Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
2.
Ecotoxicol Environ Saf ; 271: 115994, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38262094

RESUMO

Chronic exposure to crystalline silica (CS) contributes to pulmonary fibrosis. Airway epithelium dysfunction and fibroblast activation have both been recognized as pivotal players, alongside disturbances in ferroptosis and glycolysis reprogramming. However, the mechanisms involved remain unclear. In this study, we investigated the crosstalk between airway epithelium and fibroblast in the context of CS-induced pulmonary fibrosis. CS was employed in vivo and the in vitro co-culture system of airway epithelium and fibroblast. Spatial transcriptome analysis of CS-induced fibrotic lung tissue was conducted as well. Results showed that epithelium ferroptosis caused by CS enhanced TGFß1-induced fibroblast activation through paracrine signaling. tPA was further identified to be the central mediator that bridges epithelium ferroptosis and fibroblast activation. And increased fibroblast glycolysis reprogramming was evidenced to promote fibroblast activation. By inhibition of epithelium ferroptosis or silencing tPA of airway epithelium, fibroblast AMPK phosphorylation was inhibited. Moreover, we revealed that tPA secreted by ferroptotic epithelium transmits paracrine signals to fibroblasts by governing glycolysis via p-AMPK/AMPK mediated Glut1 accumulation. Collectively, our study demonstrated the regulation of airway epithelium ferroptosis on fibroblast activation in CS-induced pulmonary fibrosis, which would shed light on the complex cellular crosstalk within pulmonary fibrosis and identify potential therapeutic targets.


Assuntos
Fibrose Pulmonar , Humanos , Fibrose Pulmonar/induzido quimicamente , Fibrose Pulmonar/tratamento farmacológico , Dióxido de Silício/toxicidade , Comunicação Parácrina , Proteínas Quinases Ativadas por AMP , Epitélio , Fibroblastos , Glicólise
3.
J Affect Disord ; 320: 247-253, 2023 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-36195169

RESUMO

BACKGROUND: With the pandemic of COVID, the public are faced with tremendous threatens both physically and mentally. Postpartum depression (PPD) is one of the most serious complications of childbearing, bringing severe impact on a woman's mental state and mood after birth. Research has shown that maternal mental state is closely correlated with PPD, those undergo the emergency or significant life changes during the postpartum period are more likely to suffer from PPD. In this study, we conducted the meta-analysis to estimate the association between PPD and COVID-19 pandemic. METHODS: PubMed, Web of Science, PsycINFO, ScienceDirect, CNKI, China Science and Technology Journal Database, and WANFANG Database were searched for potentially relevant articles published before April 2022. Review Manager 5.2 was used to perform a meta-analysis and subgroup analysis to compute the pooled odds ratio. RESULTS: A total of 26 studies were included in this review. The overall pooled prevalence of PPD in the review was 24 % (95 % CI: 0.19-0.29), with China's at 22 % (95 % CI 0.16-0.28) and other countries at 25 % (95 % CI 0.18-0.32) during the COVID-19 pandemic. Moreover, compared to those who did not experience COVID-19, those who experienced it had an increased risk of PPD[OR:1.83(95 % CI 1.70-1.97)]. CONCLUSIONS: According to this analysis, there was a significantly higher prevalence and odds of PPD in those who suffered from the COVID-19 pandemic. Additionally, we also found that China had a lower prevalence of postpartum depression than other countries during the COVID-19 pandemic. Our study may provide the instruction for the care of new mother under the situation of COVID-19 prevalence.


Assuntos
COVID-19 , Depressão Pós-Parto , Humanos , Feminino , Depressão Pós-Parto/epidemiologia , Depressão Pós-Parto/etiologia , COVID-19/epidemiologia , Depressão/epidemiologia , Pandemias , Período Pós-Parto , Fatores de Risco
4.
J Nutr Biochem ; 110: 109148, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36049670

RESUMO

Silicosis is one of the severest occupational diseases worldwide, manifesting as infiltration of inflammatory cells, excessive secretion of pro-inflammatory mediators and pulmonary diffuse fibrosis. Macrophages polarization to M2 is one of the major strategies that attenuates inflammatory response. Our previous study found that vitamin D could protect against silica-induced lung injury by damping the secretion of pro-inflammatory cytokines. Here we further identified that vitamin D attenuated silica particles-induced lung inflammation by regulating macrophage polarization in a KLF4-STAT6 manner. Myeloid-specific Stat6 knockout (cKO) mice were generated for in vivo studies. Primary macrophages purified from bronchoalveolar lavage fluid (BALF) of wildtype or Stat6 cKO mice and differentiated THP-1 cells were used for in vitro studies. Vitamin D was found to promote alveolar macrophage polarizing to M2 phenotype through the STAT6 signaling pathway, as demonstrated by worse lung inflammation and ablated protection of vitamin D in silica particles-instilled Stat6 cKO mice. Mechanismly, vitamin D upregulated KLF4 expression in the alveolar macrophage, which synergistically activated STAT6. Additionally, KLF4 was found to upregulate macrophages autophagy, which protected them from silica particles-induced oxidative stress and cell apoptosis. The protective effects of vitamin D were dismissed by silencing KLF4. Our study demonstrates the potential mechanism of vitamin D-mediated macrophage polarization and reveals the therapeutic application of vitamin D in inflammatory disease.


Assuntos
Lesão Pulmonar , Pneumonia , Animais , Camundongos , Lesão Pulmonar/induzido quimicamente , Lesão Pulmonar/prevenção & controle , Ativação de Macrófagos , Macrófagos/metabolismo , Camundongos Endogâmicos C57BL , Pneumonia/tratamento farmacológico , Dióxido de Silício/toxicidade , Dióxido de Silício/metabolismo , Fator de Transcrição STAT6/metabolismo , Fator de Transcrição STAT6/farmacologia , Vitamina D/metabolismo
5.
Toxicol Res (Camb) ; 11(3): 391-401, 2022 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-35782637

RESUMO

Nonylphenol (NP) is an endocrine disrupting chemical, which widely exists in environment and can result in multiple system dysfunction. Pancreas as one of the most important organs is sensitive to NP, while the detail toxic effect is still less studied. Previously, we unveiled nonylphenol causes pancreatic damage in rats, herein, we further explore the potential mechanism and seek protection strategy in vitro. Insulinoma (INS-1) cells exposed to NP were observed to suffer oxidative stress and mitochondrial dysfunction, as reflected by the abnormal levels of reactive oxygen species, malonic dialdehyde, superoxide dismutase, Ca2+, and mitochondrial membrane potential. Melatonin (MT) was found to alleviate NP-induced mitochondrial dysfunction and oxidative stress, further inhibit apoptosis and restore pancreas function. Mechanically, MT induced the MDM2-P53-P21 signaling, which upregulated the Nrf2 signaling pathway. In summary, our study clarified NP-induced INS-1 cells mitochondrial dysfunction and oxidative stress, which could be ameliorated by MT through MDM2-P53-P21 axis.

6.
Cell Death Dis ; 13(6): 530, 2022 06 06.
Artigo em Inglês | MEDLINE | ID: mdl-35668064

RESUMO

Compelling evidences have revealed the emerging role of ferroptosis in the pathophysiological process of acute lung injury (ALI), but its modulation is not clear. Here, we identified that STAT6 acted as a critical regulator of epithelium ferroptosis during ALI. Firstly, STAT6 expression and activity were increased in the ALI mice models caused by crystalline silica (CS), LPS and X-ray exposure. Followed by confirming the contribution of ferroptosis in the above ALI with ferrostatin-1 and deferoxamine intervention, bioinformatic analyses revealed that STAT6 expression was negatively correlated with ferroptosis. Consistently, lung epithelium-specific depletion of STAT6 in mice or STAT6 knockdown in cultured epithelial cells exacerbated ferroptosis in the above ALI. While overexpression of STAT6 in lung epithelial cells attenuated the ferroptosis. Mechanistically, SLC7A11 is a typical ferroptosis-related gene and negatively regulated by P53. CREB-binding protein (CBP) is a critical acetyltransferase of P53 acetylation, showing valuable regulation on targets' transcription. Herein, we found that STAT6 negatively regulates ferroptosis through competitively binding with CBP, which inhibits P53 acetylation and transcriptionally restores SLC7A11 expression. Finally, pulmonary-specific STAT6 overexpression decreased the ferroptosis and attenuated CS and LPS induced lung injury. Our findings revealed that STAT6 is a pivotal regulator of ferroptosis, which may be a potential therapeutic target for the treatment of acute lung injury.


Assuntos
Lesão Pulmonar Aguda , Ferroptose , Lesão Pulmonar Aguda/induzido quimicamente , Lesão Pulmonar Aguda/genética , Animais , Lipopolissacarídeos/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Fator 2 Relacionado a NF-E2/metabolismo , Fator de Transcrição STAT6/genética , Fator de Transcrição STAT6/metabolismo , Proteína Supressora de Tumor p53/genética
7.
Cell Death Dis ; 13(1): 66, 2022 01 19.
Artigo em Inglês | MEDLINE | ID: mdl-35046382

RESUMO

Lipid metabolism, especially fatty acid oxidation (FAO) dysfunction, is a major driver of renal fibrosis; however, the detailed regulatory mechanisms involved remain unclear. In this study, we showed that there existed an association between the signal transducer and activator of transcription 6 (STAT6) and tubular lipid metabolism in fibrotic kidneys. Specifically, STAT6 was activated along with the accumulation of lipids via the downregulation of FAO-related genes when mice were subjected to unilateral ureteral obstruction (UUO) or high-fat diet challenge. Tubular-specific depletion, or pharmacologic inhibitor of Stat6 in mice, and Stat6 knockdown in cultured tubular cells attenuated lipid accumulation and renal fibrosis by enhancing FAO. Mechanistically, STAT6 transcriptionally inhibited the expression of PPARα and its FAO-related target genes through a sis-inducible element located in the promoter region of the protein. In conclusion, our study demonstrates the mechanistic details of STAT6-mediated FAO dysregulation in the progression of renal fibrosis and provides a preclinical rationale for efforts to improve the management of renal fibrosis brought about by FAO dysregulation.


Assuntos
Nefropatias , PPAR alfa , Fator de Transcrição STAT6 , Obstrução Ureteral , Animais , Ácidos Graxos/metabolismo , Fibrose , Rim/patologia , Nefropatias/patologia , Metabolismo dos Lipídeos , Camundongos , Camundongos Endogâmicos C57BL , PPAR alfa/metabolismo , Fator de Transcrição STAT6/metabolismo , Obstrução Ureteral/patologia
8.
Oxid Med Cell Longev ; 2022: 2485250, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35047105

RESUMO

Lung inflammatory injury is a global public health concern. It is characterized by infiltration of diverse inflammatory cells and thickening of pulmonary septum along with oxidative stress to airway epithelial cells. STAT6 is a nuclear transcription factor that plays a crucial role in orchestrating the immune response, but its function in tissue inflammatory injury has not been comprehensively studied. Here, we demonstrated that STAT6 activation can protect against particle-induced lung inflammatory injury by resisting oxidative stress. Specifically, genetic ablation of STAT6 was observed to worsen particle-induced lung injury mainly by disrupting the lungs' antioxidant capacity, as reflected by the downregulation of the Nrf2 signaling pathway, an increase in malondialdehyde levels, and a decrease in glutathione levels. Vitamin D receptor (VDR) has been previously proved to positively regulate Nrf2 signals. In this study, silencing VDR expression in human bronchial epithelial BEAS-2B cells consistently suppressed autophagy-mediated activation of the Nrf2 signaling pathway, thereby aggravating particle-induced cell damage. Mechanically, STAT6 activation promoted the nuclear translocation of VDR, which increased the transcription of autophagy-related genes and induced Nrf2 signals, and silencing VDR abolished these effects. Our research provides important insights into the role of STAT6 in oxidative damage and reveals its potential underlying mechanism. This information not only deepens the appreciation of STAT6 but also opens new avenues for the discovery of therapies for inflammatory respiratory system disorders.


Assuntos
Lesão Pulmonar/terapia , Pulmão/patologia , Fator 2 Relacionado a NF-E2/metabolismo , Fator de Transcrição STAT6/metabolismo , Animais , Humanos , Camundongos , Transdução de Sinais
9.
Front Immunol ; 13: 1094556, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36685533

RESUMO

The dysregulation of autophagy contributes to renal fibrosis. N6-Methyladenosine (m6A) RNA modification is a critical mediator of autophagy. Our previous studies have reported that the disorder of the PPARα/fatty acid oxidation (FAO) axis in renal tubular cells is suppressed by STAT6, which is involved in the regulation of renal fibrotic processes. Here, we found that canagliflozin significantly upregulates SQSTM1/P62, promoting PPARα-mediated FAO by inducing autophagy-dependent STAT6 degradation both in TGF-ß1-treated HK2 cells and in unilateral ureteral occlusion (UUO) and ischemia-reperfusion (I/R) renal fibrosis mouse models. Knockdown of P62/SQSTM1 led to the impairment autophagic flux and the dysregulation of the STAT6/PPARα axis, which was confirmed by SQSTM1/P62cKO mice with UUO treatment along with bioinformatics analysis. Furthermore, SQSTM1/P62 deficiency in renal tubular cells inhibited canagliflozin's effects that prevent FAO disorder in renal tubular cells and renal fibrosis. Mechanistically, the level of m6A eraser FTO, which interacted with SQSTM1 mRNA, decreased in the renal tubular cells both in vitro and in vivo after canagliflozin administration. Decrease in FTO stabilized SQSTM1 mRNA, which induced autophagosome formation. Collectively, this study uncovered a previously unrecognized function of canagliflozin in FTO in the autophagy modulation through the regulation of SQSTM1 mRNA stability in the renal tubular STAT6/PPARα/FAO axis and renal fibrosis.


Assuntos
Nefropatias , Obstrução Ureteral , Camundongos , Animais , Proteína Sequestossoma-1/genética , Proteína Sequestossoma-1/metabolismo , Canagliflozina/farmacologia , PPAR alfa/genética , PPAR alfa/metabolismo , Nefropatias/prevenção & controle , Autofagia , RNA Mensageiro , Fibrose , Dioxigenase FTO Dependente de alfa-Cetoglutarato/genética , Dioxigenase FTO Dependente de alfa-Cetoglutarato/metabolismo
10.
Ecotoxicol Environ Saf ; 225: 112730, 2021 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-34478973

RESUMO

Crystalline silica (CS) is a universal environmental pollutant, which causes a typical inflammatory lung injury. Vitamin D shows huge potential against particles-induced lung injury, while little known about the molecular mechanism involved in macrophage autophagy. In this study, we aim to identify the protective effects of vitamin D on CS caused lung inflammatory injury and clarify the detail mechanism. After exposure to CS (3 mg/mice in 50 µl PBS), wildtype and Atg7flox/flox Lyz2-cre mice were treated with or without vitamin D3 (40,000 IU/kg). The results indicated that exposure to CS caused an obvious lung injury, manifesting as pathological structural changes, macrophage-dominated inflammatory cell infiltration and increased pro-inflammatory cytokines. Remarkably, these damages were more serious in Atg7flox/flox Lyz2-cre mice. Vitamin D was found to inverse CS-induced inflammatory cell infiltration and restored anti-inflammatory M2 macrophages by inducing autophagy, which attenuated lung injury, as determined by decreased levels of apoptosis and inflammatory response. While, this effects of vitamin D were slashed in Atg7flox/flox Lyz2-cre mice. This study reveals the adverse effect of CS on lung tissue and the protective mechanism of vitamin D involved in M2 macrophages autophagy, which attenuates CS-caused lung injury.


Assuntos
Dióxido de Silício , Vitamina D , Animais , Autofagia , Macrófagos , Camundongos , Camundongos Endogâmicos C57BL , Dióxido de Silício/toxicidade , Regulação para Cima
11.
Oxid Med Cell Longev ; 2021: 6610124, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33603948

RESUMO

Nonalcoholic fatty liver disease is the most common liver disease worldwide. Hepatic steatosis and oxidative stress are the main characteristics of NAFLD (nonalcoholic fatty liver disease), which also affect its prognosis. Bixin acts as novel Nrf2 (NF-E2 p45-related factor 2) activator with the cytoprotection against oxidative stress and inflammation; this study mainly focused on the mechanism of Nrf2 activation by bixin and explored its potential feasibilities in long-term high-fat diet- (HFD-) caused hepatic steatosis and inflammatory response in vitro and in vivo. Bixin was found to activate Nrf2 signals by the modification of critical Keap1 (Kelch-like ECH-associated protein 1) cystine and competitive interaction with Keap1 with upregulating P62 mRNA and protein expression. In human liver cells exposed to FFA (free fatty acid), bixin displayed a pronounced cytoprotective activity with upregulation of Nrf2-mediated gene expression, such as PPARα and its targets related with fatty acid oxidation. In HFD-fed mice, systemic administration of bixin attenuated lipid accumulation, decreased oxidant inflammatory damage in the liver, and reduced circulating lipid levels through Nrf2. Different from most of other established inducers, bixin activated Nrf2 signals through two different mechanisms with safe administration for protection of oxidant inflammatory damage and attenuation of lipid accumulation in the in vivo long-term HFD-fed mice. Bixin represents a prototype Nrf2 activator that displays cytoprotective activity upon system administration targeting hepatic steatosis and oxidant inflammation originating from long-term HFD-fed mice. And bixin-based Nrf2-directed systemic intervention may also provide therapeutic benefit in protecting other organs in the process of metabolic syndrome.


Assuntos
Carotenoides/uso terapêutico , Fígado Gorduroso/tratamento farmacológico , Fígado Gorduroso/metabolismo , Inflamação/patologia , Fator 2 Relacionado a NF-E2/metabolismo , PPAR alfa/metabolismo , Transdução de Sinais , Animais , Carotenoides/química , Carotenoides/farmacologia , Linhagem Celular , Cisteína/metabolismo , Citoproteção/efeitos dos fármacos , Dieta Hiperlipídica , Fígado Gorduroso/patologia , Comportamento Alimentar , Humanos , Inflamação/complicações , Proteína 1 Associada a ECH Semelhante a Kelch/metabolismo , Metabolismo dos Lipídeos/efeitos dos fármacos , Fígado/patologia , Camundongos Knockout , Estabilidade Proteica/efeitos dos fármacos , Ubiquitinação/efeitos dos fármacos , Regulação para Cima/efeitos dos fármacos
13.
Environ Toxicol ; 36(4): 472-483, 2021 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-33107683

RESUMO

Radon is one of the major pathogenic factors worldwide. Recently, epidemiological studies have suggested that radon exposure plays an important role in lung injury, which could further cause cancer. However, the toxic effects and underlying mechanism on lung injury are still not clear. Here, we identified the detailed toxic effects of long-term radon exposure. Specifically, the manifestations were inflammatory response and cell apoptosis in dose- and time-dependent manners. In detail, it caused the mitochondrial dysfunction and oxidative stress as determined by the abnormal levels of mitochondrial DNA copy number, adenosine triphosphate, mitochondrial membrane potential, superoxide dismutase, and cycloxygenase-2. Furthermore, we found that melatonin treatment ameliorated mitochondrial dysfunction and attenuated the levels of oxidative stress caused by long-term radon exposure, which could further inhibit the lung tissue apoptosis as determined by the decreased levels of cleaved caspase 3. Our study would provide potential therapeutic application of melatonin on lung tissue injury caused by long-term radon exposure.


Assuntos
Poluentes Radioativos do Ar/toxicidade , Antioxidantes/farmacologia , Lesão Pulmonar/prevenção & controle , Melatonina/farmacologia , Lesões Experimentais por Radiação/prevenção & controle , Radônio/toxicidade , Animais , Apoptose/efeitos dos fármacos , Caspase 3/metabolismo , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/patologia , Células Epiteliais/efeitos da radiação , Humanos , Exposição por Inalação/efeitos adversos , Pulmão/efeitos dos fármacos , Pulmão/patologia , Pulmão/efeitos da radiação , Lesão Pulmonar/metabolismo , Lesão Pulmonar/patologia , Masculino , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Camundongos Endogâmicos BALB C , Mitocôndrias/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Lesões Experimentais por Radiação/metabolismo , Lesões Experimentais por Radiação/patologia , Superóxido Dismutase/metabolismo
14.
Front Cell Dev Biol ; 8: 576988, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33313043

RESUMO

Bixin, a natural carotenoid extracted from the seeds of Bixa orellana, has antioxidant and anti-inflammation effects. However, the pharmacological effects and underlying mechanisms of bixin in kidney interstitial fibrosis remain unknown. Partial epithelial-to-mesenchymal transition (EMT) of tubular cells has been linked to renal interstitial fibrosis. Here, we found that in the unilateral ureteral obstruction model, bixin administration could ameliorate kidney interstitial fibrosis. The expression of signal transducer and activator of transcription 6 (STAT6) was dramatically increased in renal tubular cells. Bixin treatment inhibited STAT6 induction. The activation of STAT6 signaling was essential for transforming growth factor ß1, fibrotic markers, and EMT-related protein expression in HK2 cells, which was confirmed by using the Stat6-/- mice. Ubiquitination, but not the acetylation level of STAT6, was induced by bixin treatment and promoted the suppression of phosphorylation and stability of STAT6. P62-dependent autophagy might be involved in this process. The study demonstrated that bixin can be exploited therapeutically to alleviate renal interstitial fibrosis by targeting STAT6 signaling deactivation.

15.
Toxicol Res (Camb) ; 9(4): 589, 2020 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-32905226

RESUMO

[This corrects the article DOI: 10.1093/toxres/tfaa003.].

16.
Toxicol Res (Camb) ; 9(1): 55-66, 2020 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-32742635

RESUMO

Fine particulate matter, a major air pollutant across the world, causes a series of pulmonary diseases. Vitamin D is a typical vitamin with emerging roles in inflammation and fibrosis. Different situations and diseases need different doses and modes of vitamin D administration, which challenges the existing vitamin D supplementary rules. Thus, studies of vitamin D applications and their mechanisms in various diseases are important for its future therapeutic applications. In this study, the therapeutic application of vitamin D3 in chronic particle-exposure-associated lung fibrosis and tissue remodeling was investigated. In vivo studies showed that vitamin D3 significantly attenuated fibrosis effects by decreasing α-smooth muscle actin-regulated extracellular matrix deposition and restoring expressions of E-cadherin and N-cadherin. With the importance of activated macrophage in the regulation of local epithelium and fibroblast in the process of tissue fibrosis, two separate in vitro systems of co-culture of macrophages with lung epithelium or fibroblast were built. The results confirmed that vitamin D3 promoted the proliferation of lung epithelium and depressed the fibrosis effects of fibroblasts as well. In addition, our results indicated that the therapeutic effects of vitamin D3 were through Nrf2 signals. Our work provides convincing experimental evidence for vitamin D therapeutic application to promote tissue repair and improve particle-associated lung fibrosis.

17.
J Agric Food Chem ; 68(31): 8321-8329, 2020 Aug 05.
Artigo em Inglês | MEDLINE | ID: mdl-32706966

RESUMO

Bixin is a natural carotenoid isolated from the seeds of Bixa orellana, with numerous important pharmacological activities, including antioxidant and antifibrotic effects. The nuclear factor erythroid-2-related factor2 (Nrf2) signaling pathway induced by bixin is involved in the process. Excessive reactive oxygen species generation in tubular cells contributes to kidney interstitial fibrosis. The potential therapeutic strategy for bixin in alleviating kidney fibrosis remains largely unclear. In this study, we used unilateral ureteral obstruction (UUO) to establish a renal fibrotic model. Dramatic oxidative DNA damage occurs in kidneys, especially in tubular cells after UUO. In cultured tubular cells, bixin could induce Nrf2 signaling activation by suppressing Nrf2 ubiquitination and increasing its protein stability. Transforming growth factor beta 1-induced epithelial-to-mesenchymal transition (EMT) and extracellular matrix production were suppressed by bixin, and blockade of Nrf2 activation by small interfering RNA could largely reverse the protective effect of bixin. In vivo studies showed that administration of bixin induces Nrf2 signaling activation in tubular cells and markedly attenuates partial EMT of tubular cells and kidney interstitial fibrosis after subjecting to UUO. Together, this study implies that bixin may protect against kidney interstitial fibrosis through stimulating Nrf2 activation in renal tubular cells.


Assuntos
Carotenoides/administração & dosagem , Fibrose/prevenção & controle , Nefropatias/prevenção & controle , Fator 2 Relacionado a NF-E2/metabolismo , Extratos Vegetais/administração & dosagem , Obstrução Ureteral/complicações , Animais , Bixaceae/química , Fibrose/etiologia , Fibrose/genética , Fibrose/metabolismo , Humanos , Rim/metabolismo , Rim/patologia , Nefropatias/etiologia , Nefropatias/genética , Nefropatias/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fator 2 Relacionado a NF-E2/genética
18.
Life Sci ; 258: 118161, 2020 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-32730835

RESUMO

AIMS: Tubulointerstitial inflammation is recognized as a key determinant of progressive sepsis-induced acute kidney injury (AKI). Schisantherin A (SchA) has been shown to be capable of regulating inflammatory processes. In the present study, we explored the possibility of SchA in preventing lipopolysaccharide (LPS)-induced kidney inflammation and injury. MATERIALS AND METHODS: AKI was induced by a single intraperitoneal injection of LPS in CD1 mice, administration of SchA was used for treatment. The protective effect of SchA on renal function and inflammation were analyzed respectively; the NRK-52E cell line was employed for the in vitro study and relative molecular mechanism was explored. KEY FINDINGS: Administration with SchA markedly attenuated LPS-induced damage on renal function and histopathological changes of the kidney. Additionally, pretreatment with SchA could inhibit the expression of inflammatory factors in the kidneys. In NRK-52E cells, SchA treatment significantly inhibited LPS-induced NF-κB activation and pro-inflammatory cytokine expression. Moreover, SchA could promote NRF2 pathway activation, and further blockade of NRF2 activation reversed the SchA-induced inhibition of NF-κB activation. SIGNIFICANCE: These presented results indicated that SchA may have great potential for protecting against sepsis-induced AKI.


Assuntos
Injúria Renal Aguda/tratamento farmacológico , Injúria Renal Aguda/etiologia , Anti-Inflamatórios/uso terapêutico , Ciclo-Octanos/uso terapêutico , Dioxóis/uso terapêutico , Lignanas/uso terapêutico , Fator 2 Relacionado a NF-E2/metabolismo , Sepse/complicações , Injúria Renal Aguda/metabolismo , Injúria Renal Aguda/patologia , Animais , Linhagem Celular , Inflamação/tratamento farmacológico , Inflamação/metabolismo , Inflamação/patologia , Rim/efeitos dos fármacos , Rim/metabolismo , Rim/patologia , Masculino , Camundongos , Ratos , Sepse/tratamento farmacológico , Sepse/metabolismo , Sepse/patologia , Transdução de Sinais/efeitos dos fármacos
19.
Front Cell Dev Biol ; 8: 643207, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33585473

RESUMO

[This corrects the article DOI: 10.3389/fcell.2020.576988.].

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA