Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
1.
J Pineal Res ; 74(1): e12842, 2023 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-36401340

RESUMO

Endometriosis is a benign gynecological disease characterized by abnormal growth of endometrial-like cells outside the uterus. Melatonin, a hormone secreted by the pineal gland, has been shown to have therapeutic effects in various diseases, including endometriosis. However, the underlying molecular mechanisms are yet to be elucidated. The results of this study demonstrated that melatonin and dienogest administration effectively reduced surgically induced endometriotic lesions in a mouse model. Melatonin suppressed proliferation, induced apoptosis, and dysregulated calcium homeostasis in endometriotic cells and primary endometriotic stromal cells. Melatonin also caused mitochondrial dysfunction by permeating through the mitochondrial membrane to disrupt redox homeostasis in the endometriotic epithelial and stromal cells. Furthermore, melatonin affected oxidative phosphorylation systems to decrease ATP production in End1/E6E7 and VK2/E6E7 cells. This was achieved through messenger RNA-mediated downregulation of respiratory complex subunits. Melatonin inhibited the PI3K/AKT and ERK1/2 pathways and the mitochondria-associated membrane axis and further suppressed the migration of endometriotic epithelial and stromal cells. Furthermore, we demonstrated that tiRNAGluCTC and tiRNAAspGTC were associated with the proliferation of endometriosis and that melatonin suppressed the expression of these tiRNAs in primary endometriotic stromal cells and lesions in a mouse model. Thus, melatonin can be used as a novel therapeutic agent to manage endometriosis.


Assuntos
Endometriose , Melatonina , Animais , Feminino , Camundongos , Proliferação de Células , Endometriose/tratamento farmacológico , Endometriose/metabolismo , Endometriose/patologia , Endométrio/metabolismo , Endométrio/patologia , Melatonina/metabolismo , Mitocôndrias/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo
2.
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
3.
J Cell Physiol ; 236(2): 1545-1558, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33000501

RESUMO

The therapeutic potential of α,ß-thujone, a functional compound found in many medicinal plants of the Cupressaceae, Asteraceae, and Lamiaceae families, has been demonstrated, including in inflammation and cancers. However, its pharmacological functions and mechanisms of action in ovarian cancer remain unclear. We investigated the anticancer properties of α,ß-thujone in ES2 and OV90 human ovarian cancer cells and its effect on sensitization to cisplatin. α,ß-thujone inhibited cancer cell proliferation and induced cell death through caspase-dependent intrinsic apoptotic pathways. Moreover, α,ß-thujone-mediated endoplasmic reticulum stress was associated with the loss of mitochondrial functions and altered metabolic landscape of ovarian cancer cells. α,ß-Thujone attenuated blood vessel formation in transgenic zebrafish, implying it has significant antiangiogenic potential. In addition, α,ß-thujone sensitized ovarian cancer cells to cisplatin, causing synergistic pharmacological effects. Collectively, our results suggest that α,ß-thujone has therapeutic potential in human ovarian cancer and functions via regulating multiple intracellular stress-associated metabolic reprogramming and caspase-dependent apoptotic pathways.


Assuntos
Monoterpenos Bicíclicos/farmacologia , Proliferação de Células/efeitos dos fármacos , Reprogramação Celular/genética , Neoplasias Ovarianas/tratamento farmacológico , Animais , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Cisplatino/farmacologia , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Feminino , Humanos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/genética , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/patologia , Ovário/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Peixe-Zebra/genética
4.
Pestic Biochem Physiol ; 177: 104893, 2021 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-34301355

RESUMO

Pesticides, which are used in agriculture and forestry to eliminate insects, are a major cause of environmental pollution. Among them, diflubenzuron (DFB), 1-(4-chlorophenyl)-3-(2,6-difluorobenzoyl) urea, is a common benzoylurea insecticide that hinders larval development, primarily in Aedes aegypti larvae. Many experts have announced the biological toxicity of DFB in various species. However, the toxicity of benzoylurea pesticides, including DFB, to bovine mammary epithelial cells (MAC-T) is unclear. Therefore, in this study, we confirmed the cytotoxic effects of DFB on the viability and proliferation of MAC-T cells. Additionally, we observed that DFB induced lipid peroxidation through reactive oxygen species (ROS) production, resulting in an increase in transcriptional gene expression related to inflammatory response. Moreover, we demonstrated mitochondrial dysfunction including depolarization of the mitochondrial membrane, perturbation of calcium homeostasis, and, eventually, apoptosis. Furthermore, we identified DFB-triggered signaling pathways related to ROS generation and cell proliferation, as well as their interactions, by treating the cells with pharmacological inhibitors in combination with DFB. DFB attenuated the phosphorylation of AKT, P70S6K, S6, and ERK1/2 and facilitated the phosphorylation of JNK and c-Jun. These results show that DFB can induce apoptotic cell death via ROS generation and mitochondrial dysfunction in MAC-T cells.


Assuntos
Diflubenzuron , Animais , Apoptose , Bovinos , Diflubenzuron/metabolismo , Células Epiteliais , Mitocôndrias , Espécies Reativas de Oxigênio/metabolismo
5.
Pestic Biochem Physiol ; 172: 104751, 2021 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-33518044

RESUMO

Benfuresate (2,3-dihydro-3,3-dimethylbenzofuran-5-yl ethanesulphonate) is a widely used pre-emergence herbicide of the benzofurane group, which works through the inhibition of lipid synthesis. During embryonic development of zebrafish, benfuresate retards growth while causing internal changes in the body, including alteration of the expression of cell cycle regulators, induction of apoptosis, and suppression of the circulatory system. Acute toxicity towards benfuresate is seen across the range of 5-15 µM in a dose-dependent manner and contributes to pathological conditions and subsequent morphological changes. For embryos 120 h post fertilization (hpf), benfuresate exposure results in an array of malformations involving eye or otolith development, pericardial edema, yolk sac edema, and abnormal curvature of the spine. Mechanistically, benfuresate exposure altered the transcription levels of the proliferative pathway genes ccnd1, ccne1, cdk2, and cdk6, all of which sensitize cells to apoptosis. Benfuresate exposure also affected vascular formation, including the formation of various vessels (DA, SIVs, CA, CV) whose functions in lymphatic-blood circulation were disrupted following decreased vegfaa, vegfc, flt1, flt4, and kdrl expression. These findings provide evidence of embryo-larval toxicity due to benfuresate and highlight the perils of herbicide exposure for non-target organisms far removed from application sites, especially in aquatic environments.


Assuntos
Poluentes Químicos da Água , Peixe-Zebra , Animais , Apoptose , Benzofuranos , Embrião não Mamífero , Larva , Ácidos Sulfônicos , Peixe-Zebra/genética
6.
Reproduction ; 159(6): 745-756, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32240978

RESUMO

α,ß-Thujone is a natural terpenoid found in many medicinal herbs, such as Artemisia absinthium (wormwood), that exhibits antioxidant, anti-diabetic, and anti-tumorigenic effects. α,ß-Thujone has numerous functions; it serves as a food ingredient, cosmetic additive, and medicinal remedy. Although the therapeutic properties of α,ß-thujone were previously revealed, a comprehensive description of the mechanisms of its anti-cancer potential in choriocarcinoma is yet to be provided. To our knowledge, this study is the first to demonstrate that α,ß-thujone attenuates JEG3 and JAR choriocarcinoma cells through a caspase-dependent intrinsic apoptotic pathway. Moreover, α,ß-thujone was demonstrated to induce a global mitochondrial defect and ER stress in choriocarcinoma by causing mitochondrial depolarization, calcium overload, and metabolic alterations, thereby leading to energy deprivation, which eventually contributes to the increase in apoptosis of choriocarcinoma cells. Herein, we also revealed the synergistic anti-cancer activity of α,ß-thujone via its sensitization effect on paclitaxel in choriocarcinoma cells. Altogether, our findings suggest that α,ß-thujone is a novel, natural pharmacological compound that can be used to treat human placental choriocarcinoma.


Assuntos
Monoterpenos Bicíclicos/farmacologia , Proliferação de Células/efeitos dos fármacos , Coriocarcinoma/patologia , Placenta/efeitos dos fármacos , Neoplasias Uterinas/patologia , Apoptose/efeitos dos fármacos , Cálcio/metabolismo , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Coriocarcinoma/metabolismo , Feminino , Humanos , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Placenta/metabolismo , Placenta/patologia , Gravidez , Transdução de Sinais/efeitos dos fármacos , Neoplasias Uterinas/metabolismo
7.
Med Mycol ; 58(1): 71-82, 2020 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-30843033

RESUMO

Ochratoxin A (OTA) is a mycotoxin originating from Penicillium and Aspergillus. In addition to toxic effects in various tissues and cells, including neurons, immune cells, hepatocytes, and nephrons, it also causes carcinogenesis and teratogenesis. Although the negative effects of OTA with respect to the pathogenesis of diseases and the malfunction of various organs have been studied widely, the biological signaling mechanisms in testicular cells are less well known. Therefore, we determined the hazardous effect of OTA in two types of testicular cells: TM3 (mouse Leydig cells) and TM4 (mouse Sertoli cells). Treatment with OTA led to a significant decrease in the proliferation of both cell lines, as revealed by an increased proportion of cells in the sub-G1 phase. In addition, the phosphorylation of signaling molecules belonging to the PI3K (Akt, P70S6K, and S6) and MAPK (ERK1/2 and JNK) pathways was regulated by OTA in a dose-dependent manner in TM3 and TM4 cells. Furthermore, the combination treatment of OTA and signaling inhibitors (LY294002, U0126, or SP600125) exerted synergistic antiproliferative effects in TM3 and TM4 cells. OTA also reduced the concentration of calcium ions in the cytosol and mitochondria, which disrupted the calcium homeostasis necessary for maintaining the normal physiological functions of testicular cells. In conclusion, the results of the present study demonstrate the mechanism underlying the antiproliferative effects of OTA in mouse testicular cells. Exposure to OTA may result in abnormal sperm maturation and the failure of spermatogenesis, which leads to male infertility.


Assuntos
Proliferação de Células/efeitos dos fármacos , Células Intersticiais do Testículo/efeitos dos fármacos , Micotoxinas/toxicidade , Ocratoxinas/toxicidade , Células de Sertoli/efeitos dos fármacos , Animais , Ciclo Celular/efeitos dos fármacos , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Homeostase/efeitos dos fármacos , Masculino , Camundongos , Fosforilação , Transdução de Sinais
8.
Pestic Biochem Physiol ; 163: 84-93, 2020 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-31973874

RESUMO

Ivermectin (IVM) is a commercially well-known antiparasitic agent derived from the natural fermentation product avermectin. Originally used as a veterinary drug, IVM has been studied for its pharmacokinetic advantages, such as anticancer, antimigration, and antiproliferative effects, using several cell types. In the present study, we verified that IVM suppressed bovine mammary gland epithelial cell proliferation and induced the arrest of the cell cycle from the sub-G1 to the G2/M phase in these cells. Due to IVM treatment, the homeostasis of calcium ions, which play a crucial role in intracellular metabolism, deteriorated, leading to the loss of the mitochondrial membrane potential (MMP). To underpin these results, further studies using inhibitors of Ca2+ signaling were performed; combination treatment with IVM and these factors, including 2-APB, BAPTA-AM, or ruthenium red, inhibited the IVM-induced MMP disruption. Furthermore, following IVM treatment, the relationships among various cell signaling mediators were altered, and the balance between diverse cellular processes associated with cell survival or death was disturbed. In conclusion, we assessed the anti-survival effects of IVM on mammary gland epithelial cells; IVM may impede normal lactation in dairy cows.


Assuntos
Antiparasitários , Ivermectina , Animais , Apoptose , Bovinos , Células Epiteliais , Feminino , Potencial da Membrana Mitocondrial
9.
J Cell Physiol ; 233(9): 7457-7466, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29663365

RESUMO

Canine osteosarcoma is an aggressive primary bone tumor that shows metastasis to distal regions and is associated with a high mortality rate. However, the pathophysiological mechanisms of canine osteosarcoma are not well characterized. In addition, development of prognostic factors and novel therapeutic agents is necessary to efficiently treat osteosarcoma. Therefore, we studied the effects of myricetin, an antioxidant found in berries, nuts, teas, wine, and vegetables, on apoptosis and signal transduction in the canine osteosarcoma cell lines, D-17 and DSN. Results of the present study demonstrated that treatment with myricetin decreased cell proliferation and DNA replication, while it increased apoptotic DNA fragmentation in D-17 and DSN cells. In addition, it increased generation of ROS, lipid peroxidation, and depolarization of MMP in both D-17 and DSN cells. Myricetin treatment activated phosphorylation of AKT, p70S6K, ERK1/2, JNK, and p90RSK in canine osteosarcoma cells. Moreover, inhibition of PI3K and MAPK using LY294002, U0126, or SP600125, in addition to myricetin treatment, effectively suppressed cell proliferation compared to treatment with myricetin or each inhibitor alone. Therefore, we concluded that myricetin may be a potentially effective and less toxic therapeutic agent to prevent and control progression of canine osteosarcoma.


Assuntos
Apoptose/efeitos dos fármacos , Fragmentação do DNA/efeitos dos fármacos , Flavonoides/farmacologia , Homeostase , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Osteossarcoma/patologia , Animais , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Cães , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Modelos Biológicos , Osteossarcoma/enzimologia , Oxirredução , Estresse Oxidativo/efeitos dos fármacos , Fosfatidilinositol 3-Quinases/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais/efeitos dos fármacos , Bibliotecas de Moléculas Pequenas/farmacologia
10.
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.


Assuntos
Embrião não Mamífero , Espécies Reativas de Oxigênio , Transdução de Sinais , Peixe-Zebra , Animais , Peixe-Zebra/embriologia , Embrião não Mamífero/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Animais Geneticamente Modificados , Herbicidas/toxicidade , Fígado/efeitos dos fármacos , Fígado/metabolismo , Fígado/patologia , Fígado/embriologia , Doença Hepática Induzida por Substâncias e Drogas/patologia , Doença Hepática Induzida por Substâncias e Drogas/metabolismo , Éteres Difenil Halogenados/toxicidade
11.
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.


Assuntos
Animais Geneticamente Modificados , Embrião não Mamífero , Herbicidas , Peixe-Zebra , Animais , Peixe-Zebra/embriologia , Embrião não Mamífero/efeitos dos fármacos , Herbicidas/toxicidade , Apoptose/efeitos dos fármacos , Desenvolvimento Embrionário/efeitos dos fármacos , Poluentes Químicos da Água/toxicidade
12.
Sci Total Environ ; 867: 161535, 2023 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-36638995

RESUMO

Fluridone is a systemic herbicide used to control a range of invasive aquatic plants in irrigation systems, lake, and reservoirs. Since aquatic herbicides are more likely to have a hazardous impact on ecosystems than terrestrially applied herbicides, a risk assessment is needed to determine whether to expand or limit their use. The aim of this study was to investigate the developmental toxicity of fluridone using zebrafish. Diverse toxicological results were observed for the sub-lethal endpoints, including lack of hatching, reduced heartbeat and disturbed blood circulation through dysmorphic heart, and edema formation. Abnormal apoptosis was observed in the brain and yolk sac of fluridone-exposed larvae. A computational analysis was used to predict chemical properties in non-target organisms and revealed that fluridone was highly relevant in the cardiovascular system. Double transgenic zebrafish (fli1a:EGFP;cmlc2:dsRed) were used to evaluate the effects of fluridone on the cardiovascular system during embryonic development. Ectopic growth of sub-intestinal vessels and sprouting angiogenesis in the hindbrain region were highly inhibited. Additionally, essential genes involved in the VEGF signaling and heart development were differentially expressed in dose-dependent manner. Collectively, our toxicological findings in fluridone exposure highlight defects in the cardiovascular development causing embryonic lethality that could damage aquatic communities and natural ecosystems.


Assuntos
Sistema Cardiovascular , Herbicidas , Poluentes Químicos da Água , Animais , Peixe-Zebra , Herbicidas/toxicidade , Ecossistema , Embrião não Mamífero , Poluentes Químicos da Água/toxicidade , Larva
13.
Sci Total Environ ; 862: 160761, 2023 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-36502969

RESUMO

Various types of fungicides, especially triazole fungicides, are used to prevent fungal diseases on farmlands. However, the developmental toxicity of one of the triazole fungicides, triadimenol, remains unclear. Therefore, we used the zebrafish animal model, a representative toxicological model, to investigate it. Triadimenol induced morphological alterations in the eyes and body length along with yolk sac and heart edema. It also stimulated the production of reactive oxygen species and expression of inflammation-related genes and caused apoptosis in the anterior regions of zebrafish, especially in the heart. The phosphorylation levels of Akt, ERK, JNK, and p38 proteins involved in the PI3K and MAPK pathways, which are important for the development process, were also reduced by triadimenol. These changes led to malformation of the heart and vascular structures, as observed in the flk1:eGFP transgenic zebrafish models and a reduction in the heart rate. In addition, the expression of genes associated with cardiac and vascular development was also reduced. Therefore, we elucidated the mechanisms associated with triadimenol toxicity that leads to various abnormalities and developmental toxicity in zebrafish.


Assuntos
Cardiotoxicidade , Fungicidas Industriais , Espécies Reativas de Oxigênio , Peixe-Zebra , Animais , Apoptose/efeitos dos fármacos , Cardiotoxicidade/metabolismo , Embrião não Mamífero , Fungicidas Industriais/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , Triazóis/metabolismo , Peixe-Zebra/metabolismo
14.
Sci Total Environ ; 859(Pt 2): 160458, 2023 Feb 10.
Artigo em Inglês | MEDLINE | ID: mdl-36435248

RESUMO

Oxamyl, a carbamate insecticide, is mainly used to control nematodes in the agricultural field. Although oxamyl is a widely used insecticide that is associated with ecological concerns, limited studies have examined the toxic effects of oxamyl on the developmental stage and the underlying mechanisms. In this study, the developmental toxicity of oxamyl was demonstrated using zebrafish, which is a representative model as it is associated with rapid embryogenesis and a toxic response similar to that of other vertebrates. The morphological alteration of zebrafish larvae was analyzed to confirm the sub-lethal toxicity of oxamyl. Analysis of transgenic zebrafish (olig2:dsRED and flk1:eGFP line) and mRNA levels of genes associated with individual organ development revealed that oxamyl exerted toxic effects on the development of neuron, notochord, and vascular system. Next, the adverse effect of oxamyl on the mitochondrial electron transport chain was examined. Treatment with oxamyl altered the PI3K/Akt signaling and p38 Mapk signaling pathways in zebrafish. Thus, this study elucidated the mechanisms underlying the developmental toxicity of oxamyl and provided information on the parameters to assess the developmental toxicity of other environmental contaminants.


Assuntos
Inseticidas , Peixe-Zebra , Animais , Peixe-Zebra/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Transporte de Elétrons , Carbamatos/metabolismo , Inseticidas/metabolismo , Embrião não Mamífero
15.
Sci Total Environ ; 854: 158780, 2023 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-36115403

RESUMO

Ethalfluralin, of dinitroaniline herbicide family, is an effective weed controller. Following residue detection in herbicide-treated fields, ethalfluralin was reported to interfere with early stages of implantation in some vertebrate species. However, the role of ethalfluralin in the development of zebrafish embryos has not been elucidated yet. Therefore, in the present study, we investigated the morphological and physiological changes that occur in the embryonic development of zebrafish due to ethalfluralin exposure. Results indicated that ethalfluralin decreased survival rate along with reduction in the hatching ratio and heartbeat. It was observed to cause edema in the heart and yolk sac, and apoptosis in the anterior region of the developing zebrafish larvae; as visualized through acridine orange and TUNEL staining. In addition, ethalfluralin increased the expression of the apoptosis-associated genes including tp53, cyc1, casp8, casp9, and casp3. The Seahorse Mito Stress analysis revealed that ethalfluralin slightly reduced mitochondrial respiration in live zebrafish embryos. Reactive oxygen species (ROS) production was also observed to be elevated in zebrafish larvae in response to ethalfluralin. Treatment with ethalfluralin decreased blood vessel formation in brain and intestine in flk1 transgenic zebrafish embryos. The decrease in angiogenesis related gene expression was specifically observed in vegfc, flt1, and kdrl, and in the intestinal vasculature related genes apoa4a, aqp3, fabp2, and vil1. Moreover, an increase in inflammatory genes such as cox2a, cox2b, cxcl-c1c, il8, mcl1a, mcl1b, and nf-κb was observed using real-time PCR analysis. Collectively, these results indicate that oxidative stress generated by exposure to ethalfluralin induced ROS generation, apoptosis, inflammation and anti-angiogenic effects, and therefore, ethalfluralin may be toxic to the development of zebrafish embryos.


Assuntos
Herbicidas , Poluentes Químicos da Água , Animais , Peixe-Zebra , Espécies Reativas de Oxigênio/metabolismo , Estresse Oxidativo , Herbicidas/metabolismo , Inflamação/induzido quimicamente , Embrião não Mamífero , Apoptose , Poluentes Químicos da Água/metabolismo
16.
J Hazard Mater ; 443(Pt A): 130202, 2023 02 05.
Artigo em Inglês | MEDLINE | ID: mdl-36272374

RESUMO

Prometryn, 2-methylthio-4,6-bis(isopropylamino)-1,3,5-triazine, is a selective thiomethyl triazine herbicide widely used to control unwanted weeds and harmful insects by inhibiting electron transport in target organisms. Despite having various advantages, herbicides pose as a major threat to the environment and human health due to persistent contamination, bioaccumulation, and damage to non-target organisms. In this study, the developmental toxicity of 5, 10, and 20 mg/L prometryn in zebrafish (Danio rerio) embryos was evaluated and compared to that of the solvent control for 96 h. Several transgenic zebrafish models (fli1a:eGFP, flk1:eGFP, olig2:dsRed and L-fabp:dsRed) were visually assessed to detect fluorescently tagged genes. Results showed that prometryn shortened body length, and induced yolk sac, heart edema, abnormal heart rate, and loss of viability. Fluorescence microscopy revealed that prometryn exposure caused defects in organ development, reactive oxygen species accumulation, and apoptotic cell death. Mitochondrial bioenergetics were also evaluated to determine the effect of prometryn on the electron transport chain activity and metabolic alterations. Prometryn was found to interfere with mitochondrial function, ultimately inhibiting energy metabolism and embryonic development. Collectively, our findings suggest that prometryn is a potential contaminate for non-target sites and organisms, especially aquatic, and emphasize the need to consider the toxic effects of prometryn.


Assuntos
Herbicidas , Poluentes Químicos da Água , Animais , Humanos , Prometrina/metabolismo , Prometrina/farmacologia , Peixe-Zebra/metabolismo , Estresse Oxidativo , Herbicidas/metabolismo , Mitocôndrias , Organogênese , Embrião não Mamífero , Poluentes Químicos da Água/metabolismo
17.
Artigo em Inglês | MEDLINE | ID: mdl-37290698

RESUMO

The zebrafish is a prominent vertebrate model popularly used for toxicity testing because of its rapid development and transparent embryos. Fluchloralin, a dinitroaniline herbicide used to control weeds, inhibits microtubule formation and cell division. The structurally homologous substances ethalfluralin and pendimethalin, which belong to the dinitroaniline family, were found to be genotoxic and to exert developmental toxicity via mitochondrial dysfunction in a zebrafish model. To date, developmental toxicity of fluchloralin in zebrafish has not been reported. In the present study, we identified morphological changes in developing zebrafish, including decreased survival rate and body length, and increased yolk sac edema. In dose-dependent response to fluchloralin exposure, inhibition of neurogenesis in the spinal cord and motor neuron defects were observed in transgenic zebrafish models (olig2:dsRed). Zebrafish exposed to fluchloralin also displayed organ dysfunction in the heart, liver, and pancreas in cmlc2:dsRed and lfabp:dsRed;elastase:GFP transgenic models. Fluchloralin increased cell death in the brain by promoting apoptosis, visualized via acridine orange staining, and by activating apoptosis signaling proteins, including cytochrome c1, zBax, and Bcl-XL. This study provides novel evidence supporting the necessity of controlling pollutants in aquatic environments.


Assuntos
Poluentes Químicos da Água , Peixe-Zebra , Animais , Peixe-Zebra/fisiologia , Embrião não Mamífero/metabolismo , Fígado , Sistema Nervoso , Desenvolvimento Embrionário , Poluentes Químicos da Água/toxicidade , Poluentes Químicos da Água/metabolismo
18.
Sci Total Environ ; 893: 164839, 2023 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-37329922

RESUMO

Terbutryn (2-(ethylamino)-4-(tert-butylamino)-6-(methylthio)-1,3,5-triazine) is a substituted symmetrical triazine herbicide used in agricultural fields to prevent undesired vegetation growth by inhibiting photosynthesis in target weeds. Although terbutryn has various benefits, long-term exposure, misuse, or abuse of terbutryn may cause non-target toxicity and severe ecosystem pollution. To provide a detailed description of the embryonic developmental toxicity of terbutryn, zebrafish (Danio rerio) were exposed to 2, 4, and 6 mg/L of terbutryn and the morphological changes, pathological abnormalities, and developmental endpoints were assessed relative to that of a solvent control. The results showed that terbutryn induces a loss of survivability, reduction in body and eye size, and edema in the yolk sac. Through fluorescence microscopy, blood vessels, motor neurons, and liver development were investigated using transgenic zebrafish models based on fluorescently tagged genes (fllk1:eGFP, olig2:dsRed, and L-fabp:dsRed). Furthermore, cell death by apoptosis in zebrafish caused by terbutryn exposure was evaluated via acridine orange staining, which is a selective fluorescent staining agent. To support the preceding results, gene expression alterations caused by terbutryn exposure in zebrafish larvae were assessed. The overall results indicate that exposure to terbutryn induces apoptosis and disrupts organ development. These embryonic developmental toxicity results suggest that terbutryn should be applied in the right areas at the appropriate rates, concentrations, and quantities.


Assuntos
Poluentes Químicos da Água , Peixe-Zebra , Animais , Peixe-Zebra/fisiologia , Ecossistema , Triazinas/metabolismo , Apoptose , Desenvolvimento Embrionário , Embrião não Mamífero/fisiologia , Larva , Poluentes Químicos da Água/toxicidade , Poluentes Químicos da Água/metabolismo
19.
Environ Int ; 176: 107973, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-37196567

RESUMO

Thiabendazole, a benzimidazole fungicide, is widely used to prevent yield loss in agricultural land by inhibiting plant diseases derived from fungi. As thiabendazole has a stable benzimidazole ring structure, it remains in the environment for an extended period, and its toxic effects on non-target organisms have been reported, indicating the possibility that it could threaten public health. However, little research has been conducted to elucidate the comprehensive mechanisms of its developmental toxicity. Therefore, we used zebrafish, a representative toxicological model that can predict toxicity in aquatic organisms and mammals, to demonstrate the developmental toxicity of thiabendazole. Various morphological malformations were observed, including decreased body length, eye size, and increased heart and yolk sac edema. Apoptosis, reactive oxygen species (ROS) production, and inflammatory response were also triggered by thiabendazole exposure in zebrafish larvae. Furthermore, PI3K/Akt and MAPK signaling pathways important for appropriate organogenesis were significantly changed by thiabendazole. These results led to toxicity in various organs and a reduction in the expression of related genes, including cardiovascular toxicity, neurotoxicity, and hepatic and pancreatic toxicity, which were detected in flk1:eGFP, olig2:dsRED, and L-fabp:dsRed;elastase:GFP transgenic zebrafish models, respectively. Overall, this study partly determined the developmental toxicity of thiabendazole in zebrafish and provided evidence of the environmental hazards of this fungicide.


Assuntos
Fungicidas Industriais , Poluentes Químicos da Água , Animais , Peixe-Zebra/metabolismo , Tiabendazol/toxicidade , Tiabendazol/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Fungicidas Industriais/toxicidade , Embrião não Mamífero , Estresse Oxidativo , Apoptose , Poluentes Químicos da Água/metabolismo , Mamíferos/metabolismo
20.
Artigo em Inglês | MEDLINE | ID: mdl-36965842

RESUMO

Chloroxylenol is an extensively consumed anti-microbial compound. Since its usage is on the rise due to the coronavirus pandemic and ban on other antimicrobial ingredients, recent studies have suggested the necessity of estimating its potential for ecotoxicity. The detrimental effect of chloroxylenol on zebrafish (Danio rerio) viability has been reported; however, research on the mechanisms underlying its toxicity is quite limited. Therefore, we applied the zebrafish model for elucidating responses against chloroxylenol to predict its toxicity toward human health and ecology. Zebrafish exposed to chloroxylenol (0, 0.5, 1, 2.5, 5, and 10 mg/L) at the embryonic stage (from 6 h post-fertilization (hpf) to 96 hpf) showed impaired viability and hatchability, and pathological phenotypes. To address these abnormalities, cellular responses such as oxidative stress, inflammation, and apoptosis were confirmed via in vivo imaging of a fluorescent dye or measurement of the transcriptional changes related to each response. In particular, developmental defects in the cardiovascular system of zebrafish exposed to 0, 0.5, 1, and 2.5 mg/L of chloroxylenol from 6 to 96 hpf were identified by structural analyses of the system in the flk1:eGFP transgenic line. Additional experiments were conducted using human umbilical vein endothelial cells (HUVECs) to predict the adverse impacts of chloroxylenol on the human vascular system. Chloroxylenol impairs the viability and tube formation ability of HUVECs by modulating ERK signaling. The findings obtained using the zebrafish model provide evidence of the possible risks of chloroxylenol exposure and suggest the importance of more in-depth ecotoxicological studies.


Assuntos
Sistema Cardiovascular , Peixe-Zebra , Animais , Humanos , Células Endoteliais , Embrião não Mamífero/metabolismo , Estresse Oxidativo , Inflamação/induzido quimicamente , Inflamação/metabolismo , Apoptose
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa