Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 5.872
Filtrar
1.
Nat Commun ; 15(1): 7730, 2024 Sep 04.
Artículo en Inglés | MEDLINE | ID: mdl-39231983

RESUMEN

Mutations in mitochondrial energy-producing genes lead to a heterogeneous group of untreatable disorders known as primary mitochondrial diseases (MD). Leigh syndrome (LS) is the most common pediatric MD and is characterized by progressive neuromuscular affectation and premature death. Here, we show that daily cannabidiol (CBD) administration significantly extends lifespan and ameliorates pathology in two LS mouse models, and improves cellular function in fibroblasts from LS patients. CBD delays motor decline and neurodegenerative signs, improves social deficits and breathing abnormalities, decreases thermally induced seizures, and improves neuropathology in affected brain regions. Mechanistically, we identify peroxisome proliferator-activated receptor gamma (PPARγ) as a key nuclear receptor mediating CBD's beneficial effects, while also providing proof of dysregulated PPARγ expression and activity as a common feature in both mouse neurons and fibroblasts from LS patients. Taken together, our results provide the first evidence for CBD as a potential treatment for LS.


Asunto(s)
Cannabidiol , Enfermedades Mitocondriales , PPAR gamma , Animales , Femenino , Humanos , Masculino , Ratones , Encéfalo/metabolismo , Encéfalo/efectos de los fármacos , Encéfalo/patología , Cannabidiol/farmacología , Cannabidiol/uso terapéutico , Modelos Animales de Enfermedad , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Enfermedad de Leigh/tratamiento farmacológico , Enfermedad de Leigh/metabolismo , Enfermedad de Leigh/genética , Ratones Endogámicos C57BL , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Enfermedades Mitocondriales/tratamiento farmacológico , Enfermedades Mitocondriales/metabolismo , Neuronas/efectos de los fármacos , Neuronas/metabolismo , PPAR gamma/metabolismo , PPAR gamma/genética
2.
Cells ; 13(17)2024 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-39273065

RESUMEN

Prostate apoptosis response-4 (Par-4, also known as PAWR) is a ubiquitously expressed tumor suppressor protein that induces apoptosis selectively in cancer cells, while leaving normal cells unaffected. Our previous studies indicated that genetic loss of Par-4 promoted hepatic steatosis, adiposity, and insulin-resistance in chow-fed mice. Moreover, low plasma levels of Par-4 are associated with obesity in human subjects. The mechanisms underlying obesity in rodents and humans are multi-faceted, and those associated with adipogenesis can be functionally resolved in cell cultures. We therefore used pluripotent mouse embryonic fibroblasts (MEFs) or preadipocyte cell lines responsive to adipocyte differentiation cues to determine the potential role of Par-4 in adipocytes. We report that pluripotent MEFs from Par-4-/- mice underwent rapid differentiation to mature adipocytes with an increase in lipid droplet accumulation relative to MEFs from Par-4+/+ mice. Knockdown of Par-4 in 3T3-L1 pre-adipocyte cultures by RNA-interference induced rapid differentiation to mature adipocytes. Interestingly, basal expression of PPARγ, a master regulator of de novo lipid synthesis and adipogenesis, was induced during adipogenesis in the cell lines, and PPARγ induction and adipogenesis caused by Par-4 loss was reversed by replenishment of Par-4. Mechanistically, Par-4 downregulates PPARγ expression by directly binding to its upstream promoter, as judged by chromatin immunoprecipitation and luciferase-reporter studies. Thus, Par-4 transcriptionally suppresses the PPARγ promoter to regulate adipogenesis.


Asunto(s)
Células 3T3-L1 , Adipocitos , Adipogénesis , Proteínas Reguladoras de la Apoptosis , PPAR gamma , Animales , PPAR gamma/metabolismo , PPAR gamma/genética , Adipogénesis/genética , Ratones , Adipocitos/metabolismo , Adipocitos/citología , Proteínas Reguladoras de la Apoptosis/metabolismo , Proteínas Reguladoras de la Apoptosis/genética , Diferenciación Celular , Humanos , Transcripción Genética , Regiones Promotoras Genéticas/genética , Fibroblastos/metabolismo
3.
Int J Mol Sci ; 25(17)2024 Aug 23.
Artículo en Inglés | MEDLINE | ID: mdl-39273102

RESUMEN

Embryonic stem cells are crucial for studying developmental biology due to their self-renewal and pluripotency capabilities. This research investigates the differentiation of mouse ESCs into adipocytes, offering insights into obesity and metabolic disorders. Using a monolayer differentiation approach over 30 days, lipid accumulation and adipogenic markers, such as Cebpb, Pparg, and Fabp4, confirmed successful differentiation. RNA sequencing revealed extensive transcriptional changes, with over 15,000 differentially expressed genes linked to transcription regulation, cell cycle, and DNA repair. This study utilized Robust Rank Aggregation to identify critical regulatory genes like PPARG, CEBPA, and EP300. Network analysis further highlighted Atf5, Ccnd1, and Nr4a1 as potential key players in adipogenesis and its mature state, validated through RT-PCR. While key adipogenic factors showed plateaued expression levels, suggesting early differentiation events, this study underscores the value of ESCs in modeling adipogenesis. These findings contribute to our understanding of adipocyte differentiation and have significant implications for therapeutic strategies targeting metabolic diseases.


Asunto(s)
Adipocitos , Adipogénesis , Diferenciación Celular , Células Madre Embrionarias de Ratones , Animales , Adipogénesis/genética , Ratones , Células Madre Embrionarias de Ratones/metabolismo , Células Madre Embrionarias de Ratones/citología , Diferenciación Celular/genética , Adipocitos/metabolismo , Adipocitos/citología , PPAR gamma/metabolismo , PPAR gamma/genética , Proteínas Potenciadoras de Unión a CCAAT/metabolismo , Proteínas Potenciadoras de Unión a CCAAT/genética , Perfilación de la Expresión Génica , Redes Reguladoras de Genes , Transcripción Genética , Regulación de la Expresión Génica
4.
J Microbiol Biotechnol ; 34(8): 1688-1697, 2024 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-39086228

RESUMEN

The current study aimed to determine whether Strongylocentrotus intermedius (S. intermedius) extract (SIE) exerts anti-obesity potentials employing 3T3-L1 cells as in vitro model. Herein we reported that treatment of SIE for 6 days reduced lipid accretion and triglyceride content whereas it increased the release of free glycerol. The inhibited lipid accumulation and induced lipolysis were evidenced by the downregulation of lipogenesis proteins, such as fatty acid synthase and lipoprotein lipase, and the upregulation of hormone-sensitive lipase expression. Furthermore, the downregulation of adipogenic transcription factors, including peroxisome proliferator-activated receptor gamma, CCAAT/enhancer-binding protein α, and sterol regulatory element-binding protein 1, highlights that reduced lipid accumulation is supported by lowering adipocyte differentiation. Additionally, treatment activates brown adipocyte phenotype in 3T3-L1 cells by inducing expression of brown adipose tissue-specific proteins, such as uncoupling protein 1 and peroxisome proliferator-activated receptor-γ coactivator 1α. Moreover, SIE induced the phosphorylation of AMP-activated protein kinase (AMPK). The pharmacological approach using AMPK inhibitor revealed that the restraining effect of SIE on adipogenesis and promotion of adipocyte browning were blocked. In GC-MS analysis, SIE was mainly composed of cholest-5-en-3-ol (36.71%) along with saturated and unsaturated fatty acids which have favorable anti-obesity potentials. These results reveal that SIE has the possibility as a lipid-lowering agent for the intervention of obesity.


Asunto(s)
Células 3T3-L1 , Proteínas Quinasas Activadas por AMP , Adipogénesis , Animales , Adipogénesis/efectos de los fármacos , Ratones , Proteínas Quinasas Activadas por AMP/metabolismo , Lipólisis/efectos de los fármacos , PPAR gamma/metabolismo , PPAR gamma/genética , Adiposidad/efectos de los fármacos , Fármacos Antiobesidad/farmacología , Metabolismo de los Lípidos/efectos de los fármacos , Adipocitos Marrones/efectos de los fármacos , Adipocitos Marrones/metabolismo , Adipocitos/efectos de los fármacos , Adipocitos/metabolismo , Triglicéridos/metabolismo , Diferenciación Celular/efectos de los fármacos , Lipogénesis/efectos de los fármacos , Fosforilación/efectos de los fármacos
5.
Sci Rep ; 14(1): 20049, 2024 08 29.
Artículo en Inglés | MEDLINE | ID: mdl-39209927

RESUMEN

Adipogenesis involves intricate molecular mechanisms regulated by various transcription factors and signaling pathways. In this study, we aimed to identify factors specifically induced during adipogenesis in the human preadipocyte cell line, SGBS, but not in the mouse preadipocyte cell line, 3T3-L1. Microarray analysis revealed distinct gene expression profiles, with 1460 genes induced in SGBS cells and 1297 genes induced in 3T3-L1 cells during adipogenesis, with only 297 genes commonly induced. Among the genes uniquely induced in SGBS cells, we focused on GALNT15, which encodes polypeptide N-acetylgalactosaminyltransferase-15. Its expression increased transiently during adipogenesis in SGBS cells but remained low in 3T3-L1 cells. Overexpression of GALNT15 increased mRNA levels of CCAAT-enhancer binding protein (C/EBPα) and leptin but had no significant impact on adipogenesis in SGBS cells. Conversely, knockdown of GALNT15 suppressed mRNA expression of adipocyte marker genes, reduced lipid accumulation, and decreased the percentage of cells with oil droplets. The induction of C/EBPα and peroxisome proliferator-activated receptor γ during adipogenesis was promoted or suppressed in SGBS cells subjected to overexpression or knockdown of GALNT15, respectively. These data suggest that polypeptide N-acetylgalactosaminyltransferase-15 is a novel regulatory molecule that enhances adipogenesis in SGBS cells.


Asunto(s)
Células 3T3-L1 , Adipogénesis , N-Acetilgalactosaminiltransferasas , Polipéptido N-Acetilgalactosaminiltransferasa , Adipogénesis/genética , Humanos , N-Acetilgalactosaminiltransferasas/metabolismo , N-Acetilgalactosaminiltransferasas/genética , Ratones , Animales , Adipocitos/metabolismo , Adipocitos/citología , PPAR gamma/metabolismo , PPAR gamma/genética , Línea Celular , Proteínas Potenciadoras de Unión a CCAAT/metabolismo , Proteínas Potenciadoras de Unión a CCAAT/genética , Leptina/metabolismo , Leptina/genética
6.
Cells ; 13(16)2024 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-39195246

RESUMEN

Given the importance of peroxisome proliferator-activated receptor (PPAR)-gamma in epidermal inflammation and carcinogenesis, we analyzed the transcriptomic changes observed in epidermal PPARγ-deficient mice (Pparg-/-epi). A gene set enrichment analysis revealed a close association with epithelial malignancy, inflammatory cell chemotaxis, and cell survival. Single-cell sequencing of Pparg-/-epi mice verified changes to the stromal compartment, including increased inflammatory cell infiltrates, particularly neutrophils, and an increase in fibroblasts expressing myofibroblast marker genes. A comparison of transcriptomic data from Pparg-/-epi and publicly available human and/or mouse actinic keratoses (AKs) and cutaneous squamous cell carcinomas (SCCs) revealed a strong correlation between the datasets. Importantly, PPAR signaling was the top common inhibited canonical pathway in AKs and SCCs. Both AKs and SCCs also had significantly reduced PPARG expression and PPARγ activity z-scores. Smaller reductions in PPARA expression and PPARα activity and increased PPARD expression but reduced PPARδ activation were also observed. Reduced PPAR activity was also associated with reduced PPARα/RXRα activity, while LPS/IL1-mediated inhibition of RXR activity was significantly activated in the tumor datasets. Notably, these changes were not observed in normal sun-exposed skin relative to non-exposed skin. Finally, Ppara and Pparg were heavily expressed in sebocytes, while Ppard was highly expressed in myofibroblasts, suggesting that PPARδ has a role in myofibroblast differentiation. In conclusion, these data provide strong evidence that PPARγ and possibly PPARα represent key tumor suppressors by acting as master inhibitors of the inflammatory changes found in AKs and SCCs.


Asunto(s)
Carcinoma de Células Escamosas , Inflamación , Queratosis Actínica , PPAR gamma , Transducción de Señal , Neoplasias Cutáneas , PPAR gamma/metabolismo , PPAR gamma/genética , Carcinoma de Células Escamosas/patología , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/genética , Animales , Humanos , Neoplasias Cutáneas/patología , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/metabolismo , Queratosis Actínica/patología , Queratosis Actínica/metabolismo , Queratosis Actínica/genética , Ratones , Inflamación/patología , Inflamación/metabolismo , Inflamación/genética , Regulación Neoplásica de la Expresión Génica , Células del Estroma/metabolismo , Células del Estroma/patología
7.
Clin Respir J ; 18(8): e13812, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39107932

RESUMEN

BACKGROUND: Qi deficiency and phlegm dampness (QPD) is one of the most common traditional Chinese medicine (TCM) syndromes in lung adenocarcinoma (LUAD). This study aimed to identify syndrome-specific biomarkers for LUAD with QPD syndrome. METHODS: Peripheral blood mononuclear cells (PBMCs) from LUAD patients with QPD, LUAD patients with non-QPD (N-QPD), and healthy control (H) were collected and analyzed with RNA-seq to identify differentially expressed genes (DEGs). The area under the receiver operator characteristic curve (AUC) of each DEG was calculated, and the top 10 highest AUC DEGs were validated by qRT-PCR. Logistic regression analysis was used to develop a diagnostic model evaluated with AUC. RESULTS: A total of 135 individuals were enrolled in this study (training set: 15 QPD, 15 N-QPD, 15 H; validation set: 30 QPD, 30 N-QPD, 30 H). A total of 1480 DEGs were identified between QPD and N-QPD. The qRT-PCR results showed that the expression of DDR2 was downregulated, and PPARG was upregulated, which was in line with the finding of the training set. We developed a diagnostic model with these two genes. The AUC of the diagnostic model in the training cohort and validation cohort was 0.891 and 0.777, respectively. CONCLUSIONS: We identified the two genes (DDR2 and PPARG) as syndrome-specific biomarkers for LUAD with QPD syndrome and developed a novel diagnostic model, which may help to improve the accuracy and sensibility of clinical diagnosis and provide a new target for natural drug treatment of LUAD.


Asunto(s)
Adenocarcinoma del Pulmón , Biomarcadores de Tumor , Neoplasias Pulmonares , Medicina Tradicional China , Humanos , Masculino , Femenino , Adenocarcinoma del Pulmón/genética , Adenocarcinoma del Pulmón/diagnóstico , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/diagnóstico , Medicina Tradicional China/métodos , Persona de Mediana Edad , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/sangre , Biomarcadores de Tumor/metabolismo , PPAR gamma/genética , PPAR gamma/metabolismo , Anciano , Qi , Leucocitos Mononucleares/metabolismo , Curva ROC , Estudios de Casos y Controles
8.
Gene ; 931: 148892, 2024 Dec 30.
Artículo en Inglés | MEDLINE | ID: mdl-39187138

RESUMEN

Sepsis-related brain injury (SRBI) refers to brain dysfunction and structural damage caused by sepsis, which is characterized by inflammation, oxidative stress, and destruction of the blood-brain barrier. Pioglitazone is a PPAR-γ agonist in which PPAR-γ acts as an inflammatory modulator, determining the relationship between PPAR-γ and SRBI and inflammatory state is critical for the disease. This study aimed to construct a drug-target-disease network for SRBI and Pioglitazone based on network pharmacology, and to investigate the therapeutic effect and potential mechanism of Pioglitazone in SRBI induced by lipopolysaccharide (LPS) in rats through transcriptomics. To establish a rat Model of SRBI by intraperitoneal injection of LPS (10 mg/kg): SD rats were divided into Control, Model (LPS), Pioglitazone, (LPS + Pioglitazone) and GW9662 group (LPS+GW9662). The effects and potential mechanisms of Pioglitazone in the treatment of SRBI were studied using biochemical indexes, pathological changes and transcriptome-sequencing (RNA-seq). RNA-seq results showed 620 DEGs between the Model and the Pioglitazone groups. Enrichment analysis involved multiple inflammatory response processes and chemokine receptor binding functions. TLR4 and CXCL10 in the Toll signaling pathway may play an important role in SRBI as important targets. Pioglitazone may ameliorate SRBI through the PPAR-γ/TLR4/CXCL10 pathway.


Asunto(s)
Lipopolisacáridos , PPAR gamma , Pioglitazona , Ratas Sprague-Dawley , Sepsis , Transcriptoma , Pioglitazona/farmacología , Animales , Ratas , PPAR gamma/metabolismo , PPAR gamma/genética , Masculino , Transcriptoma/efectos de los fármacos , Sepsis/tratamiento farmacológico , Sepsis/genética , Sepsis/complicaciones , Sepsis/metabolismo , Receptor Toll-Like 4/metabolismo , Receptor Toll-Like 4/genética , Quimiocina CXCL10/genética , Quimiocina CXCL10/metabolismo , Modelos Animales de Enfermedad , Transducción de Señal/efectos de los fármacos , Anilidas/farmacología , Lesiones Encefálicas/tratamiento farmacológico , Lesiones Encefálicas/metabolismo , Lesiones Encefálicas/genética , Lesiones Encefálicas/etiología , Perfilación de la Expresión Génica , Encefalopatía Asociada a la Sepsis/tratamiento farmacológico , Encefalopatía Asociada a la Sepsis/genética , Encefalopatía Asociada a la Sepsis/metabolismo
9.
J Nutr Biochem ; 132: 109696, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39094217

RESUMEN

Nobiletin has been reported to protect against obesity-related metabolic disorders by enhancing the circadian rhythm; however its effects on lipid metabolism in adipose tissue are unclear. In this study, mice were fed with high-fat diet (HFD) for four weeks firstly and gavaged with 50 or 200 mg/kg bodyweight/day nobiletin at Zeitgeber time (ZT) 4 for another four weeks while still receiving HFD. At the end of the 8-week experimental period, the mice were sacrificed at ZT4 or ZT8 on the same day. Mature 3T3-L1 adipocytes were treated with nobiletin in the presence or absence of siBmal1, siRora, siRorc, SR8278 or SR9009. Nobiletin reduced the weight of white adipose tissue (WAT) and the size of adipocytes in WAT. At ZT4, nobiletin decreased the TG, TC and LDL-c levels and increased serum FFA level and glucose tolerance. Nobiletin triggered the lipolysis of mesenteric and epididymal WAT at both ZT4 and ZT16. Nobiletin increased the level of RORγ at ZT16, that of BMAL1 and PPARγ at ZT4, and that of ATGL at both ZT4 and ZT16. Nobiletin increased lipolysis and ATGL levels in 3T3-L1 adipocytes in Bmal1- or Rora/c- dependent manner. Dual luciferase assay indicated that nobiletin enhanced the transcriptional activation of RORα/γ on Atgl promoter and decreased the repression of RORα/γ on PPARγ-binding PPRE. Promoter deletion analysis indicated that nobiletin inhibited the suppression of PPARγ-mediated Atgl transcription by RORα/γ. Taken together, nobiletin elevated lipolysis in WAT by increasing ATGL levels through activating the transcriptional activity of RORα/γ and decreasing the repression of RORα/γ on PPARγ-binding PPRE.


Asunto(s)
Células 3T3-L1 , Tejido Adiposo Blanco , Relojes Circadianos , Flavonas , Lipólisis , Ratones Endogámicos C57BL , Animales , Flavonas/farmacología , Lipólisis/efectos de los fármacos , Ratones , Tejido Adiposo Blanco/metabolismo , Tejido Adiposo Blanco/efectos de los fármacos , Masculino , Relojes Circadianos/efectos de los fármacos , Factores de Transcripción ARNTL/metabolismo , Factores de Transcripción ARNTL/genética , Dieta Alta en Grasa/efectos adversos , PPAR gamma/metabolismo , PPAR gamma/genética , Miembro 1 del Grupo F de la Subfamilia 1 de Receptores Nucleares/metabolismo , Miembro 1 del Grupo F de la Subfamilia 1 de Receptores Nucleares/genética , Adipocitos/efectos de los fármacos , Adipocitos/metabolismo , Lipasa/metabolismo , Obesidad/metabolismo , Obesidad/tratamiento farmacológico , Aciltransferasas , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares
10.
Signal Transduct Target Ther ; 9(1): 218, 2024 Aug 23.
Artículo en Inglés | MEDLINE | ID: mdl-39174522

RESUMEN

Obesity is a global issue that warrants the identification of more effective therapeutic targets and a better understanding of the pivotal molecular pathogenesis. Annexin A1 (ANXA1) is known to inhibit phospholipase A2, exhibiting anti-inflammatory activity. However, the specific effects of ANXA1 in obesity and the underlying mechanisms of action remain unclear. Our study reveals that ANXA1 levels are elevated in the adipose tissue of individuals with obesity. Whole-body or adipocyte-specific ANXA1 deletion aggravates obesity and metabolic disorders. ANXA1 levels are higher in stromal vascular fractions (SVFs) than in mature adipocytes. Further investigation into the role of ANXA1 in SVFs reveals that ANXA1 overexpression induces lower numbers of mature adipocytes, while ANXA1-knockout SVFs exhibit the opposite effect. This suggests that ANXA1 plays an important role in adipogenesis. Mechanistically, ANXA1 competes with MYC binding protein 2 (MYCBP2) for interaction with PDZ and LIM domain 7 (PDLIM7). This exposes the MYCBP2-binding site, allowing it to bind more readily to the SMAD family member 4 (SMAD4) and promoting its ubiquitination and degradation. SMAD4 degradation downregulates peroxisome proliferator-activated receptor gamma (PPARγ) transcription and reduces adipogenesis. Treatment with Ac2-26, an active peptide derived from ANXA1, inhibits both adipogenesis and obesity through the mechanism. In conclusion, the molecular mechanism of ANXA1 inhibiting adipogenesis was first uncovered in our study, which is a potential target for obesity prevention and treatment.


Asunto(s)
Adipocitos , Adipogénesis , Anexina A1 , Obesidad , PPAR gamma , Anexina A1/genética , Anexina A1/metabolismo , Adipogénesis/genética , Animales , Obesidad/genética , Obesidad/metabolismo , Obesidad/patología , Humanos , Ratones , PPAR gamma/genética , PPAR gamma/metabolismo , Adipocitos/metabolismo , Adipocitos/patología , Proteína Smad4/genética , Proteína Smad4/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Células 3T3-L1 , Péptidos
11.
Zhong Nan Da Xue Xue Bao Yi Xue Ban ; 49(5): 775-783, 2024 May 28.
Artículo en Inglés, Chino | MEDLINE | ID: mdl-39174891

RESUMEN

OBJECTIVES: Non-alcoholic fatty liver disease (NAFLD) has significant genetic susceptibility. Adipocytokines play a crucial role in NAFLD development by participating in insulin resistance and hepatic steatosis. However, the association between adipocytokine pathway genes and NAFLD remains unclear. This study aims to explore the association of gene polymorphisms in the adipocytokine pathway and their interactions with NAFLD in obese children. METHODS: A case-control study was conducted, dividing obese children into NAFLD and control groups. Peripheral venous blood (2 mL) was collected from each participant for DNA extraction. A total of 14 single nucleotide polymorphisms (SNP) in the adipocytokine pathway were genotyped using multiplex PCR and high-throughput sequencing. Univariate and multivariate Logistic regression analyses were used to assess the association between SNP and NAFLD in obese children. Dominant models were used to analyze additive and multiplicative interactions via crossover analysis and Logistic regression. Generalized multifactor dimensionality reduction (GMDR) was used to detect gene-gene interactions among the 14 SNPs and their association with NAFLD in obese children. RESULTS: A total of 1 022 children were included, with 511 in the NAFLD group and 511 in the control group. After adjusting for age, gender, and BMI, multivariate Logistic regression showed that PPARG rs1801282 was associated with NAFLD in the obese children in 3 genetic models: heterozygote model (CG vs CC, OR=0.58, 95% CI 0.36 to 0.95, P=0.029), dominant model (GG+CG vs CC, OR=0.62, 95% CI 0.38 to 1.00, P=0.049), and overdominant model (CC+GG vs CG, OR=1.72, 95% CI 1.06 to 2.80, P=0.028). PRKAG2 rs12703159 was associated with NAFLD in 4 genetic models: heterozygous model (CT vs CC, OR=1.51, 95% CI 1.10 to 2.07, P=0.011), dominant model (CT+TT vs CC, OR=1.50, 95% CI 1.10 to 2.03, P=0.010), overdominant model (CC+TT vs CT, OR=0.67, 95% CI 0.49 to 0.92, P=0.012), and additive model (CC vs CT vs TT, OR=1.40, 95% CI 1.07 to 1.83, P=0.015). No significant multiplicative or additive interaction between PPARG rs1801282 and PRKAG2 rs12703159 was found in association with NAFLD. GMDR analysis, adjusted for age, gender, and BMI, revealed no statistically significant interactions among the 14 SNPs (all P>0.05). CONCLUSIONS: Mutations in PPARG rs1801282 and PRKAG2 rs12703159 are associated with NAFLD in obese children. However, no gene-gene interactions among the SNP are found to be associated with NAFLD in obese children.


Asunto(s)
Adipoquinas , Predisposición Genética a la Enfermedad , Enfermedad del Hígado Graso no Alcohólico , Polimorfismo de Nucleótido Simple , Humanos , Enfermedad del Hígado Graso no Alcohólico/genética , Enfermedad del Hígado Graso no Alcohólico/complicaciones , Niño , Estudios de Casos y Controles , Masculino , Femenino , Adipoquinas/genética , Adipoquinas/sangre , Obesidad/genética , Obesidad/complicaciones , PPAR gamma/genética , Adolescente , Obesidad Infantil/genética , Obesidad Infantil/complicaciones
12.
CNS Neurosci Ther ; 30(8): e14881, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39107960

RESUMEN

BACKGROUND: Microglia and infiltrated macrophages (M/M) are integral components of the innate immune system that play a critical role in facilitating brain repair after ischemic stroke (IS) by clearing cell debris. Novel therapeutic strategies for IS therapy involve modulating M/M phenotype shifting. This study aims to elucidate the pivotal role of S100A9 in M/M and its downstream STAT6/PPARγ signaling pathway in neuroinflammation and phagocytosis after IS. METHODS: In the clinical study, we initially detected the expression pattern of S100A9 in monocytes from patients with acute IS and investigated its association with the long-term prognosis. In the in vivo study, we generated the S100A9 conditional knockout (CKO) mice and compared the stroke outcomes with the control group. We further tested the S100A9-specific inhibitor paqunimod (PQD), for its pharmaceutical effects on stroke outcomes. Transcriptomics and in vitro studies were adopted to explore the mechanism of S100A9 in modulating the M/M phenotype, which involves the regulation of the STAT6/PPARγ signaling pathway. RESULTS: S100A9 was predominantly expressed in classical monocytes and was correlated with unfavorable outcomes in patients of IS. S100A9 CKO mitigated infarction volume and white matter injury, enhanced cerebral blood flow and functional recovery, and prompted anti-inflammation phenotype and efferocytosis after tMCAO. The STAT6/PPARγ pathway, an essential signaling cascade involved in immune response and inflammation, might be the downstream target mediated by S100A9 deletion, as evidenced by the STAT6 phosphorylation inhibitor AS1517499 abolishing the beneficial effect of S100A9 inhibition in tMCAO mice and cell lines. Moreover, S100A9 inhibition by PQD treatment protected against neuronal death in vitro and brain injuries in vivo. CONCLUSION: This study provides evidence for the first time that S100A9 in classical monocytes could potentially be a biomarker for predicting IS prognosis and reveals a novel therapeutic strategy for IS. By demonstrating that S100A9-mediated M/M polarization and phagocytosis can be reversed by S100A9 inhibition in a STAT6/PPARγ pathway-dependent manner, this study opens up new avenues for drug development in the field.


Asunto(s)
Calgranulina B , Accidente Cerebrovascular Isquémico , Macrófagos , Ratones Noqueados , Microglía , PPAR gamma , Factor de Transcripción STAT6 , Transducción de Señal , Animales , Calgranulina B/genética , Calgranulina B/metabolismo , Factor de Transcripción STAT6/metabolismo , Factor de Transcripción STAT6/deficiencia , Factor de Transcripción STAT6/genética , Microglía/metabolismo , Microglía/efectos de los fármacos , Ratones , Macrófagos/metabolismo , Macrófagos/efectos de los fármacos , Masculino , PPAR gamma/metabolismo , PPAR gamma/genética , Humanos , Accidente Cerebrovascular Isquémico/metabolismo , Accidente Cerebrovascular Isquémico/genética , Accidente Cerebrovascular Isquémico/patología , Transducción de Señal/fisiología , Transducción de Señal/efectos de los fármacos , Ratones Endogámicos C57BL , Femenino , Persona de Mediana Edad , Anciano
13.
Int J Mol Sci ; 25(16)2024 Aug 06.
Artículo en Inglés | MEDLINE | ID: mdl-39201266

RESUMEN

Obesity is a complex health condition characterized by excessive adipose tissue accumulation, leading to significant metabolic disturbances such as insulin resistance and cardiovascular diseases. Fatty acid synthase (FAS), a key enzyme in lipogenesis, has been identified as a potential therapeutic target for obesity due to its role in adipocyte differentiation and lipid accumulation. This study employed a multidisciplinary approach involving in silico and in vitro analyses to investigate the anti-adipogenic properties of maclurin, a natural phenolic compound derived from Morus alba. Using SwissDock software (ChEMBL version 23), we predicted protein interactions and demonstrated a high probability (95.6%) of maclurin targeting FAS, surpassing the interaction rates of established inhibitors like cerulenin. Docking simulations revealed maclurin's superior binding affinity to FAS, with a binding score of -7.3 kcal/mol compared to -6.7 kcal/mol for cerulenin. Subsequent in vitro assays confirmed these findings, with maclurin effectively inhibiting FAS activity in a concentration-dependent manner in 3T3-L1 adipocytes, without compromising cell viability. Furthermore, maclurin treatment resulted in significant reductions in lipid accumulation and the downregulated expression of critical adipogenic genes such as PPARγ, C/EBPα, and FAS, indicating the suppression of adipocyte differentiation. Maclurin shows potential as a novel FAS inhibitor with significant anti-adipogenic effects, offering a promising therapeutic avenue for the treatment and prevention of obesity.


Asunto(s)
Células 3T3-L1 , Adipocitos , Adipogénesis , Diferenciación Celular , Simulación del Acoplamiento Molecular , Animales , Ratones , 4-Butirolactona/análogos & derivados , Adipocitos/efectos de los fármacos , Adipocitos/metabolismo , Adipocitos/citología , Adipogénesis/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Ácido Graso Sintasas/metabolismo , Ácido Graso Sintasas/antagonistas & inhibidores , Metabolismo de los Lípidos/efectos de los fármacos , PPAR gamma/metabolismo , PPAR gamma/genética
14.
Free Radic Biol Med ; 223: 237-249, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39094710

RESUMEN

Chronic prostatitis-induced excessive inflammation and oxidative stress (OS) damage substantially affect men's quality of life. However, its treatment remains a major clinical challenge. Therefore, the identification of drugs that can decrease chronic prostatitis and oxidative stress targets is urgent and essential. CXCR4 is a classic chemokine receptor that is crucially associated with the occurrence and development of inflammation. This investigation aimed to elucidate how CXCR4 affects prostatitis regression and progression. The effect of CXCR4 on chronic prostatitis was evaluated by HE staining, immunohistochemistry, immunofluorescence, PCR, and TUNEL analyses. Furthermore, CXCR4 influence on metabolism was also evaluated by monitoring body weight, body temperature, food intake, and LC/MS. Additionally, chromatin immunoprecipitation, Western blot, and double luciferase reporter gene assays were carried out to elucidate the mechanism by which CXCR4 modulates Fads2 transcription by PPARγ. Lastly, ROS, DHE, mito-tracker, and ATP were utilized to validate the α-linolenic acid's protective effect against OS in prostate epithelial cells. It was revealed that the inhibition of CXCR4 can effectively alleviate prostatitis in mice. Furthermore, downregulating CXCR4 expression can markedly reduce the inflammatory cell infiltration in mouse prostates, decrease the elevated levels of DNA damage markers,MDA and 4-HNE, and mitigate apoptosis of prostatic epithelial cells. Moreover, treatment of CXCR4 knockdown mice with a PPARγ inhibitor revealed different degrees of changes in the above phenotypes. Mechanistically, the PPARγ protein translocates to the nucleus and serves as a transcription factor to regulate Fads2 expression, thereby altering PUFA metabolism. Additionally, in vitro experiments indicated that α-linolenic acid can effectively alleviate OS damage and RWPE-1 cell apoptosis by protecting mitochondrial function and enhancing the antioxidant capacity of prostatic epithelial cells. In conclusion, reducing the levels of CXCR4 can alleviate inflammation and OS damage in chronic prostatitis.


Asunto(s)
Ácido Graso Desaturasas , Estrés Oxidativo , PPAR gamma , Prostatitis , Receptores CXCR4 , Masculino , Animales , Receptores CXCR4/metabolismo , Receptores CXCR4/genética , Ratones , Prostatitis/metabolismo , Prostatitis/patología , Prostatitis/genética , Prostatitis/tratamiento farmacológico , PPAR gamma/metabolismo , PPAR gamma/genética , Ácido Graso Desaturasas/genética , Ácido Graso Desaturasas/metabolismo , Humanos , Modelos Animales de Enfermedad , Apoptosis , Ácidos Grasos Insaturados/metabolismo , Ácido alfa-Linolénico/farmacología , Ácido alfa-Linolénico/metabolismo , Próstata/patología , Próstata/metabolismo , Próstata/efectos de los fármacos , Ratones Endogámicos C57BL , Regulación de la Expresión Génica
15.
Nat Commun ; 15(1): 6622, 2024 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-39103342

RESUMEN

Sex steroids modulate the distribution of mammalian white adipose tissues. Moreover, WAT remodeling requires adipocyte progenitor cells. Nevertheless, the sex-dependent mechanisms regulating adipocyte progenitors remain undetermined. Here, we uncover Cxcr4 acting in a sexually dimorphic manner to affect a pool of proliferating cells leading to restriction of female fat mass. We find that deletion of Cxcr4 in Pparγ-expressing cells results in female, not male, lipodystrophy, which cannot be restored by high-fat diet consumption. Additionally, Cxcr4 deletion is associated with a loss of a pool of proliferating adipocyte progenitors. Cxcr4 loss is accompanied by the upregulation of estrogen receptor alpha in adipose-derived PPARγ-labelled cells related to estradiol hypersensitivity and stalled adipogenesis. Estrogen removal or administration of antiestrogens restores WAT accumulation and dynamics of adipose-derived cells in Cxcr4-deficient mice. These findings implicate Cxcr4 as a female adipogenic rheostat, which may inform strategies to target female adiposity.


Asunto(s)
Adipocitos , Adipogénesis , Adiposidad , PPAR gamma , Receptores CXCR4 , Células Madre , Animales , Receptores CXCR4/metabolismo , Receptores CXCR4/genética , Femenino , Masculino , Ratones , Adipocitos/metabolismo , Adipocitos/citología , Células Madre/metabolismo , Células Madre/citología , PPAR gamma/metabolismo , PPAR gamma/genética , Ratones Noqueados , Tejido Adiposo Blanco/metabolismo , Tejido Adiposo Blanco/citología , Dieta Alta en Grasa/efectos adversos , Receptor alfa de Estrógeno/metabolismo , Receptor alfa de Estrógeno/genética , Ratones Endogámicos C57BL , Estradiol/farmacología , Estradiol/metabolismo , Proliferación Celular , Factores Sexuales , Caracteres Sexuales
16.
Int Immunopharmacol ; 141: 112917, 2024 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-39137630

RESUMEN

PURPOSE: This study aimed to explore novel targets for hepatocellular carcinoma (HCC) treatment by investigating the role of fatty acid metabolism. METHODS: RNA-seq and clinical data of HCC were obtained from the Gene Expression Omnibus (GEO) and The Cancer Genome Atlas (TCGA) databases. Bioinformatic analyses were employed to identify differentially expressed genes (DEGs) related to prognosis. A signature was then constructed using the Least Absolute Shrinkage and Selection Operator (LASSO) Cox regression to classify HCC patients from the TCGA database into low-risk and high-risk groups. The predictive performance of the signature was evaluated through principal components analysis (PCA), Kaplan Meier (KM) survival analysis, receiver operating characteristics (ROC) curves, nomogram, genetic mutations, drug sensitivity analysis, immunological correlation analysis, and enrichment analysis. Single-cell maps were constructed to illustrate the distribution of core genes. Immunohistochemistry (IHC), quantitative real-time PCR (qRT-PCR), and western blot were employed to verify the expression of core genes. The function of one core gene was validated through a series of in vitro assays, including cell viability, colony formation, wound healing, trans-well migration, and invasion assays. The results were analyzed in the context of relevant signaling pathways. RESULTS: Bioinformatic analyses identified 15 FAMGs that were related to prognosis. A 4-gene signature was constructed, and patients were divided into high- and low-risk groups according to the signature. The high-risk group exhibited a poorer prognosis compared to the low-risk group in both the training (P < 0.001) and validation (P = 0.020) sets. Furthermore, the risk score was identified as an independent predictor of OS (P < 0.001, HR = 8.005). The incorporation of the risk score and clinicopathologic features into a nomogram enabled the effective prediction of patient prognosis. The model was able to effectively predict the immune microenvironment, drug sensitivity to chemotherapy, and gene mutation for each group. Single-cell maps demonstrated that FAMGs in the model were distributed in tumor cells. Enrichment analyses revealed that the cell cycle, fatty acid ß oxidation and PPAR signaling pathways were the most significant pathways. Among the four key prognostically related FAMGs, Spermine Synthase (SMS) was selected and validated as a potential oncogene affecting cell cycle, PPAR-γ signaling pathway and fatty acid ß oxidation in HCC. CONCLUSIONS: The risk characteristics based on FAMGs could serve as independent prognostic indicators for predicting HCC prognosis and could also serve as evaluation criteria for gene mutations, immunity, and chemotherapy drug therapy in HCC patients. Meanwhile, targeted fatty acid metabolism could be used to treat HCC through related signaling pathways.


Asunto(s)
Carcinoma Hepatocelular , Ácidos Grasos , Regulación Neoplásica de la Expresión Génica , Neoplasias Hepáticas , PPAR gamma , Transducción de Señal , Humanos , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patología , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patología , Carcinoma Hepatocelular/mortalidad , Ácidos Grasos/metabolismo , PPAR gamma/metabolismo , PPAR gamma/genética , Oxidación-Reducción , Línea Celular Tumoral , Progresión de la Enfermedad , Pronóstico , Masculino , Femenino , Persona de Mediana Edad
17.
J Agric Food Chem ; 72(36): 19766-19785, 2024 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-39186442

RESUMEN

Colorectal cancer (CRC) is the third-largest cancer worldwide. Lactobacillus can regulate the intestinal barrier and gut microbiota. However, the mechanisms of Lactobacillus that alleviate CRC remained unknown. This study aimed to explore the regulatory effect of Lactobacillus plantarum on CRC and its potential mechanism. CCFM8661 treatment significantly ameliorated CRC compared with phosphate-buffered solution (PBS) treatment in ApcMin/+ mice. In addition, conjugated linoleic acid (CLA) was proved to be the key metabolite for CCFM8661 in ameliorating CRC by molecular biology techniques. Peroxisome proliferator-activated receptor γ (PPAR-γ) was proved to be the key receptor in ameliorating CRC by inhibitor intervention experiments. Moreover, supplementation with CCFM8661 ameliorated CRC by producing CLA to inhibit NF-κB pathway and pro-inflammatory cytokines, up-regulate ZO-1, Claudin-1, and MUC2, and promote tumor cell apoptosis in a PPAR-γ-dependent manner. Metagenomic analysis showed that CCFM8661 treatment significantly increased Odoribacter splanchnicus, which could ameliorate CRC by repairing the intestinal barrier. Clinical results showed that intestinal CLA, butyric acid, PPAR-γ, and Lactobacillus were significantly decreased in CRC patients, and these indicators were significantly negatively correlated with CRC. CCFM8661 alleviated CRC by ameliorating the intestinal barrier through the CLA-PPAR-γ axis. These results will promote the development of dietary probiotic supplements for CRC.


Asunto(s)
Neoplasias Colorrectales , Microbioma Gastrointestinal , Mucosa Intestinal , Lactobacillus plantarum , Ácidos Linoleicos Conjugados , Ratones Endogámicos C57BL , PPAR gamma , Probióticos , Lactobacillus plantarum/metabolismo , PPAR gamma/metabolismo , PPAR gamma/genética , Animales , Ratones , Neoplasias Colorrectales/metabolismo , Humanos , Probióticos/administración & dosificación , Probióticos/farmacología , Masculino , Ácidos Linoleicos Conjugados/farmacología , Ácidos Linoleicos Conjugados/metabolismo , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiología , Femenino , FN-kappa B/metabolismo , FN-kappa B/genética , Apoptosis/efectos de los fármacos , Claudina-1/metabolismo , Claudina-1/genética , Proteína de la Zonula Occludens-1/metabolismo , Proteína de la Zonula Occludens-1/genética
18.
Int Immunopharmacol ; 140: 112729, 2024 Oct 25.
Artículo en Inglés | MEDLINE | ID: mdl-39098229

RESUMEN

ADORA3 is mainly expressed in intestinal tract, and has the potential to promote the expression of mucin 2 (MUC2), the function-related factor of goblet cells, under asthma conditions. This study aims to confirm the induction and mechanisms of ADORA3 activation on goblet cells in ulcerative colitis (UC). A significant decrease in ADORA3 expression was found in mucosal biopsies from UC patients and in the colons of colitis mice. This reduction correlated negatively with disease severity and positively with goblet cell number. ADORA3 activation mitigated dextran sulfate sodium (DSS)-induced colitis and facilitated ATOH1-mediated goblet cell differentiation in both in vivo and in vitro. Metabolomics analysis unveiled that ADORA3 activation bolstered ketogenesis, leading to elevated levels of the metabolite BHB. Subsequently, BHB heightened the activity of HDAC1/2, augmenting histone acetylation at the H3K9ac site within the promoter region of the ATOH1 gene. Furthermore, the reason for ADORA3 activation to enhance ketogenesis was attributed to controlling the competitive binding among ß-arrestin2, SHP1 and PPARγ. This results in the non-ligand-dependent activation of PPARγ, thereby promoting the transcription of HMGCS2. The exact mechanisms by which ADORA3 promoted goblet cell differentiation and alleviated UC were elucidated using MRS1191 and shHMGCS2 plasmid. Collectively, ADORA3 activation promoted goblet cell differentiation and alleviated UC by enhancing ketogenesis via the "BHB-HDAC1/2-H3K9ac" pathway.


Asunto(s)
Diferenciación Celular , Colitis Ulcerosa , Células Caliciformes , Hidroximetilglutaril-CoA Sintasa , Adulto , Animales , Femenino , Humanos , Masculino , Ratones , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Ácido Butírico/farmacología , Colitis Ulcerosa/patología , Colitis Ulcerosa/metabolismo , Colon/patología , Colon/metabolismo , Sulfato de Dextran , Células Caliciformes/patología , Células Caliciformes/metabolismo , Histona Desacetilasa 1/metabolismo , Histona Desacetilasa 1/genética , Histona Desacetilasa 2/metabolismo , Histona Desacetilasa 2/genética , Hidroximetilglutaril-CoA Sintasa/metabolismo , Hidroximetilglutaril-CoA Sintasa/genética , Ratones Endogámicos C57BL , PPAR gamma/metabolismo , PPAR gamma/genética
19.
J Clin Invest ; 134(16)2024 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-39145452

RESUMEN

T cells rewire their metabolic activities to meet the demand of immune responses, but how to coordinate the immune response by metabolic regulators in activated T cells is unknown. Here, we identified autocrine VEGF-B as a metabolic regulator to control lipid synthesis and maintain the integrity of the mitochondrial inner membrane for the survival of activated T cells. Disruption of autocrine VEGF-B signaling in T cells reduced cardiolipin mass, resulting in mitochondrial damage, with increased apoptosis and reduced memory development. The addition of cardiolipin or modulating VEGF-B signaling improved T cell mitochondrial fitness and survival. Autocrine VEGF-B signaling through GA-binding protein α (GABPα) induced sentrin/SUMO-specific protease 2 (SENP2) expression, which further de-SUMOylated PPARγ and enhanced phospholipid synthesis, leading to a cardiolipin increase in activated T cells. These data suggest that autocrine VEGF-B mediates a signal to coordinate lipid synthesis and mitochondrial fitness with T cell activation for survival and immune response. Moreover, autocrine VEGF-B signaling in T cells provides a therapeutic target against infection and tumors as well as an avenue for the treatment of autoimmune diseases.


Asunto(s)
Comunicación Autocrina , Cardiolipinas , Mitocondrias , Transducción de Señal , Linfocitos T , Factor B de Crecimiento Endotelial Vascular , Mitocondrias/metabolismo , Mitocondrias/inmunología , Animales , Ratones , Comunicación Autocrina/inmunología , Linfocitos T/inmunología , Linfocitos T/metabolismo , Transducción de Señal/inmunología , Cardiolipinas/inmunología , Cardiolipinas/metabolismo , Factor B de Crecimiento Endotelial Vascular/genética , Factor B de Crecimiento Endotelial Vascular/metabolismo , Factor B de Crecimiento Endotelial Vascular/inmunología , Activación de Linfocitos , PPAR gamma/metabolismo , PPAR gamma/inmunología , PPAR gamma/genética , Humanos
20.
FASEB J ; 38(16): e70014, 2024 Aug 31.
Artículo en Inglés | MEDLINE | ID: mdl-39183544

RESUMEN

End-ischemic normothermic mechanical perfusion (NMP) could provide a curative treatment to reduce cholestatic liver injury from donation after circulatory death (DCD) in donors. However, the underlying mechanism remains elusive. Our previous study demonstrated that air-ventilated NMP could improve functional recovery of DCD in a preclinical NMP rat model. Here, metabolomics analysis revealed that air-ventilated NMP alleviated DCD- and cold preservation-induced cholestatic liver injury, as shown by the elevated release of alanine aminotransferase (ALT), aspartate aminotransferase (AST), bilirubin, and γ-glutamyl transferase (GGT) in the perfusate (p < .05) and the reduction in the levels of bile acid metabolites, including ω-muricholic acid, glycohyodeoxycholic acid, glycocholic acid, and glycochenodeoxycholate (GCDC) in the perfused livers (p < .05). In addition, the expression of the key bile acid metabolism enzyme UDP-glucuronosyltransferase 1A1 (UGT1A1), which is predominantly expressed in hepatocytes, was substantially elevated in the DCD rat liver, followed by air-ventilated NMP (p < .05), and in vitro, this increase was induced by decreased GCDC and hypoxia-reoxygenation in the hepatic cells HepG2 and L02 (p < .05). Knockdown of UGT1A1 in hepatic cells by siRNA aggravated hepatic injury caused by GCDC and hypoxia-reoxygenation, as indicated by the ALT and AST levels in the supernatant. Mechanistically, UGT1A1 is transcriptionally regulated by peroxisome proliferator-activator receptor-γ (PPAR-γ) under hypoxia-physoxia. Taken together, our data revealed that air-ventilated NMP could alleviate DCD- and cold preservation-induced cholestatic liver injury through PPAR-γ/UGT1A1 axis. Based on the results from this study, air-ventilated NMP confers a promising approach for predicting and alleviating cholestatic liver injury through PPAR-γ/UGT1A1 axis.


Asunto(s)
PPAR gamma , Animales , Ratas , PPAR gamma/metabolismo , PPAR gamma/genética , Masculino , Humanos , Glucuronosiltransferasa/metabolismo , Glucuronosiltransferasa/genética , Hígado/metabolismo , Hígado/patología , Colestasis/metabolismo , Perfusión , Ratas Sprague-Dawley , Preservación de Órganos/métodos , Trasplante de Hígado
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA