Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 315
Filtrar
1.
Comp Biochem Physiol C Toxicol Pharmacol ; 281: 109909, 2024 Apr 02.
Artigo em Inglês | MEDLINE | ID: mdl-38570177

RESUMO

Acifluorfen, a selective herbicide from the diphenyl ether family, targets broad leaf weeds. Diphenyl ether inhibits chlorophyll production in green plants by inhibiting protoporphyrinogen oxidase (PPO), causing cellular damage. Despite its known impacts on plants, the influence of acifluorfen on zebrafish embryo development remains unclear. In this study, we explored the LC50 of acifluorfen in early-stage wild-type zebrafish, determining it to be 54.99 mg/L. Subsequent examinations revealed morphological changes in zebrafish, including reduced body length. Using the cmlc2:dsRED transgenic model, we observed heart dysfunction in acifluorfen-exposed zebrafish, marked by an enlarged heart area, edema, and decreased heart rate. In response to dose-dependent acifluorfen exposure, the inhibition of angiogenesis in the brain was observed in transgenic zebrafish models (fli1a:eGFP). Organ malformations, specifically in the liver and pancreas, were noted, in lfabp:dsRED;elastase:eGFP transgenic models, indicating reduced organ size in acifluorfen-exposed zebrafish. Furthermore, acifluorfen heightened the expression of apoptosis-related genes (casp8, casp9, and tp53) in zebrafish embryos. We then determined whether acifluorfen affected the viability of zebrafish liver (ZFL) cells based on its effects on liver development in vivo. The results indicated that the proliferation of ZFL cells decreased significantly in a dose-dependent manner. Additionally, acifluorfen-treated ZFL cells exhibited a slight increase in apoptotic cells stained with annexin V and propidium iodide. In summary, these findings establish a baseline concentration for acifluorfen's effects on aquatic ecosystems and non-target organisms.

2.
Comp Biochem Physiol C Toxicol Pharmacol ; 281: 109918, 2024 Apr 05.
Artigo em Inglês | MEDLINE | ID: mdl-38583696

RESUMO

Existing evidence shows that currently used pesticides pose toxicological risks to exposed wildlife. Chemically, bifenox belongs to diphenyl ethers, a well-known group of herbicides. Its mechanism of action primarily involves inducing lipid peroxidation and blocking protoporphyrinogen oxidases. Toxicity of diphenyl ether herbicides has been elucidated in animal cells; however, in vivo toxicological evaluations of bifenox are required to determine its unexpected effects. This study aimed to determine the negative effects of bifenox, and its effects on higher eukaryotes. We found that early stages of zebrafish embryo exposed to bifenox demonstrated increased mortality and physiological defects, based on the LC50 value. Bifenox severely inhibited blood vessel growth by reducing key elements of complex connectivity; fluorescently tagged transgenic lines (fli1a:EGFP) showed morphological changes. Additionally, transgenic lines that selectively identified hepatocytes (fabp10a:DsRed) showed reduced fluorescence, indicating that bifenox may inhibit liver development. To evaluate the level of oxidative stress, we used 2',7'-dichlorofluorescein diacetate (DCFH-DA) probes in zebrafish embryos to identify the underlying mechanisms causing developmental damage. Our findings demonstrate that exposure to bifenox causes abnormalities in the hepatic and cardiovascular systems during zebrafish embryogenesis. Therefore, this study provides new information for the evaluation of toxicological risks of bifenox in vertebrates.

3.
Food Chem Toxicol ; 186: 114559, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38432436

RESUMO

Norflurazon, an inhibitor of carotenoid synthesis, is a pre-emergent herbicide that prevents growth of weeds. The norflurazon is known to hamper embryo development in non-mammals. However, specific toxic effects of norflurazon on mammalian maternal and fetal cells have not been elucidated. Thus, the hypothesis of this study is that norflurazon may influence the toxic effects between maternal and fetal cells during early pregnancy in pigs. We aimed to examine the toxic effects of norflurazon in porcine trophectoderm (Tr) and uterine luminal epithelium (LE) cells. Norflurazon, administered at 0, 20, 50 or 100 µM for 48 h was used to determine its effects on cell proliferation and cell-cycle arrest. For both uterine LE and Tr cell lines, norflurazone caused mitochondrial dysfunction by inhibiting mitochondrial respiration and ATP production, and down-regulated expression of mRNAs of mitochondrial complex genes. Norflurazon increased cell death by increasing intracellular calcium and regulating PI3K and MAPK cell signaling pathways, as well as endoplasmic reticulum (ER) stress, ER-mitochondrial contact, and autophagy-related target proteins. Norflurazone also inhibited expression of genes required for implantation of blastocysts, including SMAD2, SMAD4, and SPP1. These findings indicate that norflurazon may induce implantation failure in pigs and other mammals through adverse effects on both Tr and uterine LE cells.


Assuntos
Implantação do Embrião , Piridazinas , Útero , Gravidez , Feminino , Suínos , Animais , Útero/metabolismo , Morte Celular , Células Epiteliais , Endométrio/metabolismo , Mamíferos
4.
Sci Total Environ ; 924: 171524, 2024 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-38453072

RESUMO

Meptyldinocap is a dinitrophenol fungicide used to control powdery mildew. Although other dinitrophenol pesticides have been found to exhibit reproductive toxicity, studies of meptyldinocaps are scarce. This study investigated the adverse effects of meptyldinocap on porcine trophectoderm (pTr) and porcine endometrial luminal epithelial (pLE) cells, which play crucial roles in implantation. We confirmed that meptyldinocap decreased cell viability, induced apoptosis, and inhibited proliferation by decreasing proliferation-related gene expression and inducing changes in the cell cycle. Furthermore, meptyldinocap treatment caused mitochondrial dysfunction, endoplasmic reticulum stress, and disruption of calcium homeostasis. Moreover, it induces alterations in mitogen-activated protein kinase signaling cascades and reduces the migration ability, leading to implantation failure. Our findings suggest that meptyldinocap reduces the cellular functions of pTr and pLE cells, which are important for the implantation process, and interferes with interactions between the two cell lines, potentially leading to implantation failure. We also propose a mechanism by which the understudied fungicide meptyldinocap exerts its cytotoxicity.


Assuntos
Dinitrobenzenos , Fungicidas Industriais , Doenças Mitocondriais , Suínos , Animais , Fungicidas Industriais/metabolismo , Proliferação de Células , Apoptose , Pontos de Checagem do Ciclo Celular , Estresse do Retículo Endoplasmático , Células Epiteliais , Dinitrofenóis/metabolismo , Dinitrofenóis/farmacologia , Doenças Mitocondriais/metabolismo
5.
Artigo em Inglês | MEDLINE | ID: mdl-38423198

RESUMO

Hexaconazole is a highly effective triazole fungicide that is frequently applied in various countries to elevate crop productivity. Given its long half-life and high water solubility, this fungicide is frequently detected in the environment, including water sources. Moreover, hexaconazole exerts hazardous effects on nontarget organisms. However, little is known about the toxic effects of hexaconazole on animal development. Thus, this study aimed to investigate the developmental toxicity of hexaconazole to zebrafish, a valuable animal model for toxicological studies, and elucidate the underlying mechanisms. Results showed that hexaconazole affected the viability and hatching rate of zebrafish at 96 h postfertilization. Hexaconazole-treated zebrafish showed phenotypic defects, such as reduced size of head and eyes and enlarged pericardiac edema. Moreover, hexaconazole induced apoptosis, DNA fragmentation, and inflammation in developing zebrafish. Various organ defects, including neurotoxicity, cardiovascular toxicity, and hepatotoxicity, were observed in transgenic zebrafish models olig2:dsRed, fli1:eGFP, and l-fabp:dsRed. Furthermore, hexaconazole treatment altered the Akt and MAPK signaling pathways, which possibly triggered the organ defects and other toxic mechanisms. This study demonstrated the developmental toxicity of hexaconazole to zebrafish and elucidated the underlying mechanisms.


Assuntos
Fungicidas Industriais , Peixe-Zebra , Animais , Peixe-Zebra/metabolismo , Fungicidas Industriais/toxicidade , Proteínas Proto-Oncogênicas c-akt/metabolismo , Triazóis/toxicidade , Inflamação/induzido quimicamente , Apoptose , Água/metabolismo , Embrião não Mamífero/metabolismo
6.
Biomed Pharmacother ; 173: 116284, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38394847

RESUMO

Until non-hormonal therapeutic targets for endometriosis are suggested, we focused on mitochondrial function and autophagy regulation in the disease. Transcrocetin is a carotenoid and retinoic acid with high antioxidant potency and antiproliferative effects in several diseases. In this study, we demonstrated the therapeutic mechanisms of transcrocetin in endometriosis using the End1/E6E7 and VK2/E6E7 cell lines. Transcrocetin suppressed the viability and proliferation of these cell lines and did not affect the proliferation of normal uterine stromal cells. p21 Waf1/Cip1 as a cell cycle regulator and target of p53, were increased by transcrocetin and caused the G1 arrest via inhibition of cyclin-dependent kinase activity, which might further cause cell death. Furthermore, we confirmed endoplasmic reticulum stress and calcium ion dysregulation in the cytosol and mitochondrial matrix, disrupting the mitochondrial membrane potential. Mitochondrial bioenergetics were suppressed by transcrocetin, and oxidative phosphorylation-related gene expression was downregulated. Moreover, the proliferation of End1/E6E7 and VK2/E6E7 cells was regulated by transcrocetin-induced oxidative stress. Finally, we verified the impairment of autophagic flux following pre-treatment with chloroquine. Therefore, transcrocetin may be a potent therapeutic alternative for endometriosis.


Assuntos
Endometriose , Vitamina A/análogos & derivados , Humanos , Feminino , Endometriose/metabolismo , Carotenoides/farmacologia , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Oxirredução , Autofagia , Apoptose
7.
Phytomedicine ; 125: 155383, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38295666

RESUMO

BACKGROUND: Osthole is active constituent of Cnidium monnieri (L.) Cuss. with various physiological functions including anti-inflammation and anti-lipedemic effects. However, the regulatory activity of osthole in colorectal cancer development, focusing on mitochondrial metabolism, is not well known. HYPOTHESIS/PURPOSE: We hypothesized that osthole may suppress progression of colorectal cancer and aimed to determine the underlying mitochondrial metabolism and the autophagic flux. STUDY DESIGN: In this study, we elucidated the mechanism of action of osthole in colorectal cancer using an in vivo azoxymethane/dextran sodium sulfate (AOM/DSS) mouse model and an in vitro cell culture system. METHODS: AOM/DSS mouse model was established and analyzed the effects of osthole on survival rate, diseases activity index, number of tumor and histopathology. Then, cell based assays including viability, cell cycle, reactive oxygen species (ROS), apoptosis, calcium efflux, and mitochondrial function were analyzed. Moreover, osthole-mediated signaling was demonstrated by western blot analyses. RESULTS: Osthole effectively suppressed the growth of colorectal tumors and alleviated AOM/DSS-induced intestinal injury. Osthole restored the function of goblet cells and impaired the expression of Claudin1 and Axin1 impaired by AOM/DSS. In addition, osthole specifically showed cytotoxicity in colorectal carcinoma cells, but not in normal colon cells. Osthole decreased the ASC/caspase-1/IL-1ß inflammasome pathway and induced mitochondrial dysfunction in redox homeostasis, calcium homeostasis. Furthermore, osthole inhibited both oxidative phosphorylation (OXPHOS) and glycolysis, leading to the suppression of ATP production. Moreover, via combination treatment with chloroquine (CQ), we demonstrated that osthole impaired autophagic flux, leading to apoptosis of HCT116 and HT29 cells. Finally, we elucidated that the functional role of tiRNAHisGTG regulated by osthole directly affects the cellular fate of colon cancer cells. CONCLUSION: These results suggest that osthole has the potential to manage progression of colorectal cancer by regulating autophagy- and mitochondria-mediated signal transduction.


Assuntos
Cálcio , Neoplasias Colorretais , Cumarínicos , Camundongos , Animais , Mitocôndrias , Neoplasias Colorretais/patologia , Azoximetano , Autofagia , Sulfato de Dextrana
8.
Exp Mol Med ; 56(1): 40-50, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38172597

RESUMO

Dynamic interactions between organelles are responsible for a variety of intercellular functions, and the endoplasmic reticulum (ER)-mitochondrial axis is recognized as a representative interorganelle system. Several studies have confirmed that most proteins in the physically tethered sites between the ER and mitochondria, called mitochondria-associated ER membranes (MAMs), are vital for intracellular physiology. MAM proteins are involved in the regulation of calcium homeostasis, lipid metabolism, and mitochondrial dynamics and are associated with processes related to intracellular stress conditions, such as oxidative stress and unfolded protein responses. Accumulating evidence has shown that, owing to their extensive involvement in cellular homeostasis, alterations in the ER-mitochondrial axis are one of the etiological factors of tumors. An in-depth understanding of MAM proteins and their impact on cell physiology, particularly in cancers, may help elucidate their potential as diagnostic and therapeutic targets for cancers. For example, the modulation of MAM proteins is utilized not only to target diverse intracellular signaling pathways within cancer cells but also to increase the sensitivity of cancer cells to anticancer reagents and regulate immune cell activities. Therefore, the current review summarizes and discusses recent advances in research on the functional roles of MAM proteins and their characteristics in cancers from a diagnostic perspective. Additionally, this review provides insights into diverse therapeutic strategies that target MAM proteins in various cancer types.


Assuntos
Membranas Mitocondriais , Neoplasias , Humanos , Membranas Mitocondriais/metabolismo , Mitocôndrias/metabolismo , Retículo Endoplasmático/metabolismo , Neoplasias/diagnóstico , Neoplasias/etiologia , Neoplasias/terapia , Resposta a Proteínas não Dobradas , Estresse do Retículo Endoplasmático
9.
Pestic Biochem Physiol ; 198: 105755, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38225098

RESUMO

Pyridaben is a widely used pyridazinone insecticide used to protect crops against insects and mites. The toxicity of pyridaben has been reported in mice, zebrafish, the human reproductive system, nervous system, and respiratory system. Pyridaben can also be ingested by dairy cattle through feed. However, the toxicity of pyridaben in cattle has not been investigated on. Thus, this study focuses on demonstrating the toxicity of pyridaben in the bovine mammary glands and with the generation milk in the bovine mammary epithelial cells, as it is crucial to the continuance of the amount and the quality of the milk produced. We started by analyzing the intracellular toxicity along with the impact of pyridaben on the cell cycle distribution and the transcription of associated genes. Pyridaben treatment induced cell cycle arrest accompanied the disruption in G1 and S phases with imbalanced cytosolic and mitochondrial calcium ion homeostasis, and caused a destruction of mitochondrial membrane potential. This eventually led to apoptosis of MAC-T cells. We also investigated in the impact that pyridaben has on MAPK signaling proteins, where phosphorylation of ERK1/2, JNK, and p38 were upregulateed. Moreover, examination of the effect of pyridaben in the inflammatory genes revealed hyperactivation of the inflammatory gene transcription. This is the first research to assess the negative outcomes that pyridaben could impose on dairy cattle and milk production.


Assuntos
Cálcio , Sistema de Sinalização das MAP Quinases , Piridazinas , Bovinos , Animais , Humanos , Camundongos , Cálcio/metabolismo , Regulação para Cima , Peixe-Zebra , Apoptose , Células Epiteliais , Inflamação/metabolismo , Homeostase
10.
Reprod Sci ; 31(2): 404-412, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38180609

RESUMO

This study is aimed at identifying variations in the effect of endometriosis on fecundity in a mouse model based on prior pregnancy experience. Endometriosis is one of the most prevalent gynecological diseases and is known to impact female fecundity adversely. In this study, an endometriosis mouse model was established by allografting uterine horn tissue using Pelch's method. The effect of endometriosis on fecundity was confirmed in primiparous and multiparous female mice. As fecundity indicators, the pregnancy rate, number of litters, pregnancy period, and survival rate of the pups were investigated. As a result of the experiment, the pregnancy rate decreased, and the pregnancy period tended to be shorter in primiparous female mice. However, there was no significant change in the multiparous mice. In addition, it has been established that correlations exist between the size of lesions and certain fecundity indicators of the lesion, even among primiparous and multiparous females with endometriosis. The study attempted to demonstrate a link between pregnancy experience and fecundity changes caused by endometriosis by experimentally reproducing clinical results using mouse models. These results suggest strategies for identifying several pathophysiological characteristics of endometriosis.


Assuntos
Endometriose , Infertilidade Feminina , Gravidez , Humanos , Feminino , Camundongos , Animais , Endometriose/patologia , Fertilidade/fisiologia , Infertilidade Feminina/etiologia , Taxa de Gravidez , Paridade , Modelos Animais de Doenças
11.
Phytomedicine ; 123: 155187, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-37984125

RESUMO

BACKGROUND: Fraxetin, a phytochemical obtained from Fraxinus rhynchophylla, is well known for its anti-inflammatory and anti-fibrotic properties. However, fraxetin regulates the progression of endometriosis, which is a benign reproductive disease that results in low quality of life and infertility. HYPOTHESIS/PURPOSE: We hypothesized that fraxetin may have therapeutic effects on endometriosis and aimed to elucidate the underlying mechanisms of mitochondrial function and tiRNA regulation. STUDY DESIGN: Endometriotic animal models and cells (End1/E6E7 and VK2/E6E7) were used to identify the mode of action of fraxetin. METHODS: An auto-implanted endometriosis animal model was established and the effects of fraxetin on lesion size reduction were analyzed. Cell-based assays including proliferation, cell cycle, migration, apoptosis, mitochondrial function, calcium efflux, and reactive oxygen species (ROS) were performed. Moreover, fraxetin signal transduction was demonstrated by western blotting and qPCR analyses. RESULTS: Fraxetin inhibited proliferation and migration by inactivating the P38/JNK/ERK mitogen-activated protein kinase (MAPK) and AKT/S6 pathways. Fraxetin dissipates mitochondrial membrane potential, downregulates oxidative phosphorylation (OXPHOS), and disrupts redox and calcium homeostasis. Moreover, it triggered endoplasmic reticulum stress and intrinsic apoptosis. Furthermore, we elucidated the functional role of tiRNAHisGTG in endometriosis by transfection with its inhibitor. Finally, we established an endometriosis mouse model and verified endometriotic lesion regression and downregulation of adhesion molecules with inflammation. CONCLUSION: This study suggests that fraxetin is a novel therapeutic agent that targets mitochondria and tiRNAs. This is the first study to demonstrate the mechanisms of tiRNAHisGTG with mitochondrial function and cell fates and can be applied as a non-hormonal method against the progression of endometriosis.


Assuntos
Cumarínicos , Endometriose , Humanos , Feminino , Animais , Camundongos , Espécies Reativas de Oxigênio/metabolismo , Endometriose/metabolismo , Cálcio/metabolismo , Qualidade de Vida , Proliferação de Células , Linhagem Celular , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Mitocôndrias , Apoptose
12.
Artigo em Inglês | MEDLINE | ID: mdl-37993010

RESUMO

Environmental pollution caused by pesticides is a growing concern. Pyridaben, a widely used organochlorine insecticide, is a representative water pollutant. Owing to its extensive usage, it has been detected in various aquatic ecosystems, including rivers and oceans. Pyridaben is highly toxic to aquatic organisms; however, the mechanism of its toxicity in the liver, which is important in toxicant metabolism, has not been studied. Therefore, we employed zebrafish and its well-characterized liver cell line, ZFL to assess pyridaben hepatotoxicity and explore its potential mechanisms of action. Pyridaben led to reduction of the liver size and fluorescence intensity of dsRed-labeled Tg (fabp10a:dsRed) zebrafish. It reduced the viability and proliferation of ZFL cells in vitro by inducing apoptosis and cell cycle arrest. These changes might be primarily linked to uncontrolled intracellular calcium flow in ZFL cells exposed to pyridaben. Additionally, it also downregulates the PI3K/Akt signaling cascade, leading to the inactivation of Gsk3ß and nuclear translocation of ß-catenin. Taken together, our findings suggest that pyridaben could have hepatotoxic effects on aquatic organisms. This study is the first to provide insight into the hepatotoxic mechanism of pyridaben using both in vivo and in vitro models.


Assuntos
Proteínas Proto-Oncogênicas c-akt , Peixe-Zebra , Animais , Peixe-Zebra/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Cálcio/metabolismo , Ecossistema , Hepatócitos/metabolismo , Pontos de Checagem do Ciclo Celular , Homeostase
13.
In Vivo ; 38(1): 431-436, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38148064

RESUMO

BACKGROUND/AIM: Vasopressin injected during myomectomy is known to effectively reduce bleeding but is sometimes associated with intraoperative vasoconstriction and hypertension due to systemic absorption. Although there is a growing preference for the use of diluted vasopressin, evidence of its effect and safety is still lacking. PATIENTS AND METHODS: We performed a randomized controlled pilot trial to evaluate the effect and safety of vasopressin diluted in a constant volume during robot-assisted laparoscopic myomectomy (RALM), where a total of 39 women with uterine fibroids were randomly assigned into the following three groups (group 1, 0.2 IU/ml; group 2, 0.1 IU/ml; group 3, 0.05 IU/ml with a total of 100 ml of normal saline). The primary endpoint was to compare estimated blood loss (EBL), and the secondary endpoints were to compare postoperative value and drop ratio of hemoglobin, operation time, transfusion, hospitalization, and complications among the three groups. RESULTS: There were no differences in the number and largest size of uterine fibroids, total weight of uterine fibroids, console time, and volumes of intravenous fluid administered during RALM among the three groups, whereas combined operation was performed more commonly in group 2 than in groups 1 and 3 (53.9% vs. 0 to 7.7%; p=0.01). The primary and secondary endpoints were also not different among the three groups. However, two patients in group 1 (15.4%) showed vasopressin-related hypertension. CONCLUSION: Vasopressin diluted in a volume of 100 ml showed an effective hemostatic effect and safety during RALM (Trial No. NCT04874246 in ClinicalTrial.gov).


Assuntos
Hipertensão , Laparoscopia , Leiomioma , Robótica , Miomectomia Uterina , Neoplasias Uterinas , Humanos , Feminino , Miomectomia Uterina/efeitos adversos , Neoplasias Uterinas/tratamento farmacológico , Neoplasias Uterinas/cirurgia , Projetos Piloto , Leiomioma/tratamento farmacológico , Leiomioma/cirurgia , Vasopressinas , Perda Sanguínea Cirúrgica/prevenção & controle , Laparoscopia/efeitos adversos , Hipertensão/etiologia
14.
Pestic Biochem Physiol ; 196: 105637, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37945236

RESUMO

Bifenthrin is one of the widely used synthetic pyrethroid insecticides, employed for various purposes worldwide. As lipophilic pyrethroids can easily bind to soil particles, which is why their residues are detected in various environments. Consequently, the toxicity of bifenthrin to non-target organisms can be regarded as an environmental concern. The toxic effects of bifenthrin have been studied in various animal models and cell lines; however, its toxic effects on cattle remain unclear. In particular, gaining insights into the toxic effects of bifenthrin on the mammary lactation system is crucial for the dairy industry. Therefore, we proceeded to investigate the toxic effects of bifenthrin on the bovine mammary epithelial cells (MAC-T cells). We established that bifenthrin inhibited cell proliferation and triggered apoptosis in MAC-T cells. Additionally, bifenthrin induced mitochondrial dysfunction and altered inflammatory gene expression by disrupting mitochondrial membrane potential (MMP) and generating excessive reactive oxygen species (ROS). We also demonstrated that bifenthrin disrupted both cytosolic and mitochondrial calcium ion homeostasis. Furthermore, bifenthrin altered mitogen-activated protein kinase (MAPK) signaling cascades and downregulated casein-related genes. Collectively, we confirmed the multiple toxic effects of bifenthrin on MAC-T cells, which could potentially reduce milk yield and quality.


Assuntos
Cálcio , Piretrinas , Feminino , Bovinos , Animais , Espécies Reativas de Oxigênio/metabolismo , Cálcio/metabolismo , Células Epiteliais , Piretrinas/farmacologia , Homeostase , Apoptose
15.
Pestic Biochem Physiol ; 196: 105626, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37945260

RESUMO

Bifenox (methyl 5-(2,4-dichlorophenoxy)-2-nitrobenzoate), a nitrophenyl ether herbicide, was first introduced in the 1980s to control broadleaf weeds. As a result of its wide and frequent application in diverse agricultural settings and reports on residual traces, potential adverse effects of bifenox have been studied extensively in rat hepatocytes, bovine peripheral lymphocytes, and mice. Despite the reported risks of bifenox exposure in dairy cows, the toxicity of bifenox on bovine lactation system has not been extensively investigated. Therefore, we used bovine mammary epithelial (MAC-T) cells to study the toxic effects of bifenox on mammary glands. We found that bifenox inhibited MAC-T cells proliferation and disturbed the cell cycle, especially in the sub-G1 and G1 phases. Bifenox also disrupted the calcium homeostasis within the cell and impaired mitochondrial membrane potential. We also examined phosphoinositide 3-kinase/protein kinase B (PI3K/AKT) and mitogen-activated protein kinase (MAPK) signaling cascades. The findings indicated hyperactivation of phosphorylated protein kinase B (AKT), p70 ribosomal S6 kinase (p70S6K), S6, extracellular signal-regulated kinases 1 and 2 (ERK1/2), p38, c-Jun N-terminal kinase (JNK), and c-Jun, as well as endoplasmic reticulum (ER) stress caused by bifenox treatment. In conclusion, based on our in vitro study employing MAC-T cells, we report that bifenox can induce damage to the bovine mammary glands, potentially impacting milk production.


Assuntos
Proteínas Quinases Ativadas por Mitógeno , Proteínas Proto-Oncogênicas c-akt , Feminino , Bovinos , Animais , Camundongos , Ratos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Proteínas Quinases Ativadas por Mitógeno/farmacologia , Fosfatidilinositol 3-Quinases , Cálcio/metabolismo , Células Epiteliais , Apoptose , Homeostase
16.
Pestic Biochem Physiol ; 196: 105628, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37945262

RESUMO

Bifenox is a widely used herbicide that contains a diphenyl ether group. However its global usage, the cell physiological effects that induce toxicity have not been elucidated. In this study, the effect of bifenox was examined in porcine trophectoderm and uterine epithelial cells to investigate the potential toxicity of the implantation process. To uncover the toxic effects of bifenox, cell viability and apoptosis following treatment with bifenox were evaluated. To investigate the underlying cellular mechanisms, mitochondrial and calcium homeostasis were investigated in both cell lines. In addition, the dysregulation of cell signal transduction and transcriptional alterations were also demonstrated. Bifenox reduced cell viability and significantly increased the number of cells arrested at the sub-G1 stage. Moreover, bifenox depolarized the mitochondrial membrane and upregulated the calcium flux into the mitochondria in both cell lines. Cytosolic calcium flux increased in porcine trophectoderm (pTr) cells and decreased in porcine luminal epithelium (pLE) cells. In addition, bifenox activated the mitogen-activated protein kinase and phosphoinositide 3-kinase signaling pathways. Furthermore, bifenox inhibited the expression of retinoid receptor genes, such as RXRA, RXRB, and RXRG. Chemokine CCL8 was also downregulated at the mRNA level, whereas CCL5 expression remained unchanged. Overall, the results of this study suggest that bifenox deteriorates cell viability by arresting cell cycle progression, damaging mitochondria, and controlling calcium levels in pTr and pLE cells. The present study indicates the toxic potential of bifenox in the trophectoderm and luminal epithelial cells, which can lead to implantation disorders in early pregnancy.


Assuntos
Cálcio , Fosfatidilinositol 3-Quinases , Gravidez , Feminino , Suínos , Animais , Cálcio/metabolismo , Proliferação de Células , Éteres Fenílicos/metabolismo , Éteres Fenílicos/farmacologia , Mitocôndrias/metabolismo , Células Epiteliais , Ciclo Celular , Homeostase
17.
Artigo em Inglês | MEDLINE | ID: mdl-37659609

RESUMO

Release of agrochemicals from agricultural fields could unintentionally harm organisms that not targeted by pesticides. Flufenacet is one of the oxyacetamide herbicide applied in cultivation fields of crops and this has a possibility of unintentional exposure to diverse ecosystems including streams and surface water. Despite these environmental risks, limited information regarding toxicity of flufenacet on vertebrates is available. This study is aimed to assess environmental hazards and underlying toxic mechanisms of flufenacet by using a zebrafish model. Mortality measurements and morphological observations after the treatment of flufenacet suggested developmental toxicity of flufenacet in zebrafish. In addition, its toxicity on specific organs was evaluated using transgenic fluorescent zebrafish embryo. Adverse effects of flufenacet on vascular and hepatopancreatic development were demonstrated using Tg(flk1:EGFP) and Tg(fabp10a:DsRed; ela3l:EGFP) respectively. To address intracellular actions of flufenacet in zebrafish, cellular responses including apoptosis, cell cycle modulation, and Mapk and Akt signaling pathway were verified in transcriptional and protein levels. These results demonstrated developmental toxicity of flufenacet using the zebrafish model, providing essential information for assessing its potential hazards on vertebrates that are not directly targeted by the pesticide and for elucidating molecular mechanisms.


Assuntos
Fosfatidilinositol 3-Quinases , Peixe-Zebra , Animais , Proteínas Proto-Oncogênicas c-akt , Ecossistema , Fígado , Pâncreas , Apoptose , Transdução de Sinais
18.
Artigo em Inglês | MEDLINE | ID: mdl-37689173

RESUMO

Dimethenamid, one of the acetamide herbicides, is widely used on soybeans and corns to inhibit weed growth. Although other acetamide herbicides have been reported to have several toxicities in non-target organisms including developmental toxicity, the toxicity of dimethenamid has not yet been studied. In this research, we utilized the zebrafish animal model to verify the developmental toxicity of dimethenamid. It not only led to morphological abnormalities in zebrafish larvae but also reduced their viability. ROS production and inflammation responses were promoted in zebrafish larvae. Also, uncontrolled apoptosis occurred when the gene expression level related to the cell cycle and apoptosis was altered by dimethenamid. These changes resulted in toxicities in the cardiovascular system, liver, and pancreas are observed in transgenic zebrafish models including fli1a:EGFP and L-fabp:dsRed;elastase:GFP. Dimethenamid triggered morphological defects in the heart and vasculature by altering the mRNA levels related to cardiovascular development. The liver and pancreas were also damaged through not only the changes of their morphology but also through the dysregulation in their function related to metabolic activity. This study shows the developmental defects induced by dimethenamid in zebrafish larvae and the possibility of toxicity in other non-target organisms.


Assuntos
Sistema Cardiovascular , Peixe-Zebra , Animais , Pâncreas , Fígado , Estresse Oxidativo , Acetamidas , Apoptose
19.
Pharmaceuticals (Basel) ; 16(9)2023 Aug 29.
Artigo em Inglês | MEDLINE | ID: mdl-37765029

RESUMO

Olfactory receptors are expressed in multiple extra-nasal tissues and these ectopic olfactory receptors mediate tissue-specific functions and regulate cellular physiology. Ectopic olfactory receptors may play key roles in tissues constantly exposed to odorants, thus the functionality of these receptors in genital tissues is of particular interest. The functionality of ectopic olfactory receptors expressed in VK2/E6E7 human vaginal epithelial cells was investigated. OR2H2 was the most highly expressed olfactory receptor expressed in VK2/E6E7 cells, and activation of OR2H2 by aldehyde 13-13, a ligand of OR2H2, increased the intracellular calcium and cAMP concentrations. Immunoblotting demonstrated that activation of OR2H2 by aldehyde 13-13 stimulated the CAMKKß-AMPK-mTORC1-autophagy signaling axis, and that these effects were negated by OR2H2 knockdown. AMPK is known to regulate senescence; consequently, we investigated further the effect of aldehyde 13-13 on senescence. In H2O2-induced senescent cells, activation of OR2H2 by aldehyde 13-13 restored proliferation, and reduced the expression of senescence markers, P16 and P19. Additionally, aldehyde 13-13 induced apoptosis of H2O2-induced senescent cells, compared with non-senescent normal cells. In vivo, aldehyde 13-13 increased the lifespan of Caenorhabditis elegans and budding yeast. These findings demonstrate that OR2H2 is a functional receptor in VK2/E6E7 cells, and that activation of OR2H2 activates the AMPK-autophagy axis, and suppresses cellular aging and senescence, which may increase cellular health.

20.
Artigo em Inglês | MEDLINE | ID: mdl-37597713

RESUMO

The dinitroaniline herbicide benfluralin is used weed control in conventional systems and poses a high risk of accumulation in aquatic systems. Previous studies have shown the toxic effects of benfluralin on non-target organisms; however, its developmental toxicity in vertebrates has not yet been reported. This study demonstrated the developmental toxicity of benfluralin and its mechanism of action, using zebrafish as an aquatic vertebrate model. Benfluralin induces morphological and physiological alterations in body length, yolk sac, and heart edema. We also demonstrated a reactive oxygen species (ROS) increase of approximately 325.53 % compared with the control group after 20 µM benfluralin-treatment. In addition, the malformation of the heart and vascular structures was identified using transgenic flk1:eGFP zebrafish models at 20 µM concentration benfluralin exposure. Moreover, benfluralin induced small livers, approximately 59.81 % of normal liver size, via abnormal development of the liver as observed in the transgenic L-fabp:dsRed zebrafish. Benfluralin also inhibits normal growth via abnormal expression of cell cycle regulatory genes and increases oxidative stress, inflammation, and apoptosis. Collectively, we elucidated the mechanisms associated with benfluralin toxicity, which lead to various abnormalities and developmental toxicities in zebrafish. Therefore, this study provides information on the parameters used to assess developmental toxicity in other aquatic organisms, such as herbicides, pesticides, and environmental contaminants.


Assuntos
Herbicidas , Peixe-Zebra , Animais , Espécies Reativas de Oxigênio , Apoptose , Animais Geneticamente Modificados , Desenvolvimento Embrionário , Herbicidas/toxicidade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...