Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 320
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
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
2.
Reproduction ; 165(5): 491-505, 2023 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-36847425

RESUMO

In brief: Pendimethalin as a dinitroaniline herbicide is used to eliminate weeds during the cultivation of various crops such as grains, fruits, and vegetables. This study reveals that pendimethalin exposure at various concentrations led to disruption in Ca2+ homeostasis and mitochondrial membrane potential, as well as dysregulation of the mitogen-activated protein kinase signaling pathway and implantation-related genes in porcine trophectoderm and uterine luminal epithelial cells. Abstract: The use of herbicides is a major control method in agriculture. Pendimethalin (PDM) has been increasingly used as a herbicide for approximately 30 years. PDM has been reported to cause various reproductive problems, but its toxicity mechanism in the pre-implantation stage has not been investigated in detail. Herein, we studied the effects of PDM on porcine trophectoderm (pTr) and uterine luminal epithelial (pLE) cells and identified a PDM-mediated anti-proliferative effect in both cell types. PDM exposure generated intracellular reactive oxygen species, induced excessive Ca2+ influx into mitochondria, and activated mitogen-activated protein kinase signaling pathway. Ca2+ burden resulted in the dysfunction of mitochondria and eventual disruption of Ca2+ homeostasis. Further, PDM-exposed pTr and pLE cells showed cell cycle arrest and programmed cell death. In addition, a decrease in migration ability and dysregulated expression of genes related to the functioning of pTr and pLE cells was evaluated. This study provides insight into time-dependent transitions within the cell environment after PDM exposure and elucidates a detailed mechanism of induced adverse effects. These results imply that PDM exposure can potentially cause toxic effects on the implantation-related process in pigs. Moreover, to the best of our knowledge, this is the first study to describe the mechanism by which PDM induces these effects, enhancing our understanding of the toxicity of this herbicide.


Assuntos
Cálcio , Herbicidas , Suínos , Animais , Cálcio/metabolismo , Proliferação de Células , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Mitocôndrias/metabolismo , Herbicidas/toxicidade
3.
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
4.
Pestic Biochem Physiol ; 189: 105314, 2023 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-36549811

RESUMO

Tebufenpyrad is classified as a pyrazole acaricide and insecticide. It is widely used for several crops, especially in greenhouses, in several countries. While its unfavorable effects on non-target organisms have already been established, relatively little is known about its reproductive toxicity. Therefore, we demonstrated the biochemical effects of tebufenpyrad using porcine trophectoderm and porcine luminal epithelial cells, which are involved in implantation. We found that tebufenpyrad had antiproliferative effects and reduced cell viability. Tebufenpyrad also triggered apoptosis and excessive reactive oxygen species production. Furthermore, it induced cell cycle arrest in the G1 phase and disrupted calcium homeostasis in the cytosol and mitochondria. MAPK signaling pathways and the crosstalk among them were altered following tebufenpyrad treatment. In addition, the migration ability of cells was reduced after treatment with tebufenpyrad. Lastly, tebufenpyrad influenced the expression of genes related to pregnancy. Collectively, these results reveal the mechanism of the biochemical and physiological effects of tebufenpyrad to both trophectoderm and uterine cells and suggest that tebufenpyrad reduces the potential of successful implantation.


Assuntos
Cálcio , Pirazóis , Gravidez , Feminino , Suínos , Animais , Proliferação de Células , Cálcio/metabolismo , Pontos de Checagem do Ciclo Celular , Pirazóis/toxicidade , Pirazóis/metabolismo , Células Epiteliais , Homeostase
5.
Pestic Biochem Physiol ; 193: 105460, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-37248025

RESUMO

As the use of herbicides in agriculture has increased worldwide, the importance of identifying unexpected toxic effects on non-target organisms is emerging. Bensulide is used on various agricultural crops as an organophosphate herbicide; however, it can pose a high risk to non-target organisms because of its long half-life and accumulative potential. Despite its high risk, the hazardous effects of bensulide on implantation and mechanisms in cells have not been reported. Therefore, in this study, intracellular mechanisms and potential risk of implantation failure were identified in porcine trophectoderm (pTr) and uterine luminal epithelial (pLE) cells derived from pigs with human-like molecular mechanisms in implantation. The LC50 values of bensulide were 5.21 mg/L in pTr cells and 6.49 mg/L in pLE cells. Both cell lines were exposed to bensulide at concentrations <5 mg/L in subsequent experiments. Treatment with 5 mg/L bensulide activated ERK1/2 and JNK. Disrupted mitochondrial membrane potentials of both cell types were identified. In addition, mitochondrial Ca2+ concentration increased to 261.24% and 228.04% in pTr and pLE cells, respectively, and cytoplasmic Ca2+ concentrations decreased by approximately 50% in both cell types. The abnormal regulation of various intracellular environments by bensulide causes cell division cycle arrest and apoptosis. Finally, 5 mg/L bensulide inhibited transcription of implantation-related genes. Collectively, our results suggest that bensulide may interrupt implantation during early pregnancy by disrupting maternal-fetal interaction.


Assuntos
Apoptose , Células Epiteliais , Gravidez , Feminino , Suínos , Humanos , Animais , Proliferação de Células , Ciclo Celular
6.
Pestic Biochem Physiol ; 191: 105366, 2023 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-36963936

RESUMO

Tetraconazole is a type of fungicide that eliminates pathogens in plants and fruit. To date, studies have focused on the direct exposure of plants and fruits to residual tetraconazole, but no studies have been conducted on the indirect effects of tetraconzaole. Given the importance of cows as milk-producing animals and their potential exposure to pesticides via plant consumption, we analyzed the mechanism by which tetraconazole influences milk production. Here, we confirmed that tetraconazole-induced apoptosis and inhibited cell viability and proliferation by regulating the cell cycle in bovine mammary epithelial cells (MAC-T). In addition, Ca2+ homeostasis in mitochondria was disrupted by tetraconazole, leading to the depolarization of mitochondrial membrane potential. Consistent with the proliferation-related findings, tetraconazole downregulated AKT, ERK1/2, P38, and JNK signaling pathways and proliferation-related proteins such as CCND1 and PCNA in MAC-T cells. Meanwhile, it upregulated cleaved caspase 3, BAX, and Cytochrome c under the same conditions in MAC-T cells. Furthermore, MAC-T exposed to tetraconazole causes a failure of proper autophagy functioning. In summary, the results of this study indicated that tetraconazole exposure may lead to a failure of milk production from bovine mammary epithelial cells by disrupting calcium homeostasis and mitochondrial function.


Assuntos
Cálcio , Glândulas Mamárias Animais , Feminino , Bovinos , Animais , Cálcio/metabolismo , Apoptose , Células Epiteliais , Mitocôndrias
7.
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
8.
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
9.
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
10.
Pestic Biochem Physiol ; 193: 105461, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-37248026

RESUMO

Oxyfluorfen, a phenoxy phenyl-type herbicide, causes significant damage to ecosystems through chronically effecting invertebrates, fish, and mammals. Considering its adverse effect on ecosystem conservation, it is necessary to investigate its toxic effects on animals. However, the mechanisms of oxyfluorfen toxicity on bovines are not well established. This study investigated the cytotoxic effect of oxyfluorfen on bovine mammary epithelial cells (MAC-T). We conducted several functional experiments to examine the response of MAC-T to oxyfluorfen under various concentrations (0, 1, 2, 5, and 10 ppm). Oxyfluorfen decreased cell viability and increased apoptotic cells by regulating the expression of apoptotic genes and proteins in MAC-T. In addition, oxyfluorfen-treated cells exhibited reduced PCNA expression with a low 3D spheroid formation as compared to that of control cells. Furthermore, oxyfluorfen treatment suppressed cell cycle progression with a decrease in cyclin D1 and cyclin A2 in MAC-T. Next, we performed western blot analysis to verify intercellular signaling changes in oxyfluorfen-treated MAC-T. The phosphor-AKT protein was increased, whereas MAPK signal pathways were decreased. Particularly, the combination of oxyfluorfen with U0126 or SP600125 completely blocked the ERK1/2 and JNK pathways leading to cell viability in MAC-T. Moreover, oxyfluorfen induced inflammatory gene expression and autophagy by increasing phosphorylation of P62 and LC3B in MAC-T. These results demonstrated that oxyfluorfen has cytotoxic effect on MAC-T, implying that the milk production capacity in cows may eventually harm humans.


Assuntos
Ecossistema , Fosfatidilinositol 3-Quinases , Humanos , Feminino , Bovinos , Animais , Pontos de Checagem do Ciclo Celular , Células Epiteliais , Ruminantes/metabolismo , Autofagia , Apoptose
11.
J Cell Biochem ; 123(2): 469-480, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34816480

RESUMO

Fraxetin is a natural compound extracted from Fraxinus spp. and has various functions such as antibacterial, antioxidant, neuroprotective, and antifibrotic effects. Although studies have reported its anticancer properties in lung and breast cancer, little is known about colon cancer, the most frequent type of cancer. Thus, we used two colon cancer cell lines, HT29 and HCT116 cells, to investigate whether fraxetin could inhibit the capabilities acquired during tumor development. In this study, fraxetin suppressed cell viability and induced apoptotic cell death in HT29 and HCT116 cells. Furthermore, fraxetin regulated the expression of proteins involved in apoptosis in HT29 and HCT116 cells. Additionally, fraxetin induced reactive oxygen species levels and calcium influx with loss of mitochondrial membrane potential (ΔΨm) and endoplasmic reticulum stress. Moreover, fraxetin induced G2/M arrest and modulated the intracellular signaling pathway, including AKT, ERK1/2, JNK, and P38. Nevertheless, we found no cause-effect correlation between the antiproliferative action of fraxetin and modulation of the phosphorylation state of signaling proteins. Fraxetin-induced inhibitory effect on colon cancer cell viability was synergistic with 5-fluorouracil (5-FU) or irinotecan even in 5-FU resistant-HCT116 cells. Collectively, our results suggest that fraxetin can be effectively used as a therapeutic agent for targeting colon cancer, although it is necessary to further elucidate the relationship between the hallmark capabilities that fraxetin inhibits and the intracellular regulatory mechanism.


Assuntos
Neoplasias do Colo , Cumarínicos/farmacologia , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Fluoruracila/farmacologia , Mitocôndrias/metabolismo , Morte Celular/efeitos dos fármacos , Neoplasias do Colo/tratamento farmacológico , Neoplasias do Colo/metabolismo , Células HCT116 , Células HT29 , Humanos
12.
Mar Drugs ; 20(8)2022 Jul 25.
Artigo em Inglês | MEDLINE | ID: mdl-35892941

RESUMO

Pancreatic ductal adenocarcinoma (PDAC) is one of the most aggressive types of cancer and exhibits a devastating 5-year survival rate. The most recent procedure for the treatment of PDAC is a combination of several conventional chemotherapeutic agents, termed FOLFIRINOX, that includes irinotecan, leucovorin, oxaliplatin, and 5-fluorouracil (5-FU). However, ongoing treatment using these agents is challenging due to their severe side effects and limitations on the range of patients available for PDAC. Therefore, safer and more innovative anticancer agents must be developed. The anticarcinoma activity of matairesinol that can be extracted from seagrass has been reported in various types of cancer, including prostate, breast, cervical, and pancreatic cancer. However, the molecular mechanism of effective anticancer activity of matairesinol against pancreatic cancer remains unclear. In the present study, we confirmed the inhibition of cell proliferation and progression induced by matairesinol in representative human pancreatic cancer cell lines (MIA PaCa-2 and PANC-1). Additionally, matairesinol triggers apoptosis and causes mitochondrial impairment as evidenced by the depolarization of the mitochondrial membrane, disruption of calcium, and suppression of cell migration and related intracellular signaling pathways. Finally, matairesinol exerts a synergistic effect with 5-FU, a standard anticancer agent for PDAC. These results demonstrate the therapeutic potential of matairesinol in the treatment of PDAC.


Assuntos
Antineoplásicos , Carcinoma Ductal Pancreático , Neoplasias Pancreáticas , Antineoplásicos/uso terapêutico , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Carcinoma Ductal Pancreático/tratamento farmacológico , Carcinoma Ductal Pancreático/metabolismo , Carcinoma Ductal Pancreático/patologia , Linhagem Celular Tumoral , Fluoruracila/farmacologia , Furanos , Humanos , Lignanas , Masculino , Mitocôndrias , Neoplasias Pancreáticas/patologia , Neoplasias Pancreáticas
13.
Pestic Biochem Physiol ; 184: 105063, 2022 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-35715071

RESUMO

Alachlor is a widely used herbicide for the cultivation of various grains employed as food for cattle. The mechanisms leading to the toxic effects of alachlor on epithelial cells of the bovine mammary gland are not well known. Thus, this study was conducted to clarify the toxicological effects of alachlor on the immortalized epithelial cell line of the bovine mammary gland (MAC-T) cells. After treatment, many factors related to cell viability, proliferation, and cellular homeostasis were evaluated. Alachlor arrested cell cycle progression by blocking the expression of cyclin and cyclin-dependent kinases, and induced the breakdown of Ca2+ homeostasis. The cytosolic and mitochondrial levels of Ca2+ were also abnormally increased after the treatment of cells with alachlor, ultimately leading to the depolarization of mitochondrial membrane potential in MAC-T cells. The signaling cascade was found to be dysregulated by the abnormal phosphorylation of signaling molecules involved in PI3K/AKT (AKT, p70S6K, and S6) and MAPK/JNK (JNK and c-Jun) pathways. In these mechanisms, exposure to alachlor led to a reduction in the viability and proliferation of MAC-T cells. Altogether, the toxic effects of alachlor can lead to abnormal conditions in epithelial cells of the bovine mammary gland, which might hinder these cells from performing their main role, such as producing milk.


Assuntos
Glândulas Mamárias Animais , Fosfatidilinositol 3-Quinases , Acetamidas , Animais , Cálcio/metabolismo , Bovinos , Pontos de Checagem do Ciclo Celular , Células Epiteliais , Homeostase , Glândulas Mamárias Animais/metabolismo , Proteínas Proto-Oncogênicas c-akt
14.
Pestic Biochem Physiol ; 188: 105254, 2022 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-36464334

RESUMO

Herbicides are chemicals that have been established to have adverse impacts. However, they are still widely used in agriculture. Pendimethalin (PDM) is an herbicide that is widely used in many countries to control annual grasses. The possibility of livestock being exposed to PDM is relatively high, considering the half-life of PDM and its residues in water, soil and crops. However, the toxicity of PDM in cattle, especially in the mammary glands, has not been reported. Therefore, we investigated whether PDM has toxic effects in the mammary epithelial cells (MAC-T) of cattle. MAC-T cells were treated with various doses (0, 2.5, 5 and 10 µM) of PDM. We found that PDM affected cell viability and cell proliferation and causes cell cycle arrest. Furthermore, PDM triggered cell apoptosis, induced excessive ROS production and mitochondrial membrane potential (MMP) loss, and disrupted calcium homeostasis. In addition, PDM altered the activation of proteins associated with the endoplasmic reticulum (ER) stress response and modified PI3K and MAPK signaling cascades. In conclusion, our current study unveiled the mechanism of PDM in MAC-T cells and we suggest that PDM might be harmful to the mammary gland system of cattle, possibly affecting milk production.


Assuntos
Herbicidas , Fosfatidilinositol 3-Quinases , Bovinos , Animais , Espécies Reativas de Oxigênio , Células Epiteliais , Herbicidas/toxicidade , Transdução de Sinais
15.
Pestic Biochem Physiol ; 188: 105288, 2022 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-36464341

RESUMO

Aclonifen is a diphenyl-ether herbicide that is used to control the growth of weeds while growing crops such as corn and wheat. Although the biochemical effects of aclonifen are well characterized, including its ability to inhibit protoporphyrinogen oxidase and carotenoid synthesis, the toxicity of aclonifen in embryonic implantation and development during early pregnancy, has not been reported. Thus, in this study, we investigated the potential interference of aclonifen in embryonic implantation using porcine trophectoderm (pTr) and uterine luminal epithelial (pLE) cells isolated during implantation period of early pregnancy. Cell viability in both pTr and pLE cells significantly decreased in a dose-dependent manner following aclonifen treatment. Moreover, the proportion of cells in the sub-G1 phase of the cell cycle gradually increased upon treatment with increasing concentrations of aclonifen, which in turn led to an increase in the number of apoptotic cells, as determined by annexin V and propidium iodide staining. Aclonifen treatment caused mitochondrial dysfunction by increasing the depolarization of the mitochondrial membrane potential and the mitochondrial calcium concentration. Aclonifen inhibited cell mobility by suppressing the expression of implantation-related genes in pTr and pLE cells. To explore the underlying mechanism, we evaluated the phosphorylation of PI3K and MAPK signaling molecules. The phosphorylation of AKT, S6, JNK, and ERK1/2 were significantly increased by aclonifen. Collectively, our results suggest that aclonifen may interrupt implantation during early pregnancy by disrupting maternal-fetal interaction.


Assuntos
Desenvolvimento Embrionário , Células Epiteliais , Feminino , Gravidez , Suínos , Animais , Útero , Apoptose
16.
Pestic Biochem Physiol ; 180: 105003, 2022 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-34955186

RESUMO

Fluroxypyr-1-methylheptyl ester (FPMH) is an auxin herbicide that is widely applied to crops and pastures to block growth of post-emergence weeds. Several studies have reported the toxicity of FPMH in aquatic vertebrates. However, the adverse impacts of FPMH on mammals, including domestic animals, have not been reported. The purpose of our current study is to assess the impact of FPMH on the bovine mammary system and milk production. To evaluate the toxicity of FPMH on the mammary glands of lactating cows, the bovine mammary gland epithelial cell line, MAC-T, was exposed to various concentrations (0, 5, 7.5, 10, 15, and 20 µM) of FPMH for 24 h, and then various assessments were performed. The results showed that FPMH dose-dependently reduced MAC-T cell viability following exposure to FPMH and induced mitochondrial depolarization and apoptosis. FPMH also modulated signaling through the PI3K and MAPK pathways. In addition, the expression levels of proteins related to endoplasmic reticulum (ER) stress were upregulated, indicating induction of ER stress, and calcium homeostasis was disrupted following FPMH treatment. In conclusion, our investigation suggests that FPMH may be toxic to the bovine mammary system and may decrease dairy production.


Assuntos
Apoptose/efeitos dos fármacos , Estresse do Retículo Endoplasmático , Células Epiteliais/efeitos dos fármacos , Transdução de Sinais , Acetatos , Animais , Bovinos , Células Cultivadas , Ésteres , Lactação , Sistema de Sinalização das MAP Quinases , Glândulas Mamárias Animais/citologia , Fosfatidilinositol 3-Quinases , Piridinas
17.
Pestic Biochem Physiol ; 181: 105011, 2022 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-35082034

RESUMO

Herbicides play key roles in agriculture. Aclonifen is a diphenyl ether herbicide that is widely used for sunflower, potato, corn, and wheat crops. Since it has a long half-life, it is considered persistent and can easily accumulate in the environment. Therefore, livestock and humans are at risk of exposure to aclonifen. Importantly, aclonifen is toxic to several mammals such as rats, mice, and dogs. However, the toxicity of aclonifen in cattle remains unclear. Therefore, we sought to investigate its toxicity in cattle using bovine mammary gland epithelial cells (MAC-T). We found that aclonifen induces sub-G1 phase arrest and represses MAC-T proliferation. In addition, aclonifen caused mitochondrial dysfunction, as evidenced by excessive ROS production and loss of mitochondrial membrane potential. Furthermore, cytosolic and mitochondrial calcium homeostases were disrupted after aclonifen treatment. Moreover, aclonifen treatment caused alterations in the PI3K/AKT and MAPK signaling pathways, which are involved in the regulation of cell survival and death. In conclusion, aclonifen causes MAC-T cell death through mitochondrial dysfunction and the collapse of calcium homeostasis.


Assuntos
Cálcio , Glândulas Mamárias Humanas , Compostos de Anilina , Animais , Apoptose , Bovinos , Morte Celular , Cães , Células Epiteliais , Homeostase , Humanos , Camundongos , Fosfatidilinositol 3-Quinases , Ratos , Espécies Reativas de Oxigênio
18.
Pestic Biochem Physiol ; 187: 105196, 2022 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-36127068

RESUMO

FPMH (Fluroxypyr-1-methylheptyl ester) is a synthetic auxin herbicide used in agriculture. The mechanism by which FPMH induces adverse effects in porcine trophectoderm (pTr) and porcine uterine luminal epithelial (pLE) cells, which are involved in porcine implantation, have not been studied yet. Therefore, the present study investigates the toxicological effects of FPMH on pTr and pLE cells. We confirmed that FPMH induced cytotoxic effects on the cells, including apoptosis induction, mitochondrial membrane potential (MMP) depolarization, and ROS production. The phosphorylation of the MAPK pathway (ERK1/2, JNK, and p38) was dysregulated by FPMH administration. In addition, FPMH could suppress cell-cell adhesion and migration abilities of pTr and pLE, which are crucial for implantation. Therefore, exposure to FPMH induced adverse effects in pTr and pLE cells and could result in implantation failure.


Assuntos
Herbicidas , Sistema de Sinalização das MAP Quinases , Acetatos , Animais , Apoptose , Células Epiteliais , Ésteres/metabolismo , Ésteres/farmacologia , Glicolatos , Herbicidas/farmacologia , Ácidos Indolacéticos/metabolismo , Piridinas , Espécies Reativas de Oxigênio/metabolismo , Suínos
19.
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
20.
J Cell Physiol ; 236(2): 1025-1042, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-32697363

RESUMO

Osthole is a natural coumarin found in a variety of plants and has been reported to have diverse biological functions, including antimicrobial, antiviral, immunomodulatory, and anticancer effects. Here, we investigated the natural derivative osthole as a promising anticancer compound against ovarian cancer and evaluated its ability to suppress and abrogate tumor progression. In addition, we found the endoplasmic reticulum-mitochondrial axis-mediated anticancer mechanisms of osthole against ES2 and OV90 ovarian cancer cells and demonstrated its calcium-dependent pharmacological potential. Mechanistically, osthole was found to target the phosphatidylinositol 3-kinase/mitogen-activated protein kinase signaling pathway to facilitate tumor suppression in ovarian cancer. Furthermore, we identified the effects of osthole in a three-dimensional tumor-formation model using the zebrafish xenograft assay, providing convincing evidence of the pharmacological effects of osthole within the anchorage-independent tumor microenvironment. These findings suggest that osthole has strong potential as a pharmacological agent for targeting ovarian cancer.


Assuntos
Cumarínicos/farmacologia , Mitocôndrias/genética , Neoplasias Ovarianas/tratamento farmacológico , Proteínas Supressoras de Tumor/genética , Animais , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Retículo Endoplasmático/efeitos dos fármacos , Feminino , Humanos , Mitocôndrias/efeitos dos fármacos , Proteínas Quinases Ativadas por Mitógeno/genética , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/patologia , Fosfatidilinositol 3-Quinase/genética , Transdução de Sinais/efeitos dos fármacos , Microambiente Tumoral/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto , Peixe-Zebra/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA