Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
1.
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
2.
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
3.
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
4.
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
5.
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
6.
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
7.
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
8.
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
9.
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
10.
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
11.
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
12.
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
13.
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
14.
Artigo em Inglês | MEDLINE | ID: mdl-39106914

RESUMO

Flusilazole is a well-known triazole fungicide applied to various crops and fruits worldwide. Flusilazole residues are frequently detected in the environment, and many researchers have reported the hazardous effects of flusilazole on non-target organisms; however, the developmental toxicity of flusilazole has not been fully elucidated. In this study, we investigated flusilazole-induced developmental defects in zebrafish, which are used in toxicology studies to assess the toxic effects of chemicals on aquatic species or vertebrates. We confirmed that flusilazole exposure affected the viability and hatching rate of zebrafish larvae, and resulted in morphological defects, reduced body length, diminished eye and head sizes, and inflated pericardial edema. Apoptosis, oxidative stress, and inflammation were also observed. These factors interrupted the normal organ formation during early developmental stages, and transgenic models were used to identify organ defects. We confirmed the effects of flusilazole on the nervous system using olig2:dsRed transgenic zebrafish, and on the cardiovascular system using cmlc2:dsRed and fli1:eGFP transgenic zebrafish. Our results demonstrate the developmental toxicity of flusilazole and its mechanisms in zebrafish as well as the detrimental effects of flusilazole.


Assuntos
Animais Geneticamente Modificados , Apoptose , Fungicidas Industriais , Estresse Oxidativo , Triazóis , Peixe-Zebra , Animais , Estresse Oxidativo/efeitos dos fármacos , Apoptose/efeitos dos fármacos , Triazóis/toxicidade , Fungicidas Industriais/toxicidade , Embrião não Mamífero/efeitos dos fármacos , Silanos
15.
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
16.
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
17.
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
18.
Sci Total Environ ; 934: 173097, 2024 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-38729356

RESUMO

Pyrethroid insecticides, such as beta-cyfluthrin, are used extensively globally, including in households and agriculture, and have been detected in the milk and urine of humans and cattle. Beta-cyfluthrin exhibits toxic effects, including neurotoxicity and male reproductive toxicity; however, few studies have investigated female reproductive toxicity despite its wide environmental distribution. The present study investigates effects of beta-cyfluthrin on implantation in porcine cells (pTr from the trophectoderm and pLE from the endometrial luminal epithelium). To identify the various physiological changes induced by beta-cyfluthrin, such as apoptosis and lipid peroxidation, flow cytometry analysis and immunofluorescence were performed with various reagents. In addition, the expression of genes and proteins associated with intracellular changes was confirmed using qRT-PCR and western blotting. Beta-cyfluthrin induced cell-cycle arrest and altered intracellular calcium flux. It also disrupted the mitochondrial function and promoted reactive oxygen species (ROS) production, leading to lipid peroxidation. Moreover, ROS induced by beta-cyfluthrin altered mitogen-activated protein kinase (MAPK) pathways and decreased cell migration capability. The expression levels of genes that are significant during early pregnancy were altered by beta-cyfluthrin in both cell lines. The changes resulted in apoptosis and diminished cell proliferation of pTr and pLE. Collectively, the results imply that beta-cyfluthrin disrupts the implantation process by affecting the physiology of the trophectoderm and endometrial luminal epithelial cells. The present study is the first to reveal the cellular mechanisms of beta-cyfluthrin on the female reproductive system and highlights the need for further in-depth research into its hazards.


Assuntos
Células Epiteliais , Inseticidas , Mitocôndrias , Nitrilas , Piretrinas , Espécies Reativas de Oxigênio , Transdução de Sinais , Animais , Espécies Reativas de Oxigênio/metabolismo , Feminino , Piretrinas/toxicidade , Nitrilas/toxicidade , Suínos , Inseticidas/toxicidade , Células Epiteliais/efeitos dos fármacos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Transdução de Sinais/efeitos dos fármacos , Implantação do Embrião/efeitos dos fármacos , Útero/efeitos dos fármacos , Apoptose/efeitos dos fármacos , Trofoblastos/efeitos dos fármacos
19.
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
20.
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
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa