , Ribonucleosides , Animals , Breast Neoplasms/metabolism , Endothelial Cells/metabolism , Female , Humans , Mice , Nucleosides/metabolism , Nucleotides/metabolism , Pyridones , Ribonucleosides/pharmacology
4.
Eur J Med Chem ; 237: 114367, 2022 Jul 05.
Article in English | MEDLINE | ID: mdl-35533570

ABSTRACT

Leishmaniasis causes high mortality and morbidity in tropical and subtropical regions of Africa, Asia, the Americas and southern Europe, and is characterized by diverse clinical manifestations. As a neglected tropical disease, limited resources are allocated for antileishmanial drug discovery. The Leishmania parasite is deficient in de novo purine synthesis, and therefore acquires purines from the host and processes these using a purine salvage pathway. By making use of purine transport systems and interfering with this salvage pathway, purine (nucleoside) analogues might exert a selective detrimental impact on its growth and survival. In vitro screening of an in-house purine nucleoside library and analogue synthesis afforded the 6-methyl-7-(2-pyridyl)-7-deazapurine ribonucleoside analogue 18 as a promising hit. Optimization of the 7-substituent afforded 31 and 32 which displayed potent activity against wild-type and resistant L. infantum, intracellular amastigote and extracellular promastigote forms, and favorable selectivity versus primary mouse macrophages (Mφ) and MRC-5 cells. Encouraged by the favorable in vitro metabolic stability of 32, an in vivo study was performed using an early curative L. infantum hamster model. When orally administrated at 50 mg/kg once daily (s.i.d) for 10 days, 32 was devoid of side effects, however, it only poorly reduced amastigote burdens in the major target organs.


Subject(s)
Antiprotozoal Agents , Leishmania , Leishmaniasis , Purines , Ribonucleosides , Animals , Antiprotozoal Agents/pharmacology , Antiprotozoal Agents/therapeutic use , Cricetinae , Leishmania/drug effects , Leishmania/metabolism , Leishmaniasis/drug therapy , Mice , Nucleosides/pharmacology , Nucleosides/therapeutic use , Purine Nucleosides/pharmacology , Purine Nucleosides/therapeutic use , Purines/pharmacology , Purines/therapeutic use , Ribonucleosides/pharmacology , Ribonucleosides/therapeutic use
5.
Toxicol In Vitro ; 82: 105355, 2022 Aug.
Article in English | MEDLINE | ID: mdl-35390475

ABSTRACT

One of the promising approaches in the development of nucleoside prodrugs is to use the nucleoside analogs containing lipophilic biodegradable residues, which are cleaved to biologically active forms after metabolic transformations in the cell. The introduction of such fragments makes it possible to reduce the general toxicity of the drug candidate and increase its stability in the cell. In order to study the influence of biodegradable lipophilic groups on antiviral activity and cytotoxicity, in this work we synthesized N6-benzyl-2',3',5'-tri-O-nicotinoyl adenosine and N6-(3-fluorobenzyl)-2',3',5'-tri-O-nicotinoyl adenosine, derivatives of N6-benzyladenosine (BAR) and N6-(3-fluorobenzyl)adenosine (FBAR), which had previously shown prominent antiviral activity against human enterovirus EV-A71 but appeared to be cytotoxic. The obtained fully-O-nicotinoylated BAR and FBAR inhibited reproduction of EV-A71 strains BrCr and 46973 and manifested significantly lower cytotoxicity compared to non-protected compounds. In addition, we performed enzymatic hydrolysis of the fully-O-nicotinoylated FBAR in the presence of esterases (CalB and PLE) to investigate metabolic degradation of O-nicotinoylated compounds in cells. Both enzymes hydrolyzed the tested substrate to form the corresponding O-deprotected nucleoside that may suggest the role of hydrolase-type enzymes as general participants of metabolic activation of O-nicotinoylated prodrugs in different cells.


Subject(s)
Enterovirus A, Human , Prodrugs , Ribonucleosides , Adenosine/pharmacology , Antiviral Agents/toxicity , Benzyl Compounds , Enterovirus A, Human/physiology , Humans , Nucleosides , Prodrugs/pharmacology , Purines , Ribonucleosides/pharmacology
6.
Drugs ; 82(3): 335-340, 2022 Feb.
Article in English | MEDLINE | ID: mdl-35147913

ABSTRACT

Maribavir (LIVTENCITYTM), a cytomegalovirus (CMV) enzyme pUL97 kinase inhibitor, is being developed by Takeda Pharmaceuticals for the treatment of CMV infections. Maribavir was recently approved in the USA for the treatment of post-transplant CMV infection/disease that is refractory to treatment (with or without genotypic resistance) with ganciclovir, valganciclovir, cidofovir or foscarnet in adults and paediatric (≥ 12 years of age and weighing ≥ 35 kg) patients. This article summarizes the milestones in the development of maribavir leading to this first approval for CMV infections.


Subject(s)
Cytomegalovirus Infections , Ribonucleosides , Adult , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Benzimidazoles/pharmacology , Benzimidazoles/therapeutic use , Child , Cytomegalovirus Infections/drug therapy , Drug Resistance, Viral , Humans , Ribonucleosides/pharmacology , Ribonucleosides/therapeutic use
7.
Lipids Health Dis ; 20(1): 151, 2021 Nov 02.
Article in English | MEDLINE | ID: mdl-34727932

ABSTRACT

BACKGROUND: Acylcarnitine is an intermediate product of fatty acid oxidation. It is reported to be closely associated with the occurrence of diabetic cardiomyopathy (DCM). However, the mechanism of acylcarnitine affecting myocardial disorders is yet to be explored. This current research explores the different chain lengths of acylcarnitines as biomarkers for the early diagnosis of DCM and the mechanism of acylcarnitines for the development of DCM in-vitro. METHODS: In a retrospective non-interventional study, 50 simple type 2 diabetes mellitus patients and 50 DCM patients were recruited. Plasma samples from both groups were analyzed by high throughput metabolomics and cluster heat map using mass spectrometry. Principal component analysis was used to compare the changes occurring in the studied 25 acylcarnitines. Multivariable binary logistic regression was used to analyze the odds ratio of each group for factors and the 95% confidence interval in DCM. Myristoylcarnitine (C14) exogenous intervention was given to H9c2 cells to verify the expression of lipid metabolism-related protein, inflammation-related protein expression, apoptosis-related protein expression, and cardiomyocyte hypertrophy and fibrosis-related protein expression. RESULTS: Factor 1 (C14, lauroylcarnitine, tetradecanoyldiacylcarnitine, 3-hydroxyl-tetradecanoylcarnitine, arachidic carnitine, octadecanoylcarnitine, 3-hydroxypalmitoleylcarnitine) and factor 4 (octanoylcarnitine, hexanoylcarnitine, decanoylcarnitine) were positively correlated with the risk of DCM. Exogenous C14 supplementation to cardiomyocytes led to increased lipid deposition in cardiomyocytes along with the obstacles in adenosine 5'-monophosphate (AMP)-activated protein kinase (AMPK) signaling pathways and affecting fatty acid oxidation. This further caused myocardial lipotoxicity, ultimately leading to cardiomyocyte hypertrophy, fibrotic remodeling, and increased apoptosis. However, this effect was mitigated by the AMPK agonist acadesine. CONCLUSIONS: The increased plasma levels in medium and long-chain acylcarnitine extracted from factors 1 and 4 are closely related to the risk of DCM, indicating that these factors can be an important tool for DCM risk assessment. C14 supplementation associated lipid accumulation by inhibiting the AMPK/ACC/CPT1 signaling pathway, aggravated myocardial lipotoxicity, increased apoptosis apart from cardiomyocyte hypertrophy and fibrosis were alleviated by the acadesine.


Subject(s)
Carnitine/analogs & derivatives , Diabetes Mellitus, Type 2/complications , Diabetic Cardiomyopathies/metabolism , Lipid Metabolism , Adult , Aminoimidazole Carboxamide/analogs & derivatives , Aminoimidazole Carboxamide/pharmacology , Animals , Biomarkers/blood , Carnitine/blood , Carnitine/chemistry , Carnitine/pharmacology , Cell Line , Diabetes Mellitus, Type 2/metabolism , Female , Humans , Lipid Metabolism/drug effects , Male , Mass Spectrometry , Middle Aged , Myoblasts, Cardiac/drug effects , Myoblasts, Cardiac/metabolism , Myristic Acids/pharmacology , Rats , Retrospective Studies , Ribonucleosides/pharmacology , Risk Factors
8.
Life Sci Alliance ; 4(12)2021 12.
Article in English | MEDLINE | ID: mdl-34610973

ABSTRACT

DNA damage is a double-edged sword for cancer cells. On the one hand, DNA damage-induced genomic instability contributes to cancer development; on the other hand, accumulating damage compromises proliferation and survival of cancer cells. Understanding the key regulators of DNA damage repair machinery would benefit the development of cancer therapies that induce DNA damage and apoptosis. In this study, we found that isoprenylcysteine carboxylmethyltransferase (ICMT), a posttranslational modification enzyme, plays an important role in DNA damage repair. We found that ICMT suppression consistently reduces the activity of MAPK signaling, which compromises the expression of key proteins in the DNA damage repair machinery. The ensuing accumulation of DNA damage leads to cell cycle arrest and apoptosis in multiple breast cancer cells. Interestingly, these observations are more pronounced in cells grown under anchorage-independent conditions or grown in vivo. Consistent with the negative impact on DNA repair, ICMT inhibition transforms the cancer cells into a "BRCA-like" state, hence sensitizing cancer cells to the treatment of PARP inhibitor and other DNA damage-inducing agents.


Subject(s)
Breast Neoplasms/metabolism , DNA Damage/genetics , DNA Repair/genetics , MAP Kinase Signaling System/genetics , Protein Methyltransferases/metabolism , Animals , Apoptosis/drug effects , Apoptosis/genetics , Benzamides/pharmacology , Breast Neoplasms/pathology , Cell Cycle Checkpoints/drug effects , Cell Cycle Checkpoints/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/genetics , DNA Damage/drug effects , DNA Repair/drug effects , Female , Genetic Vectors , HEK293 Cells , Humans , Indazoles/pharmacology , MAP Kinase Signaling System/drug effects , Mice , Mice, SCID , Piperidines/pharmacology , Poly (ADP-Ribose) Polymerase-1/antagonists & inhibitors , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Protein Methyltransferases/genetics , RNA, Small Interfering/genetics , Ribonucleosides/pharmacology , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
9.
J Mater Sci Mater Med ; 32(11): 133, 2021 Oct 23.
Article in English | MEDLINE | ID: mdl-34689241

ABSTRACT

OBJECTIVE: In our previous study, tantalum nanoparticle (Ta-NPs) was demonstrated to promote osteoblast proliferation via autophagy induction, but the specific mechanism remains unclear. In the present study, we will explore the potential mechanism. METHODS: Ta-NPs was characterized by transmission electron microscopy, scanning electron microscopy, dynamic light scattering, and BET specific surface area test. MC3T3-E1 were treated with 0 or 20 µg/mL Ta-NPs with or without pretreatment with 10 µM LY294002, Triciribine, Rapamycin (PI3K/Akt/mTOR pathway inhibitors) for 1 h respectively. Western blotting was used to detect the expressions of pathway proteins and LC3B. CCK-8 assay was used to assess cell viability. Flow cytometry was used to detect apoptosis and cell cycle. RESULTS: After pretreatment with LY294002, Triciribine and Rapamycin, the p-Akt/Akt ratio of pathway protein in Triciribine and Rapamycin groups decreased (P < 0.05), while the autophagy protein LC3-II/LC3-I in the Rapamycin group was upregulated obviously (P < 0.001). In all pretreated groups, apoptosis was increased (LY294002 group was the most obvious), G1 phase cell cycle was arrested (Triciribine and Rapamycin groups were more obvious), and MC3T3-E1 cells were proliferated much more (P < 0.01, P < 0.001, P < 0.05). CONCLUSION: Pretreatment with Triciribine or Rapamycin has a greater effect on pathway protein Akt, cell cycle arrest, autophagy protein, and cell proliferation but with inconsistent magnitude, which may be inferred that the Akt/mTOR pathway, as well as its feedback loop, were more likely involved in these processes.


Subject(s)
Apoptosis/drug effects , Cell Proliferation/drug effects , Metal Nanoparticles/toxicity , Tantalum/chemistry , 3T3 Cells , Animals , Biocompatible Materials , Chromones/pharmacology , Gene Expression Regulation/drug effects , Materials Testing , Metal Nanoparticles/chemistry , Mice , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Morpholines/pharmacology , Ribonucleosides/pharmacology , Sirolimus/pharmacology
10.
Neuropharmacology ; 197: 108745, 2021 10 01.
Article in English | MEDLINE | ID: mdl-34375627

ABSTRACT

The voltage-sensitive sodium channel NaV1.1 plays a critical role in regulating excitability of GABAergic neurons and mutations in the corresponding gene are associated to Dravet syndrome and other forms of epilepsy. The activity of this channel is regulated by several protein kinases. To identify novel regulatory kinases we screened a library of activated kinases and we found that AKT1 was able to directly phosphorylate NaV1.1. In vitro kinase assays revealed that the phosphorylation site was located in the C-terminal part of the large intracellular loop connecting domains I and II of NaV1.1, a region that is known to be targeted by other kinases like PKA and PKC. Electrophysiological recordings revealed that activated AKT1 strongly reduced peak Na+ currents and displaced the inactivation curve to more negative potentials in HEK-293 cell stably expressing NaV1.1. These alterations in current amplitude and steady-state inactivation were mimicked by SC79, a specific activator of AKT1, and largely reverted by triciribine, a selective inhibitor. Neurons expressing endogenous NaV1.1 in primary cultures were identified by expressing a fluorescent protein under the NaV1.1 promoter. There, we also observed a strong decrease in the current amplitude after addition of SC79, but small effects on the inactivation parameters. Altogether, we propose a novel mechanism that might regulate the excitability of neural networks in response to AKT1, a kinase that plays a pivotal role under physiological and pathological conditions, including epileptogenesis.


Subject(s)
NAV1.1 Voltage-Gated Sodium Channel/physiology , Proto-Oncogene Proteins c-akt/physiology , Animals , Electrophysiological Phenomena , Epilepsies, Myoclonic/genetics , HEK293 Cells , Humans , NAV1.1 Voltage-Gated Sodium Channel/genetics , Nerve Net/drug effects , Neurons/metabolism , Phosphorylation , Primary Cell Culture , Proto-Oncogene Proteins c-akt/agonists , Proto-Oncogene Proteins c-akt/genetics , Rats , Ribonucleosides/pharmacology , Sodium Channel Agonists/pharmacology , Sodium Channel Blockers/pharmacology
11.
Antiviral Res ; 193: 105139, 2021 09.
Article in English | MEDLINE | ID: mdl-34273445

ABSTRACT

Because ganciclovir resistance mutations in the cytomegalovirus UL97 gene most commonly occur at codons 460, 520 and 590-607, diagnostic genotyping for drug resistance has often omitted the analysis of codons below 440. However, the UL97 kinase inhibitor maribavir selects for distinctive resistance mutations at codons 409 and 411, and ganciclovir/maribavir resistance mutations have also been described in the ATP binding region starting at codon 335. Expanded genotypic testing of UL97 codons 335-440 in 1535 clinical specimens disclosed 10 uncharacterized sequence variants that were phenotyped for ganciclovir and maribavir susceptibility. Notable findings included low-grade ganciclovir resistance conferred by amino acid substitutions K359N and E362D, decreased maribavir susceptibility of L348V, and maribavir hypersensitivity of V345I and E362D. Recently published substitutions F342Y and K359E/Q were also confirmed. The data indicate that mutations in the UL97 ATP binding region may arise in clinical specimens to affect the interpretation of ganciclovir and maribavir resistance. This region should now be included in the standard diagnostic genotyping of UL97, especially with the introduction of maribavir into therapeutic use.


Subject(s)
Benzimidazoles/pharmacology , Cytomegalovirus Infections/drug therapy , Cytomegalovirus/drug effects , Drug Resistance, Viral/genetics , Ganciclovir/pharmacology , Ribonucleosides/pharmacology , Adenosine Triphosphate , Amino Acid Substitution , Antiviral Agents/pharmacology , Codon , Genotyping Techniques , Humans , Mutation , Phenotype , Phosphotransferases (Alcohol Group Acceptor)/genetics
12.
Am J Physiol Cell Physiol ; 321(1): C82-C93, 2021 07 01.
Article in English | MEDLINE | ID: mdl-34038245

ABSTRACT

The resolution of inflammation is closely linked with tissue repair. Recent studies have revealed that macrophages suppress inflammatory reactions by producing lipid mediators, called specialized proresolving mediators (SPMs); however, the biological significance of SPMs in tissue repair remains to be fully elucidated in the heart. In this study, we focused on maresin-1 (MaR1) and examined the reparative effects of MaR1 in cardiomyocytes. The treatment with MaR1 increased cell size in cultured neonatal rat cardiomyocytes. Since the expression of fetal cardiac genes was unchanged by MaR1, physiological hypertrophy was induced by MaR1. SR3335, an inhibitor of retinoic acid-related orphan receptor α (RORα), mitigated MaR1-induced cardiomyocyte hypertrophy, consistent with the recent report that RORα is one of MaR1 receptors. Importantly, in response to MaR1, cardiomyocytes produced IGF-1 via RORα. Moreover, MaR1 activated phosphoinositide 3-kinase (PI3K)/Akt signaling pathway and wortmannin, a PI3K inhibitor, or triciribine, an Akt inhibitor, abrogated MaR1-induced cardiomyocyte hypertrophy. Finally, the blockade of IGF-1 receptor by NVP-AEW541 inhibited MaR-1-induced cardiomyocyte hypertrophy as well as the activation of PI3K/Akt pathway. These data indicate that MaR1 induces cardiomyocyte hypertrophy through RORα/IGF-1/PI3K/Akt pathway. Considering that MaR1 is a potent resolving factor, MaR1 could be a key mediator that orchestrates the resolution of inflammation with myocardial repair.


Subject(s)
Cardiomegaly/genetics , Cardiotonic Agents/pharmacology , Docosahexaenoic Acids/adverse effects , Insulin-Like Growth Factor I/genetics , Myocardial Infarction/genetics , Myocytes, Cardiac/drug effects , Paracrine Communication/genetics , Animals , Cardiomegaly/chemically induced , Cardiomegaly/pathology , Cardiomegaly/prevention & control , Disease Models, Animal , Docosahexaenoic Acids/antagonists & inhibitors , Gene Expression Regulation , Insulin-Like Growth Factor I/antagonists & inhibitors , Insulin-Like Growth Factor I/metabolism , Male , Mice , Mice, Inbred C57BL , Myocardial Infarction/chemically induced , Myocardial Infarction/pathology , Myocardial Infarction/prevention & control , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Nuclear Receptor Subfamily 1, Group F, Member 1/antagonists & inhibitors , Nuclear Receptor Subfamily 1, Group F, Member 1/genetics , Nuclear Receptor Subfamily 1, Group F, Member 1/metabolism , Paracrine Communication/drug effects , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Primary Cell Culture , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Pyrimidines/pharmacology , Pyrroles/pharmacology , Rats , Ribonucleosides/pharmacology , Signal Transduction , Sulfonamides/pharmacology , Thiophenes/pharmacology , Wortmannin/pharmacology
13.
Bioorg Med Chem Lett ; 40: 127957, 2021 05 15.
Article in English | MEDLINE | ID: mdl-33741462

ABSTRACT

Trypanosoma brucei parasites cause Human African Trypanosomiasis and the current drugs for its treatment are often inefficient and toxic. This urges the need to development of new antitrypanosomal agents. We report the synthesis and biological profiling of 3'-deoxy-3'-fluororibonucleosides derived from 7-deazaadenine nucleosides bearing diverse substituents at position 7. They were synthesized through glycosylation of 6-chloro-7-bromo- or -7-iodo-7-deazapurine with protected 3'-fluororibose followed by cross-coupling reactions at position 7 and/or deprotection. Most of the title nucleosides displayed micromolar or submicromolar activity against Trypanosoma brucei brucei. The most active were the 7-bromo- and 7-iododerivatives which exerted double-digit nanomolar activity against T. b. brucei and T. b. gambiense and no cytotoxicity and thus represent promising candidates for further development.


Subject(s)
Ribonucleosides/pharmacology , Trypanocidal Agents/pharmacology , Cell Line, Tumor , Fibroblasts/drug effects , Humans , Molecular Structure , Parasitic Sensitivity Tests , Ribonucleosides/chemical synthesis , Ribonucleosides/toxicity , Trypanocidal Agents/chemical synthesis , Trypanocidal Agents/toxicity , Trypanosoma brucei brucei/drug effects , Trypanosoma brucei gambiense/drug effects
14.
Int J Mol Sci ; 22(2)2021 Jan 08.
Article in English | MEDLINE | ID: mdl-33430060

ABSTRACT

Human cytomegalovirus (HCMV) is a human pathogenic herpesvirus associated with a variety of clinical symptoms. Current antiviral therapy is not always effective, so that improved drug classes and drug-targeting strategies are needed. Particularly host-directed antivirals, including pharmaceutical kinase inhibitors (PKIs), may help to overcome problems of drug resistance. Here, we focused on utilizing a selection of clinically relevant PKIs and determined their anticytomegaloviral efficacies. Particularly, PKIs directed to host or viral cyclin-dependent kinases, i.e., abemaciclib, LDC4297 and maribavir, exerted promising profiles against human and murine cytomegaloviruses. The anti-HCMV in vitro activity of the approved anti-cancer drug abemaciclib was confirmed in vivo using our luciferase-based murine cytomegalovirus (MCMV) animal model in immunocompetent mice. To assess drug combinations, we applied the Bliss independence checkerboard and Loewe additivity fixed-dose assays in parallel. Results revealed that (i) both affirmative approaches provided valuable information on anti-CMV drug efficacies and interactions, (ii) the analyzed combinations comprised additive, synergistic or antagonistic drug interactions consistent with the drugs' antiviral mode-of-action, (iii) the selected PKIs, especially LDC4297, showed promising inhibitory profiles, not only against HCMV but also other α-, ß- and γ-herpesviruses, and specifically, (iv) the combination treatment with LDC4297 and maribavir revealed a strong synergism against HCMV, which might open doors towards novel clinical options in the near future. Taken together, this study highlights the potential of therapeutic drug combinations of current developmental/preclinical PKIs.


Subject(s)
Cytomegalovirus Infections/drug therapy , Drug Resistance, Viral/drug effects , Protein Kinase Inhibitors/pharmacology , Virus Replication/genetics , Aminopyridines/pharmacology , Animals , Antiviral Agents/pharmacology , Benzimidazoles/pharmacology , Cell Line , Cytomegalovirus/drug effects , Cytomegalovirus/pathogenicity , Cytomegalovirus Infections/genetics , Cytomegalovirus Infections/virology , Drug Combinations , Ganciclovir/pharmacology , Humans , Mice , Pyrazoles/pharmacology , Ribonucleosides/pharmacology , Triazines/pharmacology , Virus Replication/drug effects
15.
J Infect Dis ; 224(6): 1024-1028, 2021 09 17.
Article in English | MEDLINE | ID: mdl-33475730

ABSTRACT

Two transplant recipients (1 kidney and 1 hematopoietic stem cell) received maribavir (MBV) after cytomegalovirus (CMV) infection clinically resistant to standard therapy. Both patients achieved CMV DNA clearance within 30 and 18 days; however, the UL97 C480F variant emerged, causing recurrent CMV infection after a cumulative 2 months of MBV and 15 or 4 weeks of ganciclovir treatment, respectively. C480F was not detected under ganciclovir before MBV treatment. Recombinant phenotyping showed that C480F conferred the highest level of MBV resistance and ganciclovir cross-resistance, with impaired viral growth. Clinical follow-up and genotypic and phenotypic studies are essential for the assessment and optimization of patients with suspected MBV resistance.


Subject(s)
Benzimidazoles/therapeutic use , Cytomegalovirus Infections/drug therapy , Cytomegalovirus/drug effects , Drug Resistance, Viral/genetics , Ganciclovir/therapeutic use , Hematopoietic Stem Cell Transplantation/adverse effects , Kidney Transplantation/adverse effects , Ribonucleosides/therapeutic use , Transplant Recipients , Adult , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Benzimidazoles/pharmacology , Cytomegalovirus/genetics , Drug Resistance, Viral/drug effects , Female , Ganciclovir/pharmacology , Hematopoietic Stem Cells , Humans , Mutation/drug effects , Organ Transplantation , Phosphotransferases (Alcohol Group Acceptor)/genetics , Phosphotransferases (Alcohol Group Acceptor)/therapeutic use , Ribonucleosides/pharmacology , Treatment Outcome
16.
Int J Mol Sci ; 22(1)2021 Jan 02.
Article in English | MEDLINE | ID: mdl-33401686

ABSTRACT

Senescent cells secrete pro-inflammatory factors, and a hallmark feature of senescence is senescence-associated secretory phenotype (SASP). The aim of this study is to investigate the protein kinase CK2 (CK2) effects on SASP factors expression in cellular senescence and organism aging. Here CK2 down-regulation induced the expression of SASP factors, including interleukin (IL)-1ß, IL-6, and matrix metalloproteinase (MMP) 3, through the activation of nuclear factor-κB (NF-κB) signaling in MCF-7 and HCT116 cells. CK2 down-regulation-mediated SIRT1 inactivation promoted the degradation of inhibitors of NF-κB (IκB) by activating the AKT-IκB kinase (IKK) axis and increased the acetylation of lysine 310 on RelA/p65, an important site for the activity of NF-κB. kin-10 (the ortholog of CK2ß) knockdown increased zmp-1, -2, and -3 (the orthologs of MMP) expression in nematodes, but AKT inhibitor triciribine and SIRT activator resveratrol significantly abrogated the increased expression of these genes. Finally, antisense inhibitors of miR-186, miR-216b, miR-337-3p, and miR-760 suppressed CK2α down-regulation, activation of the AKT-IKK-NF-κB axis, RelA/p65 acetylation, and expression of SASP genes in cells treated with lipopolysaccharide. Therefore, this study indicated that CK2 down-regulation induces the expression of SASP factors through NF-κB activation, which is mediated by both activation of the SIRT1-AKT-IKK axis and RelA/p65 acetylation, suggesting that the mixture of the four miRNA inhibitors can be used as anti-inflammatory agents.


Subject(s)
Gene Expression Regulation/genetics , Protein Serine-Threonine Kinases/metabolism , Resveratrol/pharmacology , Ribonucleosides/pharmacology , Signal Transduction/genetics , Animals , Caenorhabditis elegans , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Casein Kinase II/genetics , Casein Kinase II/metabolism , Cell Line, Tumor , Cellular Senescence , Down-Regulation , Gene Expression Regulation/drug effects , Humans , Interleukin-1beta/metabolism , Interleukin-6/metabolism , Lipopolysaccharides/pharmacology , Matrix Metalloproteinase 3/metabolism , Metalloendopeptidases/genetics , Metalloendopeptidases/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , RNA Interference , Signal Transduction/drug effects , Sirtuin 1/metabolism , Transcription Factor RelA/metabolism , NF-kappaB-Inducing Kinase
17.
ACS Infect Dis ; 7(2): 471-478, 2021 02 12.
Article in English | MEDLINE | ID: mdl-33395259

ABSTRACT

A series of 7-deazaadenine ribonucleosides bearing alkyl, alkenyl, alkynyl, aryl, or hetaryl groups at position 7 as well as their 5'-O-triphosphates and two types of monophosphate prodrugs (phosphoramidates and S-acylthioethanol esters) were prepared and tested for antiviral activity against selected RNA viruses (Dengue, Zika, tick-borne encephalitis, West Nile, and SARS-CoV-2). The modified triphosphates inhibited the viral RNA-dependent RNA polymerases at micromolar concentrations through the incorporation of the modified nucleotide and stopping a further extension of the RNA chain. 7-Deazaadenosine nucleosides bearing ethynyl or small hetaryl groups at position 7 showed (sub)micromolar antiviral activities but significant cytotoxicity, whereas the nucleosides bearing bulkier heterocycles were still active but less toxic. Unexpectedly, the monophosphate prodrugs were similarly or less active than the corresponding nucleosides in the in vitro antiviral assays, although the bis(S-acylthioethanol) prodrug 14h was transported to the Huh7 cells and efficiently released the nucleoside monophosphate.


Subject(s)
Antiviral Agents/pharmacology , Prodrugs/pharmacology , Purines/pharmacology , RNA Viruses/drug effects , Ribonucleosides/pharmacology , COVID-19/virology , Cell Line, Tumor , Dengue Virus/drug effects , Encephalitis Viruses, Tick-Borne/drug effects , Humans , Phosphates/pharmacology , Purine Nucleosides , RNA-Dependent RNA Polymerase/metabolism , SARS-CoV-2/drug effects , West Nile virus/drug effects , Zika Virus/drug effects , COVID-19 Drug Treatment
18.
Circ Heart Fail ; 14(1): e005890, 2021 01.
Article in English | MEDLINE | ID: mdl-33356364

ABSTRACT

BACKGROUND: We recently reported that treatment with rhBDNF (recombinant human brain-derived neurotrophic factor) improved the reduced exercise capacity of mice with heart failure (HF) after myocardial infarction (MI). Since BDNF is reported to enhance fatty acid oxidation, we herein conducted an in vivo investigation to determine whether the improvement in exercise capacity is due to the enhancement of the fatty acid oxidation of skeletal muscle via the AMPKα-PGC1α (adenosine monophosphate-activated protein kinase-ɑ-proliferator-activated receptor-r coactivator-1ɑ) axis. METHODS: MI and sham operations were conducted in C57BL/6J mice. Two weeks postsurgery, we randomly divided the MI mice into groups treated with rhBDNF or vehicle for 2 weeks. AMPKα-PGC1α signaling and mitochondrial content in the skeletal muscle of the mice were evaluated by Western blotting and transmission electron microscopy. Fatty acid ß-oxidation was examined by high-resolution respirometry using permeabilized muscle fiber. BDNF-knockout mice were treated with 5-aminoimidazole-4-carboxamide-1-beta-d-riboruranoside, an activator of AMPK. RESULTS: The rhBDNF treatment significantly increased the expressions of phosphorylated AMPKα and PGC1α protein and the intermyofibrillar mitochondrial density in the MI mice. The lowered skeletal muscle mitochondrial fatty acid oxidation was significantly improved in the rhBDNF-treated MI mice. The reduced exercise capacity and mitochondrial dysfunction of the BDNF-knockout mice were improved by 5-aminoimidazole-4-carboxamide-1-beta-d-riboruranoside. CONCLUSIONS: Beneficial effects of BDNF on the exercise capacity of mice with HF are mediated through an enhancement of fatty acid oxidation via the activation of AMPKα-PGC1α in skeletal muscle. BDNF may become a therapeutic option to improve exercise capacity as an alternative or adjunct to exercise training.


Subject(s)
AMP-Activated Protein Kinases/drug effects , Brain-Derived Neurotrophic Factor/pharmacology , Exercise Tolerance/drug effects , Fatty Acids/metabolism , Heart Failure/metabolism , Muscle, Skeletal/drug effects , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/drug effects , AMP-Activated Protein Kinases/metabolism , Aminoimidazole Carboxamide/analogs & derivatives , Aminoimidazole Carboxamide/pharmacology , Animals , Brain-Derived Neurotrophic Factor/genetics , Heart Failure/genetics , Heart Failure/physiopathology , Humans , Male , Mice , Mice, Knockout , Microscopy, Electron, Transmission , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondria/ultrastructure , Muscle, Skeletal/metabolism , Muscle, Skeletal/ultrastructure , Myocardial Infarction/metabolism , Myocardial Infarction/physiopathology , Oxidation-Reduction/drug effects , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Recombinant Proteins , Ribonucleosides/pharmacology
19.
Front Immunol ; 11: 556838, 2020.
Article in English | MEDLINE | ID: mdl-33329515

ABSTRACT

Recent study in our laboratory has demonstrated that BEFV-induced autophagy via activation of the PI3K/Akt/NF-κB and Src/JNK pathways and suppression of the PI3K-AKt-mTORC1 pathway is beneficial for virus replication. In the current study, we found that both aspirin and 5-aminoimidazole-4-carboxamide-1-ß-riboside (AICAR) siginificantly attenuated virus replication by inhibiting BEFV-induced autophagy via suppressing the BEFV-activated PI3K/Akt/NF-κB and Src/JNK pathways as well as inducing reversion of the BEFV-suppressed PI3K-Akt-mTORC1 pathway. AICAR reversed the BEFV-activated PI3K/Akt/NF-κB and Src/JNK pathways at the early to late stages of infection and induced reversion of the BEFV-suppressed PI3K-AKt-mTORC1 pathway at the late stage of infection. Our findings reveal that inhibition of BEFV-induced autophagy by AICAR is independent of AMPK. Furthermore, we found that AICAR transcriptionally downregulates the ATG related genes ULK1, Beclin 1, and LC3 and enhances Atg7 degradation by the proteasome pathway. Aspirin suppresses virus replication by inhibiting BEFV-induced autophagy. It directly suppressed the NF-κB pathway and reversed the BEFV-activated Src/JNK pathway at the early stage of infection and reversed the BEFV-suppressed PI3K/Akt/mTOR pathway at the late stage of infection. The current study provides mechanistic insights into the effects of aspirin and AICAR on BEFV replication through suppression of BEFV-induced autophagy.


Subject(s)
Aminoimidazole Carboxamide/analogs & derivatives , Aspirin/pharmacology , Autophagy/drug effects , Ephemeral Fever Virus, Bovine/drug effects , Ephemeral Fever Virus, Bovine/physiology , Ephemeral Fever/virology , Ribonucleosides/pharmacology , Virus Replication/drug effects , Aminoimidazole Carboxamide/pharmacology , Animals , Biomarkers , Cattle , Cell Line , Cell Survival/drug effects , Cyclooxygenase 2/metabolism , Ephemeral Fever/metabolism , Gene Expression Regulation/drug effects , NF-kappa B/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , RNA, Small Interfering
20.
PLoS One ; 15(12): e0244307, 2020.
Article in English | MEDLINE | ID: mdl-33362238

ABSTRACT

RATIONALE: Age-related macular degeneration (AMD) is the most prevalent form of irreversible blindness in the developed world. Aging, inflammation and complement dysregulation affecting the retinal pigment epithelium (RPE), are considered significant contributors in its pathogenesis and several evidences have linked tumor necrosis factor alpha (TNF-α) and complement component 3 (C3) with AMD. Acadesine, an analog of AMP and an AMP-activated protein kinase (AMPK) activator, has been shown to have cytoprotective effects in human clinical trials as well as having anti-inflammatory and anti-vascular exudative effects in animals. The purpose of this study was to evaluate if acadesine is able to suppress TNF-α induced C3 in RPE cells. METHODS: ARPE-19 and human primary RPE cells were cultured and allowed to grow to confluence. TNF-α was used for C3 induction in the presence or absence of acadesine. Small molecule inhibitors and siRNA were used to determine if acadesine exerts its effect via the extracellular or intracellular pathway and to evaluate the importance of AMPK for these effects. The expression level of C3 was determined by immunoblot analysis. RESULTS: Acadesine suppresses TNF-α induced C3 in a dose dependent manner. When we utilized the adenosine receptor inhibitor dipyridamole (DPY) along with acadesine, acadesine's effects were abolished, indicating the necessity of acadesine to enter the cell in order to exert it's action. However, pretreatment with 5-iodotubericidin (5-Iodo), an adenosine kinase (AK) inhibitor, didn't prevent acadesine from decreasing TNF-α induced C3 expression suggesting that acadesine does not exert its effect through AMP conversion and subsequent activation of AMPK. Consistent with this, knockdown of AMPK α catalytic subunit did not affect the inhibitory effect of acadesine on TNF-α upregulation of C3. CONCLUSIONS: Our results suggest that acadesine suppresses TNF-α induced C3, likely through an AMPK-independent pathway, and could have potential use in complement over activation diseases.


Subject(s)
Aminoimidazole Carboxamide/analogs & derivatives , Complement C3/metabolism , Retinal Pigment Epithelium/metabolism , Ribonucleosides/pharmacology , AMP-Activated Protein Kinases/metabolism , Adenosine/metabolism , Aminoimidazole Carboxamide/metabolism , Aminoimidazole Carboxamide/pharmacology , Animals , Cell Line , Cells, Cultured , Complement Activation/drug effects , Complement C3/drug effects , Humans , Macular Degeneration/metabolism , Phosphorylation , Retinal Pigment Epithelium/drug effects , Retinal Pigments/metabolism , Ribonucleosides/metabolism , Ribonucleotides/pharmacology , Tumor Necrosis Factor-alpha/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...