Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 101
Filter
1.
J Appl Toxicol ; 44(6): 846-852, 2024 06.
Article in English | MEDLINE | ID: mdl-38291012

ABSTRACT

Trovafloxacin is a quinolone antibiotic drug with broad-spectrum activity, which was withdrawn from a global market relatively soon after approval because of serious liver injury. The characteristics of trovafloxacin-induced liver injury are consistent with an idiosyncratic reaction; however, the details of the mechanism have not been elucidated. We examined whether trovafloxacin induces the release of damage-associated molecular patterns (DAMPs) that activate inflammasomes. We also tested ciprofloxacin, levofloxacin, gatifloxacin, and grepafloxacin for their ability to activate inflammasomes. Drug bioactivation was performed with human hepatocarcinoma functional liver cell-4 (FLC-4) cells, and THP-1 cells (human monocyte cell line) were used for the detection of inflammasome activation. The supernatant from the incubation of trovafloxacin with FLC-4 cells for 7 days increased caspase-1 activity and production of IL-1ß by THP-1 cells. In the supernatant of FLC-4 cells that had been incubated with trovafloxacin, heat shock protein (HSP) 40 was significantly increased. Addition of a cytochrome P450 inhibitor to the FLC-4 cells prevented the release of HSP40 from the FLC-4 cells and inflammasome activation in THP-1 cells by the FLC-4 supernatant. These results suggest that reactive metabolites of trovafloxacin can cause the release of DAMPs from hepatocytes that can activate inflammasomes. Inflammasome activation may be an important step in the activation of the immune system by trovafloxacin, which, in some patients, can cause immune-related liver injury.


Subject(s)
Chemical and Drug Induced Liver Injury , Fluoroquinolones , Inflammasomes , Naphthyridines , Humans , Inflammasomes/metabolism , Inflammasomes/drug effects , Fluoroquinolones/toxicity , Chemical and Drug Induced Liver Injury/metabolism , Naphthyridines/toxicity , Naphthyridines/pharmacology , THP-1 Cells , Anti-Bacterial Agents/toxicity , Cell Line, Tumor , Interleukin-1beta/metabolism
2.
Toxicol In Vitro ; 82: 105374, 2022 Aug.
Article in English | MEDLINE | ID: mdl-35537566

ABSTRACT

Drug-induced liver injury (DILI) is an adverse hepatic reaction and a serious concern for public healthcare systems and pharmaceutical companies. DILI is frequently caused by a combination of direct toxic stresses and subsequent immune damage to hepatocytes. However, little is known about the mechanism by which drugs facilitate the activation of the innate immune system. Here, we aimed to decipher the inflammatory events in trovafloxacin (TVX)-induced reactions using liver macrophages. We showed that proinflammatory M1-like macrophages mainly contributed to hepatotoxicity mediated by TVX, a DILI drug. Additionally, transcriptome results showed that the interferon type I pathway, cytokines, and apoptosis pathway were involved in the initiation of synergistic effects resulting in TVX-induced liver injury. We hypothesized that DILI drugs could drive liver injury by altering the activation and phenotype of hepatic macrophages. Furthermore, drug treatment-induced transcriptional changes such as Traf1 and 2, Socs3, and Hbegf in macrophage polarization could be used to assess drug-specific immune-mediated reactions. Therefore, we proposed that transcriptional change in the genes related to macrophage polarization index could be an indicator to reflect the severity of DILI in a preclinical setting during drug development.


Subject(s)
Chemical and Drug Induced Liver Injury , Drug-Related Side Effects and Adverse Reactions , Chemical and Drug Induced Liver Injury/metabolism , Fluoroquinolones , Humans , Inflammation/chemically induced , Inflammation/metabolism , Liver/metabolism , Macrophages , Naphthyridines/metabolism , Naphthyridines/toxicity
3.
Toxicol Sci ; 186(2): 323-337, 2022 03 28.
Article in English | MEDLINE | ID: mdl-35134999

ABSTRACT

Izencitinib (TD-1473), an oral, gut-selective pan-Janus kinase (JAK) inhibitor under investigation for treatment of inflammatory bowel diseases, was designed for optimal efficacy in the gastrointestinal tract while minimizing systemic exposures and JAK-related safety findings. The nonclinical safety of izencitinib was evaluated in rat and dog repeat-dose and rat and rabbit reproductive and developmental toxicity studies. Systemic exposures were compared with JAK inhibitory potency to determine effects at or above pharmacologic plasma concentrations (≥1× plasma average plasma concentration [Cave]:JAK 50% inhibitory concentration [IC50] ratio). In rats and dogs, 1000 and 30 mg/kg/day izencitinib, respectively, produced minimal systemic findings (ie, red/white cell changes) and low systemic concentrations (approximately 1× plasma Cave:JAK IC50 ratio) with an 8× nonclinical:clinical systemic area under the curve (AUC) margin compared with exposures at the highest clinically tested dose (300 mg, quaque die, once daily, phase 1 study in healthy volunteers). In dogs, it was possible to attain sufficient systemic exposures to result in immunosuppression characteristic of systemic JAK inhibition, but at high AUC margins (43×) compared with systemic exposures observed at the highest tested dose in humans. No adverse findings were observed in the gastrointestinal tract or systemic tissues. Izencitinib did not affect male or female fertility. Izencitinib did not affect embryonic development in rats and rabbits as commonly reported with systemic JAK inhibition, consistent with low maternal systemic concentrations (2-6× plasma Cave:JAK IC50 ratio, 10-33× nonclinical:clinical AUC margin) and negligible fetal exposures. In conclusion, the izencitinib gut-selective approach resulted in minimal systemic findings in nonclinical species at pharmacologic, clinically relevant systemic exposures, highlighting the impact of organ-selectivity in reducing systemic safety findings.


Subject(s)
Janus Kinases , Naphthyridines , Nitriles , Administration, Oral , Animals , Dogs , Embryonic Development/drug effects , Female , Healthy Volunteers , Humans , Inflammatory Bowel Diseases , Janus Kinases/antagonists & inhibitors , Male , Naphthyridines/pharmacology , Naphthyridines/toxicity , Nitriles/pharmacology , Nitriles/toxicity , Pregnancy , Rabbits , Rats , Reproduction/drug effects , Toxicity Tests
4.
Toxicol Lett ; 342: 73-84, 2021 May 15.
Article in English | MEDLINE | ID: mdl-33609687

ABSTRACT

Trovafloxacin (TVX) is associated with idiosyncratic drug-induced liver injury (iDILI) and inflammation-mediated hepatotoxicity. However, the inflammatory stress-regulated mechanisms in iDILI remain unclear. Herein, we elucidated the novel role of tumor-necrosis factor alpha (TNFα), an inflammatory stress factor, in TVX-induced in vitro hepatotoxicity and synergistic toxicity. TVX specifically induced synergistic toxicity in HepG2 cells with TNFα, which inhibits autophagy. TVX-treated HepG2 cells induced protective autophagy by inhibiting the expression of mTOR signaling proteins, while ATG5 knockdown in HepG2 cells, responsible for the impairment of autophagy, enhanced TVX-induced toxicity due to the increase in cytochrome C release and JNK pathway activation. Interestingly, the expression of mTOR signal proteins, which were suppressed by TVX, disrupted the negative feedback of the PI3K/AKT pathway and TNFα rebounded p70S6K phosphorylation. Co-treatment with TVX and TNFα inhibited protective autophagy by maintaining p70S6K activity, which enhanced TVX-induced cytotoxicity. Phosphorylation of p70S6K was inhibited by siRNA knockdown and rapamycin to restore TNFα-inhibited autophagy, which prevented the synergistic effect on TVX-induced cytotoxicity. These results indicate that TVX activates protective autophagy in HepG2 cells exposed to toxicity and an imbalance in negative feedback regulation of autophagy by TNFα synergistically enhanced the toxicity. The finding from this study may contribute to a better understanding of the mechanisms underlying iDILI associated with inflammatory stress.


Subject(s)
Autophagy/drug effects , Fluoroquinolones/toxicity , Hepatocytes/drug effects , Naphthyridines/toxicity , Tumor Necrosis Factor-alpha/pharmacology , Antimalarials/toxicity , Cell Survival , Chloroquine/toxicity , Gene Expression Regulation/drug effects , Hep G2 Cells , Humans , Levofloxacin/pharmacology , MAP Kinase Kinase 4/genetics , MAP Kinase Kinase 4/metabolism , Piperazines/toxicity , Ribosomal Protein S6 Kinases, 70-kDa/genetics , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Serotonin and Noradrenaline Reuptake Inhibitors/toxicity , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism , Triazoles/toxicity
5.
Genetics ; 215(3): 609-621, 2020 07.
Article in English | MEDLINE | ID: mdl-32414869

ABSTRACT

New anticancer therapeutics require extensive in vivo characterization to identify endogenous and exogenous factors affecting efficacy, to measure toxicity and mutagenicity, and to determine genotypes that result in therapeutic sensitivity or resistance. We used Caenorhabditis elegans as a platform with which to characterize properties of the anticancer therapeutic CX-5461. To understand the processes that respond to CX-5461-induced damage, we generated pharmacogenetic profiles for a panel of C. elegans DNA replication and repair mutants with common DNA-damaging agents for comparison with the profile of CX-5461. We found that multiple repair pathways, including homology-directed repair, microhomology-mediated end joining, nucleotide excision repair, and translesion synthesis, were needed for CX-5461 tolerance. To determine the frequency and spectrum of CX-5461-induced mutations, we used a genetic balancer to capture CX-5461-induced mutations. We found that CX-5461 is mutagenic, resulting in both large copy number variations and a high frequency of single-nucleotide variations (SNVs), which are consistent with the pharmacogenetic profile for CX-5461. Whole-genome sequencing of CX-5461-exposed animals found that CX-5461-induced SNVs exhibited a distinct mutational signature. We also phenocopied the CX-5461 photoreactivity observed in clinical trials and demonstrated that CX-5461 generates reactive oxygen species when exposed to UVA radiation. Together, the data from C. elegans demonstrate that CX-5461 is a multimodal DNA-damaging anticancer agent.


Subject(s)
Antineoplastic Agents/toxicity , Benzothiazoles/toxicity , Caenorhabditis elegans/genetics , Carcinogenicity Tests/methods , Genome-Wide Association Study/methods , Mutagens/toxicity , Naphthyridines/toxicity , Pharmacogenomic Variants , Animals , Caenorhabditis elegans/drug effects , DNA Repair , Drug Resistance, Neoplasm , Genome, Helminth , Mutation , Polymorphism, Single Nucleotide
6.
EBioMedicine ; 54: 102711, 2020 Apr.
Article in English | MEDLINE | ID: mdl-32279056

ABSTRACT

BACKGROUND: Cystic echinococcosis (CE), a condition caused by the larval stage of the dog tapeworm Echinococcus granulosus sensu stricto, is a globally distributed zoonotic disease. Current treatment options for CE are limited, and an effective and safe anti-echinococcal drug is urgently required. METHODS: Drug repurposing strategy was employed to identify new therapeutic agents against echinococcal cysts. An in vitro protoscolicidal assay along with in vivo murine models was applied in the drug screening. A microinjection procedure was employed to mimic the clinical PAIR (puncture, aspiration, injection and reaspiration) technique to evaluate the potential application of the candidate drug in clinical practice. FINDINGS: We repurposed pyronaridine, an approved antimalarial drug, for the treatment of CE. Following a three-dose intraperitoneal regimen (57 mg/kg, q.d. for 3 days), pyronaridine caused 100% cyst mortality. Oral administration of pyronaridine at 57 mg/kg, q.d. for 30 days significantly reduced the parasitic burden in the pre-infected mice compared with albendazole group (p < 0.001). Using a microinjection of drug into cysts, pyronaridine (200 µM) showed highly effective in term of inhibition of cyst growth (p < 0.05, compared with saline group). Pharmacokinetic analysis revealed that pyronaridine was highly distributed in the liver and lungs, the most affected organs of CE. Function analysis showed that pyronaridine inhibited the activity of topoisomerase I (IC50 = 209.7 ± 1.1 µM). In addition, classical apoptotic hallmarks, including DNA fragmentation and caspase activation, were triggered. INTERPRETATION: Given its approved clinical safety, the repurposing of pyronaridine offers a rapidly translational option for treating CE including PAIR. FUND: National Natural Science Foundation of China and International Cooperation Project of the Qinghai Science and Technology Department.


Subject(s)
Antimalarials/therapeutic use , Echinococcosis/drug therapy , Naphthyridines/therapeutic use , Topoisomerase Inhibitors/therapeutic use , Animals , Antimalarials/administration & dosage , Antimalarials/pharmacokinetics , Antimalarials/toxicity , DNA Fragmentation , DNA Topoisomerases, Type I/metabolism , Drug Repositioning , Echinococcus granulosus/drug effects , Echinococcus granulosus/pathogenicity , Female , Liver/metabolism , Liver/parasitology , Lung/metabolism , Lung/parasitology , Mice , Mice, Inbred BALB C , Naphthyridines/administration & dosage , Naphthyridines/pharmacokinetics , Naphthyridines/toxicity , Tissue Distribution , Topoisomerase Inhibitors/administration & dosage , Topoisomerase Inhibitors/pharmacokinetics , Topoisomerase Inhibitors/toxicity
7.
Toxicol Appl Pharmacol ; 391: 114915, 2020 03 15.
Article in English | MEDLINE | ID: mdl-32035082

ABSTRACT

Idiosyncratic drug-induced liver injury (IDILI) is a severe disease that cannot be detected during drug development. It has been shown that hepatotoxicity of some compounds associated with IDILI becomes apparent when these are combined in vivo and in vitro with LPS or TNF. Among these compounds trovafloxacin (TVX) induced apoptosis in the liver and increased pro-inflammatory cytokines in mice exposed to LPS/TNF. The hepatocyte survival and the cytokine release after TNF/LPS stimulation relies on a pulsatile activation of NF-κB. We set out to evaluate the dynamic activation of NF-κB in response to TVX + TNF or LPS models, both in mouse and human cells. Remarkably, TVX prolonged the first translocation of NF-κB induced by TNF both in vivo and in vitro. The prolonged p65 translocation caused by TVX was associated with an increased phosphorylation of IKK and MAPKs and accumulation of inhibitors of NF-κB such as IκBα and A20 in HepG2. Coherently, TVX suppressed further TNF-induced NF-κB translocations in HepG2 leading to decreased transcription of ICAM-1 and inhibitors of apoptosis. TVX prolonged LPS-induced NF-κB translocation in RAW264.7 macrophages increasing the secretion of TNF. In summary, this study presents new, relevant insights into the mechanism of TVX-induced liver injury underlining the resemblance between mouse and human models. In this study we convincingly show that regularly used toxicity models provide a coherent view of relevant pathways for IDILI. We propose that assessment of the kinetics of activation of NF-κB and MAPKs is an appropriate tool for the identification of hepatotoxic compounds during drug development.


Subject(s)
Anti-Bacterial Agents/toxicity , Chemical and Drug Induced Liver Injury/pathology , Fluoroquinolones/toxicity , Lipopolysaccharides/pharmacology , Naphthyridines/toxicity , Transcription Factor RelA/drug effects , Transcription Factor RelA/genetics , Translocation, Genetic/drug effects , Tumor Necrosis Factor-alpha/pharmacology , Animals , Apoptosis/drug effects , Chemical and Drug Induced Liver Injury/genetics , Cytokines/metabolism , Humans , I-kappa B Proteins/metabolism , Mice , Mitogen-Activated Protein Kinases/metabolism , RAW 264.7 Cells , Tumor Necrosis Factor-alpha/metabolism
8.
J Inorg Biochem ; 203: 110872, 2020 02.
Article in English | MEDLINE | ID: mdl-31689592

ABSTRACT

New polynuclear silver(I) complexes with 1,5-naphthyridine (1,5-naph), [Ag(NO3)(1,5-naph)]n (Ag1), [Ag(CF3COO)(1,5-naph)]n (Ag2) and [Ag(CF3SO3)(1,5-naph)]n (Ag3) were synthesized by the reaction of the corresponding silver(I) salt and 1,5-naph in ethanol at room temperature. These complexes were characterized by NMR, IR and UV-Vis spectroscopy, while their crystal structures were determined by single-crystal X-ray diffraction analysis. In all these complexes, 1,5-naph acts as a bridging ligand between two Ag(I) ions, while the remaining coordination sites are occupied by oxygen atom(s) of the corresponding anion. The antimicrobial efficiency of these silver(I) complexes was evaluated against the broad panel of Gram-positive and Gram-negative bacteria and fungi. The complexes showed good to moderate antibacterial activity with the minimal inhibitory concentration (MIC) values being in the range 2.5-100 µg/mL (6.5-333.3 µM), while their antifungal activity against the investigated Candida spp. was significantly higher (MIC = 0.78-6.25 µg/mL; 2.6-20.8 µM). Moreover, complexes Ag1 and Ag2 effectively inhibited C. albicans biofilms formation, while Ag1 was also shown to inhibit the formation of mixed C. albicans/Pseudomonas aeruginosa biofilms. Toxicological evaluations on zebrafish (Danio rerio) embryos revealed that all silver(I) complexes could be applied as antifungal agents, whereas Ag3 had the best therapeutic potential showing both the lowest MIC values against the tested Candida strains and the non-toxic in vivo response in the zebrafish embryos at these doses.


Subject(s)
Anti-Bacterial Agents/pharmacology , Antifungal Agents/pharmacology , Coordination Complexes/pharmacology , Naphthyridines/pharmacology , Animals , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/toxicity , Antifungal Agents/chemical synthesis , Antifungal Agents/toxicity , Bacteria/drug effects , Biofilms/drug effects , Candida albicans/drug effects , Candida albicans/physiology , Coordination Complexes/chemical synthesis , Coordination Complexes/toxicity , Microbial Sensitivity Tests , Naphthyridines/chemical synthesis , Naphthyridines/toxicity , Silver/chemistry , Zebrafish
9.
Eur J Med Chem ; 162: 176-193, 2019 Jan 15.
Article in English | MEDLINE | ID: mdl-30445266

ABSTRACT

[1,2,3]Triazolo[4,5-h][1,6]naphthyridines and [1,3]oxazolo[5,4-h][1,6]naphthyridines were synthesized with the aim to investigate their photocytotoxic activity. Upon irradiation, oxazolo-naphtapyridines induced light-dependent cell death at nanomolar/low micromolar concentrations (EC50 0.01-6.59 µM). The most photocytotoxic derivative showed very high selectivity and photocytotoxicity indexes (SI = 72-86, PTI>5000), along with a triplet excited state with exceptionally long lifetime (18.0 µs) and high molar absorptivity (29781 ±â€¯180 M-1cm-1 at λmax 315 nm). The light-induced production of ROS promptly induced an unquenchable apoptotic process selectively in tumor cells, with mitochondrial and lysosomal involvement. Altogether, these results demonstrate that the most active compound acts as a promising singlet oxygen sensitizer for biological applications.


Subject(s)
Cell Death/drug effects , Naphthyridines/pharmacology , Photochemotherapy/methods , Apoptosis , Cell Line, Tumor , Humans , Lysosomes/drug effects , Mitochondria/drug effects , Naphthyridines/chemical synthesis , Naphthyridines/toxicity , Photosensitizing Agents/chemistry , Photosensitizing Agents/pharmacology , Reactive Oxygen Species , Singlet Oxygen
10.
Toxicol Sci ; 167(2): 385-396, 2019 02 01.
Article in English | MEDLINE | ID: mdl-30247740

ABSTRACT

The fluoroquinolone trovafloxacin (TVX) is associated with a high risk of drug-induced liver injury (DILI). Although part of the liver damage by TVX+TNF relies on neutrophils, we have recently demonstrated that liver recruitment of monocytes and neutrophils is delayed by TVX. Here we show that the delayed leukocyte recruitment is caused by a combination of effects which are linked to the capacity of TVX to block the hemichannel pannexin 1. TVX inhibited find-me signal release in apoptotic HepG2 hepatocytes, decelerated freshly isolated human neutrophils toward IL-8 and f-MLF, and decreased the liver expression of ICAM-1. In blood of TVX+TNF-treated mice, we observed an accumulation of activated neutrophils despite an increased MIP-2 release by the liver. Depletion of monocytes and neutrophils caused increased serum concentrations of TNF, IL-6, and MIP-2 in TVX-treated mice as well as in mice treated with the fluoroquinolone levofloxacin, known to have a lower DILI-inducing profile. This supports the idea that early leukocyte recruitment regulates inflammation. In conclusion, disrupted regulation by leukocytes appears to constitute a fundamental step in the onset of TVX-induced liver injury, acting in concert with the capability of TVX to induce hepatocyte cell death. Interference of leukocyte-mediated regulation of inflammation represents a novel mechanism to explain the onset of DILI.


Subject(s)
Anti-Infective Agents/toxicity , Chemical and Drug Induced Liver Injury/immunology , Fluoroquinolones/toxicity , Naphthyridines/toxicity , Neutrophil Infiltration/drug effects , Neutrophils/drug effects , Nucleotides/metabolism , Tumor Necrosis Factor-alpha/toxicity , Animals , Chemical and Drug Induced Liver Injury/metabolism , Connexins/metabolism , Hep G2 Cells , Humans , Inflammation , Intercellular Adhesion Molecule-1/metabolism , Male , Mice, Inbred C57BL , Nerve Tissue Proteins/metabolism , Neutrophil Infiltration/immunology , Neutrophils/immunology
11.
J Appl Toxicol ; 38(5): 753-765, 2018 05.
Article in English | MEDLINE | ID: mdl-29377180

ABSTRACT

Idiosyncratic drug-induced liver injury (iDILI) has a poorly understood pathogenesis. However, iDILI is often associated with inflammatory stress signals in human patients as well as animal models. Tumor necrosis factor (TNF) and neutrophils play a key role in onset of trovafloxacin (TVX)-induced iDILI, but the exact role of neutrophils and other leukocytes remains to be defined. We therefore set out to study the kinetics of immunological changes during the development of TVX-induced iDILI in the established murine model of acute liver injury induced by administration of TVX and TNF. Initially, TNF stimulated the appearance of leukocytes, in particular neutrophils, into the liver of TVX-treated mice, but even more so in control mice treated with the non-DILI inducing analogue levofloxacin (LVX) or saline as vehicle (Veh). This difference was apparent at 2 hours after TNF administration, but at 4 hours, the relative neutrophil amounts were reduced again in Veh- and LVX-treated mice whereas the amounts in TVX-treated mice remained at the same increased level as at 2 hours. The influx of monocytes/macrophages, which was unaffected in Veh- and LVX-treated mice was markedly reduced or even absent in TVX-treated mice. Unlike controls, mice receiving TVX + TNF display severe hepatotoxicity with clear pathology and apoptosis, coagulated hepatic vessels and increased alanine aminotransferase levels and interleukin 6/10 ratios. Findings indicate that TVX delays the acute influx of neutrophils and monocytes/macrophages. Considering their known anti-inflammatory functions, the disruption of influx of these innate immune cells may hamper the resolution of initial cytotoxic effects of TVX and thus contribute to liver injury development.


Subject(s)
Chemical and Drug Induced Liver Injury/metabolism , Fluoroquinolones/toxicity , Monocytes/drug effects , Naphthyridines/toxicity , Neutrophils/drug effects , Tumor Necrosis Factor-alpha/toxicity , Alanine Transaminase/blood , Animals , Chemical and Drug Induced Liver Injury/immunology , Chemical and Drug Induced Liver Injury/pathology , Cytokines/blood , Flow Cytometry , Leukocytes/drug effects , Levofloxacin/pharmacology , Liver/drug effects , Liver/immunology , Liver/pathology , Male , Mice , Mice, Inbred C57BL
12.
Article in English | MEDLINE | ID: mdl-29208543

ABSTRACT

Dysfunction of copper homeostasis can lead to a host of disorders, which might be toxic sometimes. 4-Methoxy-5-hydroxy-canthin-6-one (CAN) is one of the major constituents from Picrasma quassioides and responsible for its therapeutic effects. In this work, we evaluated the toxic effect of CAN (7.5µM) on zebrafish embryos. CAN treatment decreased survival, delayed hatching time and induced malformations (loss of pigmentation, pericardial edema, as well as hematologic and neurologic abnormalities). Besides, exogenous copper supplementation rescued the pigmentation and cardiovascular defects in CAN-treated embryos. Further spectroscopic studies revealed a copper-chelating activity of CAN. Then its regulation on the expressions of copper homeostasis related genes also be analyzed. In addition, CAN lowered the total activity of SOD, elevated the ROS production and altered the oxidative related genes transcriptions, which led to oxidative stress. In conclusion, we demonstrated that CAN (7.5µM) might exert its toxic effects in zebrafish embryos by causing copper dyshomeostasis and oxidative stress. It will give insight into the risk assessment and prevention of CAN-mediated toxicity.


Subject(s)
Copper/metabolism , Embryo, Nonmammalian/drug effects , Homeostasis/drug effects , Indoles/toxicity , Naphthyridines/toxicity , Oxidative Stress/drug effects , Zebrafish/embryology , Animals , Lethal Dose 50 , Molecular Structure
13.
Birth Defects Res ; 109(14): 1075-1126, 2017 Aug 15.
Article in English | MEDLINE | ID: mdl-28646540

ABSTRACT

The World Health Organization currently recommends quinine+clindamycin for use against malaria in the first trimester. This may soon change to recommending artemisinin-based combination therapies (standard duration of dosing = 3 days). The non-artemisinin partner drugs include amodiaquine, lumefantrine, mefloquine, piperaquine, sulfadoxine+pyrimethamine, and pyronaridine. For quinine, clindamycin, and mefloquine and the combinations of sulfadoxine+pyrimethamine and artemether+lumefantrine, there are reports (including studies without internal comparison groups) that combined describe 304 to >1100 exposures of women in the first trimester for each drug with no conclusive evidence of adverse effects on pregnancy at therapeutic doses. This is despite the fact that all of these drugs or drug combinations caused embryo deaths and/or malformations in at least one animal species and all except lumefantrine had at least one exposure ratio <1. It now seems that these animal studies overestimated the risk of developmental toxicity in women with malaria. Three other non-artemisinins (amodiaquine, piperaquine, and pyronaridine) have few or no reported exposures in women in the first trimester and have exposure ratios ≤2 based on studies in pregnant rats and rabbits with dosing throughout organogenesis. However, none of these drugs caused embryo deaths or malformations in pregnant rats and rabbits with the exception of pyronaridine, which caused embryo deaths only at a dose that was excessively toxic to the mothers. Thus, for amodiaquine, piperaquine, and pyronaridine, the testing in animals did not reveal findings of concern and the exposure ratios were in the range of the other non-artemisinin antimalarials described above. Birth Defects Research 109:1075-1126, 2017. © 2017 The Authors. Birth Defects Research Published by Wiley Periodicals, Inc.


Subject(s)
Antimalarials/pharmacology , Antimalarials/toxicity , Amodiaquine/pharmacology , Amodiaquine/therapeutic use , Amodiaquine/toxicity , Animals , Antimalarials/administration & dosage , Antimalarials/metabolism , Artemisinins , Drug Combinations , Female , Humans , Malaria/drug therapy , Malaria, Falciparum/drug therapy , Naphthyridines/pharmacology , Naphthyridines/toxicity , Pregnancy , Pregnancy Trimester, First/drug effects , Pyrimethamine/therapeutic use , Pyrimethamine/toxicity , Quinine/administration & dosage , Quinolines/pharmacology , Quinolines/toxicity , Rabbits , Rats , Sulfadoxine/therapeutic use , Sulfadoxine/toxicity , Teratogens/analysis
14.
Cancer Lett ; 385: 168-178, 2017 01 28.
Article in English | MEDLINE | ID: mdl-27793693

ABSTRACT

A Pt(IV) prodrug, Cx-platin, containing CX-4945 (a CK2 inhibitor) as an axial ligand was designed and prepared by targeting CK2 to disrupt DNA damage response. In vitro study indicated that Cx-platin had superior cytotoxicity to cisplatin against a number of cancer cell lines with distinct CK2-expressed levels, caused CK2-overexpressed cancer cells death via suppressing CK2-mediated DNA damage repair and reversed cisplatin resistance. Mechanistic investigation suggested that the potent antitumor activity of Cx-platin resulted from its major suppression of CK2-phosphorylated MDC1 to combine FHA domain of aprataxin to DNA double strand breaks (DSBs) caused by improved cellular uptakes of Pt and ATM deactivation. Further in vivo tests exhibited that Cx-platin displayed high tumor inhibition rates, increased weight gain, and hardly toxicity effects in contrast to cisplatin.


Subject(s)
Casein Kinase II/antagonists & inhibitors , Cisplatin/pharmacology , DNA Damage , DNA Repair/drug effects , Naphthyridines/pharmacology , Neoplasms/drug therapy , Organoplatinum Compounds/pharmacology , Prodrugs/pharmacology , Protein Kinase Inhibitors/pharmacology , Adaptor Proteins, Signal Transducing , Animals , Antineoplastic Agents/toxicity , Ataxia Telangiectasia Mutated Proteins/metabolism , Casein Kinase II/metabolism , Cell Cycle Proteins , Cisplatin/pharmacokinetics , Cisplatin/toxicity , DNA-Binding Proteins/metabolism , Dose-Response Relationship, Drug , Drug Combinations , Drug Design , Female , HCT116 Cells , Hep G2 Cells , Humans , Inhibitory Concentration 50 , MCF-7 Cells , Male , Mice, Inbred BALB C , Mice, Nude , Molecular Targeted Therapy , Naphthyridines/pharmacokinetics , Naphthyridines/toxicity , Neoplasms/enzymology , Neoplasms/pathology , Nuclear Proteins/metabolism , Organoplatinum Compounds/pharmacokinetics , Organoplatinum Compounds/toxicity , Phenazines , Phosphorylation , Prodrugs/pharmacokinetics , Prodrugs/toxicity , Protein Interaction Domains and Motifs , Protein Kinase Inhibitors/pharmacokinetics , Protein Kinase Inhibitors/toxicity , Rats, Sprague-Dawley , Signal Transduction/drug effects , Time Factors , Trans-Activators/metabolism , Xenograft Model Antitumor Assays
15.
Toxicol Lett ; 263: 34-43, 2016 Nov 30.
Article in English | MEDLINE | ID: mdl-27713024

ABSTRACT

Trovafloxacin develops severe hepatotoxicity; however, the underlying mechanism of the trovafloxacin-induced liver injury has not been cleared. It has been shown that microRNAs (miRNAs) can be involved in the development of drug-induced liver injuries. We performed a miRNA microarray analysis to identify hepatic miRNAs that were induced or reduced by trovafloxacin in mice. It was demonstrated that miR-877-5p was the most increased miRNA in the mouse liver 24h after the trovafloxacin administration. To investigate the role of miR-877-5p in the liver, we established miR-877-5p-overexpressed HepG2 cells. Microarray analysis detected altered expressions in 2077 (>2-fold) and 1547 (<0.5-fold) genes in the miR-877-5p overexpressing cells compared to the mock cells. Especially, SLCO4C1, PEPCK, MT1M, HIST1H2BM, LGI1, and PLA2G2A were markedly increased or decreased in the miR-877-5p overexpressing cells. We conducted a correlation analysis between the expression levels of miR-877-5p and the six genes in eight miR-877-5p stably-expressed clones. It was shown that the PEPCK expression levels were correlated with miR-877-5p expression levels. PEPCK is associated with development of apoptotic cell death; therefore, the increased miR- 877-5p-induced PEPCK can be a trigger that is involved in the development of trovafloxacin-induced liver injury.


Subject(s)
Anti-Bacterial Agents/toxicity , Chemical and Drug Induced Liver Injury/genetics , Fluoroquinolones/toxicity , MicroRNAs/genetics , Naphthyridines/toxicity , Animals , Apoptosis/drug effects , Cell Line , Chemical and Drug Induced Liver Injury/metabolism , Gene Expression Regulation/drug effects , Humans , Male , Mice , Mice, Inbred C57BL , MicroRNAs/metabolism , Microarray Analysis , Phosphoenolpyruvate Carboxykinase (ATP)/biosynthesis , Phosphoenolpyruvate Carboxykinase (ATP)/genetics
16.
Biol Pharm Bull ; 39(10): 1604-1610, 2016.
Article in English | MEDLINE | ID: mdl-27725437

ABSTRACT

Trovafloxacin is an antibiotic that was withdrawn from the market relatively soon after its release due to the risk of hepatotoxicity. Trovafloxacin is mainly metabolized to its acyl-glucuronide by uridine 5'-diphosphate (UDP)-glucuronosyltransferase (UGT) 1A1. In this study, we examined whether the acyl-glucuronide is involved in the development of hepatotoxicity. A UGT1A1-induced cell model was developed and the toxicity of trovafloxacin acyl-glucuronide was evaluated. The UGT1A1-induced cell model was developed by treating HepG2 cells with chrysin for 48 h. Chemokine (C-X-C motif) ligand 2, a cytokine involved in drug-induced liver injury, was uniquely induced by trovafloxacin in the UGT1A1-induced HepG2 cells. Induction of UGT1A1 resulted in a decrease in cell viability. An in vivo animal study further demonstrated the importance of UGT1A1 in the trovafloxacin-induced liver toxicity. Although the complete mechanism of trovafloxacin-induced liver injury is still unknown, trovafloxacin acyl-glucuronide can be involved in the development of toxic reactions in vitro and in vivo.


Subject(s)
Chemical and Drug Induced Liver Injury/metabolism , Chemokine CXCL2/metabolism , Fluoroquinolones/toxicity , Glucuronides/metabolism , Naphthyridines/toxicity , Animals , Chemical and Drug Induced Liver Injury/genetics , Flavonoids/pharmacology , Gene Expression Regulation/drug effects , Glucuronosyltransferase/metabolism , Hep G2 Cells , Humans , Mice, Transgenic , RNA, Messenger/metabolism
17.
Arch Insect Biochem Physiol ; 93(4): 202-209, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27588824

ABSTRACT

Gemifloxacin mesylate (GEM) is a synthetic, fourth-generation fluoroquinolone antibacterial antibiotic that has a broad spectrum of activity against bacteria. GEM inhibits DNA synthesis by inhibiting DNA gyrase and topoisomerase IV activities. Recent research into insect nutrition and mass-rearing programs, in which antibiotics are incorporated into the culture media to maintain diet quality, raised a question of whether clinical antibiotics influence the health or biological performance of the insects that ingest these compounds. Because some antibiotics are pro-oxidant compounds, we addressed the question with experiments designed to assess the effects of GEM (mesylate salt) on oxidative stress indicators, using Galleria mellonella larvae. The insects were reared from first-instar larvae to adulthood on artificial diets amended with GEM at 0.001, 0.01, 0.1, or 1.0%. Feeding on the 1% diets led to significantly increased hemolymph contents of the lipid peroxidation product, malondialdehyde and protein oxidation products, protein carbonyl. All GEM concentrations led to increased hemolymph glutathione S-transferase activity. We inferred that although it was not directly lethal to G. mellonella larvae, dietary exposure to GEM exerts measurable oxidative damage, possibly on insects generally. Long-term, multigenerational effects remain unknown.


Subject(s)
Fluoroquinolones/toxicity , Glutathione Transferase/metabolism , Hemolymph/metabolism , Moths/drug effects , Naphthyridines/toxicity , Animals , Anti-Bacterial Agents/toxicity , Enzyme Activation/drug effects , Gemifloxacin , Hemolymph/drug effects , Hemolymph/enzymology , Larva/drug effects , Larva/growth & development , Larva/physiology , Moths/growth & development , Moths/physiology , Oxidation-Reduction/drug effects , Oxidative Stress/drug effects
18.
J Neurosci ; 36(22): 5933-45, 2016 06 01.
Article in English | MEDLINE | ID: mdl-27251616

ABSTRACT

UNLABELLED: Autophagy is an essential degradative pathway that maintains neuronal homeostasis and prevents axon degeneration. Initial observations suggest that autophagy is spatially regulated in neurons, but how autophagy is regulated in distinct neuronal compartments is unclear. Using live-cell imaging in mouse hippocampal neurons, we establish the compartment-specific mechanisms of constitutive autophagy under basal conditions, as well as in response to stress induced by nutrient deprivation. We find that at steady state, the cell soma contains populations of autophagosomes derived from distinct neuronal compartments and defined by differences in maturation state and dynamics. Axonal autophagosomes enter the soma and remain confined within the somatodendritic domain. This compartmentalization likely facilitates cargo degradation by enabling fusion with proteolytically active lysosomes enriched in the soma. In contrast, autophagosomes generated within the soma are less mobile and tend to cluster. Surprisingly, starvation did not induce autophagy in either the axonal or somatodendritic compartment. While starvation robustly decreased mTORC1 signaling in neurons, this decrease was not sufficient to activate autophagy. Furthermore, pharmacological inhibition of mammalian target of rapamycin with Torin1 also was not sufficient to markedly upregulate neuronal autophagy. These observations suggest that the primary physiological function of autophagy in neurons may not be to mobilize amino acids and other biosynthetic building blocks in response to starvation, in contrast to findings in other cell types. Rather, constitutive autophagy in neurons may function to maintain cellular homeostasis by balancing synthesis and degradation, especially within distal axonal processes far removed from the soma. SIGNIFICANCE STATEMENT: Autophagy is an essential homeostatic process in neurons, but neuron-specific mechanisms are poorly understood. Here, we compare autophagosome dynamics within neuronal compartments. Axonal autophagy is a vectorial process that delivers cargo from the distal axon to the soma. The soma, however, contains autophagosomes at different maturation states, including input received from the axon combined with locally generated autophagosomes. Once in the soma, autophagosomes are confined to the somatodendritic domain, facilitating cargo degradation and recycling of biosynthetic building blocks to primary sites of protein synthesis. Neuronal autophagy is not robustly upregulated in response to starvation or mammalian target of rapamycin inhibition, suggesting that constitutive autophagy in neurons maintains homeostasis by playing an integral role in regulating the quality of the neuronal proteome.


Subject(s)
Autophagy/physiology , Neurons/cytology , Neurons/physiology , Amino Acids/deficiency , Animals , Autophagy/drug effects , Axons/drug effects , Axons/physiology , Biological Transport/drug effects , Biological Transport/physiology , Cells, Cultured , Dendrites/drug effects , Dendrites/physiology , Embryo, Mammalian , Enzyme Inhibitors/toxicity , Female , Hippocampus/cytology , Homeostasis/drug effects , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Lysosomal Membrane Proteins/genetics , Lysosomal Membrane Proteins/metabolism , Macrolides/toxicity , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Naphthyridines/toxicity , Neurons/drug effects , Phagosomes/metabolism
19.
N Z Vet J ; 64(3): 179-81, 2016 May.
Article in English | MEDLINE | ID: mdl-26503546

ABSTRACT

CASE HISTORY: A one-year-old female goat presented with acute onset of recumbency, seizures and vocalisation approximately 5 hours after being given access to branch trimmings from a neighbour's garden. The plant from which the pruned branches came was subsequently identified as wintersweet (Chimonanthus praecox). Three other goats kept in the same paddock displayed similar clinical signs over a period of 4 hours following the initial presentation. CLINICAL FINDINGS: All four goats were ataxic, displayed tetanic seizures and were in lateral recumbency; they had dilated pupils and were hyperaesthetic, with elevated heart and respiratory rates. After symptomatic treatment, including sedation with diazepam, one of the three goats continued to deteriorate and was subjected to euthanasia. The remaining three goats recovered over 1-14 days with nursing care and physiotherapy. DIAGNOSIS: Toxicity due to ingestion of wintersweet, which contains the alkaloid calycanthine. CLINICAL RELEVANCE: Calycanthine is a central nervous system toxin, causing convulsions. Wintersweet shrubs are present in many New Zealand gardens. Practitioners should be aware that the seeds and flowers, and possibly the leaves, of this plant are highly toxic with signs of toxicity including ataxia, hyperaesthesia and seizures.


Subject(s)
Calycanthaceae/toxicity , Goat Diseases/chemically induced , Naphthyridines/toxicity , Plant Poisoning/veterinary , Animals , Central Nervous System Diseases/chemically induced , Central Nervous System Diseases/veterinary , Female , Goats , Male , Naphthyridines/chemistry , Plant Poisoning/etiology , Plants, Toxic/chemistry , Plants, Toxic/toxicity
20.
Bioorg Med Chem ; 24(1): 42-52, 2016 Jan 01.
Article in English | MEDLINE | ID: mdl-26678175

ABSTRACT

Recently numerous non-fluoroquinolone-based bacterial type II topoisomerase inhibitors from both the GyrA and GyrB classes have been reported as antibacterial agents. Inhibitors of the GyrA class include aminopiperidine-based novel bacterial type II topoisomerase inhibitors (NBTIs). However, inhibition of the cardiac ion channel remains a serious liability for the aminopiperidine based NBTIs. In this paper we replaced central aminopiperidine linker with piperazine moiety and tested for its biological activity. We developed a series of twenty four compounds with a piperazine linker 1-(2-(piperazin-1-yl)ethyl)-1,5-naphthyridin-2(1H)-one, by following a multistep protocol. Among them compound 4-(2-(7-methoxy-2-oxo-1,5-naphthyridin-1(2H)-yl)ethyl)-N-(4-nitrophenyl)piperazine-1-carboxamide (11) was the most promising inhibitor with Mycobacterium tuberculosis (MTB) DNA gyrase enzyme supercoiling IC50 of 0.29±0.22µM, with a good MTB MIC of 3.45µM. These kind of compounds retains good potency and showed reduced cardiotoxicity compared to aminopiperidines.


Subject(s)
Antitubercular Agents/pharmacology , Cardiotoxicity/drug therapy , Mycobacterium tuberculosis/enzymology , Naphthyridines/pharmacology , Piperazines/pharmacology , Topoisomerase II Inhibitors/pharmacology , Animals , Antitubercular Agents/chemical synthesis , Antitubercular Agents/toxicity , Atrioventricular Block/drug therapy , DNA Gyrase/metabolism , Enzyme Assays , Ether-A-Go-Go Potassium Channels/antagonists & inhibitors , Heart Rate/drug effects , Naphthyridines/chemical synthesis , Naphthyridines/toxicity , Novobiocin/pharmacology , Piperazines/chemical synthesis , Piperazines/toxicity , Terfenadine/pharmacology , Topoisomerase II Inhibitors/chemical synthesis , Topoisomerase II Inhibitors/toxicity , Zebrafish , Zebrafish Proteins/antagonists & inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL
...