Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 723
Filter
1.
Ecotoxicol Environ Saf ; 280: 116521, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-38850708

ABSTRACT

The aim of this study is to investigate the role of estrogen receptor ß (ERß) in nonylphenol (NP) - induced depression - like behavior in rats and its impact on the regulation of the TPH2/5-HT pathway. In the in vitro experiment, rat basophilic leukaemia cells (RBL-2H3) cells were divided into the four groups: blank group, NP group (20 µM), ERß agonist group (0.01 µM), and NP+ERß agonist group (20 µM+0.01 µM). For the in vivo experiment, 72 adult male Sprague-Dawley rats were randomly divided into following six groups: the Control, NP (40 mg/kg) group, ERß agonist (2 mg/kg, Diarylpropionitrile (DPN)) group, ERß inhibitor (0.1 mg/kg, 4-(2-phenyl-5,7-bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-3-yl) phenol (PHTPP)) group, NP+ERß agonist (40 mg/kg NP + 2 mg/kg DPN) group, and NP+ERß inhibitor (40 mg/kg NP + 0.1 mg/kg PHTPP) group, with 12 rats in each group. Each rat in drug group were given NP by gavage and/or received a single intraperitoneal injection of DPN 2 mg/kg or PHTPP 0.1 mg/kg. Both in vivo and in vitro, NP group showed a decrease in the expression levels of ERß, tryptophan hydroxylase (TPH1), and tryptophan hydroxylase-2 (TPH2) genes and proteins, and reduced levels of DA, NE, and 5-hydroxytryptophan (5-HT) neurotransmitters. RBL-2H3 cells showed signs of cell shrinkage, with rounded cells, increased suspension and more loosely arranged cells. The effectiveness of the ERß agonist stimulation exhibited an increase exceeding 60% in RBL-2H3 cells. The application of ERß agonist resulted in an alleviation the aforementioned alterations. ERß agonist activated the TPH2/5-HT signaling pathways. Compared to the control group, the NP content in the brain tissue of the NP group was significantly increased. The latency to eat for the rats was longer and the amount of food consumed was lower, and the rats had prolonged immobility time in the behavioral experiment of rats. The expression levels of ERß, TPH1, TPH2, 5-HT and 5-HITT proteins were decreased in the NP group, suggesting NP-induced depression-like behaviours as well as disturbances in the secretion of serum hormones and monoamine neurotransmitters. In the NP group, the midline raphe nucleus showed an elongated nucleus with a dark purplish-blue colour, nuclear atrophy, displacement and pale cytoplasm. ERß might ameliorate NP-induced depression-like behaviors, and secretion disorders of serum hormones and monoamine neurotransmitters via activating TPH2/5-HT signaling pathways.


Subject(s)
Depression , Estrogen Receptor beta , Phenols , Rats, Sprague-Dawley , Serotonin , Tryptophan Hydroxylase , Animals , Tryptophan Hydroxylase/metabolism , Estrogen Receptor beta/metabolism , Phenols/toxicity , Male , Rats , Serotonin/metabolism , Depression/chemically induced , Depression/drug therapy , Depression/metabolism , Neurotransmitter Agents/metabolism , Signal Transduction/drug effects , Cell Line, Tumor , Nitriles/toxicity , Nitriles/pharmacology , Propionates/toxicity , Propionates/pharmacology , Pyrazoles , Pyrimidines
2.
Chem Biol Interact ; 395: 111026, 2024 May 25.
Article in English | MEDLINE | ID: mdl-38679115

ABSTRACT

In the pursuit of novel antioxidant therapies for the prevention and treatment of neurodegenerative diseases, three new arylpiperazine derivatives (LQFM181, LQFM276, and LQFM277) were synthesized through a molecular hybridization approach involving piribedil and butylated hydroxytoluene lead compounds. To evaluate the antioxidant and neuroprotective activities of the arylpiperazine derivatives, we employed an integrated approach using both in vitro (SH-SY5Y cells) and in vivo (neurotoxicity induced by 3-nitropropionic acid in Swiss mice) models. In the in vitro tests, LQFM181 showed the most promising antioxidant activity at the neuronal membrane and cytoplasmic levels, and significant neuroprotective activity against the neurotoxicity induced by 3-nitropropionic acid. Hence, this compound was further subjected to in vivo evaluation, which demonstrated remarkable antioxidant capacity such as reduction of MDA and carbonyl protein levels, increased activities of succinate dehydrogenase, catalase, and superoxide dismutase. Interestingly, using the same in vivo model, LQFM181 also reduced locomotor behavior and memory dysfunction through its ability to decrease cholinesterase activity. Consequently, LQFM181 emerges as a promising candidate for further investigation into its neuroprotective potential, positioning it as a new therapeutic agent for neuroprotection.


Subject(s)
Antioxidants , Neuroprotective Agents , Nitro Compounds , Piperazines , Propionates , Animals , Propionates/toxicity , Nitro Compounds/toxicity , Neuroprotective Agents/pharmacology , Neuroprotective Agents/chemistry , Mice , Piperazines/pharmacology , Piperazines/chemistry , Humans , Cell Line, Tumor , Antioxidants/pharmacology , Male , Succinate Dehydrogenase/metabolism , Superoxide Dismutase/metabolism , Catalase/metabolism , Neurons/drug effects , Neurons/metabolism , Malondialdehyde/metabolism , Oxidative Stress/drug effects
3.
Sci Total Environ ; 929: 172640, 2024 Jun 15.
Article in English | MEDLINE | ID: mdl-38670351

ABSTRACT

Bees are important for maintaining ecosystems, pollinating crops and producing marketable products. In recent years, a decline in bee populations has been reported, with multifactorial causes, including the intensification of pesticide use in agriculture. Among pesticides, cyflumetofen is an insecticide and acaricide used in apple, coffee and citrus crops, whose main pollinator is the honey bee Apis mellifera. Therefore, this bee is a potential target of cyflumetofen during foraging. This study evaluated the histopathological and cytological damage in the midgut, hypopharyngeal glands and fat body of A. mellifera workers exposed to LC50 of cyflumetofen. The midgut epithelium of exposed bees presented cytoplasmic vacuolization, release of vesicles and cell fragments, which indicate autophagy, increased production of digestive enzymes and cell death, respectively. The cytological analysis of the midgut revealed the dilation of the basal labyrinth and the presence of spherocrystals in the digestive cells. The hypopharyngeal glands produced greater amounts of secretion in treated bees, whereas no changes were observed in the fat body. The results indicate that acute exposure to cyflumetofen negatively affect A. mellifera, causing damage to the midgut and changes in the hypopharyngeal glands, which may compromise the survival and foraging of this pollinator.


Subject(s)
Acaricides , Animals , Bees/drug effects , Acaricides/toxicity , Propionates/toxicity , Fat Body/drug effects , Insecticides/toxicity
4.
Toxicol Sci ; 200(1): 165-182, 2024 Jun 26.
Article in English | MEDLINE | ID: mdl-38574381

ABSTRACT

Like many per- or polyfluorinated alkyl substances (PFAS), toxicity studies with HFPO-DA (ammonium, 2,3,3,3-tetrafluoro-2-(heptafluoropropoxy)-propanoate), a short-chain PFAS used in the manufacture of some types of fluorinated polymers, indicate that the liver is the primary target of toxicity in rodents following oral exposure. Although the current weight of evidence supports the PPARα mode of action (MOA) for liver effects in HFPO-DA-exposed mice, alternate MOAs have also been hypothesized including PPARγ or cytotoxicity. To further evaluate the MOA for HFPO-DA in rodent liver, transcriptomic analyses were conducted on samples from primary mouse, rat, and pooled human hepatocytes treated for 12, 24, or 72 h with various concentrations of HFPO-DA, or agonists of PPARα (GW7647), PPARγ (rosiglitazone), or cytotoxic agents (ie, acetaminophen or d-galactosamine). Concordance analyses of enriched pathways across chemicals within each species demonstrated the greatest concordance between HFPO-DA and PPARα agonist GW7647-treated hepatocytes compared with the other chemicals evaluated. These findings were supported by benchmark concentration modeling and predicted upstream regulator results. In addition, transcriptomic analyses across species demonstrated a greater transcriptomic response in rodent hepatocytes treated with HFPO-DA or agonists of PPARα or PPARγ, indicating rodent hepatocytes are more sensitive to HFPO-DA or PPARα/γ agonist treatment. These results are consistent with previously published transcriptomic analyses and further support that liver effects in HFPO-DA-exposed rodents are mediated through rodent-specific PPARα signaling mechanisms as part of the MOA for PPARα activator-induced rodent hepatocarcinogenesis. Thus, effects observed in mouse liver are not appropriate endpoints for toxicity value development for HFPO-DA in human health risk assessment.


Subject(s)
Hepatocytes , PPAR alpha , PPAR gamma , Transcriptome , Animals , Hepatocytes/drug effects , Hepatocytes/metabolism , PPAR alpha/agonists , PPAR alpha/genetics , PPAR alpha/metabolism , Humans , PPAR gamma/genetics , PPAR gamma/agonists , PPAR gamma/metabolism , Transcriptome/drug effects , Male , Mice , Fluorocarbons/toxicity , Rats , Propionates/toxicity , Cells, Cultured , Gene Expression Profiling , Rosiglitazone/pharmacology , Rosiglitazone/toxicity , Rats, Sprague-Dawley , Mice, Inbred C57BL , Species Specificity , Dose-Response Relationship, Drug , Butyrates , Phenylurea Compounds
5.
Toxicol Sci ; 200(1): 183-198, 2024 Jun 26.
Article in English | MEDLINE | ID: mdl-38574385

ABSTRACT

Recent in vitro transcriptomic analyses for the short-chain polyfluoroalkyl substance, HFPO-DA (ammonium, 2,3,3,3-tetrafluoro-2-(heptafluoropropoxy)-propanoate), support conclusions from in vivo data that HFPO-DA-mediated liver effects in mice are part of the early key events of the peroxisome proliferator-activated receptor alpha (PPARα) activator-induced rodent hepatocarcinogenesis mode of action (MOA). Transcriptomic responses in HFPO-DA-treated rodent hepatocytes have high concordance with those treated with a PPARα agonist and lack concordance with those treated with PPARγ agonists or cytotoxic agents. To elucidate whether HFPO-DA-mediated transcriptomic responses in mouse liver are PPARα-dependent, additional transcriptomic analyses were conducted on samples from primary PPARα knockout (KO) and wild-type (WT) mouse hepatocytes exposed for 12, 24, or 72 h with various concentrations of HFPO-DA, or well-established agonists of PPARα (GW7647) and PPARγ (rosiglitazone), or cytotoxic agents (acetaminophen or d-galactosamine). Pathway and predicted upstream regulator-level responses were highly concordant between HFPO-DA and GW7647 in WT hepatocytes. A similar pattern was observed in PPARα KO hepatocytes, albeit with a distinct temporal and concentration-dependent delay potentially mediated by compensatory responses. This delay was not observed in PPARα KO hepatocytes exposed to rosiglitazone, acetaminophen, d-galactosamine. The similarity in transcriptomic signaling between HFPO-DA and GW7647 in both the presence and absence of PPARα in vitro indicates these compounds share a common MOA.


Subject(s)
Hepatocytes , Mice, Knockout , PPAR alpha , PPAR gamma , Transcriptome , Animals , Hepatocytes/drug effects , Hepatocytes/metabolism , PPAR alpha/agonists , PPAR alpha/genetics , PPAR alpha/metabolism , PPAR gamma/agonists , PPAR gamma/genetics , PPAR gamma/metabolism , Transcriptome/drug effects , Mice , Fluorocarbons/toxicity , Propionates/pharmacology , Propionates/toxicity , Mice, Inbred C57BL , Male , Cells, Cultured , Gene Expression Profiling , Acetaminophen/toxicity , Cytotoxins/toxicity , Butyrates , Phenylurea Compounds
6.
Toxicol Appl Pharmacol ; 485: 116910, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38521372

ABSTRACT

3-nitropropionic acid (3-NPA), a toxic metabolite produced by mold, is mainly found in moldy sugarcane. 3-NPA inhibits the activity of succinate dehydrogenase that can induce oxidative stress injury in cells, reduce ATP production and induce oxidative stress in mouse ovaries to cause reproductive disorders. Ursolic acid (UA) has a variety of biological activities and is a pentacyclic triterpene compound found in many plants. This experiment aimed to investigate the cytotoxicity of 3-NPA during mouse oocyte in vitro maturation and the protective effects of UA on oocytes challenged with 3-NPA. The results showed that UA could alleviate 3-NPA-induced oocyte meiotic maturation failure. Specifically, 3-NPA induced a decrease in the first polar body extrusion rate of oocytes, abnormal distribution of cortical granules, and an increase in the proportion of spindle abnormalities. In addition, 3-NPA caused mitochondrial dysfunction and induced oxidative stress, including decreases in the GSH, mitochondrial membrane potential and ATP levels, and increases in the ROS levels, and these effects led to apoptosis and autophagy. The addition of UA could significantly improve the adverse effects caused by 3-NPA. In general, our data show that 3-NPA affects the normal development of oocytes during the in vitro culture, and the addition of UA can effectively repair the damage caused by 3-NPA to oocytes.


Subject(s)
Meiosis , Nitro Compounds , Oocytes , Oxidative Stress , Propionates , Triterpenes , Ursolic Acid , Animals , Nitro Compounds/toxicity , Propionates/toxicity , Oocytes/drug effects , Oocytes/metabolism , Female , Meiosis/drug effects , Mice , Triterpenes/pharmacology , Oxidative Stress/drug effects , Membrane Potential, Mitochondrial/drug effects , Reactive Oxygen Species/metabolism , Apoptosis/drug effects , Mitochondria/drug effects , Mitochondria/metabolism , Autophagy/drug effects , Adenosine Triphosphate/metabolism , Mice, Inbred ICR
7.
Neurotoxicology ; 102: 12-28, 2024 May.
Article in English | MEDLINE | ID: mdl-38453033

ABSTRACT

Huntington's disease (HD) is a progressive neurodegenerative condition characterized by a severe motor incoordination, cognitive decline, and psychiatric complications. However, a definitive cure for this devastating disorder remains elusive. Agmatine, a biogenic amine, has gain attention for its reported neuromodulatory and neuroprotective properties. The present study was designed to examine the influence of agmatine on the behavioral, biochemical, and molecular aspects of HD in an animal model. A mitochondrial toxin, 3-nitro propionic acid (3-NP), was used to induce HD phenotype and similar symptoms such as motor incoordination, memory impairment, neuro-inflammation, and depressive-like behavior in rats. Rats were pre-treated with 3-NP (10 mg/kg, i.p.) on days 1, 3, 5, 7, and 9 and then continued on agmatine treatment (5 - 20 µg/rat, i.c.v.) from day-8 to day-27 of the treatment protocol. 3-NP-induced cognitive impairment was associated with declined in agmatine levels within prefrontal cortex, striatum, and hippocampus. Further, the 3-NP-treated rats showed an increase in IL-6 and TNF-α and a reduction in BDNF immunocontent within these brain areas. Agmatine treatment not only improved the 3-NP-induced motor incoordination, depression-like behavior, rota-rod performance, and learning and memory impairment but also normalized the GABA/glutamate, BDNF, IL-6, and TNF-α levels in discrete brain areas. Similarly, various agmatine modulators, which increase the endogenous agmatine levels in the brain, such as L-arginine (biosynthetic precursor), aminoguanidine (diamine oxidase inhibitor), and arcaine (agmatinase inhibitor) also demonstrated similar effects exhibiting the importance of endogenous agmatinergic pathway in the pathogenesis of 3-NP-induced HD like symptoms. The present study proposed the possible role of agmatine in the pathogenesis and treatment of HD associated motor incoordination, and psychiatric and cognitive complications.


Subject(s)
Agmatine , Huntington Disease , Nitro Compounds , Propionates , Animals , Nitro Compounds/toxicity , Propionates/toxicity , Agmatine/pharmacology , Huntington Disease/chemically induced , Huntington Disease/metabolism , Huntington Disease/drug therapy , Male , Rats , Behavior, Animal/drug effects , Brain/drug effects , Brain/metabolism , Rats, Wistar , Disease Models, Animal , Motor Activity/drug effects , Rats, Sprague-Dawley , Neuroprotective Agents/pharmacology
8.
J Huntingtons Dis ; 13(1): 55-66, 2024.
Article in English | MEDLINE | ID: mdl-38489193

ABSTRACT

Background: Huntington's disease (HD) is a neurodegenerative disorder characterized by motor, cognitive, and psychiatric dysfunction caused by a mutant huntingtin protein. Compromised metabolic activity resulting from systemic administration of the mitochondrial toxin, 3-nitropropionic acid (3-NP), is known to mimic the pathology of HD and induce HD-like symptoms in rats. N-hexanoic-Tyr-Ile-(6)-amino hexanoic amide (PNB-0408), also known as Dihexa, has been shown to have neuroprotective and procognitive properties in animal models of Alzheimer's and Parkinson's diseases. Given the mechanism of action and success in other neurodegenerative diseases, we felt it an appropriate compound to investigate further for HD. Objective: The present study was designed to test if PNB-0408, an angiotensin IV analog, could attenuate 3-NP-induced HD-like symptoms in rats and serve as a potential therapeutic agent. Methods: Forty male Wistar rats were randomized into three groups consisting of a "vehicle" group, a "3-NP" group, and a "3-NP + PNB-0408" group. PNB-0408 was administered along with chronic exposure to 3-NP. Animal body weight, motor function, and cognitive abilities were measured for five weeks, before euthanasia and histopathological analysis. Results: Exposure to 3-NP decreased the amount of weight rats gained, impaired spatial learning and memory consolidation, and led to marked motor dysfunction. From our observations and analysis, PNB-0408 did not protect rats from the deficits induced by 3-NP neurotoxicity. Conclusions: Our findings suggest that PNB-0408 may not be an efficacious treatment strategy for preventing 3-NP-induced HD-like symptoms in a preclinical model. These data highlight the need for further research of this compound in alternate models and/or alternative approaches to managing this disorder.


Subject(s)
Angiotensin II/analogs & derivatives , Huntington Disease , Neuroprotective Agents , Rats , Male , Animals , Rats, Wistar , Huntington Disease/chemically induced , Huntington Disease/drug therapy , Huntington Disease/metabolism , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Nitro Compounds/toxicity , Nitro Compounds/therapeutic use , Propionates/toxicity , Propionates/therapeutic use , Disease Models, Animal
11.
Environ Toxicol Pharmacol ; 105: 104354, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38151218

ABSTRACT

Fescue toxicosis (FT) is produced by an ergot alkaloid (i.e., ergovaline [EV])-producing fungus residing in toxic fescue plants. Associations between EV, decreased weight gain and ruminal volatile fatty acids are unclear. Feces, rumen fluid, and blood were collected from 12 steers that grazed non-toxic (NT) or toxic (E +) fescue for 28 days. The E + group exhibited decreased propionate (P), increased acetate (A), and increased ruminal A:P ratio, with similar trends in feces. Plasma GASP-1 (G-Protein-Coupled-Receptor-Associated-Sorting-Protein), a myostatin inhibitor, decreased (day 14) only in E + steers. Ergovaline was present only in E + ruminal fluid and peaked on day 14. The lower ruminal propionate and higher A:P ratio might contribute to FT while reduced GASP-1 might be a new mechanism linked to E + -related weight gain reduction. Day 14 ergovaline zenith likely reflects ruminal adaptations favoring EV breakdown and its presence only in rumen points to local, rather than systemic effects.


Subject(s)
Festuca , Propionates , Animals , Propionates/toxicity , Ergotamines , Festuca/microbiology , Fatty Acids, Volatile , Weight Gain , Animal Feed/analysis
12.
Aquat Toxicol ; 261: 106572, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37307698

ABSTRACT

Ethyl 3-(N-butylacetamido) propanoate (EBAAP) is one of the most widely used mosquito repellents worldwide, and is also commonly used to produce cosmetics. Residues have recently been detected in surface and groundwater in many countries, and their potential to harm the environment is unknown. Therefore, more studies are needed to fully assess the toxicity of EBAAP. This is the first investigation into the developmental toxicity and cardiotoxicity of EBAAP on zebrafish embryos. EBAAP was toxic to zebrafish, with a lethal concentration 50 (LC50) of 140 mg/L at 72 hours post fertilization (hpf). EBAAP exposure also reduced body length, slowed the yolk absorption rate, induced spinal curvature and pericardial edema, decreased heart rate, promoted linear lengthening of the heart, and diminished cardiac pumping ability. The expression of heart developmental-related genes (nkx2.5, myh6, tbx5a, vmhc, gata4, tbx2b) was dysregulated, intracellular oxidative stress increased significantly, the activities of catalase (CAT) and superoxide dismutase (SOD) decreased, and malondialdehyde (MDA) content increased significantly. The expression of apoptosis-related genes (bax/bcl2, p53, caspase9, caspase3) was significantly upregulated. In conclusion, EBAAP induced abnormal morphology and heart defects during the early stages of zebrafish embryo development by potentially inducing the generation and accumulation of reactive oxygen species (ROS) in vivo and activating the oxidative stress response. These events dysregulate the expression of several genes and activate endogenous apoptosis pathways, eventually leading to developmental disorders and heart defects.


Subject(s)
Cardiotoxicity , Insect Repellents , Water Pollutants, Chemical , Animals , Embryo, Nonmammalian/metabolism , Oxidative Stress/genetics , Propionates/toxicity , Propionates/metabolism , Water Pollutants, Chemical/toxicity , Zebrafish/metabolism , Insect Repellents/toxicity , Toxicity Tests
13.
Toxicol Lett ; 381: 48-59, 2023 May 15.
Article in English | MEDLINE | ID: mdl-37116597

ABSTRACT

Redox homeostasis, mitochondrial functions, and mitochondria-endoplasmic reticulum (ER) communication were evaluated in the striatum of rats after 3-nitropropionic acid (3-NP) administration, a recognized chemical model of Huntington's disease (HD). 3-NP impaired redox homeostasis by increasing malondialdehyde levels at 28 days, decreasing glutathione (GSH) concentrations at 21 and 28 days, and the activities of glutathione peroxidase (GPx), superoxide dismutase (SOD) and glutathione S-transferase at 7, 21, and 28 days, catalase at 21 days, and glutathione reductase at 21 and 28 days. Impairment of mitochondrial respiration at 7 and 28 days after 3-NP administration was also observed, as well as reduced activities of succinate dehydrogenase (SDH) and respiratory chain complexes. 3-NP also impaired mitochondrial dynamics and the interactions between ER and mitochondria and induced ER-stress by increasing the levels of mitofusin-1, and of DRP1, VDAC1, Grp75 and Grp78. Synaptophysin levels were augmented at 7 days but reduced at 28 days after 3-NP injection. Finally, bezafibrate prevented 3-NP-induced alterations of the activities of SOD, GPx, SDH and respiratory chain complexes, DCFH oxidation and on the levels of GSH, VDAC1 and synaptophysin. Mitochondrial dysfunction and synaptic disruption may contribute to the pathophysiology of HD and bezafibrate may be considered as an adjuvant therapy for this disorder.


Subject(s)
Huntington Disease , Rats , Animals , Huntington Disease/chemically induced , Huntington Disease/drug therapy , Huntington Disease/metabolism , Rats, Wistar , Bezafibrate/adverse effects , Bezafibrate/metabolism , Synaptophysin/metabolism , Models, Chemical , Oxidative Stress , Glutathione/metabolism , Superoxide Dismutase/metabolism , Mitochondria/metabolism , Propionates/toxicity , Nitro Compounds/toxicity , Nitro Compounds/metabolism
14.
Neurochem Res ; 48(6): 1631-1647, 2023 Jun.
Article in English | MEDLINE | ID: mdl-36738367

ABSTRACT

Animal models are used to better understand the various mechanisms involved in the pathogenesis of diseases and explore potential pathways that will aid in discovering therapeutic targets. 3-Nitropropionic Acid (3-NPA) is a neurotoxin used to induce Huntington's disease (HD)-like symptoms in experimental animals. The 3-NPA is a fungus toxin that impairs the complex II (succinate dehydrogenase) activity of the mitochondria and reduces ATP synthesis, leading to excessive production of free radicals resulting in the degeneration of GABAergic medium spiny neurons (MSNs) in the striatum. This is characterized by motor impairments a key clinical manifestation of HD. 3-NPA has the potential to alter several cellular processes, including mitochondrial functions, oxidative stress, apoptosis, and neuroinflammation mimicking HD-like pathogenic conditions in animals. This review strives to provide a new insight towards the 3-NPA induced molecular dysfunctioning in developing an animal model of HD. Moreover, we summarise several preclinical studies that support the use of the 3-NPA-induced models for drug discovery and development in HD. This review is a collection of various articles that were published from 1977 to 2022 on Pubmed (1639), Web of Science (2139), and Scopus (2681), which are related to the 3-NPA induced animal model.


Subject(s)
Huntington Disease , Animals , Huntington Disease/chemically induced , Huntington Disease/metabolism , Neurotoxins/toxicity , Disease Models, Animal , Nitro Compounds/toxicity , Propionates/toxicity , Drug Discovery
15.
Pestic Biochem Physiol ; 188: 105235, 2022 Nov.
Article in English | MEDLINE | ID: mdl-36464352

ABSTRACT

The two-spotted spider mite, Tetranychus urticae Koch (Acari: Tetranychidae) is the most economically important mite pest in agricultural areas and chemical acaricides are widely used to control T. urticae populations. Cyflumetofen is a recently introduced acaricide that inhibits the mitochondrial electron transport chain at complex II (succinate dehydrogenase, SDH), which represents the most recently developed mode of action for mite control worldwide. In the present study, started upon the launch of cyflumetofen in Turkey, a five-year survey was performed to monitor cyflumetofen susceptibility in 28 T. urticae populations collected from agricultural fields across the country. The first resistance case that might cause control failure in practical field conditions was uncovered in 2019, three years after the registration of cyflumetofen. In addition, an extremely resistant population (1722-fold resistance) was also detected towards the end of 2019. Cyflumetofen resistance did not decrease in the laboratory after relaxation of selection pressure for over one year in field-collected populations, suggesting the absence of a fitness cost associated with resistance in these populations. Next to phenotypic resistance, metabolic and physiological mechanisms underlying the decreased susceptibility were also investigated. Synergism assays showed the involvement of P450 monooxygenases in cyflumetofen resistance. Downregulation of carboxylesterases as resistance mechanism, is underpinned by the fact that pre-treatment with esterase inhibitor DEF decreased cyflumetofen toxicity in field-collected strains. Furthermore, a novel H258L substitution in the subunit B of complex II was uncovered in a field population. In silico modeling of the new mutation suggested that the mutation might indeed influence toxicity to complex II inhibitors cyenopyrafen and pyflubumide, but most likely not cyflumetofen. However, further studies are needed to uncover the exact role of this mutation in resistance to this new class of complex II inhibitors.


Subject(s)
Acaricides , Tetranychidae , Animals , Tetranychidae/genetics , Turkey , Propionates/toxicity , Acaricides/pharmacology
16.
Neurotoxicol Teratol ; 93: 107123, 2022.
Article in English | MEDLINE | ID: mdl-36150581

ABSTRACT

Propionate is an effective mould inhibitor widely used as a food preservative. In this study, we used zebrafish to explore the adverse effects of long-term exposure to low concentrations of sodium propionate and the underlying molecular mechanisms (from larvae to adult). When exposed for 3 months, we found that blood glucose, total cholesterol, and triglyceride levels increased, and zebrafish developed a hyperglycaemic state. New tank test results showed depression in zebrafish reduced 5-hydroxytryptamine levels in the brain and damaged the dopamine system. At the same time, the results of the color preference test showed that zebrafish had cognitive impairments. In addition, Hypothalamic-Pituitary-Adrenal axis analysis revealed abnormal gene expression, increased cortisol levels, and reduced glucocorticoid receptor mRNA levels, which were consistent with depressive behavior. We also observed abnormal transcription of inflammatory and apoptotic factors. Overall, we found that chronic exposure to sodium propionate induces depressive symptoms. This may be related to the activation of the HPA axis by the hyperglycaemic state, thereby inducing inflammation and disrupting the dopaminergic system. In summary, this study provides theoretical and technical support for the overlap of the emotional pathogenesis associated with diabetes.


Subject(s)
Hyperglycemia , Nervous System Diseases , Animals , Blood Glucose/metabolism , Cholesterol , Dopamine/metabolism , Food Preservatives/metabolism , Food Preservatives/pharmacology , Hydrocortisone/metabolism , Hyperglycemia/chemically induced , Hyperglycemia/metabolism , Hypothalamo-Hypophyseal System/metabolism , Pituitary-Adrenal System/metabolism , Propionates/metabolism , Propionates/toxicity , RNA, Messenger/metabolism , Receptors, Glucocorticoid/metabolism , Serotonin/metabolism , Triglycerides/metabolism , Triglycerides/pharmacology , Zebrafish/metabolism
19.
Toxicology ; 477: 153259, 2022 07.
Article in English | MEDLINE | ID: mdl-35850385

ABSTRACT

The toxicity induced by the persistent organic pollutants per- and polyfluoroalkyl substances (PFAS) is dependent on the length of their polyfluorinated tail. Long-chain PFASs have significantly longer half-lives and profound toxic effects compared to their short-chain counterparts. Recently, production of a short-chain PFAS substitute called ammonium 2,3,3,3-tetrafluoro-2-(heptafluoropropoxy) propanoate, also known as GenX, has significantly increased. However, the adverse health effects of GenX are not completely known. In this study, we investigated the dose-dependent effects of GenX on primary human hepatocytes (PHH). Freshly isolated PHH were treated with either 0.1, 10, or 100 µM of GenX for 48 and 96 h; then, global transcriptomic changes were determined using Human Clariom™ D arrays. GenX-induced transcriptional changes were similar at 0.1 and 10 µM doses but were significantly different at the 100 µM dose. Genes involved in lipid, monocarboxylic acid, and ketone metabolism were significantly altered following exposure of PHH at all doses. However, at the 100 µM dose, GenX caused changes in genes involved in cell proliferation, inflammation and fibrosis. A correlation analysis of concentration and differential gene expression revealed that 576 genes positively (R > 0.99) and 375 genes negatively (R < -0.99) correlated with GenX concentration. The upstream regulator analysis indicated HIF1α was inhibited at the lower doses but were activated at the higher dose. Additionally, VEGF, PPARα, STAT3, and SMAD4 signaling was induced at the 100 µM dose. These data indicate that at lower doses GenX can interfere with metabolic pathways and at higher doses can induce fibroinflammatory changes in human hepatocytes.


Subject(s)
Fluorocarbons , Fluorocarbons/toxicity , Gene Expression , Hepatocytes , Humans , Propionates/toxicity
20.
Ecotoxicol Environ Saf ; 241: 113791, 2022 Aug.
Article in English | MEDLINE | ID: mdl-35753272

ABSTRACT

Sodium propionate is widely used as a preservative in food. The widespread use of preservatives is known to cause both environmental and public health problems. This study aimed to investigate the effects of sodium propionate on the developmental behavior and glucose metabolism of zebrafish. Our results showed that sodium propionate had no significant effect on the embryonic morphological development of zebrafish embryos but changed the head eye area. Then we found sodium propionate disturbed the thigmotaxis behavior, impaired neural development. Moreover, changes in clock gene expression disrupted the circadian rhythm of zebrafish. Circadian genes regulated insulin sensitivity and secretion in various tissues. Then our results showed that the disorder of circadian rhythm in zebrafish affected glucose metabolism and insulin resistance, which damaged the development of retina. Therefore, the safety of propionate should be further evaluated.


Subject(s)
Insulin Resistance , Zebrafish , Animals , Circadian Rhythm , Glucose/metabolism , Propionates/toxicity , Zebrafish/metabolism , Zebrafish Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...