Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 22.201
Filtrer
1.
Mol Med Rep ; 30(4)2024 Oct.
Article de Anglais | MEDLINE | ID: mdl-39092554

RÉSUMÉ

Alcohol­related liver disease (ALD) is a major health concern worldwide. In recent years, there has been growing interest in natural products and functional foods for preventing and treating ALD due to their potential antioxidant and hepatoprotective properties. Rosa roxburghii Tratt, known for its rich content of bioactive compounds, has demonstrated promising health benefits, including anti­inflammatory and antioxidant effects. Fermentation has been utilized as a strategy to enhance the bioavailability and efficacy of natural products. In the present study, using a mixture of Rosa roxburghii Tratt juice, lotus leaf extract and grape seed proanthocyanidins fermented by Lactobacillus plantarum HH­LP56, a novel fermented Rosa roxburghii Tratt (FRRT) juice was discovered that can prevent and regulate ethanol­induced liver cell damage. Following fermentation, the pH was significantly decreased, and the content of VC and superoxide dismutase (SOD) were significantly increased, along with a noticeable enhancement in hydroxyl and 2,2­diphenyl­1­picrylhydrazyl free radical scavenging abilities. Alpha Mouse liver 12 cells were exposed to ethanol for 24 h to establish an in vitro liver cell injury model. The present study evaluated the effects of FRRT on cell damage, lipid accumulation and oxidative stress markers. The results revealed that FRRT pretreatment (cells were pre­treated with 2.5 and 5 mg/ml FRRT for 2 h) significantly reduced lipid accumulation and oxidative stress in liver cells. Mechanistically, FRRT regulated lipid metabolism by influencing key genes and proteins, such as AMP­activated protein kinase, sterol regulatory element binding transcription factor 1 and Stearyl­CoA desaturase­1. Furthermore, FRRT enhanced antioxidant activity by increasing SOD activity, glutathione and catalase levels, while reducing reactive oxygen species and malondialdehyde levels. It also reversed the expression changes of ethanol­induced oxidative stress­related genes and proteins. In conclusion, a novel functional food ingredient may have been discovered with extensive potential applications. These findings indicated that FRRT has antioxidant properties and potential therapeutic benefits in addressing ethanol­induced liver cell damage through its effects on liver lipid metabolism and oxidative stress.


Sujet(s)
AMP-Activated Protein Kinases , Éthanol , Fermentation , Hépatocytes , Facteur-2 apparenté à NF-E2 , Extraits de plantes , Rosa , Transduction du signal , Animaux , Souris , Rosa/composition chimique , Transduction du signal/effets des médicaments et des substances chimiques , Hépatocytes/effets des médicaments et des substances chimiques , Hépatocytes/métabolisme , Facteur-2 apparenté à NF-E2/métabolisme , Extraits de plantes/pharmacologie , Extraits de plantes/composition chimique , AMP-Activated Protein Kinases/métabolisme , Stress oxydatif/effets des médicaments et des substances chimiques , Lignée cellulaire , Antioxydants/pharmacologie , Jus de fruits et de légumes , Agents protecteurs/pharmacologie
2.
Nat Commun ; 15(1): 6602, 2024 Aug 04.
Article de Anglais | MEDLINE | ID: mdl-39097583

RÉSUMÉ

Broadening gene therapy applications requires manufacturable vectors that efficiently transduce target cells in humans and preclinical models. Conventional selections of adeno-associated virus (AAV) capsid libraries are inefficient at searching the vast sequence space for the small fraction of vectors possessing multiple traits essential for clinical translation. Here, we present Fit4Function, a generalizable machine learning (ML) approach for systematically engineering multi-trait AAV capsids. By leveraging a capsid library that uniformly samples the manufacturable sequence space, reproducible screening data are generated to train accurate sequence-to-function models. Combining six models, we designed a multi-trait (liver-targeted, manufacturable) capsid library and validated 88% of library variants on all six predetermined criteria. Furthermore, the models, trained only on mouse in vivo and human in vitro Fit4Function data, accurately predicted AAV capsid variant biodistribution in macaque. Top candidates exhibited production yields comparable to AAV9, efficient murine liver transduction, up to 1000-fold greater human hepatocyte transduction, and increased enrichment relative to AAV9 in a screen for liver transduction in macaques. The Fit4Function strategy ultimately makes it possible to predict cross-species traits of peptide-modified AAV capsids and is a critical step toward assembling an ML atlas that predicts AAV capsid performance across dozens of traits.


Sujet(s)
Protéines de capside , Capside , Dependovirus , Vecteurs génétiques , Foie , Dependovirus/génétique , Animaux , Humains , Souris , Vecteurs génétiques/génétique , Capside/métabolisme , Protéines de capside/génétique , Protéines de capside/métabolisme , Foie/métabolisme , Transduction génétique , Techniques de transfert de gènes , Apprentissage machine , Thérapie génétique/méthodes , Macaca , Hépatocytes/métabolisme , Cellules HEK293 , Génie génétique/méthodes
3.
Biol Pharm Bull ; 47(8): 1422-1428, 2024.
Article de Anglais | MEDLINE | ID: mdl-39111864

RÉSUMÉ

CYP2D6 variants contain various single nucleotide polymorphisms as well as differing levels of metabolic activity. Among these, one of the less active variants CYP2D6*10 (100C > T) is the most prevalent mutation in East Asians, including Japanese. This mutation leads to an amino acid substitution from proline to serine, which reduces the stability of CYP2D6 and consequently decreases its metabolic activity. In this study, we used a genome editing technology called the Precise Integration into Target Chromosome (PITCh) system to stably express six drug-metabolizing enzymes (CYP3A4, POR, uridine diphosphate glucuronosyltransferase 1A1 (UGT1A1), CYP1A2, CYP2C19, CYP2C9, and CYP2D6*10) in HepG2 (CYP2D6*10 KI-HepG2) cells to examine the effect of CYP2D6*10 on drug metabolism prediction. The protein expression levels of CYP2D6 in CYP2D6*10 KI-HepG2 cells were reduced relative to those in the CYP3A4-POR-UGT1A1-CYP1A2-CYP2C19-CYP2C9-CYP2D6 knock-in-HepG2 (CYPs-UGT1A1 KI-HepG2) cells. Consistent with the CYP2D6 protein expression results, CYP2D6 metabolic activity in CYP2D6*10 KI-HepG2 cells was reduced relative to CYPs-UGT1A1 KI-HepG2 cells. We successfully generated CYP2D6*10 KI-HepG2 cells with highly expressed, functional CYP2D6*10, as well as CYP1A2, 2C9, 2C19 and 3A4. CYP2D6*10 KI-HepG2 cells could be an invaluable model for hepatic metabolism and hepatotoxicity studies in East Asians, including Japanese.


Sujet(s)
Cytochrome P-450 CYP2D6 , Hépatocytes , Humains , Cytochrome P-450 CYP2D6/génétique , Cytochrome P-450 CYP2D6/métabolisme , Cellules HepG2 , Hépatocytes/métabolisme , Édition de gène/méthodes , Glucuronosyltransferase/génétique , Glucuronosyltransferase/métabolisme , Polymorphisme de nucléotide simple , Modèles biologiques
4.
Sci Rep ; 14(1): 18189, 2024 08 06.
Article de Anglais | MEDLINE | ID: mdl-39107343

RÉSUMÉ

Desmosomes are intercellular adhesion complexes providing mechanical coupling and tissue integrity. Previously, a correlation of desmosomal molecule expression with invasion and metastasis formation in several tumor entities was described together with a relevance for circulating tumor cell cluster formation. Here, we investigated the contribution of the desmosomal core adhesion molecule desmoglein-2 (DSG2) to the initial steps of liver metastasis formation by pancreatic cancer cells using a novel ex vivo liver perfusion mouse model. We applied the pancreatic ductal adenocarcinoma cell line AsPC-1 with and without a knockout (KO) of DSG2 and generated mouse lines with a hepatocyte-specific KO of the known interacting partners of DSG2 (DSG2 and desmocollin-2). Liver perfusion with DSG2 KO AsPC-1 cells led to smaller circulating cell clusters and a reduced number of cells adhering to murine livers compared to control cells. While this was independent of the expression levels of desmosomal adhesion molecules in hepatocytes, we show that increased cluster size of cancer cells, which correlates with stronger cell-cell adhesion and expression of desmosomal molecules, is a major factor contributing to the early phase of metastatic spreading. In conclusion, impaired desmosomal adhesion results in reduced circulating cell cluster size, which is relevant for seeding and attachment of metastatic cells to the liver.


Sujet(s)
Adhérence cellulaire , Desmogléine-2 , Desmosomes , Tumeurs du foie , Tumeurs du pancréas , Animaux , Desmosomes/métabolisme , Tumeurs du pancréas/anatomopathologie , Tumeurs du pancréas/métabolisme , Tumeurs du pancréas/génétique , Souris , Tumeurs du foie/secondaire , Tumeurs du foie/anatomopathologie , Tumeurs du foie/métabolisme , Lignée cellulaire tumorale , Humains , Desmogléine-2/métabolisme , Desmogléine-2/génétique , Carcinome du canal pancréatique/anatomopathologie , Carcinome du canal pancréatique/métabolisme , Carcinome du canal pancréatique/génétique , Hépatocytes/métabolisme , Hépatocytes/anatomopathologie , Souris knockout , Cellules tumorales circulantes/métabolisme , Cellules tumorales circulantes/anatomopathologie
5.
FASEB J ; 38(15): e23880, 2024 Aug 15.
Article de Anglais | MEDLINE | ID: mdl-39132919

RÉSUMÉ

Long noncoding RNAs (lncRNAs) are strongly associated with glucose homeostasis, but their roles remain largely unknown. In this study, the potential role of lncRNA-Snhg3 in glucose metabolism was evaluated both in vitro and in vivo. Here, we found a positive relationship between Snhg3 and hepatic glycogenesis. Glucose tolerance improved in hepatocyte-specific Snhg3 knock-in (Snhg3-HKI) mice, while it worsened in hepatocyte-specific Snhg3 knockout (Snhg3-HKO) mice. Furthermore, hepatic glycogenesis had shown remarkable increase in Snhg3-HKI mice and reduction in Snhg3-HKO mice, respectively. Mechanistically, Snhg3 increased mRNA and protein expression levels of PPP1R3B through inducing chromatin remodeling and promoting the phosphorylation of protein kinase B. Collectively, these results suggested that lncRNA-Snhg3 plays a critical role in hepatic glycogenesis.


Sujet(s)
Foie , ARN long non codant , Animaux , ARN long non codant/génétique , ARN long non codant/métabolisme , Souris , Foie/métabolisme , Souris knockout , Glucose/métabolisme , Mâle , Hépatocytes/métabolisme , Souris de lignée C57BL , Glycogène hépatique/métabolisme
6.
Cells ; 13(15)2024 Aug 04.
Article de Anglais | MEDLINE | ID: mdl-39120332

RÉSUMÉ

Hepatocyte organoids (HOs) have superior hepatic functions to cholangiocyte-derived organoids but suffer from shorter lifespans. To counteract this, we co-cultured pig HOs with adipose-derived mesenchymal stem cells (A-MSCs) and performed transcriptome analysis. The results revealed that A-MSCs enhanced the collagen synthesis pathways, which are crucial for maintaining the three-dimensional structure and extracellular matrix synthesis of the organoids. A-MSCs also increased the expression of liver progenitor cell markers (KRT7, SPP1, LGR5+, and TERT). To explore HOs as a liver disease model, we exposed them to alcohol to create an alcoholic liver injury (ALI) model. The co-culture of HOs with A-MSCs inhibited the apoptosis of hepatocytes and reduced lipid accumulation of HOs. Furthermore, varying ethanol concentrations (0-400 mM) and single-versus-daily exposure to HOs showed that daily exposure significantly increased the level of PLIN2, a lipid storage marker, while decreasing CYP2E1 and increasing CYP1A2 levels, suggesting that CYP1A2 may play a critical role in alcohol detoxification during short-term exposure. Moreover, daily alcohol exposure led to excessive lipid accumulation and nuclear fragmentation in HOs cultured alone. These findings indicate that HOs mimic in vivo liver regeneration, establishing them as a valuable model for studying liver diseases, such as ALI.


Sujet(s)
Apoptose , Techniques de coculture , Hépatocytes , Régénération hépatique , Cellules souches mésenchymateuses , Organoïdes , Cellules souches mésenchymateuses/métabolisme , Animaux , Hépatocytes/métabolisme , Hépatocytes/anatomopathologie , Organoïdes/métabolisme , Apoptose/effets des médicaments et des substances chimiques , Suidae , Tissu adipeux/cytologie , Tissu adipeux/métabolisme , Éthanol , Stéatose hépatique/anatomopathologie , Stéatose hépatique/métabolisme , Maladies alcooliques du foie/anatomopathologie , Maladies alcooliques du foie/métabolisme , Métabolisme lipidique
7.
Nutrients ; 16(15)2024 Jul 23.
Article de Anglais | MEDLINE | ID: mdl-39125267

RÉSUMÉ

The VLCKD is a diet recognized to promote rapid fat mobilization and reduce inflammation, hepatic steatosis, and liver fibrosis. Extracellular vesicles (EVs) mediate cell-to-cell communication. The aim of the study is to investigate the role of circulating EVs in cell proliferation, ketone bodies, and ROS production in patients on an 8-week VLCKD regimen. Participants were classified as responders (R) or non-responders (NR) to VLCKD treatment based on their fibroscan results. In vitro experiments with the hepatic cell lines HEPA-RG (normal hepatocytes) and LX-2 (stellate cells) were conducted to investigate the effects of circulating EVs on cell viability, ROS production, and ketone body presence. The findings reveal a notable reduction in cell viability in both cell lines when treated with exosomes (EXOs). In contrast, treatment with microvesicles (MVs) did not appear to affect cell viability, which remained unchanged. Additionally, the levels of ketone bodies measured in urine were not consistently correlated with the reduction of fibrosis in responders (R). Similarly, an increase in ketone bodies was observed in non-responders (NR), which was also not aligned with the expected reduction in fibrosis. This inconsistency stands in stark contrast to the levels of Reactive Oxygen Species (ROS), which exhibited a clear and consistent pattern in accordance with the dietary intervention. Finally, in this preliminary study, ROS has been identified as a potential diet adherence marker for VLCKD patients; the ROS levels reliably follow the progression of the fibrosis response, providing a more accurate reflection of the therapeutic effects.


Sujet(s)
Survie cellulaire , Régime cétogène , Vésicules extracellulaires , Hépatocytes , Corps cétoniques , Espèces réactives de l'oxygène , Humains , Espèces réactives de l'oxygène/métabolisme , Régime cétogène/méthodes , Vésicules extracellulaires/métabolisme , Mâle , Femelle , Corps cétoniques/métabolisme , Hépatocytes/métabolisme , Adulte , Adulte d'âge moyen , Lignée cellulaire , Cirrhose du foie/métabolisme , Cirrhose du foie/diétothérapie , Exosomes/métabolisme
8.
Int J Mol Sci ; 25(15)2024 Jul 24.
Article de Anglais | MEDLINE | ID: mdl-39125617

RÉSUMÉ

Progression of metabolic dysfunction-associated steatites liver disease (MASLD) to steatohepatitis (MASH) is driven by stress-inducing lipids that promote liver inflammation and fibrosis, and MASH can lead to cirrhosis and hepatocellular carcinoma. Previously, we showed coordinated defenses regulated by transcription factors, nuclear factor erythroid 2-related factor-1 (Nrf1) and -2 (Nrf2), protect against hepatic lipid stress. Here, we investigated protective effects of hepatocyte Nrf1 and Nrf2 against MASH-linked liver fibrosis and tumorigenesis. Male and female mice with flox alleles for genes encoding Nrf1 (Nfe2l1), Nrf2 (Nfe2l2), or both were fed a MASH-inducing diet enriched with high fat, fructose, and cholesterol (HFFC) or a control diet for 24-52 weeks. During this period, hepatocyte Nrf1, Nrf2, or combined deficiency for ~7 days, ~7 weeks, and ~35 weeks was induced by administering mice hepatocyte-targeting adeno-associated virus (AAV) expressing Cre recombinase. The effects on MASH, markers of liver fibrosis and proliferation, and liver tumorigenesis were compared to control mice receiving AAV-expressing green fluorescent protein. Also, to assess the impact of Nrf1 and Nrf2 induction on liver fibrosis, HFFC diet-fed C57bl/6J mice received weekly injections of carbon tetrachloride, and from week 16 to 24, mice were treated with the Nrf2-activating drug bardoxolone, hepatocyte overexpression of human NRF1 (hNRF1), or both, and these groups were compared to control. Compared to the control diet, 24-week feeding with the HFFC diet increased bodyweight as well as liver weight, steatosis, and inflammation. It also increased hepatocyte proliferation and a marker of liver damage, p62. Hepatocyte Nrf1 and combined deficiency increased liver steatosis in control diet-fed but not HFFC diet-fed mice, and increased liver inflammation under both diet conditions. Hepatocyte Nrf1 deficiency also increased hepatocyte proliferation, whereas combined deficiency did not, and this also occurred for p62 level in control diet-fed conditions. In 52-week HFFC diet-fed mice, 35 weeks of hepatocyte Nrf1 deficiency, but not combined deficiency, resulted in more liver tumors in male mice, but not in female mice. In contrast, hepatocyte Nrf2 deficiency had no effect on any of these parameters. However, in the 15-week CCL4-exposed and 24-week HFFC diet-fed mice, Nrf2 induction with bardoxolone reduced liver steatosis, inflammation, fibrosis, and proliferation. Induction of hepatic Nrf1 activity with hNRF1 enhanced the effect of bardoxolone on steatosis and may have stimulated liver progenitor cells. Physiologic Nrf1 delays MASLD progression, Nrf2 induction alleviates MASH, and combined enhancement synergistically protects against steatosis and may facilitate liver repair.


Sujet(s)
Hépatocytes , Facteur-2 apparenté à NF-E2 , Animaux , Facteur-2 apparenté à NF-E2/métabolisme , Facteur-2 apparenté à NF-E2/génétique , Souris , Hépatocytes/métabolisme , Mâle , Femelle , Évolution de la maladie , Souris de lignée C57BL , Stéatose hépatique/métabolisme , Stéatose hépatique/anatomopathologie , Stéatose hépatique/génétique , Cirrhose du foie/métabolisme , Cirrhose du foie/anatomopathologie , Cirrhose du foie/génétique , Tumeurs du foie/métabolisme , Tumeurs du foie/génétique , Tumeurs du foie/anatomopathologie , Facteur-1 apparenté à NF-E2/métabolisme , Facteur-1 apparenté à NF-E2/génétique , Facteur nucléaire-1 respiratoire/métabolisme , Facteur nucléaire-1 respiratoire/génétique , Alimentation riche en graisse/effets indésirables , Foie/métabolisme , Foie/anatomopathologie , Humains
9.
Int J Mol Sci ; 25(15)2024 Jul 26.
Article de Anglais | MEDLINE | ID: mdl-39125712

RÉSUMÉ

Liver lipid metabolism disruption significantly contributes to excessive fat buildup in waterfowl. Research suggests that the supplementation of Threonine (Thr) in the diet can improve liver lipid metabolism disorder, while Thr deficiency can lead to such metabolic disorders in the liver. The mechanisms through which Thr regulates lipid metabolism remain unclear. STAT3 (signal transducer and activator of transcription 3), a crucial transcription factor in the JAK-STAT (Janus kinase-signal transducer and activator of transcription) pathway, participates in various biological processes, including lipid and energy metabolism. This research investigates the potential involvement of STAT3 in the increased lipid storage seen in primary duck hepatocytes as a result of a lack of Thr. Using small interfering RNA and Stattic, a specific STAT3 phosphorylation inhibitor, we explored the impact of STAT3 expression patterns on Thr-regulated lipid synthesis metabolism in hepatocytes. Through transcriptome sequencing, we uncovered pathways related to lipid synthesis and metabolism jointly regulated by Thr and STAT3. The results showed that Thr deficiency increases lipid deposition in primary duck hepatocytes (p < 0.01). The decrease in protein and phosphorylation levels of STAT3 directly caused this deposition (p < 0.01). Transcriptomic analysis revealed that Thr deficiency and STAT3 knockdown jointly altered the mRNA expression levels of pathways related to long-chain fatty acid synthesis and energy metabolism (p < 0.05). Thr deficiency, through mediating STAT3 inactivation, upregulated ELOVL7, PPARG, MMP1, MMP13, and TIMP4 mRNA levels, and downregulated PTGS2 mRNA levels (p < 0.01). In summary, these results suggest that Thr deficiency promotes lipid synthesis, reduces lipid breakdown, and leads to lipid metabolism disorders and triglyceride deposition by downregulating STAT3 activity in primary duck hepatocytes.


Sujet(s)
Canards , Hépatocytes , Facteur de transcription STAT-3 , Thréonine , Triglycéride , Animaux , Facteur de transcription STAT-3/métabolisme , Hépatocytes/métabolisme , Phosphorylation , Thréonine/métabolisme , Triglycéride/métabolisme , Métabolisme lipidique , Cellules cultivées
10.
Elife ; 122024 Aug 13.
Article de Anglais | MEDLINE | ID: mdl-39137024

RÉSUMÉ

Hepatic factors secreted by the liver promote homeostasis and are pivotal for maintaining the liver-gut axis. Bile acid metabolism is one such example wherein, bile acid synthesis occurs in the liver and its biotransformation happens in the intestine. Dysfunctional interactions between the liver and the intestine stimulate varied pathological outcomes through its bidirectional portal communication. Indeed, aberrant bile acid metabolism has been reported in inflammatory bowel disease (IBD). However, the molecular mechanisms underlying these crosstalks that perpetuate intestinal permeability and inflammation remain obscure. Here, we identify a novel hepatic gene program regulated by Rela and Stat3 that accentuates the inflammation in an acute experimental colitis model. Hepatocyte-specific ablation of Rela and Stat3 reduces the levels of primary bile acids in both the liver and the gut and shows a restricted colitogenic phenotype. On supplementation of chenodeoxycholic acid (CDCA), knock-out mice exhibit enhanced colitis-induced alterations. This study provides persuasive evidence for the development of multi-organ strategies for treating IBD and identifies a hepatocyte-specific Rela-Stat3 network as a promising therapeutic target.


Sujet(s)
Acides et sels biliaires , Colite , Modèles animaux de maladie humaine , Hépatocytes , Souris knockout , Facteur de transcription STAT-3 , Facteur de transcription RelA , Animaux , Facteur de transcription STAT-3/métabolisme , Facteur de transcription STAT-3/génétique , Colite/induit chimiquement , Colite/métabolisme , Colite/génétique , Colite/anatomopathologie , Hépatocytes/métabolisme , Facteur de transcription RelA/métabolisme , Facteur de transcription RelA/génétique , Souris , Acides et sels biliaires/métabolisme , Régulation de l'expression des gènes , Foie/métabolisme , Foie/anatomopathologie , Souris de lignée C57BL
11.
Front Immunol ; 15: 1414594, 2024.
Article de Anglais | MEDLINE | ID: mdl-39091506

RÉSUMÉ

Hepatitis B Virus (HBV) is a stealthy and insidious pathogen capable of inducing chronic necro-inflammatory liver disease and hepatocellular carcinoma (HCC), resulting in over one million deaths worldwide per year. The traditional understanding of Chronic Hepatitis B (CHB) progression has focused on the complex interplay among ongoing virus replication, aberrant immune responses, and liver pathogenesis. However, the dynamic progression and crucial factors involved in the transition from HBV infection to immune activation and intrahepatic inflammation remain elusive. Recent insights have illuminated HBV's exploitation of the sodium taurocholate co-transporting polypeptide (NTCP) and manipulation of the cholesterol transport system shared between macrophages and hepatocytes for viral entry. These discoveries deepen our understanding of HBV as a virus that hijacks hepatocyte metabolism. Moreover, hepatic niche macrophages exhibit significant phenotypic and functional diversity, zonal characteristics, and play essential roles, either in maintaining liver homeostasis or contributing to the pathogenesis of chronic liver diseases. Therefore, we underscore recent revelations concerning the importance of hepatic niche macrophages in the context of viral hepatitis. This review particularly emphasizes the significant role of HBV-induced metabolic changes in hepatic macrophages as a key factor in the transition from viral infection to immune activation, ultimately culminating in liver inflammation. These metabolic alterations in hepatic macrophages offer promising targets for therapeutic interventions and serve as valuable early warning indicators, shedding light on the disease progression.


Sujet(s)
Virus de l'hépatite B , Hépatite B chronique , Foie , Macrophages , Humains , Virus de l'hépatite B/immunologie , Virus de l'hépatite B/physiologie , Macrophages/immunologie , Macrophages/métabolisme , Macrophages/virologie , Animaux , Foie/immunologie , Foie/virologie , Foie/métabolisme , Foie/anatomopathologie , Hépatite B chronique/immunologie , Hépatite B chronique/métabolisme , Hépatite B chronique/virologie , Inflammation/immunologie , Inflammation/métabolisme , Hépatocytes/métabolisme , Hépatocytes/immunologie , Hépatocytes/virologie
12.
BMC Endocr Disord ; 24(1): 135, 2024 Aug 01.
Article de Anglais | MEDLINE | ID: mdl-39090659

RÉSUMÉ

BACKGROUND: Prevalence of metabolic dysfunction-associated steatotic liver disease (MASLD) is higher in men than in women. Hormonal and genetic causes may account for the sex differences in MASLD. Current human in vitro liver models do not sufficiently take the influence of biological sex and sex hormones into consideration. METHODS: Primary human hepatocytes (PHHs) were isolated from liver specimen of female and male donors and cultured with sex hormones (17ß-estradiol, testosterone and progesterone) for up to 72 h. mRNA expression levels of 8 hepatic lipid metabolism genes were analyzed by RT-qPCR. Sex hormones and their metabolites were determined in cell culture supernatants by LC-MS analyses. RESULTS: A sex-specific expression was observed for LDLR (low density lipoprotein receptor) with higher mRNA levels in male than female PHHs. All three sex hormones were metabolized by PHHs and the effects of hormones on gene expression levels varied depending on hepatocyte sex. Only in female PHHs, 17ß-estradiol treatment affected expression levels of PPARA (peroxisome proliferator-activated receptor alpha), LIPC (hepatic lipase) and APOL2 (apolipoprotein L2). Further changes in mRNA levels of female PHHs were observed for ABCA1 (ATP-binding cassette, sub-family A, member 1) after testosterone and for ABCA1, APOA5 (apolipoprotein A-V) and PPARA after progesterone treatment. Only the male PHHs showed changing mRNA levels for LDLR after 17ß-estradiol and for APOA5 after testosterone treatment. CONCLUSIONS: Male and female PHHs showed differences in their expression levels of hepatic lipid metabolism genes and their responsiveness towards sex hormones. Thus, cellular sex should be considered, especially when investigating the pathophysiological mechanisms of MASLD.


Sujet(s)
Hormones sexuelles stéroïdiennes , Hépatocytes , Métabolisme lipidique , Humains , Mâle , Femelle , Hépatocytes/métabolisme , Hépatocytes/effets des médicaments et des substances chimiques , Métabolisme lipidique/génétique , Métabolisme lipidique/effets des médicaments et des substances chimiques , Hormones sexuelles stéroïdiennes/pharmacologie , Hormones sexuelles stéroïdiennes/métabolisme , Cellules cultivées , Adulte d'âge moyen , Testostérone/pharmacologie , Testostérone/métabolisme , Oestradiol/pharmacologie , Adulte , Progestérone/pharmacologie , Progestérone/métabolisme , Facteurs sexuels
13.
Signal Transduct Target Ther ; 9(1): 214, 2024 Aug 08.
Article de Anglais | MEDLINE | ID: mdl-39117631

RÉSUMÉ

Nonalcoholic fatty liver disease (NAFLD) is a serious threat to public health, but its underlying mechanism remains poorly understood. In screening important genes using Gene Importance Calculator (GIC) we developed previously, ribosomal modification protein rimK-like family member A (RIMKLA) was predicted as one essential gene but its functions remained largely unknown. The current study determined the roles of RIMKLA in regulating glucose and lipid metabolism. RIMKLA expression was reduced in livers of human and mouse with NAFLD. Hepatic RIMKLA overexpression ameliorated steatosis and hyperglycemia in obese mice. Hepatocyte-specific RIMKLA knockout aggravated high-fat diet (HFD)-induced dysregulated glucose/lipid metabolism in mice. Mechanistically, RIMKLA is a new protein kinase that phosphorylates betaine-homocysteine S-methyltransferase 1 (BHMT1) at threonine 45 (Thr45) site. Upon phosphorylation at Thr45 and activation, BHMT1 eliminated homocysteine (Hcy) to inhibit the activity of transcription factor activator protein 1 (AP1) and its induction on fatty acid synthase (FASn) and cluster of differentiation 36 (CD36) gene transcriptions, concurrently repressing lipid synthesis and uptake in hepatocytes. Thr45 to alanine (T45A) mutation inactivated BHMT1 to abolish RIMKLA's repression on Hcy level, AP1 activity, FASn/CD36 expressions, and lipid deposition. BHMT1 overexpression rescued the dysregulated lipid metabolism in RIMKLA-deficient hepatocytes. In summary, RIMKLA is a novel protein kinase that phosphorylates BHMT1 at Thr45 to repress lipid synthesis and uptake. Under obese condition, inhibition of RIMKLA impairs BHMT1 activity to promote hepatic lipid deposition.


Sujet(s)
Betaine-homocysteine S-methyltransferase , Métabolisme lipidique , Stéatose hépatique non alcoolique , Animaux , Souris , Humains , Betaine-homocysteine S-methyltransferase/génétique , Betaine-homocysteine S-methyltransferase/métabolisme , Stéatose hépatique non alcoolique/génétique , Stéatose hépatique non alcoolique/métabolisme , Métabolisme lipidique/génétique , Alimentation riche en graisse/effets indésirables , Hépatocytes/métabolisme , Mâle , Souris knockout , Phosphorylation/génétique
14.
Sci Transl Med ; 16(760): eadl0715, 2024 Aug 14.
Article de Anglais | MEDLINE | ID: mdl-39141698

RÉSUMÉ

Extracellular acyl-coenzyme A binding protein [ACBP encoded by diazepam binding inhibitor (DBI)] is a phylogenetically ancient appetite stimulator that is secreted in a nonconventional, autophagy-dependent fashion. Here, we show that low ACBP/DBI plasma concentrations are associated with poor prognosis in patients with anorexia nervosa, a frequent and often intractable eating disorder. In mice, anorexia induced by chronic restraint stress (CRS) is accompanied by a reduction in circulating ACBP/DBI concentrations. We engineered a chemical-genetic system for the secretion of ACBP/DBI through a biotin-activatable, autophagy-independent pathway. In transgenic mice expressing this system in hepatocytes, biotin-induced elevations in plasma ACBP/DBI concentrations prevented anorexia induced by CRS or chemotherapeutic agents including cisplatin, doxorubicin, and paclitaxel. ACBP/DBI reversed the CRS or cisplatin-induced increase in plasma lipocalin-2 concentrations and the hypothalamic activation of anorexigenic melanocortin 4 receptors, for which lipocalin-2 is an agonist. Daily intravenous injections of recombinant ACBP/DBI protein or subcutaneous implantation of osmotic pumps releasing recombinant ACBP/DBI mimicked the orexigenic effects of the chemical-genetic system. In conclusion, the supplementation of extracellular and peripheral ACBP/DBI might constitute a viable strategy for treating anorexia.


Sujet(s)
Anorexie , Inhibiteur de la liaison au diazépam , Animaux , Inhibiteur de la liaison au diazépam/métabolisme , Anorexie/traitement médicamenteux , Anorexie/métabolisme , Humains , Souris transgéniques , Souris , Anorexie mentale/métabolisme , Anorexie mentale/traitement médicamenteux , Lipocaline-2/métabolisme , Lipocaline-2/sang , Hypothalamus/métabolisme , Mâle , Femelle , Souris de lignée C57BL , Contention physique , Hépatocytes/métabolisme , Hépatocytes/effets des médicaments et des substances chimiques
15.
Commun Biol ; 7(1): 991, 2024 Aug 14.
Article de Anglais | MEDLINE | ID: mdl-39143151

RÉSUMÉ

Liver fibrosis progressing to cirrhosis is a major risk factor for liver cancer, impacting surgical treatment and survival. Our study focuses on the role of extracellular nicotinamide adenine dinucleotide (eNAD+) in liver fibrosis, analyzing liver disease patients undergoing surgery. Additionally, we explore NAD+'s therapeutic potential in a mouse model of extended liver resection and in vitro using 3D hepatocyte spheroids. eNAD+ correlated with aspartate transaminase (AST) and bilirubin after liver resection (AST: r = 0.2828, p = 0.0087; Bilirubin: r = 0.2584, p = 0.0176). Concordantly, post-hepatectomy liver failure (PHLF) was associated with higher eNAD+ peaks (n = 10; p = 0.0063). Post-operative eNAD+ levels decreased significantly (p < 0.05), but in advanced stages of liver fibrosis or cirrhosis, this decline not only diminished but actually showed a trend towards an increase. The expression of NAD+ biosynthesis rate-limiting enzymes, nicotinamide phosphoribosyltransferase (NAMPT) and nicotinamide mononucleotide adenylyltransferase 3 (NMNAT3), were upregulated significantly in the liver tissue of patients with higher liver fibrosis stages (p < 0.0001). Finally, the administration of NAD+ in a 3D hepatocyte spheroid model rescued hepatocytes from TNFalpha-induced cell death and improved viability (p < 0.0001). In a mouse model of extended liver resection, NAD+ treatment significantly improved survival (p = 0.0158) and liver regeneration (p = 0.0186). Our findings reveal that eNAD+ was upregulated in PHLF, and rate-limiting enzymes of NAD+ biosynthesis demonstrated higher expressions under liver fibrosis. Further, eNAD+ administration improved survival after extended liver resection in mice and enhanced hepatocyte viability in vitro. These insights may offer a potential target for future therapies.


Sujet(s)
Hépatectomie , Défaillance hépatique , NAD , NAD/métabolisme , Animaux , Humains , Souris , Défaillance hépatique/étiologie , Défaillance hépatique/métabolisme , Défaillance hépatique/anatomopathologie , Défaillance hépatique/chirurgie , Mâle , Hépatocytes/métabolisme , Adulte d'âge moyen , Femelle , Souris de lignée C57BL , Cirrhose du foie/métabolisme , Cirrhose du foie/chirurgie , Modèles animaux de maladie humaine , Sujet âgé
16.
Front Endocrinol (Lausanne) ; 15: 1404318, 2024.
Article de Anglais | MEDLINE | ID: mdl-39145310

RÉSUMÉ

Background: Hepatocyte nuclear factor 4 alpha (HNF4α) is the master regulator of hepatic differentiation. Recent studies have also revealed the role of HNF4α in hepatocyte proliferation via negatively regulating the expression of proto-mitogenic genes, including cMyc. Here, we aimed to study the interaction between HNF4α-cMyc during liver regeneration after partial hepatectomy (PHX). Methods: Wild-type (WT), hepatocyte-specific knockout of HNF4α (HNF4α-KO), cMyc (cMyc-KO), and HNF4α-cMyc double knockout (DKO) mice were subjected to PHX to induce liver regeneration. Blood and liver tissue samples were collected at 0h, 24h, 48h, 7D, and 14D after PHX for further analysis. Results: WT, HNF4α-KO, cMyc-KO and DKO mice regained liver weight by 14 days after PHX. The deletion of cMyc did not affect liver regeneration, which was similar to the WT mice. WT and cMyc-KO mice started regaining liver weight as early as 24 hours after PHX, with a peak proliferation response at 48 hours after PHX. HNF4α- KO and DKO showed a delayed response with liver weight increase by day 7 after PHX. The overall hepatocyte proliferation response by DKO mice following PHX was lower than that of other genotypes. Interestingly, the surviving HNF4α-KO and DKO mice showed re-expression of HNF4α at mRNA and protein levels on day 14 after PHX. This was accompanied by a significant increase in the expression of Krt19 and Epcam, hepatic progenitor cell markers, in the DKO mice on day 14 after PHX. Conclusion: These data indicate that, in the absence of HNF4α, cMyc contributes to hepatocyte-driven proliferation to compensate for the lost tissue mass. Furthermore, in the absence of both HNF4α and cMyc, HPC-driven proliferation occurs to support liver regeneration.


Sujet(s)
Hépatectomie , Facteur nucléaire hépatocytaire HNF-4 , Régénération hépatique , Souris knockout , Animaux , Régénération hépatique/physiologie , Facteur nucléaire hépatocytaire HNF-4/métabolisme , Facteur nucléaire hépatocytaire HNF-4/génétique , Souris , Prolifération cellulaire , Protéines proto-oncogènes c-myc/métabolisme , Protéines proto-oncogènes c-myc/génétique , Hépatocytes/métabolisme , Foie/métabolisme , Mâle , Souris de lignée C57BL
17.
Proc Natl Acad Sci U S A ; 121(34): e2409262121, 2024 Aug 20.
Article de Anglais | MEDLINE | ID: mdl-39145929

RÉSUMÉ

Insig-1 and Insig-2 are endoplasmic reticulum (ER) proteins that inhibit lipid synthesis by blocking transport of sterol regulatory element-binding proteins (SREBP-1 and SREBP-2) from ER to Golgi. In the Golgi, SREBPs are processed proteolytically to release their transcription-activating domains, which enhance the synthesis of fatty acids, triglycerides, and cholesterol. Heretofore, the two Insigs have redundant functions, and there is no rationale for two isoforms. The current data identify a specific function for Insig-2. We show that eicosapentaenoic acid (EPA), a polyunsaturated fatty acid, inhibits fatty acid synthesis in human fibroblasts and rat hepatocytes by activating adenylate cyclase, which induces protein kinase A (PKA) to phosphorylate serine-106 in Insig-2. Phosphorylated Insig-2 inhibits the proteolytic processing of SREBP-1, thereby blocking fatty acid synthesis. Phosphorylated Insig-2 does not block the processing of SREBP-2, which activates cholesterol synthesis. Insig-1 lacks serine-106 and is not phosphorylated at this site. EPA inhibition of SREBP-1 processing was reduced by the replacement of serine-106 in Insig-2 with alanine or by treatment with KT5720, a PKA inhibitor. Inhibition did not occur in mutant human fibroblasts that possess Insig-1 but lack Insig-2. These data provide an Insig-2-specific mechanism for the long-known inhibition of fatty acid synthesis by polyunsaturated fatty acids.


Sujet(s)
Cyclic AMP-Dependent Protein Kinases , Fibroblastes , Protéines et peptides de signalisation intracellulaire , Protéines membranaires , Protéine-1 de liaison à l'élément de régulation des stérols , Humains , Protéines membranaires/métabolisme , Protéines membranaires/génétique , Animaux , Phosphorylation , Rats , Protéine-1 de liaison à l'élément de régulation des stérols/métabolisme , Protéine-1 de liaison à l'élément de régulation des stérols/génétique , Protéines et peptides de signalisation intracellulaire/métabolisme , Protéines et peptides de signalisation intracellulaire/génétique , Cyclic AMP-Dependent Protein Kinases/métabolisme , Fibroblastes/métabolisme , Acides gras insaturés/métabolisme , Acides gras/métabolisme , Acides gras/biosynthèse , Acide eicosapentanoïque/pharmacologie , Protéine-2 de liaison à l'élément de régulation des stérols/métabolisme , Hépatocytes/métabolisme
18.
Front Biosci (Landmark Ed) ; 29(7): 241, 2024 Jun 28.
Article de Anglais | MEDLINE | ID: mdl-39082347

RÉSUMÉ

BACKGROUND: The involvement of gut microbiota in carcinogenesis has gradually been highlighted in past decades. Bacteria could play its role by the secretion of extracellular vesicles (EVs); however, interrelationship between bacterial EVs and hepatocellular carcinoma (HCC) development has not been investigated much. METHODS: Diethylnitrosamine (DEN) was utilized to produce HCC model in mice, of which fecal was collected for detecting Bifidobacterium longum (B.longum) with real-time polymerase chain reaction (PCR). EV isolated from B.longum (B.longum-EV) with ultracentrifugation were stained with PKH26 to investigate the cellular uptake of murine hepatocytes (AML12). After treatment with B.longum-EV, TGF-ß1-induced AML12 cells were subjected to morphological observation, fibrosis- and apoptosis-related marker detection with western blot, apoptotic ratio and reactive oxygen species (ROS) level analysis with flow cytometry, and oxidative stress biomarker assessment with enzyme-linked immunosorbent assay (ELISA); meanwhile, animal studies including liver function, tumor formation rate, and histological analysis, were also performed to investigate the role of B.longum-EV in the fibrosis, apoptosis, oxidative stress, and carcinogenesis of the liver in vivo. RESULTS: The levels of B.longum were significantly reduced in HCC model mice. B.longum-EV could enter AML12 cells and effectively attenuate TGF-ß1-induced fibrosis, apoptosis, and oxidative stress in AML12 cells. In vivo studies showed that B.longum-EV administration alleviated DEN-induced liver fibrosis, apoptosis, and oxidative stress at the early stage. Moreover, B.longum-EV administration also effectively reduced the tumor formation rate and liver function injury in DEN-induced mice and down-regulated TGF-ß1 expression and Smad3 phosphorylation of mouse liver. CONCLUSIONS: B.longum-EVs protect hepatocytes against fibrosis, apoptosis, and oxidative damage, which exert a potential of preventing HCC development.


Sujet(s)
Apoptose , Bifidobacterium longum , Carcinome hépatocellulaire , Vésicules extracellulaires , Tumeurs du foie , Transduction du signal , Facteur de croissance transformant bêta-1 , Animaux , Facteur de croissance transformant bêta-1/métabolisme , Vésicules extracellulaires/métabolisme , Carcinome hépatocellulaire/métabolisme , Carcinome hépatocellulaire/prévention et contrôle , Carcinome hépatocellulaire/anatomopathologie , Carcinome hépatocellulaire/induit chimiquement , Tumeurs du foie/métabolisme , Tumeurs du foie/prévention et contrôle , Tumeurs du foie/anatomopathologie , Souris , Bifidobacterium longum/métabolisme , Apoptose/effets des médicaments et des substances chimiques , Stress oxydatif , Mâle , N-Éthyl-N-nitroso-éthanamine/toxicité , Protéines Smad/métabolisme , Hépatocytes/métabolisme , Espèces réactives de l'oxygène/métabolisme , Cirrhose du foie/métabolisme , Cirrhose du foie/prévention et contrôle , Cirrhose du foie/induit chimiquement , Cirrhose du foie/anatomopathologie
19.
J Agric Food Chem ; 72(30): 17099-17120, 2024 Jul 31.
Article de Anglais | MEDLINE | ID: mdl-39033510

RÉSUMÉ

Utilization of in vitro (cellular) techniques, like Cell Painting and transcriptomics, could provide powerful tools for agrochemical candidate sorting and selection in the discovery process. However, using these models generates challenges translating in vitro concentrations to the corresponding in vivo exposures. Physiologically based pharmacokinetic (PBPK) modeling provides a framework for quantitative in vitro to in vivo extrapolation (IVIVE). We tested whether in vivo (rat liver) transcriptomic and apical points of departure (PODs) could be accurately predicted from in vitro (rat hepatocyte or human HepaRG) transcriptomic PODs or HepaRG Cell Painting PODs using PBPK modeling. We compared two PBPK models, the ADMET predictor and the httk R package, and found httk to predict the in vivo PODs more accurately. Our findings suggest that a rat liver apical and transcriptomic POD can be estimated utilizing a combination of in vitro transcriptome-based PODs coupled with PBPK modeling for IVIVE. Thus, high content in vitro data can be translated with modest accuracy to in vivo models of ultimate regulatory importance to help select agrochemical analogs in early stage discovery program.


Sujet(s)
Agrochimie , Animaux , Rats , Humains , Agrochimie/pharmacocinétique , Agrochimie/toxicité , Hépatocytes/métabolisme , Foie/métabolisme , Modèles biologiques , Mâle , Transcriptome , Lignée cellulaire , Appréciation des risques
20.
Proc Natl Acad Sci U S A ; 121(28): e2403442121, 2024 Jul 09.
Article de Anglais | MEDLINE | ID: mdl-38968107

RÉSUMÉ

Plasmodium falciparum causes severe malaria and assembles a protein translocon (PTEX) complex at the parasitophorous vacuole membrane (PVM) of infected erythrocytes, through which several hundred proteins are exported to facilitate growth. The preceding liver stage of infection involves growth in a hepatocyte-derived PVM; however, the importance of protein export during P. falciparum liver infection remains unexplored. Here, we use the FlpL/FRT system to conditionally excise genes in P. falciparum sporozoites for functional liver-stage studies. Disruption of PTEX members ptex150 and exp2 did not affect sporozoite development in mosquitoes or infectivity for hepatocytes but attenuated liver-stage growth in humanized mice. While PTEX150 deficiency reduced fitness on day 6 postinfection by 40%, EXP2 deficiency caused 100% loss of liver parasites, demonstrating that PTEX components are required for growth in hepatocytes to differing degrees. To characterize PTEX loss-of-function mutations, we localized four liver-stage Plasmodium export element (PEXEL) proteins. P. falciparum liver specific protein 2 (LISP2), liver-stage antigen 3 (LSA3), circumsporozoite protein (CSP), and a Plasmodium berghei LISP2 reporter all localized to the periphery of P. falciparum liver stages but were not exported beyond the PVM. Expression of LISP2 and CSP but not LSA3 was reduced in ptex150-FRT and exp2-FRT liver stages, suggesting that expression of some PEXEL proteins is affected directly or indirectly by PTEX disruption. These results show that PTEX150 and EXP2 are important for P. falciparum development in hepatocytes and emphasize the emerging complexity of PEXEL protein trafficking.


Sujet(s)
Hépatocytes , Foie , Paludisme à Plasmodium falciparum , Plasmodium falciparum , Protéines de protozoaire , Sporozoïtes , Plasmodium falciparum/croissance et développement , Plasmodium falciparum/génétique , Plasmodium falciparum/métabolisme , Animaux , Protéines de protozoaire/métabolisme , Protéines de protozoaire/génétique , Sporozoïtes/métabolisme , Sporozoïtes/croissance et développement , Souris , Foie/parasitologie , Foie/métabolisme , Humains , Hépatocytes/parasitologie , Hépatocytes/métabolisme , Paludisme à Plasmodium falciparum/parasitologie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE