Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Sci Total Environ ; 922: 171234, 2024 Apr 20.
Artigo em Inglês | MEDLINE | ID: mdl-38428612

RESUMO

As a ubiquitous pollutant in the environment, hexafluoropropylene oxide trimer acid (HFPO-TA) has been proven to have strong hepatotoxicity. However, the underlying mechanism is still unclear. Consequently, in vivo and in vitro models of HFPO-TA exposure were established to investigate the detrimental effects of HFPO-TA on the liver. In vivo, we discovered that HFPO-TA enhanced endoplasmic reticulum (ER)-mitochondrial association, caused mitochondrial oxidative damage, activated ER stress, and induced apoptosis in mouse livers. In vitro experiments confirmed that IP3R overexpression on ER structure increased mitochondrial calcium levels, which led to mitochondrial damage and mitochondria-dependent apoptosis in HepG2 cells exposed to HFPO-TA. Subsequently, damaged mitochondria released a large amount of mitochondrial ROS, which activated ER stress and ER stress-dependent apoptosis. In conclusion, this study demonstrates that HFPO-TA can induce apoptosis by regulating the crosstalk between ER and mitochondria, ultimately leading to liver damage. These findings reveal the significant hepatotoxicity of HFPO-TA and its potential mechanisms.


Assuntos
Doença Hepática Induzida por Substâncias e Drogas , Fluorocarbonos , Mitocôndrias , Propionatos , Animais , Camundongos , Apoptose , Retículo Endoplasmático/metabolismo , Doença Hepática Induzida por Substâncias e Drogas/metabolismo
5.
Ecotoxicol Environ Saf ; 264: 115459, 2023 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-37703808

RESUMO

Aluminum is a neurotoxic food contaminant. Aluminum trichloride (AlCl3) causes hippocampal mitochondrial damage, leading to hippocampal injury. Damaged mitochondria can release mitochondrial reactive oxygen species (mtROS) and activate nucleotide-binding oligomerization domain-like receptor-containing 3 (NLRP3) inflammasomes and apoptosis. E3 ubiquitin ligase PARK2 (Parkin)-mediated mitophagy can attenuate mitochondrial damage. However, the role of mitophagy in AlCl3-induced mice hippocampal damage and its regulatory mechanism remain elusive. First, C57BL/6 N mice were treated with 0, 44.825, 89.65, and 179.3 mg/kg body weight AlCl3 drinking water for 90 d. Apoptosis, NLRP3-inflammasome activation and mitochondrial damage were increased in AlCl3-induced hippocampal damage. In addition, Parkin-mediated mitophagy peaked in the middle-dose group and was slightly attenuated in the high-dose group. Subsequently, we used wild-type and Parkin knockout (Parkin-/-) mice to investigate the AlCl3-induced hippocampal damage. The results showed that Parkin-/- inhibited mitophagy, and aggravated AlCl3-induced mitochondrial damage, NLRP3-inflammasome activation, apoptosis and hippocampal damage. Finally, we administered MitoQ (mtROS inhibitor) and MCC950 (NLRP3 inhibitor) to AlCl3-treated Parkin-/- mice to investigate the mechanism of Parkin-mediated mitophagy. The results showed that inhibition of mtROS and NLRP3 attenuated hippocampal NLRP3-inflammasome activation, apoptosis, and damage in AlCl3-treated Parkin-/- mice. These findings indicate that Parkin-mediated mitophagy protects against AlCl3-induced hippocampal apoptosis in mice via the mtROS-NLRP3 pathway.


Assuntos
Cloreto de Alumínio , Hipocampo , Inflamassomos , Mitofagia , Animais , Camundongos , Cloreto de Alumínio/toxicidade , Apoptose , Hipocampo/efeitos dos fármacos , Hipocampo/patologia , Camundongos Endogâmicos C57BL , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética
6.
Sci Total Environ ; 905: 167033, 2023 Dec 20.
Artigo em Inglês | MEDLINE | ID: mdl-37709082

RESUMO

Hexafluoropropylene oxide trimer acid (HFPO-TA), an emerging alternative to perfluorooctanoic acid (PFOA), has recently been identified as a significant environmental pollutant. Nevertheless, there is a scarcity of studies regarding the hepatotoxic effects of HFPO-TA. Here, we investigated the types and potential mechanisms of liver damage caused by HFPO-TA. Initially, we validated that the introduction of HFPO-TA resulted in the Wnt/ß-catenin signaling (W/ß signaling) activation, as well as the induction of necroptosis and inflammation, both in the liver of mice and in HepG2 cells. Subsequently, we established that the W/ß signaling mediated the necroptosis and inflammation observed in the liver and HepG2 cells exposed to HFPO-TA. Finally, we demonstrated that the phosphorylated form of NF-κB p65 (p-NF-κB p65) played a role in mediating the necroptosis and inflammation, and its activity could be regulated by the W/ß signaling pathway in the liver of mice and HepG2 cells exposed to HFPO-TA. In conclusion, our investigation elucidates the role of HFPO-TA in inducing necroptosis and inflammation in the liver, which is facilitated through the activation of the W/ß/NF-κB axis.


Assuntos
NF-kappa B , beta Catenina , Humanos , Necroptose , Fígado , Inflamação/induzido quimicamente
7.
Food Chem Toxicol ; 178: 113915, 2023 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-37393014

RESUMO

Aluminum (Al) is a common environmental pollutant that can induce kidney damage. However, the mechanism is not clear. In the present study, to explored the exact mechanism of AlCl3-induced nephrotoxicity, C57BL/6 N male mice and HK-2 cells were used as experimental subjects. Our results showed that Al induced reactive oxygen species (ROS) overproduction, c-Jun N-terminal kinase (JNK) signaling activation, RIPK3-dependent necroptosis, NLRP3 inflammasome activation, and kidney damage. In addition, inhibiting JNK signaling could downregulate the protein expressions of necroptosis and NLRP3 inflammasome, thereby alleviating kidney damage. Meanwhile, clearing ROS effectively inhibited JNK signaling activation, which in turn inhibited necroptosis and NLRP3 inflammasome activation, ultimately alleviating kidney damage. In conclusion, these findings suggest that necroptosis and NLPR3 inflammasome activation mediated by ROS/JNK pathway participate in AlCl3-induced kidney damage.


Assuntos
Inflamassomos , Sistema de Sinalização das MAP Quinases , Camundongos , Animais , Masculino , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Necroptose , Camundongos Endogâmicos C57BL , Rim/metabolismo
8.
Sci Total Environ ; 897: 165500, 2023 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-37442457

RESUMO

Dibutyl phthalate (DBP) is one of the most employed plasticizers pervading the environment. DBP is a newly identified global organic pollutant that can activate NLRP3 inflammasomes and induce inflammatory liver injury. However, its hepatotoxicity remains poorly understood. The objective of this investigation was to investigate the probable pathways underlying DBP-induced liver injury. First, C57BL/6N mice were orally administered DBP at 10 and 50 mg/kg B.W. doses for 28 days. The observed results indicated a significant increase in liver collagen deposition and upregulated protein expression of fibrosis markers in mice. In addition, the p38MAPK/NF-κB signaling pathway and pyroptosis-related protein expression were upregulated. To establish a correlation between these changes, we conducted a conditioned medium co-culture of human hepatocellular carcinoma (HepG2) and human hepatic stellate (LX-2) cells. We performed inhibitor interventions to validate the mechanism of DBP-induced liver fibrosis in vitro. After treatment with p38MAPK (SB203580), NF-κB (PDTC), and NLRP3 (MCC950) inhibitors, the activation of LX-2 cells, the p38MAPK/NF-κB signaling pathway and pyroptosis due to DBP were alleviated. Therefore, DBP exposure leads to NLRP3-mediated pyroptosis of hepatocytes via the p38MAPK/NF-κB signaling pathway, activating LX-2 cells and causing liver fibrosis. Our findings offer a conceptual framework to understand the pathological underpinnings of DBP-induced liver injury while proposing novel ideas to prevent and treat DBP hepatotoxicity. Thus, targeting p38MAPK, NF-κB, and NLRP3 may prevent DBP-induced liver fibrosis.


Assuntos
Doença Hepática Crônica Induzida por Substâncias e Drogas , NF-kappa B , Camundongos , Humanos , Animais , Proteína 3 que Contém Domínio de Pirina da Família NLR , Dibutilftalato/toxicidade , Piroptose/fisiologia , Proteínas Quinases p38 Ativadas por Mitógeno , Camundongos Endogâmicos C57BL , Cirrose Hepática/induzido quimicamente
9.
Sci Total Environ ; 892: 164620, 2023 Sep 20.
Artigo em Inglês | MEDLINE | ID: mdl-37270010

RESUMO

Dibutyl phthalate (DBP) is a typical plasticizer and is widely used in industrial manufacturing. DBP has been reported to be cardiotoxic, manifested by oxidative stress and inflammatory damage. However, the potential mechanism of heart damage caused by DBP remains unclear. By in vivo and in vitro experiments, first, this study demonstrated that DBP induced endoplasmic reticulum (ER) stress, mitochondrial damage, and pyroptosis in cardiomyocytes; second, it was confirmed that the ER stress increased mitochondrial-associated ER membrane (MAM), which led to mitochondrial damage by abnormalizing Ca2+ transfer within MAMs; finally, it was confirmed that mitochondrial reactive oxygen species (mtROS) production was increased after mitochondrial damage, which activated NLRP3 inflammasome and pyroptosis in cardiomyocytes. In summary, ER stress is the initiation of DBP cardiotoxicity, which leads to mitochondrial damage by disrupting Ca2+ transfer from ER to mitochondria. Subsequently, released mtROS promotes the activation of NLRP3 inflammasome and pyroptosis, eventually leading to heart damage.


Assuntos
Dibutilftalato , Traumatismos Cardíacos , Humanos , Dibutilftalato/metabolismo , Piroptose , Inflamassomos/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR , Mitocôndrias , Retículo Endoplasmático/metabolismo , Traumatismos Cardíacos/metabolismo
10.
Food Chem Toxicol ; 176: 113762, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-37028746

RESUMO

Aluminium (Al) accumulates in the spleen and causes spleen apoptosis. Mitochondrial dyshomeostasis represents primary mechanisms of spleen apoptosis induced by Al. Apoptosis-inducing factor (AIF) is located in the gap of the mitochondrial membrane and can be released into the nucleus, leading to apoptosis. Phosphatase and tensin homolog (PTEN)-induced putative kinase1 (PINK1)/E3 ubiquitin ligase PARK2 (Parkin)-mediated mitophagy maintains mitochondrial homeostasis by removing damaged mitochondria, but its function in AIF-mediated spleen apoptosis induced by Al is not clear. In our study, aluminium trichloride (AlCl3) was diluted in water for 90 d and administered to 75 male C57BL/6N mice at 0, 44.8, 59.8, 89.7, and 179.3 mg/kg body weight. AlCl3 triggered PINK1/Parkin pathway-mediated mitophagy, induced AIF release and AIF-mediated spleen apoptosis. AlCl3 was administered to sixty male C57BL/6N mice of wild type and Parkin knockout for 90 d at 0 and 179.3 mg/kg body weight. The results indicated that Parkin deficiency decreased mitophagy, aggravated mitochondrial damage, AIF release and AIF-mediated spleen apoptosis induced by AlCl3. According to our results, PINK1/Parkin-mediated mitophagy and AIF-mediated spleen apoptosis are caused by AlCl3, whereas mitophagy is protective in AIF-mediated apoptosis induced by AlCl3.


Assuntos
Fator de Indução de Apoptose , Mitofagia , Animais , Masculino , Camundongos , Apoptose , Peso Corporal , Camundongos Endogâmicos C57BL , Proteínas Quinases/genética , Proteínas Quinases/metabolismo , Baço/metabolismo , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/metabolismo , Cloreto de Alumínio
11.
Ecotoxicol Environ Saf ; 256: 114846, 2023 May.
Artigo em Inglês | MEDLINE | ID: mdl-37018856

RESUMO

Aflatoxin B1 (AFB1) is an extremely hazardous and unavoidable pollutant for cereals and feedstuff. AFB1 can cause testicular lesion, and how to alleviate its testicular toxicity has received much attention in recent years. Lycopene (LYC), a foodborne nutrient derived from red fruits and vegetables, has protective effects against sperm abnormality and testicular lesions. To confirm the beneficial effects and mechanisms of LYC on AFB1-induced testicular lesion, 48 male mice were exposed to 0.75 mg/kg AFB1 or/and 5 mg/kg LYC for consecutive 30 days. Results demonstrated the LYC significantly restored the lesions of testicular microstructure and ultrastructure, and sperm abnormalities in AFB1-exposed mice. Furthermore, LYC effectively attenuated AFB1-induced oxidative stress and mitochondrial damage, including ameliorative mitochondrial structural, and elevated mitochondrial biogenesis for maintaining mitochondrial function. Meanwhile, LYC resisted AFB1-induced mitochondrial-dependent apoptosis. In addition, LYC promoted nuclear factor erythroid 2-related factor 2 (Nrf2) nuclear translocation, and upregulated the Nrf2 signaling pathway. Collectively, our findings demonstrate LYC ameliorates AFB1-induced testicular lesion by attenuating oxidative stress and mitochondrial damage, which is related to the activation of Nrf2.


Assuntos
Aflatoxina B1 , Fator 2 Relacionado a NF-E2 , Masculino , Animais , Camundongos , Licopeno/metabolismo , Licopeno/farmacologia , Aflatoxina B1/toxicidade , Fator 2 Relacionado a NF-E2/metabolismo , Sementes/metabolismo , Estresse Oxidativo
12.
Food Chem Toxicol ; 174: 113706, 2023 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-36871880

RESUMO

Hexafluoropropylene oxide trimer acid (HFPO-TA) causes hepatotoxicity, however, its underlying mechanisms have not been conclusively determined. We investigated the effects of HFPO-TA on mice liver after 28 days of orally administered 0 or 0.5 mg/kg/d HFPO-TA. Administration of HFPO-TA induced mitochondrial ROS (mtROS) overexpression, cGAS-STING signaling activation, pyroptosis and fibrosis in mice liver. To determine the HFPO-TA-associated hepatotoxic mechanisms, mtROS, cGAS-STING signaling and pyroptosis intervention assays were performed in HFPO-TA-exposed mice liver. First, mtROS was found to be an upstream regulatory target of cGAS-STING signaling, pyroptosis and fibrosis. Second, cGAS-STING signaling was established to be an upstream regulatory mechanism of pyroptosis and fibrosis. Finally, pyroptosis was shown to regulate fibrosis. The above results confirm that HFPO-TA causes mice liver fibrosis via mtROS/cGAS-STING/NLRP3-mediated pyroptosis.


Assuntos
Proteína 3 que Contém Domínio de Pirina da Família NLR , Piroptose , Animais , Camundongos , Espécies Reativas de Oxigênio/farmacologia , Nucleotidiltransferases/farmacologia , Fígado , Fibrose
13.
Chem Biol Interact ; 369: 110266, 2023 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-36402210

RESUMO

Mycotoxins are most frequent contaminants in environment and agricultural production globally. The T-2 toxin of Fusarium species is the most toxic type of A trichothecene mycotoxins. T-2 toxin can accumulate in bone and cause bone development disorders. Osteoblast is the functional cell responsible for bone formation. Whereas, the mechanism of T-2 toxin toxicity on osteoblast remains unknown. In present study, MC3T3-E1 cells were treated with 0, 2, 4, and 8 nM T-2 toxin for 24h to explore the effect of T-2 toxin on the differentiation and mineralization of osteoblasts. Subsequently, autophagy and Wnt intervention agents were used to explore the roles of autophagy and Wnt signaling pathway in T-2 toxin-induced osteoblastic differentiation and mineralization disorders, respectively. The results showed that 2 nM of T-2 toxin had no significant effect on cell vitality, but 4 and 8 nM of T-2 significantly inhibited cell viability. All doses of T-2 toxin inhibited both osteoblastic differentiation and mineralization, as assessed by alkaline phosphatase staining, Alizarin red S staining, and protein expressions of osteogenic proteins. In addition, the activation of Wnt signaling pathway mitigated T-2 toxin-induced osteoblast impairment, while the inhibition of autophagy exacerbated it. Our results also indicated that there was a positive feedback loop between the Wnt signaling pathway and autophagy.


Assuntos
Toxina T-2 , Via de Sinalização Wnt , Linhagem Celular , Toxina T-2/metabolismo , Diferenciação Celular , Osteogênese , Osteoblastos , Autofagia
14.
Chem Biol Interact ; 368: 110229, 2022 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-36354081

RESUMO

Aluminum (Al) exposure can lead to oxidative stress and liver inflammatory injury in mice. The overproduction of reactive oxygen species (ROS) activates the NLRP3 inflammasome and further induces liver pyroptosis. However, the participation of pyroptosis in inducing Al-mediated liver injury and the underlying regulatory mechanisms remain largely unclear. Herein, a mice model of subchronic Al exposure was established to investigate the role of pyroptosis in Al-induced liver injury. Then, MCC950 and N-acetylcysteine were used to inhibit NLRP3 inflammasome-mediated pyroptosis and ROS production for exploring the role and the underlying mechanisms of pyroptosis in determining Al-induced liver injury. It was confirmed that Al induced hepatocyte pyroptosis in mice, and that NLRP3 inflammasome-mediated pyroptosis plays a damaging role in Al-induced liver injury. ROS promotes pyroptosis in an Al-induced liver injury model by activating the NLRP3 inflammasome. Collectively, it was shown that ROS promotes pyroptosis to aggravate Al-induced liver injury by activating the NLRP3 inflammasome.


Assuntos
Doença Hepática Crônica Induzida por Substâncias e Drogas , Inflamassomos , Camundongos , Animais , Piroptose , Espécies Reativas de Oxigênio , Proteína 3 que Contém Domínio de Pirina da Família NLR , Alumínio
15.
J Agric Food Chem ; 70(42): 13765-13777, 2022 Oct 26.
Artigo em Inglês | MEDLINE | ID: mdl-36239691

RESUMO

T-2 toxin causes kidney fibrosis. Wnt/ß-catenin signaling promotes kidney fibrosis when sustained and activated. However, whether T-2-induced kidney fibrosis involves Wnt/ß-catenin signaling activation has not been explored yet. T-2 toxin causes renal mitochondrial damage, leading to mitochondrial reactive oxygen species (mtROS) overproduction and NLRP3-inflammasome activation. The activated NLRP3-inflammasome can mediate fibrosis. However, whether the NLRP3-inflammasome can be mediated by mtROS and further regulate T-2-induced kidney fibrosis through Wnt/ß-catenin signaling is unclear. In this study, first, we confirmed that T-2 toxin caused Wnt/ß-catenin signaling activation in mice kidneys and HK-2 cells. Second, we confirmed that mtROS activated the NLRP3-inflammasome in T-2-exposed mice kidneys and HK-2 cells. Third, we confirmed that the NLRP3-inflammasome regulated the Wnt/ß-catenin signaling in T-2 toxin-exposed mice kidneys and HK-2 cells. Finally, we confirmed that Wnt/ß-catenin signaling regulated fibrosis in T-2 toxin-exposed mice kidneys and HK-2 cells. The above results confirm that T-2 toxin induces kidney fibrosis via the mtROS-NLRP3-Wnt/ß-catenin axis.


Assuntos
Nefropatias , Toxina T-2 , Animais , Camundongos , beta Catenina/genética , Fibrose , Inflamassomos , Rim/patologia , Nefropatias/induzido quimicamente , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética , Espécies Reativas de Oxigênio , Toxina T-2/toxicidade , Proteínas Wnt/metabolismo
16.
Toxicol Lett ; 366: 33-44, 2022 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-35810998

RESUMO

Aflatoxin B1 (AFB1) can cause mitochondrial malfunction and immunosuppression in spleen. Mitochondrial damage can lead to oxidative stress and aggravate immune cell dysfunction. Phosphatase and tensin homolog (PTEN)-induced putative kinase1 (PINK1)/ E3 ubiquitin ligase PARK2 (Parkin)-mediated mitophagy can scavenge damaged mitochondria and alleviate oxidative stress to maintain cellular homeostasis. However, the role of PINK1/Parkin-mediated mitophagy in AFB1-induced immunosuppression in spleen is unclear. In this study, sixty male mice were sensibilized orally with AFB1 at different concentrations [0, 0.5, 0.75, and 1 mg/kg body weight (BW)] for 28 days, and AFB1 caused splenic structure injury and immunosuppression, also led to upregulation of PINK1/Parkin-mediated mitophagy in a dose-dependent manner. Subsequently, thirty male WT C57BL/6 N mice and thirty male Parkin knockout (Parkin-/-) C57BL/6 N mice were sensibilized orally with AFB1 at 0 or 1 mg/kg BW for 28 days, and Parkin-/- inhibited mitophagy and further aggravated AFB1-induced splenic structure injury, immunosuppression, mitochondrial damage and oxidative stress. Collectively, these results indicate that AFB1 exposure activates PINK1/Parkin-mediated mitophagy, which protects against immunosuppression in spleen.


Assuntos
Aflatoxina B1/farmacologia , Mitofagia , Baço , Animais , Terapia de Imunossupressão , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Quinases , Ubiquitina-Proteína Ligases/genética
17.
Environ Toxicol ; 37(10): 2341-2353, 2022 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-35716031

RESUMO

Dibutyl phthalate (DBP) is a plasticizer widely used in daily production, which causes serious environmental pollution, and damage to brain, liver, kidney, and lung by producing excessive reactive oxygen species (ROS) after entering the body. DBP can also cause skeletal dysplasia, but it is unclear whether ROS is involved. In addition, overproduction of ROS can activate mitophagy, which is an important mechanism for regulating mitochondrial quality and cell homeostasis. In order to investigate whether DBP can damage MC3T3-E1 cells (osteoblast cell line) and whether ROS and mitophagy are involved, DBP toxicity experiment, Parkin gene silencing experiment, and N-acetylcysteine (NAC) intervention experiment were performed on MC3T3-E1 cells in turn. First, we found that DBP caused MC3T3-E1 cell viability decline and osteogenic dysfunction, accompanied by the overproduction of ROS and the activation of mitophagy. Then, we found that silencing Parkin expression alleviated DBP-induced apoptosis and osteogenic dysfunction of MC3T3-E1 cells. In addition, NAC treatment inhibited the PINK1/Parkin-mediated mitophagy and alleviated the apoptosis and osteogenic dysfunction of MC3T3-E1 cells caused by DBP. Our research results showed that DBP could cause MC3T3-E1 cell damage by increasing ROS to promote the PINK1/Parkin-mediated mitophagy.


Assuntos
Dibutilftalato , Mitofagia , Acetilcisteína/farmacologia , Dibutilftalato/toxicidade , Mitofagia/genética , Proteínas Quinases/genética , Proteínas Quinases/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/metabolismo
18.
Food Chem Toxicol ; 164: 113078, 2022 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-35489469

RESUMO

T-2 toxin can cause mitochondrial impairment and subsequent renal damage. PINK1/Parkin-mediated mitophagy can mitigate renal impairment by alleviating mitochondrial damage. Nevertheless, the impact of PINK1/Parkin-mediated mitophagy in T-2 toxin-induced renal injury remains unclear. Here, we studied the role of PINK1/Parkin-mediated mitophagy in T-2 toxin-induced nephrotoxicity. Mitochondrial damage was accompanied by NLRP3-inflammasome activation and PINK1/Parkin-mediated mitophagy in the kidney of T-2 toxin-exposed C57BL/6N mice. Knocking out Parkin inhibited the mitophagy but aggravated the structural and functional damage, NLRP3-inflammasome activation, mitochondrial damage, and apoptosis. Correlation analysis revealed that NLRP3-inflammasome activation was correlated with apoptosis. These results show that PINK1/Parkin-mediated mitophagy mitigates T-2 toxin-induced nephrotoxicity.


Assuntos
Mitofagia , Toxina T-2 , Animais , Inflamassomos , Camundongos , Camundongos Endogâmicos C57BL , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética , Proteínas Quinases , Toxina T-2/toxicidade , Ubiquitina-Proteína Ligases/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...