Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Pharmacol Res ; 191: 106754, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-37019194

RESUMEN

Pulmonary fibrosis is a progressive and fatal fibrotic lung disease with mysterious pathogenesis and limited effective therapies. G protein-coupled receptors (GPRs) participate in a variety of physiologic functions, and several GPRs have critical fibrosis-promoting or -inhibiting roles in pulmonary fibrosis. Here, we explored the role of GPR41 in the pathobiology of pulmonary fibrosis. We found that GPR41 expression was elevated in lung tissues of mice with bleomycin-induced pulmonary fibrosis and lung fibroblasts treated with transforming growth factor-ß1 (TGF-ß1). Knockout of GPR41 attenuated pulmonary fibrosis in mice, as evidenced by improved lung morphology, decreased lung weight and collagen secretion, and down-regulated α-SMA, collagen type I alpha and fibronectin expression in lungs. Additionally, GPR41 knockout inhibited the differentiation of fibroblasts to myofibroblasts, and decreased myofibroblast migration. By further mechanistic analysis, we demonstrated that GPR41 regulated TGF-ß1-induced fibroblast-to-myofibroblast differentiation and Smad2/3 and ERK1/2 phosphorylation via its Gαi/o subunit but not Gßγ subunit. Together, our data indicate that GPR41 is involved in pulmonary fibroblast activation and fibrosis, and GPR41 represents a potential therapeutic target for pulmonary fibrosis.


Asunto(s)
Fibrosis Pulmonar , Animales , Ratones , Bleomicina , Diferenciación Celular , Fibroblastos/metabolismo , Proteínas de Unión al GTP/metabolismo , Pulmón , Sistema de Señalización de MAP Quinasas , Ratones Endogámicos C57BL , Ratones Noqueados , Miofibroblastos/metabolismo , Fosforilación , Fibrosis Pulmonar/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo
2.
Acta Pharmacol Sin ; 44(3): 573-583, 2023 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-36056082

RESUMEN

Long-term treatment with adriamycin (ADR) is associated with higher incidences of cumulative cardiotoxicity manifest as heart failure. ADR-induced cardiomyopathy is characterized by extensive fibrosis that is caused by cardiac fibroblast activation. To date, however, no specific treatment is available to alleviate ADR-induced cardiotoxicity. Protein arginine methyltransferase 5 (PRMT5), a major enzyme responsible for methylation of arginine, regulates numerous cellular processes such as cell differentiation. In the present study we investigated the role of PRMT5 in cardiac fibrosis. Mice were administered ADR (3 mg/kg, i.p., every 2 days) for 2 weeks. We showed that aberrant PRMT5 expression was largely co-localized with α-SMA-positive activated cardiac fibroblasts in ADR-injected mice and in ADR-treated cardiac fibroblasts in vitro. PRMT5-overexpression exacerbated, whereas PRMT5 knockdown alleviated ADR-induced cardiac fibrosis in vivo and TGF-ß1-induced cardiac fibroblast activation in vitro. We demonstrated that PRMT5-overexpression enhanced methylated-Smad3 levels in vivo and in vitro. Pretreatment with a specific PRMT5 inhibitor EPZ015666 (5 nM) or overexpression of a catalytically inactive mutant of PRMT5, PRMT5(E444Q), reduced PRMT5-induced methylation of Smad3, thus suppressing PRMT5-mediated cardiac fibroblast activation in vitro. Furthermore, ADR activated cardiac fibroblasts was depending on autocrine TGF-ß1. Taken together, our results demonstrate that PRMT5 promotes ADR-induced cardiac fibrosis via activating cardiac fibroblasts, suggesting that it may be a potential therapeutic target of ADR-caused cardiotoxicity.


Asunto(s)
Cardiomiopatías , Factor de Crecimiento Transformador beta1 , Ratones , Animales , Factor de Crecimiento Transformador beta1/metabolismo , Doxorrubicina , Cardiotoxicidad/metabolismo , Fibrosis , Fibroblastos/metabolismo , Cardiomiopatías/patología , Proteína smad3/metabolismo
3.
Crit Rev Food Sci Nutr ; : 1-13, 2022 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-36168918

RESUMEN

Type 1 diabetes (T1D) is an autoimmune disorder characterized by the destruction of insulin-producing pancreatic ß cell. It contributes to high mortality, frequent diabetic complications, poor quality of life in patients and also puts a significant economic burden on health care systems. Therefore, the development of new therapeutic strategies is urgently needed. Recently, certain dietary compounds with potential applications in food industry, particularly polyphenols and polysaccharides, have gained increasing attention with their prominent anti-diabetic effects on T1D by modulating ß cell function, the gut microbiota and/or the immune system. In this review, we critically discuss the recent findings of several dietary polyphenols and polysaccharides with the potential to protect against T1D and the underlying anti-diabetic mechanisms. More importantly, we highlight the current trends, major issues, and future directions of industrial production of polyphenols- and polysaccharides-based functional foods for preventing or delaying T1D.

4.
Pharmacol Res ; 152: 104592, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31830521

RESUMEN

Pulmonary fibrosis is a progressive and fatal fibrotic lung disease with mysterious pathogenesis and limited effective therapies. The aberrantly activated lung myofibroblasts with resultant excessive accumulation of extracellular matrix is a central event in the progression of pulmonary fibrosis. Lysine-specific demethylase 1 (LSD1) has been suggested to epigenetically regulate cell differentiation, migration and invasion in tumor microenvironment. However, its function in pulmonary fibrosis remains unclear. The present study aimed to investigate the potential effect and underlying mechanisms of LSD1 in pulmonary fibrosis. Here, we found that LSD1 expression was elevated in lung tissues of mice with bleomycin-induced pulmonary fibrosis and lung fibroblasts treated with transforming growth factor-ß1 (TGF-ß1). In vivo knockdown of LSD1 by lentiviral shRNA transfection attenuated pulmonary fibrosis in mice, as evidenced by improved lung morphology, decreased lung coefficient and collagen secretion, and down-regulated α-SMA, collagen type I alpha and fibronectin expression in lungs. Additionally, in vitro knockdown of LSD1 inhibited the differentiation of fibroblasts to myofibroblasts, and decreased myofibroblast migration. By further mechanistic analysis, we demonstrated that knockdown of LSD1 prevented fibroblast--to-myofibroblast differentiation and subsequent pulmonary fibrosis by suppressing TGF-ß1/Smad3 signaling pathway through modulation of a balance between histone H3 lysine 9 methylation and histone H3 lysine 4 methylation. Together, our data indicate that LSD1 activation contributes to pulmonary myofibroblast differentiation and fibrosis by targeting TGF-ß1/Smad3 signaling, and suggest LSD1 as a therapeutic target for the treatment of pulmonary fibrosis.


Asunto(s)
Fibroblastos/metabolismo , Histona Demetilasas/metabolismo , Fibrosis Pulmonar/metabolismo , Proteína smad3/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Animales , Bleomicina , Diferenciación Celular , Fibroblastos/citología , Células HEK293 , Histona Demetilasas/genética , Humanos , Masculino , Ratones Endogámicos C57BL , Fibrosis Pulmonar/inducido químicamente , ARN Interferente Pequeño/genética
5.
Cell Physiol Biochem ; 45(1): 406-418, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29402793

RESUMEN

BACKGROUND/AIMS: Inflammation-induced injury of the endothelial barrier occurs in several pathological conditions, including atherosclerosis, ischemia, and sepsis. Endothelial cytoskeleton rearrangement is an important pathological mechanism by which inflammatory stimulation triggers an increase of vascular endothelial permeability. However, the mechanism maintaining endothelial cell barrier function against inflammatory stress is not fully understood. Glutathione S-transferase pi (GSTpi) exists in various types of cells and protects them against different stresses. In our previous study, GSTpi was found to act as a negative regulator of inflammatory responses. METHODS: We used a Transwell permeability assay to test the influence of GSTpi and its transferase activity on the increase of endothelial permeability induced by tumor necrosis factor alpha (TNF-α). TNF-α-induced actin remodeling and the influence of GSTpi were observed by using laser confocal microscopy. Western blotting was used to test the influence of GSTpi on TNF-α-activated p38 mitogen-activated protein kinase (MAPK)/MK2/heat shock protein 27 (HSP27). RESULTS: GSTpi reduced TNF-α-induced stress fiber formation and endothelial permeability increase by restraining actin cytoskeleton rearrangement, and this reduction was unrelated to its transferase activity. We found that GSTpi inhibited p38MAPK phosphorylation by directly binding p38 and influenced downstream substrate HSP27-induced actin remodeling. CONCLUSION: GSTpi inhibited TNF-α-induced actin remodeling, stress fiber formation and endothelial permeability increase by inhibiting the p38MAPK/HSP27 signaling pathway.


Asunto(s)
Actinas/metabolismo , Gutatión-S-Transferasa pi/metabolismo , Células Cultivadas , Gutatión-S-Transferasa pi/antagonistas & inhibidores , Gutatión-S-Transferasa pi/genética , Proteínas de Choque Térmico HSP27/metabolismo , Células Endoteliales de la Vena Umbilical Humana , Humanos , Inmunoprecipitación , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Microscopía Fluorescente , Permeabilidad/efectos de los fármacos , Polimerizacion , Proteínas Serina-Treonina Quinasas/metabolismo , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Transducción de Señal/efectos de los fármacos , Factor de Necrosis Tumoral alfa/farmacología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
6.
Chin J Physiol ; 61(4): 230-239, 2018 Aug 31.
Artículo en Inglés | MEDLINE | ID: mdl-30139245

RESUMEN

Glutathione S-transferase Pi (GSTpi) is the most important subtribe of GSTs protein superfamily, and plays an important role in the process of detoxification, antioxidant and antiinflammation. Here we use a GSTpi inhibitor, 6-(7-nitro-2, 1, 3-benzoxadiazol-4-ylthio) hexanol (NBDHEX), to evaluate the effect of GSTpi on the growth of mice. Mice treated with NBDHEX have heavier weight and longer length. But there is no significant difference in ratios of the weights of brain, kidney, lung, spleen, liver and heart to the whole body weight and Lee's index between NBDHEX and control mice. These data suggested that GSTpi might inhibit mouse growth. Enzyme-linked immunosorbent assay (ELISA) showed that GSTpi inhibition induced a significant increase of growth hormone (GH) levels in blood and pituitary and insulin-like growth factor (IGF-1) levels in liver and blood in mice. Further observation demonstrated that GSTpi negatively regulated GHJanus Kinase 2 (JAK2)-signal transducer and activator of transcription 5 (STAT5) axis through inhibiting STAT5 phosphorylation, and as a result of GSTpi decreased the expression of IGF-1.


Asunto(s)
Gutatión-S-Transferasa pi/metabolismo , Transducción de Señal , Animales , Inhibidores Enzimáticos , Factor I del Crecimiento Similar a la Insulina , Ratones , Factor de Transcripción STAT5
8.
J Org Chem ; 81(12): 5202-8, 2016 06 17.
Artículo en Inglés | MEDLINE | ID: mdl-27218656

RESUMEN

An efficient method for the synthesis of 6-trichloromethylphenanthridines by benzoyl peroxide promoted cyclization reaction of 2-isocyanobiphenyls with carbon tetrachloride is developed. A radical pathway is proposed and evidenced for the reaction mechanism. This reaction tolerates a wide range of functional groups and the resulting 6-trichloromethylphenanthridines can be utilized as a useful synthetic precursor for corresponding 6-substituted phenanthridines.

9.
J Org Chem ; 81(11): 4797-806, 2016 06 03.
Artículo en Inglés | MEDLINE | ID: mdl-27220261

RESUMEN

An efficient method for the synthesis of (Z)-selective α-trifluoromethyl alkenyl triflates is described. As an important fluorinated building block, it is utilized successfully for the synthesis of various trifluoromethyl derivatives such as diarylethylenes, enynes, alkynes, and benzofurans.

10.
J Urol ; 194(2): 556-62, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25752441

RESUMEN

PURPOSE: CD8(+) TILs at different tumor sites have diverse clinical attributes, which might result from distinct tumor microenvironments that promote differentiation into distinct subsets. However, only a few markers have been identified that can define CD8(+) T-cell subsets. CD103 is a marker of tissue resident memory CD8(+) T cells. In this retrospective study we investigated the cellular source and clinical significance of CD103 expression in urothelial cell carcinoma of bladder tissues in situ. MATERIALS AND METHODS: Immunohistochemistry and immunofluorescence were used to identify the cellular source of CD103 in bladder urothelial cell carcinoma tissues. Kaplan-Meier analysis and Cox proportional hazards regression models were applied to estimate overall and recurrence-free survival in 302 patients with bladder urothelial cell carcinoma. RESULTS: CD8(+) T cells but not natural killer cells accounted for most CD103 expressing cells in bladder urothelial cell carcinoma tissues. Notably CD103(+) cells were predominantly located in intratumor regions rather than in associated stroma (p < 0.0001). The density of intratumor CD103(+) TILs was inversely associated with tumor size (p < 0.0001) and could represent a favorable prognostic predictor of overall and recurrence-free survival (p = 0.002 and 0.011, respectively). Moreover, intratumor CD103(+) TILs were positively associated with the expression of cognate ligand E-cadherin in intratumor regions of bladder urothelial cell carcinoma tissues (p = 0.008). CONCLUSIONS: Our findings suggest that CD8(+) T cells might have a significant role in tumor immunity by expressing CD103 in intratumor regions of bladder urothelial cell carcinoma tissues. Intratumor CD103(+) TILs could potentially serve as a prognostic marker in patients with bladder urothelial cell carcinoma.


Asunto(s)
Antígenos CD/inmunología , Carcinoma de Células Transicionales/inmunología , Cadenas alfa de Integrinas/inmunología , Linfocitos Infiltrantes de Tumor/inmunología , Estadificación de Neoplasias , Neoplasias de la Vejiga Urinaria/inmunología , Vejiga Urinaria/inmunología , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Antígenos CD/metabolismo , Biomarcadores de Tumor/análisis , Carcinoma de Células Transicionales/mortalidad , Carcinoma de Células Transicionales/patología , China/epidemiología , Supervivencia sin Enfermedad , Femenino , Técnica del Anticuerpo Fluorescente , Humanos , Inmunohistoquímica , Cadenas alfa de Integrinas/metabolismo , Estimación de Kaplan-Meier , Linfocitos Infiltrantes de Tumor/metabolismo , Linfocitos Infiltrantes de Tumor/patología , Masculino , Persona de Mediana Edad , Pronóstico , Modelos de Riesgos Proporcionales , Estudios Retrospectivos , Tasa de Supervivencia/tendencias , Vejiga Urinaria/metabolismo , Vejiga Urinaria/patología , Neoplasias de la Vejiga Urinaria/mortalidad , Neoplasias de la Vejiga Urinaria/patología , Adulto Joven
11.
Biochem Biophys Res Commun ; 443(1): 326-32, 2014 Jan 03.
Artículo en Inglés | MEDLINE | ID: mdl-24321097

RESUMEN

Septic diseases represent the prevalent complications in intensive care units. Luteolin, a plant flavonoid, has potent anti-inflammatory properties; however, the molecular mechanism beneath luteolin mediated immune modulation remains unclear. Here in vitro investigations showed that luteolin dose-dependently inhibited LPS-triggered secretion and relocation of high mobility group B-1 (HMGB1) and LPS-induced production of tumor necrosis factor alpha (TNF-α) and nitric oxide (NO) in macrophages. The mechanism analysis demonstrated that luteolin reduced the release of HMGB1 through destabilizing c-Jun and suppressed HMGB1-induced aggravation of inflammatory cascade through reducing Akt protein level. As an inhibitor of Hsp90, luteolin destabilized Hsp90 client protein c-Jun and Akt. In vivo investigations showed that luteolin effectively protected mice from lipopolysaccharide (LPS)-induced lethality. In conclusion, the present study suggested that luteolin may act as a potential therapeutic reagent for treating septic diseases.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacología , Proteínas HSP90 de Choque Térmico/antagonistas & inhibidores , Luteolina/farmacología , Macrófagos/efectos de los fármacos , Animales , Antiinflamatorios no Esteroideos/uso terapéutico , Línea Celular , Modelos Animales de Enfermedad , Células HEK293 , Proteínas HSP90 de Choque Térmico/metabolismo , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Lipopolisacáridos , Luteolina/uso terapéutico , Ratones , Ratones Endogámicos BALB C , Proteínas Proto-Oncogénicas c-akt/metabolismo , Sepsis/tratamiento farmacológico , Transducción de Señal/efectos de los fármacos
12.
Int Immunopharmacol ; 137: 112363, 2024 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-38851161

RESUMEN

Intestinal dysfunction plays a pivotal role in the development of acute pancreatitis (AP), however, the underlying mechanisms of intestinal dysfunction on severity of hyperlipidemic acute pancreatitis (HLAP) are still unclear. Herein, we explored the role of intestinal function on the severity of HLAP. We found that HLAP patients exhibit higher lipid and inflammatory response than AP patients. Hyperlipidemia significantly elevates serum lipids and worsen pancreatic damage in AP mice. In addition, significant exacerbated intestinal barrier damage and inflammation were observed in experimental HLAP mice, as evidenced by increased serum amylase and lipase levels, and pancreatic edema. Further, RNA-Seq showed that a markedly decrease of glutathione S-transferase pi (GSTpi) in colonic tissue of HLAP mice compared with AP mice, accompanied with increased serum lipopolysaccharides level. However, colonic GSTpi overexpression by adeno-associated virus significantly attenuated intestinal damage and subsequent pancreatic inflammation in HLAP mice. Mechanistically, GSTpi mitigated HLAP-mediated colonic NLRP3 inflammasome activation and barrier dysfunction. These results suggest that intestinal GSTpi deficiency exacerbates the severity of experimental HLAP, providing new insights for the clinical treatment of HLAP.


Asunto(s)
Hiperlipidemias , Ratones Endogámicos C57BL , Pancreatitis , Animales , Pancreatitis/patología , Humanos , Ratones , Masculino , Modelos Animales de Enfermedad , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Índice de Severidad de la Enfermedad , Inflamasomas/metabolismo , Mucosa Intestinal/patología , Mucosa Intestinal/metabolismo , Mucosa Intestinal/inmunología , Intestinos/patología , Ratones Noqueados , Femenino , Colon/patología , Páncreas/patología
13.
Phytomedicine ; 128: 155403, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38564920

RESUMEN

BACKGROUND: Cardiovascular disease is one of the main causes of global mortality, and there is an urgent need for effective treatment strategies. Gut microbiota-dependent metabolite trimethylamine-N-oxide (TMAO) promotes the development of cardiovascular diseases, and shizukaol C, a natural sesquiterpene isolated from Chloranthus multistachys with various biological activities, might exhibit beneficial role in preventing TMAO-induced vascular inflammation. PURPOSE: The purpose of this study was to investigate the anti-inflammatory effects and the underlying mechanisms of shizukaol C on TMAO-induced vascular inflammation. METHODS: The effect and underlying mechanism of shizukaol C on TMAO-induced adhesion molecules expression, bone marrow-derived macrophages (BMDM) adhesion to VSMC were evaluated by western blot, cell adhesion assay, co-immunoprecipitation, immunofluorescence assay, and quantitative Real-Time PCR, respectively. To verify the role of shizukaol C in vivo, TMAO-induced vascular inflammation model were established using guidewire-induced injury on mice carotid artery. Changes in the intima area and the expression of GSTpi, VCAM-1, CD68 were examined using haematoxylin-eosin staining, and immunofluorescence assay. RESULTS: Our data demonstrated that shizukaol C significantly suppressed TMAO-induced adhesion molecule expression and the bone marrow-derived macrophages (BMDM) adhesion in vascular smooth muscle cells (VSMC). Mechanically, shizukaol C inhibited TMAO-induced c-Jun N-terminal kinase (JNK)-nuclear factor-kappa B (NF-κB)/p65 activation, and the JNK inhibition was dependent on the shizukaol C-mediated glutathione-S-transferase pi (GSTpi) expression. By further molecular docking and protein-binding analysis, we demonstrated that shizukaol C directly binds to Keap1 to induce Nrf2 nuclear translocation and upregulated GSTpi expression. Consistently, our in vivo experiment showed that shizukaol C elevated the expression level of GSTpi in carotid arteries and alleviates TMAO-induced vascular inflammation. CONCLUSION: Shizukaol C exerts anti-inflammatory effects in TMAO-treated VSMC by targeting Keap1 and activating Nrf2-GSTpi signaling and resultantly inhibits the downstream JNK-NF-κB/p65 activation and VSMC adhesion, and alleviates TMAO-induced vascular inflammation in vivo, suggesting that shizukaol C may be a potential drug for treating TMAO-induced vascular diseases.


Asunto(s)
Inflamación , Músculo Liso Vascular , Sesquiterpenos , Animales , Masculino , Ratones , Antiinflamatorios/farmacología , Adhesión Celular/efectos de los fármacos , Inflamación/inducido químicamente , Inflamación/tratamiento farmacológico , Proteína 1 Asociada A ECH Tipo Kelch/efectos de los fármacos , Proteína 1 Asociada A ECH Tipo Kelch/metabolismo , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Metilaminas/farmacología , Ratones Endogámicos C57BL , Músculo Liso Vascular/efectos de los fármacos , Miocitos del Músculo Liso/efectos de los fármacos , Factor 2 Relacionado con NF-E2/efectos de los fármacos , Factor 2 Relacionado con NF-E2/metabolismo , Sesquiterpenos/farmacología , Transducción de Señal/efectos de los fármacos , Gutatión-S-Transferasa pi/efectos de los fármacos , Gutatión-S-Transferasa pi/metabolismo
14.
Chin Med ; 18(1): 78, 2023 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-37370098

RESUMEN

BACKGROUND: Acute pancreatitis (AP) is a recurrent inflammatory disease. Studies have shown that intestinal homeostasis is essential for the treatment of AP. Formononetin is a plant-derived isoflavone with antioxidant properties that can effectively treat a variety of inflammatory diseases. This study aims to investigate the role of formononetin in protecting against AP and underlying mechanism. METHODS: Caerulein was used to induce AP. The inflammatory cytokines were detected using Quantitative real-time PCR and commercial kits. Histological examination was applied with hematoxylin and eosin staining. Western blot was conducted to detect expression of intestinal barrier protein and signaling molecular. Molecular docking was performed to assess protein-ligand interaction. RESULTS: In this study, we found formononetin administration significantly reduced pancreatic edema, the activities of serum amylase, lipase, myeloperoxidase, and serum endotoxin. The mRNA levels of inflammatory cytokines such as tumor necrosis factor α, monocyte chemoattractant protein-1, interleukin-6, and interleukin-1 beta (IL-1ß) in pancreas were also significantly decreased by formononetin. The following data showed formononetin pretreatment up-regulated the expressions of tight junction proteins in the colon, and decreased Escherichia coli translocation in the pancreas. In addition, formononetin inhibited the activation of nucleotide-binding oligomerization domain leucine-rich repeat and pyrin domain-containing 3 in pancreatic and colonic tissues of AP mice. Moreover, formononetin activated Kelch Like ECH Associated Protein 1 (Keap1) / Nuclear factor erythroid2-related factor 2 (Nrf2) signaling pathway to reduce reactive oxygen species (ROS) levels. Docking results showed that formononetin interact with Keap1 through hydrogen bond. CONCLUSIONS: These findings demonstrate that formononetin administration significantly mitigate AP through reducing oxidative stress and restoring intestinal homeostasis, and provide insights into the new treatment for AP.

15.
Cardiovasc Res ; 119(11): 2142-2156, 2023 09 05.
Artículo en Inglés | MEDLINE | ID: mdl-37201513

RESUMEN

AIMS: Accumulating evidence supports the indispensable role of protein arginine methyltransferase 5 (PRMT5) in the pathological progression of several human cancers. As an important enzyme-regulating protein methylation, how PRMT5 participates in vascular remodelling remains unknown. The aim of this study was to investigate the role and underlying mechanism of PRMT5 in neointimal formation and to evaluate its potential as an effective therapeutic target for the condition. METHODS AND RESULTS: Aberrant PRMT5 overexpression was positively correlated with clinical carotid arterial stenosis. Vascular smooth muscle cell (SMC)-specific PRMT5 knockout inhibited intimal hyperplasia with an enhanced expression of contractile markers in mice. Conversely, PRMT5 overexpression inhibited SMC contractile markers and promoted intimal hyperplasia. Furthermore, we showed that PRMT5 promoted SMC phenotypic switching by stabilizing Kruppel-like factor 4 (KLF4). Mechanistically, PRMT5-mediated KLF4 methylation inhibited ubiquitin-dependent proteolysis of KLF4, leading to a disruption of myocardin (MYOCD)-serum response factor (SRF) interaction and MYOCD-SRF-mediated the transcription of SMC contractile markers. CONCLUSION: Our data demonstrated that PRMT5 critically mediated vascular remodelling by promoting KLF4-mediated SMC phenotypic conversion and consequently the progression of intimal hyperplasia. Therefore, PRMT5 may represent a potential therapeutic target for intimal hyperplasia-associated vascular diseases.


Asunto(s)
Factor 4 Similar a Kruppel , Factores de Transcripción de Tipo Kruppel , Animales , Humanos , Ratones , Arginina , Hiperplasia/metabolismo , Factores de Transcripción de Tipo Kruppel/genética , Metilación , Miocitos del Músculo Liso/metabolismo , Neointima/metabolismo , Proteína-Arginina N-Metiltransferasas/genética , Proteína-Arginina N-Metiltransferasas/metabolismo , Proteína-Arginina N-Metiltransferasas/farmacología , Remodelación Vascular
16.
Front Cardiovasc Med ; 9: 986353, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35990954

RESUMEN

[This corrects the article DOI: 10.3389/fcvm.2021.756098.].

17.
Biochem Pharmacol ; 198: 114947, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35143753

RESUMEN

Neointima formation is characterized by the proliferation of vascular smooth muscle cells (VSMC). Although lysine-specific demethylase 1 (LSD1) has critical functions in several diseases, its role in neointima formation remains to be clarified. In this study, we aimed to explore the crucial role of LSD1 on neointima formation using a carotid artery injury model in mice. We observed that aberrant LSD1 expression was increased in human and mouse stenotic arteries and platelet-derived growth factor-BB (PDGF-BB)-treated VSMC. Furthermore, LSD1 knockdown significantly mitigated neointima formation in vivo and inhibited PDGF-BB-induced VSMC proliferation in vitro. We further uncovered that LSD1 overexpression exhibited opposite phenotypes in vivo and in vitro. Finally, LSD1 knockdown inhibited VSMC proliferation by increasing p21 expression, which is associated with LSD1 mediated di-methylated histone H3 on lysine 4 (H3K4me2) modification. Taken together, our data suggest that LSD1 may be a potential therapeutic target for the treatment of neointima formation.


Asunto(s)
Traumatismos de las Arterias Carótidas , Histona Demetilasas , Miocitos del Músculo Liso , Neointima , Animales , Becaplermina/metabolismo , Becaplermina/farmacología , Traumatismos de las Arterias Carótidas/genética , Traumatismos de las Arterias Carótidas/metabolismo , Movimiento Celular , Proliferación Celular , Células Cultivadas , Histona Demetilasas/genética , Histona Demetilasas/metabolismo , Humanos , Lisina/metabolismo , Ratones , Músculo Liso Vascular/citología , Miocitos del Músculo Liso/metabolismo , Neointima/metabolismo
18.
Mol Nutr Food Res ; 66(23): e2200300, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36208084

RESUMEN

SCOPE: Dietary fibers can alter gut microbiota and microbial metabolite profiles. SCFAs are produced by bacterial fermentation of fiber, mediating immune homeostasis through G-protein-coupled receptors (GPCRs). GPR109a, a receptor for niacin and butyrate, expressed by immune cells and non-immune cells, is a key factor regulating immune responses. However, the role and underlying mechanisms of GPR109a in type 1 diabetes (T1D) remain unclear. METHODS AND RESULTS: Experimental T1D was induced by streptozotocin in GPR109a-deficient (Gpr109a-/- ) and wild type mice. The study found that Gpr109a-/- mice were more susceptible to T1D with dysregulated immune responses, along with increased M1 macrophage polarization (from 10.55% to 21.48%). Further, an adoptive transfer experiment demonstrated that GPR109a-deficient macrophages promoted the homing of intestine-derived type 1 cytotoxic T cells to pancreas (from 18.91% to 24.24%), thus disturbing the pancreatic immune homeostasis in non-obese diabetic mice. Mechanistically, GPR109a deficiency promoted M1 macrophage polarization associated with the activation of suppressor of cytokine signaling 3-signal transducer and activator of transcription 1 signaling pathway. CONCLUSION: The findings reveal that macrophage GPR109a deficiency accelerates the development of T1D. Activation of GPR109a on macrophage by dietary components may provide a new strategy for preventing or treating T1D.


Asunto(s)
Diabetes Mellitus Experimental , Diabetes Mellitus Tipo 1 , Receptores Nicotínicos , Ratones , Animales , Receptores Nicotínicos/metabolismo , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Macrófagos/metabolismo
19.
Ying Yong Sheng Tai Xue Bao ; 33(6): 1489-1496, 2022 Jun.
Artículo en Zh | MEDLINE | ID: mdl-35729124

RESUMEN

To explore food composition of Chinese mitten crab (Eriocheir sinensis) in rice-crab integrated ecosystem in saline-alkali land of the Yellow River Delta, we analyzed carbon and nitrogen stable isotope ratios (δ13C and δ15N) in the crab and that in food sources, including plants (Elodea, Potamogeton crispus, Ceratophyllum demersum, Lemna minor, Oryza sativa stem and leaf, rice grain), animals (benthos, zooplankton), organic debris and artificial feed (compound feed, corn meal) in Kenli District, Dongying, Shandong Province in June to October of 2020. Substantial differences in δ13C and δ15N were found among food sources. The δ13C and δ15N values of different food sources were in a range of -30.09‰--11.24‰ and 0.03‰-12.78‰, respectively, while those of the crab muscle were in range of -24.61‰--20.08‰ and 4.74‰-9.21‰, respectively, indicating diverse food sources for the crab. During the experiment, the contribution rate of different food sources followed the order: plant (46.7%-57.1%)>animal (21.5%-24.5%)>artificial feed (10.9%-21.3%)>organic detritus (7.1%-7.9%). It suggested that the natural bait of the paddy field could meet the feeding needs of Chinese mitten crabs in saline-alkali land. Even the crabs were fed with non-animal artificial feed, the contribution rates of the main food sources were not altered.


Asunto(s)
Oryza , Ríos , Álcalis , Animales , Isótopos de Carbono/análisis , Ecosistema , Isótopos de Nitrógeno/análisis
20.
Br J Pharmacol ; 179(1): 141-158, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34599829

RESUMEN

BACKGROUND AND PURPOSE: There is emerging evidence for a critical role for epigenetic modifiers in the development of inflammatory bowel disease (IBD). Protein arginine methyltransferase 2 (PRMT2) is responsible for the methylation of arginine residues on histones and targets transcription factors involved in many cellular processes, including gene transcription, mRNA splicing, cell proliferation, and cell differentiation. In this study, the role and underlying mechanisms of PRMT2 in colitis were studied. EXPERIMENTAL APPROACH: A mouse dextran sulfate sodium (DSS)-induced experimental colitis model was used to study PRMT2 in colitis. Lentivirus-induced PRMT2 silencing or overexpression in vivo was applied to address the role of PRMT2 in colitis. Detailed western blot and expression analysis were done to understand epigenetic changes induced by PRMT2 in colitis. KEY RESULTS: PRMT2 is highly expressed in inflammatory bowel disease patients, in inflamed murine colon and in TNF-α stimulated murine gut epithelial cells. PRMT2 overexpression aggravates, while knockdown alleviates DSS-induced colitis, suggesting that PRMT2 is a pivotal mediator of colitis in mice. Mechanistically, PRMT2 mediates colitis by increasing repressive histone mark H3R8 asymmetric methylation (H3R8me2a) at the promoter region of the suppressor of cytokine signalling 3 promoter (SOCS3). Resultant inhibition of SOCS3 expression and inhibition of SOCS3-mediated degradation of TNF receptor associated factor 5 (TRAF5) via ubiquitination led to elevated TRAF5 expression and TRAF5-mediated downstream NF-κB/MAPK activation. CONCLUSION AND IMPLICATIONS: Our study demonstrates that PRMT2 acts as a transcriptional co-activator for proinflammatory genes during colitis. Hence, targeting PRMT2 may provide a novel therapeutic approach for colitis.


Asunto(s)
Colitis , Histonas , Animales , Colitis/inducido químicamente , Colitis/genética , Sulfato de Dextran/farmacología , Histonas/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Ratones , FN-kappa B/metabolismo , Regiones Promotoras Genéticas , Proteína-Arginina N-Metiltransferasas/genética , Proteína 3 Supresora de la Señalización de Citocinas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA