Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 79
1.
Biochem Biophys Res Commun ; 584: 46-52, 2021 12 20.
Article En | MEDLINE | ID: mdl-34768081

Efavirenz (EFV) is a non-nucleoside reverse transcriptase inhibitor (NNRTI), which is widely used for anti-HIV-1. Evidences revealed that several central nervous system side effects could be observed in mice and patients with administration of EFV. However, the detailed mechanisms are still unknown. In this study, we investigated the effects of long-term EFV treatment on cognitive functions and the potential underlying mechanisms in mice. We maintained C57BL/6 mice aged 2 months with treatment containing 40 or 80 mg/kg/day EFV for 5 months, while control group treated with saline. The cognitive functions were evaluated by novel object recognition test, Barnes maze test and Morris water maze. The results showed significant short-term memory impairment in 40 and 80 mg/kg groups, and notable spatial learning and memory impairments in 80 mg/kg group, without any spontaneous activity alteration. Moreover, EFV induced impairments in dendritic integrity and synaptic plasticity in hippocampus. Furthermore, Significant increases were observed in the expression levels of pro-IL-1ß, a similar tendency of TNF-α and phosphorylation of p65 of the 80 mg/kg group compared with control group. These results imply that long-term EFV treatment causes synaptic dysfunction resulting in cognitive deficits, which might be induced by the enhanced pro-inflammatory cytokines IL-1ß and TNF-α via activating NF-κB pathway.


Alkynes/toxicity , Benzoxazines/toxicity , Cognition/drug effects , Cognitive Dysfunction/physiopathology , Cyclopropanes/toxicity , Memory Disorders/physiopathology , Neuroinflammatory Diseases/physiopathology , Animals , Cognition/physiology , Cognitive Dysfunction/chemically induced , Hippocampus/drug effects , Hippocampus/metabolism , Humans , Maze Learning/drug effects , Maze Learning/physiology , Memory Disorders/chemically induced , Mice, Inbred C57BL , Neuroinflammatory Diseases/chemically induced , Reverse Transcriptase Inhibitors/toxicity , Spatial Learning/drug effects , Spatial Learning/physiology , Synapsins/metabolism , Synaptophysin/metabolism , Synaptotagmin I/metabolism , Time Factors
2.
ACS Appl Mater Interfaces ; 13(34): 41012-41020, 2021 Sep 01.
Article En | MEDLINE | ID: mdl-34410119

Photodynamic therapy (PDT) is a potential approach to resolve antibiotic resistance, and phenylene/thiophene-ethynylene oligomers have been widely studied as effective antibacterial reagents. Oligomers with thiophene moieties usually exhibit good antibacterial activity under light irradiation and dark conditions. In the previous study, we verified that neutral oligo-p-phenylene-ethynylenes (OPEs) exhibit better antibacterial activity than the corresponding cationic ones; however, whether this regular pattern also operates in other kinds of oligomers such as oligo-thiophene-ethynylene (OTE) is unknown. Also, the antibacterial activity comparison of OTEs bearing cyclic and acyclic amino groups will offer useful information to further understand the role of amino groups in the antibacterial process and guide the antibacterial reagent design as amino groups affect the antibacterial activity a lot. We synthesized four OTEs bearing neutral or cationic, cyclic, or acyclic amino groups and studied their antibacterial activity in detail. The experimental results indicated that the OTEs exhibited better antibacterial activity than the OPEs, the neutral OTEs exhibited better antibacterial activity in most cases, and OTEs bearing cyclic amino groups exhibited better antibacterial activity than those bearing acyclic ones in most cases. This study provides useful guidelines for further antibacterial reagent design and investigations.


Alkynes/pharmacology , Anti-Bacterial Agents/pharmacology , Thiophenes/pharmacology , Alkynes/chemistry , Alkynes/radiation effects , Alkynes/toxicity , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/radiation effects , Anti-Bacterial Agents/toxicity , Cell Line , Escherichia coli/drug effects , Humans , Hydrophobic and Hydrophilic Interactions , Light , Microbial Sensitivity Tests , Staphylococcus aureus/drug effects , Thiophenes/chemistry , Thiophenes/radiation effects , Thiophenes/toxicity
3.
Ocul Immunol Inflamm ; 28(7): 1076-1084, 2020 Oct 02.
Article En | MEDLINE | ID: mdl-32870052

Ocular lesions in patients with human immunodeficiency virus (HIV) are commonly due to underlying opportunistic infections. With highly active antiretroviral therapy (HAART), infectious lesions have reduced and noninfectious ocular manifestations including drug-related side effects have been noted. While retinal toxicity has been noted with few other HAART drugs, there are not many on the same with Efavirenz usage. We report a series of five patients with possible efavirenz-related retinal toxicity, visual function abnormalities, and its management. Efavirenz was replaced with alternate anti-retroviral drug. Reversal of ocular side effects were noted subjectively in the form of symptom amelioration of the patients. Objectively, it could be documented with electroretinogram changes and other visual function tests reverting back to normal after change in HAART regime. Early identification of this uncommon side effect in select patients can prevent irreversible vision loss due to efavirenz-associated retinal toxicity.


Alkynes/toxicity , Benzoxazines/toxicity , Cyclopropanes/toxicity , Night Blindness/chemically induced , Retina/drug effects , Retinal Diseases/chemically induced , Reverse Transcriptase Inhibitors/toxicity , Adult , Antiretroviral Therapy, Highly Active , Color Vision/physiology , Electroretinography , Evoked Potentials, Visual , Female , HIV Infections/drug therapy , Humans , Male , Middle Aged , Night Blindness/diagnostic imaging , Night Blindness/physiopathology , Ophthalmoscopy , Retina/physiopathology , Retinal Diseases/diagnostic imaging , Retinal Diseases/physiopathology , Slit Lamp Microscopy , Tomography, Optical Coherence , Visual Acuity/physiology
4.
J Neurovirol ; 26(5): 642-651, 2020 10.
Article En | MEDLINE | ID: mdl-32737860

Highly active antiretroviral treatment has led to unprecedented efficacy and tolerability in people living with HIV. This effect was also observed in the central nervous system with the nowadays uncommon observation of dementias; yet in more recent works milder forms are still reported in 20-30% of optimally treated individuals. The idea of a subclinical neuronal toxicity induced by antiretrovirals has been proposed and was somehow supported by the late-emerging effects associated with efavirenz use. In this manuscript we are reviewing all the potential mechanisms by which antiretroviral drugs have been associated with in vitro, ex vivo, or in vivo toxicity to cells pertaining to the central nervous system (neurons, astrocytes, oligodendrocytes, and endothelial cells). These include direct or indirect effects and pathological pathways such as amyloid deposition, damage to small cerebral vessels, and impairment in neurotransmission. The aim of this review is therefore to provide a detailed description of the available literature in order to guide further clinical research for improving patients' neurocognition and quality of life.


Alkynes/toxicity , Anti-HIV Agents/toxicity , Benzoxazines/toxicity , Central Nervous System/drug effects , Cognitive Dysfunction/chemically induced , Cyclopropanes/toxicity , HIV Infections/drug therapy , Neurons/drug effects , Antiretroviral Therapy, Highly Active/methods , Astrocytes/drug effects , Astrocytes/pathology , Astrocytes/virology , Atazanavir Sulfate/toxicity , Central Nervous System/pathology , Central Nervous System/virology , Cognitive Dysfunction/pathology , Cognitive Dysfunction/prevention & control , Cognitive Dysfunction/virology , Dideoxynucleosides/toxicity , Endothelial Cells/drug effects , Endothelial Cells/pathology , Endothelial Cells/virology , HIV Infections/pathology , HIV Infections/virology , Humans , Neurons/pathology , Neurons/virology , Nevirapine/toxicity , Nitriles/toxicity , Oligodendroglia/drug effects , Oligodendroglia/pathology , Oligodendroglia/virology , Pyrimidines/toxicity
5.
New Dir Child Adolesc Dev ; 2020(171): 107-133, 2020 May.
Article En | MEDLINE | ID: mdl-32657046

Efavirenz (EFV) is a well-known, effective anti-retroviral drug long used in first-line treatment for children and adults with HIV and HIV/AIDS. Due to its narrow window of effective concentrations, between 1 and 4 µg/mL, and neurological side effects at supratherapeutic levels, several investigations into the pharmacokinetics of the drug and its genetic underpinnings have been carried out, primarily with adult samples. A number of studies, however, have examined the genetic influences on the metabolism of EFV in children. Their primary goal has been to shed light on issues of appropriate pediatric dosing, as well as the manifestation of neurotoxic effects of EFV in some children. Although EFV is currently being phased out of use for the treatment of both adults and children, we share this line of research to highlight an important aspect of medical treatment that is relevant to understanding the development of children diagnosed with HIV.


Alkynes , Anti-HIV Agents , Benzoxazines , Child Development/drug effects , Cyclopropanes , Cytochrome P-450 CYP2B6/genetics , HIV Infections/drug therapy , Pharmacogenetics , Reverse Transcriptase Inhibitors , Alkynes/administration & dosage , Alkynes/metabolism , Alkynes/toxicity , Anti-HIV Agents/administration & dosage , Anti-HIV Agents/metabolism , Anti-HIV Agents/toxicity , Benzoxazines/administration & dosage , Benzoxazines/metabolism , Benzoxazines/toxicity , Child , Child, Preschool , Cyclopropanes/administration & dosage , Cyclopropanes/metabolism , Cyclopropanes/toxicity , Humans , Reverse Transcriptase Inhibitors/administration & dosage , Reverse Transcriptase Inhibitors/metabolism , Reverse Transcriptase Inhibitors/toxicity
6.
Biochem Pharmacol ; 178: 114050, 2020 08.
Article En | MEDLINE | ID: mdl-32446887

Efavirenz (EFV) is used for antiretroviral treatment of HIV infection, and successfully inhibits viral replication and mother-to-child transmission of HIV during pregnancy and childbirth. Unfortunately, the drug induces neuropsychiatric symptoms such as anxiety and depressed mood and potentially affects cognitive performance. EFV acts on, among others, the serotonin transporter and serotonin receptors that are expressed in the developing brain. Yet, how perinatal EFV exposure affects brain cytoarchitecture remains unclear. Here, we exposed pregnant and lactating rats to EFV, and examined in the medial prefrontal cortex (mPFC) of their adult offspring the effects of the maternal EFV exposure on cortical architecture. We observed a significant decrease in the number of cells, mainly mature neurons, in the infra/prelimbic and cingulate cortices of adult offspring. Next, we found an altered cortical cytoarchitecture characterized by a significant reduction in deep- and superficial-layer cells. This was accompanied by a sharp increase in programmed cell death, as we identified a significantly higher number of cleaved Caspase-3-positive cells. Finally, the serotonergic and dopaminergic innervation of the mPFC subdomains was increased. Thus, the perinatal exposure to EFV provoked in the mPFC of adult offspring cell death, significant changes in cytoarchitecture, and disturbances in serotonergic and dopaminergic innervation. Our results are important in the light of EFV treatment of HIV-positive pregnant women, and its effect on brain development and cognitive behavior.


Alkynes/toxicity , Benzoxazines/toxicity , Cyclopropanes/toxicity , Prefrontal Cortex/drug effects , Prefrontal Cortex/pathology , Prenatal Exposure Delayed Effects/chemically induced , Prenatal Exposure Delayed Effects/pathology , Reverse Transcriptase Inhibitors/toxicity , Animals , Animals, Newborn , Anti-HIV Agents/toxicity , Female , Male , Prefrontal Cortex/growth & development , Pregnancy , Rats , Rats, Wistar
7.
Toxins (Basel) ; 12(4)2020 04 09.
Article En | MEDLINE | ID: mdl-32283677

There are several reports indicating that the roots of the Carlina acaulis L. used to be commonly applied as a treatment measure in skin diseases and as an antiparasitic agent, starting from antiquity to the 19th century; however, nowadays, it has lost its importance. Currently, numerous studies are being conducted assessing the possibility of reintroducing C. acaulis-derived extracts to phytotherapy. Determining the safety profile of the main constituents of the plant material is crucial for achieving this goal. Here, we aimed to determine the toxicity profile of carlina oxide, one of the most abundant components of the C. acaulis root extract. We obtained the carlina oxide by distillation of C. acaulis roots in the Deryng apparatus. The purity of the standard was evaluated using GC-MS, and the identity was confirmed by IR, Raman, and NMR spectroscopy. In vitro cytotoxicity was assessed using a panel of human cell lines of skin origin, including BJ normal fibroblasts and UACC-903, UACC-647, and C32 melanoma cells. This was accompanied by an in vivo zebrafish acute toxicity test (ZFET). In vitro studies showed a toxic effect of carlina oxide, as demonstrated by an induction of apoptosis and necrosis in both normal and melanoma cells. Decreased expression of AKT kinase and extracellular signal-regulated kinase 1/2 (ERK1/2) was noted in the UACC-647 melanoma cell line. It was also observed that carlina oxide modified the expression of programmed cell death-ligand 1 (PD-L1) in tested cell lines. Carlina oxide exhibited high in vivo toxicity, with LC50 = 10.13 µg/mL upon the 96 h of exposure in the ZFET test. Here, we demonstrate that carlina oxide displays toxic effects to cells in culture and to living organisms. The data indicate that C. acaulis-based extracts considered for therapeutic use should be completely deprived of carlina oxide.


Alkynes/toxicity , Asteraceae/toxicity , Furans/toxicity , Oils, Volatile/toxicity , Plant Oils/toxicity , Plant Roots/toxicity , Zebrafish/embryology , Alkynes/isolation & purification , Animals , Apoptosis/drug effects , Asteraceae/chemistry , Cell Line, Tumor , Cell Survival/drug effects , Embryo, Nonmammalian/abnormalities , Embryo, Nonmammalian/drug effects , Embryonic Development/drug effects , Furans/isolation & purification , Humans , Lethal Dose 50 , Necrosis , Oils, Volatile/isolation & purification , Plant Oils/isolation & purification , Plant Roots/chemistry , Risk Assessment , Toxicity Tests, Acute
8.
Eur J Med Chem ; 177: 221-234, 2019 Sep 01.
Article En | MEDLINE | ID: mdl-31151057

Alzheimer's disease (AD) is a multifactorial neurological disorder involving complex pathogenesis. Single target directed drugs proved ineffective and since last few years' different pharmacological strategies including multi-targeting agents are being explored for the effective drug development for AD. A total of 19 dipropargyl substituted diphenylpyrimidines have been synthesized and evaluated for the monoamine oxidase (MAO) and acetylcholinesterase (AChE) inhibition potential. All the compounds were found to be selective and reversible inhibitors of MAO-B isoform. These compounds also displayed good AChE inhibition potential with IC50 values in low micromolar range. AVB4 was found to be the most potent MAO-B inhibitor with IC50 value of 1.49 ±â€¯0.09 µM and AVB1 was found to be the most potent AChE inhibitor with IC50 value of 1.35 ±â€¯0.03 µM. In the ROS protection inhibition studies, AVB1 and AVB4 displayed weak but interesting activity in SH-SY5Y cells. In the cytotoxicity studies involving SH-SY5Y cells, both AVB1 and AVB4 were found to be non-toxic to the tissue cells. In the molecular dynamic simulation studies of 30 ns, the potent compounds were found to be quite stable in the active site of MAO-B and AChE. The results suggested that AVB1 and AVB4 are promising dual inhibitors and have the potential to be developed as anti-Alzheimer's drug.


Alkynes/pharmacology , Cholinesterase Inhibitors/pharmacology , Monoamine Oxidase Inhibitors/pharmacology , Pyrimidines/pharmacology , Acetylcholinesterase/chemistry , Alkynes/chemical synthesis , Alkynes/chemistry , Alkynes/toxicity , Alzheimer Disease/drug therapy , Catalytic Domain , Cell Line, Tumor , Cholinesterase Inhibitors/chemical synthesis , Cholinesterase Inhibitors/chemistry , Cholinesterase Inhibitors/toxicity , Drug Design , Humans , Kinetics , Molecular Docking Simulation , Molecular Dynamics Simulation , Molecular Structure , Monoamine Oxidase/chemistry , Monoamine Oxidase Inhibitors/chemical synthesis , Monoamine Oxidase Inhibitors/chemistry , Monoamine Oxidase Inhibitors/toxicity , Pyrimidines/chemical synthesis , Pyrimidines/chemistry , Pyrimidines/toxicity , Structure-Activity Relationship
9.
Chembiochem ; 19(23): 2438-2442, 2018 12 04.
Article En | MEDLINE | ID: mdl-30303294

The first biologically relevant clickable probe related to the antitumor marine lipid jaspine B is reported. The concise synthetic route to both enantiomers relied on the supercritical fluid chromatography (SFC) enantiomeric resolution of racemic materials. The eutomeric dextrogyre derivative represents the first jaspine B analogue with enhanced cytotoxicity with IC50 down to 30 nm. These enantiomeric probes revealed a chiralitydependent cytoplasmic imaging of U2OS cancer cells by in situ click labeling.


Alkynes/chemistry , Antineoplastic Agents/chemistry , Fluorescent Dyes/chemistry , Molecular Probes/chemistry , Sphingosine/analogs & derivatives , Alkynes/chemical synthesis , Alkynes/toxicity , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/toxicity , Cell Line, Tumor , Click Chemistry , Fluorescent Dyes/chemical synthesis , Fluorescent Dyes/toxicity , Humans , Molecular Probes/chemical synthesis , Molecular Probes/toxicity , Sphingosine/chemical synthesis , Sphingosine/toxicity , Stereoisomerism
10.
Analyst ; 143(20): 4844-4848, 2018 Oct 08.
Article En | MEDLINE | ID: mdl-30246812

Stimulated Raman Scattering (SRS) coupled with alkyne tags has been an emerging imaging technique to visualize small-molecule species with high sensitivity and specificity. Here we describe the development of a ratiometric Raman probe for visualizing hydrogen sulfide (H2S) species in living cells as the first alkyne-based sensor for SRS microscopy. This probe uses an azide unit as a selective reactive site, and it targets mitochondria with high specificity. The SRS ratiometric images show a strong response to H2S level changes in living cells.


Alkynes/chemistry , Azides/chemistry , Hydrogen Sulfide/analysis , Luminescent Agents/chemistry , Mitochondria/metabolism , Spectrum Analysis, Raman/methods , Alkynes/chemical synthesis , Alkynes/toxicity , Animals , Azides/chemical synthesis , Azides/toxicity , COS Cells , Chlorocebus aethiops , HeLa Cells , Humans , Hydrogen Sulfide/chemistry , Hydrogen Sulfide/metabolism , Limit of Detection , Luminescent Agents/chemical synthesis , Luminescent Agents/toxicity
11.
ChemMedChem ; 13(7): 705-712, 2018 04 06.
Article En | MEDLINE | ID: mdl-29534334

Monoamine oxidase B (MAO-B) inhibitors are potential drug candidates for the treatment of various neurological disorders including Parkinson's disease. A total of 20 new propargyl-containing 2,4,6-trisubstituted pyrimidine derivatives were synthesized and screened for MAO inhibition using Amplex Red assays. All the synthesized compounds were found to be reversible and selective inhibitors of the MAO-B isoform at sub-micromolar concentrations. MVB3 was the most potent MAO-B inhibitor with an IC50 value of 0.38±0.02 µµ, whereas MVB6 (IC50 =0.51±0.04 µµ) and MVB16 (IC50 =0.48±0.06 µµ) were the most selective for MAO-B with a selectivity index of more than 100-fold. In cytotoxic studies, these compounds were found to be nontoxic to human neuroblastoma SH-SY5Y cells at concentrations of 25 µm. MVB6 was found to decrease the intracellular level of reactive oxygen species to 68 % at 10 µm concentration, whereas other compounds did not produce significant changes in reactive oxygen species levels. In molecular modeling studies, MVB3 displayed strong binding affinity for the MAO-B isoform with a dock score of -10.45, in agreement with the observed activity. All the compounds fitted well in the hydrophobic cavity of MAO-B. Thus, propargyl-substituted pyrimidine derivatives can be promising leads in the development of potent, selective and reversible MAO-B inhibitors for the treatment of Parkinson's disease.


Alkynes/pharmacology , Antiparkinson Agents/pharmacology , Pyrimidines/pharmacology , Alkynes/chemical synthesis , Alkynes/chemistry , Alkynes/toxicity , Antiparkinson Agents/chemical synthesis , Antiparkinson Agents/chemistry , Antiparkinson Agents/toxicity , Binding Sites , Cell Line, Tumor , Humans , Hydrophobic and Hydrophilic Interactions , Models, Molecular , Molecular Structure , Monoamine Oxidase/metabolism , Monoamine Oxidase Inhibitors/chemical synthesis , Monoamine Oxidase Inhibitors/chemistry , Monoamine Oxidase Inhibitors/pharmacology , Monoamine Oxidase Inhibitors/toxicity , Pyrimidines/chemical synthesis , Pyrimidines/chemistry , Pyrimidines/toxicity , Structure-Activity Relationship
12.
Biointerphases ; 12(2): 02C403, 2017 04 20.
Article En | MEDLINE | ID: mdl-28427270

Each year, the United States spends about $20 billion to treat people who have been infected with antibiotic resistant bacteria. Even so, the development of new antibiotics has slowed considerably since the mid-20th century. As a result, researchers are looking into developing synthetic compounds and materials with antimicrobial activities such as those made by the Schanze and Whitten groups [ACS Appl. Mater. Interfaces 3, 2820 (2011)]. Previously, they have demonstrated that poly(phenylene ethynylene) (PPE) based electrolytes and oligomeric end-only phenylene ethynylene (EO-OPE) based electrolytes possess strong biocidal activity. However, before the PPE and OPE can be used with humans, skin irritation tests are required to ensure their safety. In this work, in vitro skin assays are used to predict in vivo irritation. Tissues were conditioned for 24 h, exposed to test substances for 1 h, and then tested for viability using colorimetric and cytokine assays. Concentrations up to 50 µg/ml were tested. Viability assays and cytokine (IL-1α) assays demonstrated that the two polymers, three symmetric oligomers, and three "end only" oligomers were nonirritants. In addition, electrospun mats consisting of several promising compounds, including poly(caprolactone), were evaluated. Therefore, all test substances are conservatively classified as nonirritants after a 1 h exposure time period.


Alkynes/chemistry , Anti-Infective Agents/chemistry , Electrolytes/chemistry , Ethers/chemistry , Alkynes/toxicity , Cell Culture Techniques , Cell Line , Cell Survival/drug effects , Electrolytes/toxicity , Enzyme-Linked Immunosorbent Assay , Ethers/toxicity , Humans , Interleukin-1alpha/analysis , Keratinocytes/cytology , Keratinocytes/drug effects , Keratinocytes/metabolism
13.
Chembiochem ; 18(1): 81-84, 2017 Jan 03.
Article En | MEDLINE | ID: mdl-27813293

Cysteine residues play critical roles in protein function and are susceptible to numerous post-translational modifications (PTMs) that serve to modulate the activity and localization of diverse proteins. Many of these PTMs are highly transient and labile, thus necessitating methods to study these modifications directly within the context of living cells. We previously reported a caged electrophilic probe, CBK1, that can be activated by UV for temporally controlled covalent modification of cysteine residues in living cells. To improve upon the number of cysteine residues identified in cellular cysteine-profiling studies, the reactivity and uncaging efficiency of a panel of caged electrophiles were explored. We identified an optimized caged electrophilic probe, CIK4, that affords significantly improved coverage of cellular cysteine residues. The broader proteome coverage afforded by CIK4 renders it a useful tool for the biological investigation of cysteine-reactivity changes and PTMs directly within living cells and highlights design elements that are critical to optimizing photoactivatable chemical probes for cellular labeling.


Alkynes/chemistry , Cysteine/chemistry , Dioxoles/chemistry , Alkynes/toxicity , Cell Line, Tumor , Cell Survival/drug effects , Chromatography, High Pressure Liquid , Cysteine/metabolism , Dioxoles/toxicity , Electrophoresis, Polyacrylamide Gel , HeLa Cells , Humans , Ketones/chemistry , Ketones/toxicity , Protein Processing, Post-Translational , Proteins/chemistry , Proteins/metabolism , Tandem Mass Spectrometry , Ultraviolet Rays
14.
Chemistry ; 21(30): 10729-40, 2015 Jul 20.
Article En | MEDLINE | ID: mdl-26096074

The synthesis, characterization, photophysics, lipophilicity, and cellular properties of new phosphorescent ruthenium(II) polypyridine complexes functionalized with a dibenzocyclooctyne (DIBO) or amine moiety [Ru(N^N)2 (L)](PF6 )2 are reported (L=4-(13-N-(3,4:7,8-dibenzocyclooctyne-5-oxycarbonyl) amino-4,7,10-trioxa-tridecanyl-aminocarbonyl-oxy-methyl)-4'-methyl-2,2'-bipyridine bpy-DIBO, N^N=2,2'-bipyridine bpy (1 a), 1,10-phenanthroline phen (2 a); L=4-(13-amino-4,7,10-trioxa-tridecanylaminocarbonyl-oxy-methyl)-4'-methyl-2,2'-bipyridine bpy-NH2 , N^N=bpy (1 b), phen (2 b)). The strain-promoted alkyne-azide cycloaddition (SPAAC) reaction of the DIBO complexes 1 a and 2 a with benzyl azide were studied. Also, the DIBO complexes 1 a and 2 a can selectively label N-azidoglycans located on the surface of CHO-K1 and A549 cells that were pretreated with 1,3,4,6-tetra-O-acetyl-N-azidoacetyl-D-mannosamine (Ac4 ManNAz). Additionally, the intracellular trafficking and localization of these biomolecules were monitored using laser-scanning confocal microscopy. Interestingly, the biolabeling and cellular uptake efficiency of the DIBO complexes 1 a and 2 a were cell-line dependent, as revealed by flow cytometry and ICP-MS. Furthermore, the complexes showed good biocompatibility toward the Ac4 ManNAz-pretreated cells in the dark, but exhibited photoinduced cytotoxicity due to the generation of singlet oxygen.


2,2'-Dipyridyl/analogs & derivatives , Alkynes/chemistry , Benzene Derivatives/chemistry , Cell Membrane/chemistry , Luminescent Agents/chemistry , Organometallic Compounds/chemistry , Polysaccharides/analysis , 2,2'-Dipyridyl/chemistry , 2,2'-Dipyridyl/toxicity , Alkynes/toxicity , Animals , Benzene Derivatives/toxicity , CHO Cells , Cell Line , Cricetulus , Humans , Luminescent Agents/toxicity , Microscopy, Confocal , Optical Imaging , Organometallic Compounds/toxicity , Staining and Labeling
15.
Mol Med Rep ; 10(1): 341-6, 2014 Jul.
Article En | MEDLINE | ID: mdl-24756435

Sodium hydrosulfide (NaHS) is an exogenous hydrogen sulfide (H2S)-releasing molecule and has antitumor potential against a wide variety of human cancer types. The effect of exogenous H2S on the invasion of breast cancer and the possible underlying mechanisms remain unknown. The present study aimed to investigate the in vitro effects of H2S on transforming growth factor-ß1 (TGF-ß1)-induced human breast cancer cells and the associated mechanisms. MCF-7 cells were incubated with TGF-ß1 to induce epithelial-mesenchymal transition (EMT) and an MTT assay was performed to detect cell viability. Flow cytometry, using propidium iodide (PI) staining, was used to determine the stages of the cell cycle. Apoptosis was detected with Annexin V-fluorescein isothiocyanate and PI double staining. Western blotting was performed to detect the protein expression of cystathionine γ-lyase (CSE, an endogenous H2S producer), phospho-p38 (a signaling protein associated with apoptosis), and SNAI1 (Snail, associated with the induction of EMT). A Boyden chamber invasion assay was performed to detect tumor invasion. The results demonstrated that when NaHS was administered to TGF-ß1-treated MCF-7 cells, the cells exhibited decreased proliferation, G0/G1 phase cell cycle arrest and increased apoptosis. NaHS treatment following TGF-ß1 administration also resulted in decreased cell invasion and decreased EMT, which was indicated by decreased Snail protein expression. In addition, incubation with NaHS increased endogenous CSE protein expression and decreased p38 mitogen-activated protein kinase phosphorylation in MCF-7 cells stimulated by TGF-ß1. Furthermore, the inhibition of endogenous CSE by DL-propargylglycine increased EMT in the MCF-7 cells treated with NaHS and TGF-ß1. In conclusion, the present study provides insights into a novel anticancer effect of H2S on breast cancer cells through activation of the CSE/H2S pathway and decreased expression of phospho-p38.


Epithelial-Mesenchymal Transition/drug effects , Hydrogen Sulfide/toxicity , p38 Mitogen-Activated Protein Kinases/metabolism , Alkynes/toxicity , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Movement/drug effects , Cell Survival/drug effects , Cystathionine gamma-Lyase/metabolism , Female , G1 Phase Cell Cycle Checkpoints/drug effects , Glycine/analogs & derivatives , Glycine/toxicity , Humans , MCF-7 Cells , Phosphorylation/drug effects , Snail Family Transcription Factors , Transcription Factors/metabolism , Transforming Growth Factor beta1/pharmacology
16.
Toxicology ; 314(1): 100-11, 2013 Dec 06.
Article En | MEDLINE | ID: mdl-24035744

Propargyl alcohol (PA) is a high production volume chemical used in synthesis of many industrial chemicals and agricultural products. Despite the potential for prolonged or accidental exposure to PA in industrial settings, the toxicity potential of PA was not well characterized. To address the knowledge gaps relevant to the toxicity profile of PA, the National Toxicology Program (NTP) conducted 2-week, 14-week and 2-year studies in male and female F344/N rats and B6C3F1/N mice. For the 2-week inhalation study, the rats and mice were exposed to 0, 31.3, 62.5, 125, 250 or 500ppm. Significant mortality was observed in both rats and mice exposed to ≥125ppm of PA. The major target organ of toxicity in both mice and rats was the liver with exposure-related histopathological changes (250 and 500ppm). Based on the decreased survival in the 2-week study, the rats and mice were exposed to 0, 4, 8, 16, 32 or 64ppm of PA in the 14-week study. No treatment-related mortality was observed. Mean body weights of male (≥8ppm) and female mice (32 and 64ppm) were significantly decreased (7-16%). Histopathological changes were noted in the nasal cavity, and included suppurative inflammation, squamous metaplasia, hyaline droplet accumulation, olfactory epithelium atrophy, and necrosis. In the 2-year inhalation studies, the rats were exposed to 0, 16, 32 and 64ppm of PA and the mice were exposed to 0, 8, 16 and 32ppm of PA. Survival of male rats was significantly reduced (32 and 64ppm). Mean body weights of 64ppm male rats were significantly decreased relative to the controls. Both mice and rats showed a spectrum of non-neoplastic changes in the nose. Increased neoplastic incidences of nasal respiratory/transitional epithelial adenoma were observed in both rats and mice. The incidence of mononuclear cell leukemia was significantly increased in male rats and was considered to be treatment-related. In conclusion, the key findings from this study indicated that the nose was the primary target organ of toxicity for PA. Long term inhalation exposure to PA led to nonneoplastic changes in the nose, and increased incidences of respiratory/transitional epithelial adenomas in both mice and rats. Increased incidences of harderian gland adenoma may also have been related to exposure to PA in male mice.


Alkynes/toxicity , Carcinogens , Propanols/toxicity , Adenoma/chemically induced , Adenoma/pathology , Alkynes/administration & dosage , Animals , Atmosphere Exposure Chambers , Carcinogenicity Tests , Female , Hyaline Cartilage/drug effects , Inflammation/pathology , Inhalation Exposure , Kaplan-Meier Estimate , Leukemia/chemically induced , Leukemia/epidemiology , Male , Mice , Mice, Inbred Strains , Neoplasms/chemically induced , Neoplasms/pathology , Occupational Exposure , Propanols/administration & dosage , Rats , Rats, Inbred F344 , Respiratory Tract Neoplasms/chemically induced , Respiratory Tract Neoplasms/pathology , Sex Characteristics , Survival Analysis
17.
J Environ Monit ; 14(1): 181-6, 2012 Jan.
Article En | MEDLINE | ID: mdl-22105539

The present study evaluates the toxicity of 34 propargylic alcohols, including primary, primary homo-, secondary, and tertiary alcohols, based on their effects on phytoplankton. A closed-system algal toxicity test was applied because the closed-system technique presents more realistic concentration-response relationships for the above compounds than the conventional batch tests. The green alga, Pseudokirchneriella subcapitata, was the test organism and final yield and growth rate were chosen as the test endpoints. Among all the propargylic alcohols tested, 1-pentyn-3-ol is the most toxic compound with its EC50 equal to 0.50 mg L(-1), which can be classified as a "R50" compound (very toxic to aquatic organisms, EC50/LC50 < 1 mg L(-1)), following the current practice for classification of chemicals in the European Union (EU). There are several other compounds including 2-decyn-1-ol, 3-decyn-1-ol, 1-hexyn-3-ol, 3-butyn-2-ol, and 3-hexyne-2,5-diol, which deserve more attention for their possible adverse impact on the aquatic environment, because these alcohols can be classified as "R51" compounds (toxic to aquatic organisms, EC50/LC50 between 1 and 10 mg L(-1)). Compared to the base-line toxicity relationship (narcosis QSAR) derived previously, tertiary propargylic alcohols can be identified as nonpolar narcotic chemicals, while secondary alcohols and primary alcohols with low molecular weight generally exhibit obvious excess toxicity in relation to the base-line toxicity. Finally, quantitative structure-activity relationships were established for deriving a preliminary estimation of the toxicity of other propargylic alcohols.


Alkynes/toxicity , Chlorophyta/drug effects , Propanols/toxicity , Water Pollutants, Chemical/toxicity , Quantitative Structure-Activity Relationship , Risk Assessment
18.
Chem Res Toxicol ; 25(1): 170-80, 2012 Jan 13.
Article En | MEDLINE | ID: mdl-22117088

For eight acrylates, three methacrylates, and three propiolates as three subclasses of α,ß-unsaturated esters, short-term and long-term bacterial toxicity with Vibrio fischeri was determined in terms of EC(50) (effective concentration 50%) values for the 30-min bioluminescence and 24-h growth inhibition. To this end, experimental exposure concentrations were corrected for volatilization through a thermodynamic model based on Henry's law constant of the compounds. Moreover, toxicity enhancements T(e) as the ratio of narcosis-predicted over actual EC(50) were determined and discussed in terms of underlying mechanisms of reaction of the electrophiles with endogenous nucleophiles such as glutathione (GSH) and proteins. Overall, log EC(50) [M] ranges from -2.28 to -3.70 (30 min) and from -2.80 to -5.28 (24 h), respectively, indicating a significantly larger sensitivity of the growth inhibition bioassay for the reactive toxicity of these Michael acceptors. The latter is also reflected in the observed toxicity enhancements, where log T(e) > 1 was obtained for only 5 of 14 30-min EC(50) values but for 11 of 13 24-h EC(50) values. Moreover, the average long-term to short-term difference in log T(e) is 1 unit for the acrylates and 0.7 units for both methacrylates and propiolates. Methacrylates exert narcosis-level toxicity except for the methyl derivative in the long-term assay. The log EC(50) (24 h) of a subset of 10 mostly excess-toxic acrylates and a propiolate correlates with their logarithmic rate constants of reaction with GSH, log k(GSH), significantly more than with log K(ow) (r(2) 0.76 vs 0.47), yielding a respective regression rms of 0.34 log units. For allyl and propargyl acrylate as well as propargyl methacrylate, the observed excess toxicity is likely caused by initial enzymatic hydrolysis and subsequent oxidation of the α,ß-unsaturated alcohols to the respective carbonyls. The latter shows that in the context of nonanimal testing schemes such as for REACH, the metabolic capacity of in vitro screens requires attention.


Acrylates/toxicity , Aliivibrio fischeri/drug effects , Alkynes/toxicity , Propionates/toxicity , Aliivibrio fischeri/growth & development , Aliivibrio fischeri/metabolism , Glutathione/metabolism , Luminescent Measurements
19.
Chem Res Toxicol ; 24(9): 1457-9, 2011 Sep 19.
Article En | MEDLINE | ID: mdl-21812477

Previously, we determined that diethyl acetylenedicarboxylate (DAD), a protein cross-linker, was significantly more toxic than analogous monofunctional electrophiles. We hypothesized that other protein cross-linkers enhance toxicity similarly. In agreement with this hypothesis, the bifunctional electrophile divinyl sulfone (DVSF) was 6-fold more toxic than ethyl vinyl sulfone (EVSF) in colorectal carcinoma cells and greater than 10-fold more toxic in Saccharomyces cerevisiae. DVSF and DAD caused oligomerization of yeast thioredoxin 2 (Trx2p) in vitro and promoted Trx2p cross-linking to other proteins in yeast at cytotoxic doses. Our results suggest that protein cross-linking is considerably more detrimental to cellular homeostasis than simple alkylation.


Alkynes/toxicity , Cross-Linking Reagents/toxicity , Cytotoxins/toxicity , Proteins/chemistry , Sulfones/toxicity , Alkylation , Cell Line, Tumor , Humans , Saccharomyces cerevisiae/cytology
20.
Arch Toxicol ; 85(11): 1419-27, 2011 Nov.
Article En | MEDLINE | ID: mdl-21445588

Biocides are added to biodiesels to inhibit and remove microbial growth. The effects of 5-chloro-2-methyl-4-isothiazolin-3-one (CMIT), a candidate biodiesel biocide, were studied using freshly isolated rat alveolar macrophages (AM) and NR8383 cell line. CMIT markedly inhibited phagocytic oxidative burst as measured by zymosan-induced chemiluminescence, and cellular cytokine secretion as measured by zymosan-induced TNF-α secretion. The 50% inhibition concentration (LC(50)) for CMIT was 0.002-0.004 mM for both cellular functions. AM exposed to CMIT for as little as 2 min showed markedly inhibited functions that persisted for at least 5 h. Sodium metabisulfite was able to partially neutralize the inhibitory activity of CMIT. Cysteine and glutathione, when present at a molar ratio of 2-1 or higher against CMIT, were effective neutralizers, while serine, histidine, alanine, and albumin were without effect. When the AM testing system was used to compare the toxicity of CMIT against three other candidate biodiesel biocides, methylene dithiocyanate (MDC) was found to be of comparable toxicity to CMIT, 2-methyl-4-isothiazolin-3-one (MIT) was much less toxic, and dimethyl acetylenedicarboxylate (DMAD) was non-toxic. Because AM is among the first cell-type exposed to inhaled biodiesel aerosols, the result suggested that CMIT present in biodiesel may produce respiratory effects, and further investigations including animal studies are warranted.


Biofuels/toxicity , Disinfectants/toxicity , Macrophages, Alveolar/drug effects , Thiazoles/pharmacology , Alkynes/toxicity , Animals , Cell Line , Cysteine/metabolism , Cytokines/metabolism , Glutathione/metabolism , Luminescence , Macrophages, Alveolar/cytology , Male , Rats , Rats, Sprague-Dawley , Respiratory Burst/drug effects , Thiazoles/toxicity , Tumor Necrosis Factor-alpha/metabolism , Zymosan/metabolism
...