Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 40.257
Filtrer
1.
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
2.
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
3.
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
4.
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
5.
Virol J ; 21(1): 170, 2024 Aug 01.
Article de Anglais | MEDLINE | ID: mdl-39090742

RÉSUMÉ

BACKGROUND: Chronic hepatitis B virus (HBV) infection affects around 250 million people worldwide, causing approximately 887,000 deaths annually, primarily owing to cirrhosis and hepatocellular carcinoma (HCC). The current approved treatments for chronic HBV infection, such as interferon and nucleos(t)ide analogs, have certain limitations as they cannot completely eradicate covalently closed circular DNA (cccDNA). Considering that HBV replication relies on host transcription factors, focusing on host factors in the HBV genome may provide insights into new therapeutic targets against HBV. Therefore, understanding the mechanisms underlying viral persistence and hepatocyte pathogenesis, along with the associated host factors, is crucial. In this study, we investigated novel therapeutic targets for HBV infection by identifying gene and pathway networks involved in HBV replication in primary human hepatocytes (PHHs). Importantly, our study utilized cultured primary hepatocytes, allowing transcriptomic profiling in a biologically relevant context and enabling the investigation of early HBV-mediated effects. METHODS: PHHs were infected with HBV virion particles derived from HepAD38 cells at 80 HBV genome equivalents per cell (Geq/cell). For transcriptomic sequencing, PHHs were harvested 1, 2-, 3-, 5-, and 7 days post-infection (dpi). After preparing the libraries, clustering and sequencing were conducted to generate RNA-sequencing data. This data was processed using Bioinformatics tools and software to analyze DEGs and obtain statistically significant results. Furthermore, qRT-PCR was performed to validate the RNA-sequencing results, ensuring consistent findings. RESULTS: We observed significant alterations in the expression patterns of 149 genes from days 1 to 7 following HBV infection (R2 > 0.7, q < 0.05). Functional analysis of these genes identified RNA-binding proteins involved in mRNA metabolism and the regulation of alternative splicing during HBV infection. Results from qRT-PCR experiments and the analysis of two validation datasets suggest that RBM14 and RPL28 may serve as potential biomarkers for HBV-associated HCC. CONCLUSIONS: Transcriptome analysis of gene expression changes during HBV infection in PHHs provided valuable insights into chronic HBV infection. Additionally, understanding the functional involvement of host factor networks in the molecular mechanisms of HBV replication and transcription may facilitate the development of novel strategies for HBV treatment.


Sujet(s)
Virus de l'hépatite B , Hépatocytes , Réplication virale , Humains , Hépatocytes/virologie , Virus de l'hépatite B/génétique , Virus de l'hépatite B/physiologie , Analyse de profil d'expression de gènes , Interactions hôte-pathogène , Cellules cultivées , Réseaux de régulation génique , Hépatite B/virologie , Hépatite B/génétique , Hépatite B chronique/virologie
6.
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
7.
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
8.
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
9.
Emerg Microbes Infect ; 13(1): 2387448, 2024 Dec.
Article de Anglais | MEDLINE | ID: mdl-39109538

RÉSUMÉ

Therapeutics for eradicating hepatitis B virus (HBV) infection are still limited and current nucleos(t)ide analogs (NAs) and interferon are effective in controlling viral replication and improving liver health, but they cannot completely eradicate the hepatitis B virus and only a very small number of patients are cured of it. The TCR-like antibodies recognizing viral peptides presented on human leukocyte antigens (HLA) provide possible tools for targeting and eliminating HBV-infected hepatocytes. Here, we generated three TCR-like antibodies targeting three different HLA-A2.1-presented peptides derived from HBV core and surface proteins. Bispecific antibodies (BsAbs) were developed by fuzing variable fragments of these TCR-like mAbs with an anti-CD3ϵ antibody. Our data demonstrate that the BsAbs could act as T cell engagers, effectively redirecting and activating T cells to target HBV-infected hepatocytes in vitro and in vivo. In HBV-persistent mice expressing human HLA-A2.1, two infusions of BsAbs induced marked and sustained suppression in serum HBsAg levels and also reduced the numbers of HBV-positive hepatocytes. These findings highlighted the therapeutic potential of TCR-like BsAbs as a new strategy to cure hepatitis B.


Sujet(s)
Anticorps bispécifiques , Modèles animaux de maladie humaine , Virus de l'hépatite B , Hépatite B , Hépatocytes , Animaux , Anticorps bispécifiques/immunologie , Anticorps bispécifiques/pharmacologie , Hépatocytes/virologie , Hépatocytes/immunologie , Souris , Humains , Virus de l'hépatite B/immunologie , Virus de l'hépatite B/génétique , Hépatite B/immunologie , Hépatite B/virologie , Antigène HLA-A2/immunologie , Antigènes de surface du virus de l'hépatite B/immunologie , Récepteurs aux antigènes des cellules T/immunologie , Lymphocytes T/immunologie
10.
Gut Microbes ; 16(1): 2390164, 2024.
Article de Anglais | MEDLINE | ID: mdl-39154362

RÉSUMÉ

Non-alcoholic fatty liver disease (NAFLD) has emerged as a global health concern, lacking specific therapeutic strategies. Time-restricted feeding (TRF) regimen demonstrated beneficial effects in NAFLD; however, the underlying mechanisms remain unclear. In this study, we established a NAFLD mouse model through a high-fat diet (HFD) and implemented the 16:8 TRF regimen for a duration of 6 weeks. We demonstrated that TRF remarkably alleviated hepatic steatosis in HFD mice. Of note, aldehyde oxidase 1 (AOX1), a key enzyme in hepatic nicotinamide (NAM) catabolism, exhibited apparent upregulation in response to HFD, leading to abnormal accumulation of N-Methyl-6-pyridone-3-carboxamide (N-Me-6-PY, also known as 2PY) and N-Methyl-4-pyridone-5-carboxamide (N-Me-4-PY, also known as 4PY), whereas it was almost restored by TRF. Both N-Me-6-PY and N-Me-4-PY promoted de novo lipogenesis and fatty acid uptake capacities in hepatocyte, and aggravated hepatic steatosis in mice either fed chow diet or HFD. In contrast, pharmacological inhibition of AOX1 was sufficient to ameliorate the hepatic steatosis and lipid metabolic dysregulation induced by HFD. Moreover, transplantation of fecal microbiota efficiently mimicked the modulatory effect of TRF on NAM metabolism, thus mitigating hepatic steatosis and lipid metabolic disturbance, suggesting a gut microbiota-dependent manner. In conclusion, our study reveals the intricate relationship between host NAM metabolic modification and gut microbiota remodeling during the amelioration of NAFLD by TRF, providing promising insights into the prevention and treatment of NAFLD.


Sujet(s)
Alimentation riche en graisse , Microbiome gastro-intestinal , Foie , Souris de lignée C57BL , Nicotinamide , Stéatose hépatique non alcoolique , Animaux , Stéatose hépatique non alcoolique/métabolisme , Stéatose hépatique non alcoolique/microbiologie , Souris , Foie/métabolisme , Alimentation riche en graisse/effets indésirables , Mâle , Nicotinamide/métabolisme , Modèles animaux de maladie humaine , Métabolisme lipidique , Aldehyde oxidase/métabolisme , Lipogenèse , Hépatocytes/métabolisme , Humains
11.
Life Sci Alliance ; 7(11)2024 Nov.
Article de Anglais | MEDLINE | ID: mdl-39159974

RÉSUMÉ

Regeneration of insulin-producing ß-cells is an alternative avenue to manage diabetes, and it is crucial to unravel this process in vivo during physiological responses to the lack of ß-cells. Here, we aimed to characterize how hepatocytes can contribute to ß-cell regeneration, either directly or indirectly via secreted proteins or metabolites, in a zebrafish model of ß-cell loss. Using lineage tracing, we show that hepatocytes do not directly convert into ß-cells even under extreme ß-cell ablation conditions. A transcriptomic analysis of isolated hepatocytes after ß-cell ablation displayed altered lipid- and glucose-related processes. Based on the transcriptomics, we performed a genetic screen that uncovers a potential role of the molybdenum cofactor (Moco) biosynthetic pathway in ß-cell regeneration and glucose metabolism in zebrafish. Consistently, molybdenum cofactor synthesis 2 (Mocs2) haploinsufficiency in mice indicated dysregulated glucose metabolism and liver function. Together, our study sheds light on the liver-pancreas crosstalk and suggests that the molybdenum cofactor biosynthesis pathway should be further studied in relation to glucose metabolism and diabetes.


Sujet(s)
Coenzymes , Glucose , Hépatocytes , Cellules à insuline , Foie , Métalloprotéines , Cofacteurs à molybdène , Ptéridines , Danio zébré , Animaux , Cellules à insuline/métabolisme , Ptéridines/métabolisme , Coenzymes/métabolisme , Souris , Foie/métabolisme , Foie/cytologie , Métalloprotéines/métabolisme , Métalloprotéines/génétique , Hépatocytes/métabolisme , Glucose/métabolisme , Régénération/génétique , Pancréas/métabolisme , Pancréas/cytologie , Protéines de poisson-zèbre/génétique , Protéines de poisson-zèbre/métabolisme
12.
Cell Death Dis ; 15(8): 600, 2024 Aug 19.
Article de Anglais | MEDLINE | ID: mdl-39160159

RÉSUMÉ

Crizotinib carries an FDA hepatotoxicity warning, yet analysis of the FAERS database suggests that the severity of its hepatotoxicity risks, including progression to hepatitis and liver failure, might be underreported. However, the underlying mechanism remains poorly understood, and effective intervention strategies are lacking. Here, mRNA-sequencing analysis, along with KEGG and GO analyses, revealed that DEGs linked to Crizotinib-induced hepatotoxicity predominantly associate with the ferroptosis pathway which was identified as the principal mechanism behind Crizotinib-induced hepatocyte death. Furthermore, we found that ferroptosis inhibitors, namely Ferrostatin-1 and Deferoxamine mesylate, significantly reduced Crizotinib-induced hepatotoxicity and ferroptosis in both in vivo and in vitro settings. We have also discovered that overexpression of AAV8-mediated Nrf2 could mitigate Crizotinib-induced hepatotoxicity and ferroptosis in vivo by restoring the imbalance in glutathione metabolism, iron homeostasis, and lipid peroxidation. Additionally, both Stat1 deficiency and the Stat1 inhibitor NSC118218 were found to reduce Crizotinib-induced ferroptosis. Mechanistically, Crizotinib induces the phosphorylation of Stat1 at Ser727 but not Tyr701, promoting the transcriptional inhibition of Nrf2 expression after its entry into the nucleus to promote ferroptosis. Meanwhile, we found that MgIG and GA protected against hepatotoxicity to counteract ferroptosis without affecting or compromising the anti-cancer activity of Crizotinib, with a mechanism potentially related to the Stat1/Nrf2 pathway. Overall, our findings identify that the phosphorylation activation of Stat1 Ser727, rather than Tyr701, promotes ferroptosis through transcriptional inhibition of Nrf2, and highlight MgIG and GA as potential therapeutic approaches to enhance the safety of Crizotinib-based cancer therapy.


Sujet(s)
Lésions hépatiques dues aux substances , Crizotinib , Ferroptose , Facteur-2 apparenté à NF-E2 , Facteur de transcription STAT-1 , Ferroptose/effets des médicaments et des substances chimiques , Facteur-2 apparenté à NF-E2/métabolisme , Humains , Animaux , Crizotinib/pharmacologie , Crizotinib/effets indésirables , Lésions hépatiques dues aux substances/métabolisme , Lésions hépatiques dues aux substances/anatomopathologie , Lésions hépatiques dues aux substances/génétique , Facteur de transcription STAT-1/métabolisme , Facteur de transcription STAT-1/génétique , Souris , Transduction du signal/effets des médicaments et des substances chimiques , Mâle , Phénylènediamines/pharmacologie , Souris de lignée C57BL , Hépatocytes/métabolisme , Hépatocytes/effets des médicaments et des substances chimiques , Phosphorylation/effets des médicaments et des substances chimiques
13.
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
14.
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
15.
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
16.
Gene ; 929: 148812, 2024 Dec 15.
Article de Anglais | MEDLINE | ID: mdl-39116959

RÉSUMÉ

Nonalcoholic fatty liver disease (NAFLD) is the most common chronic liver disease and represents the main cause of liver cirrhosis and hepatocellular carcinoma. Cav3.2 is a T-type calcium channel that is widely present in tissues throughout the body and plays a vital role in energy and metabolic balance. However, the effects of Cav3.2 on the NFALD remain unclear. Here, we investigated the role of Cav3.2 channel in the development and progression of NAFLD. After 16 weeks on a high-fat diets (HFD), Cav3.2 knockout (Cav3.2 KO) improved hepatic steatosis, liver injury and metabolic syndrome in an NAFLD mouse model. We provided evidence that Cav3.2 KO inhibited HFD-induced hepatic oxidative stress, inflammation and hepatocyte apoptosis. In addition, Cav3.2 KO also attenuated hepatic lipid accumulation, oxidative stress, inflammation and hepatocyte apoptosis in palmitic acid/oleic acid (PAOA)-treated primary hepatocytes. These results suggest that therapeutic approaches targeting Cav3.2 provide effective approaches for treating NAFLD.


Sujet(s)
Apoptose , Canaux calciques de type T , Alimentation riche en graisse , Hépatocytes , Souris knockout , Stéatose hépatique non alcoolique , Stress oxydatif , Animaux , Stéatose hépatique non alcoolique/génétique , Stéatose hépatique non alcoolique/métabolisme , Stéatose hépatique non alcoolique/anatomopathologie , Canaux calciques de type T/génétique , Canaux calciques de type T/métabolisme , Souris , Alimentation riche en graisse/effets indésirables , Hépatocytes/métabolisme , Hépatocytes/anatomopathologie , Mâle , Souris de lignée C57BL , Modèles animaux de maladie humaine , Foie/métabolisme , Foie/anatomopathologie , Inflammation/génétique , Inflammation/métabolisme
17.
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
18.
Cell Biol Toxicol ; 40(1): 71, 2024 Aug 16.
Article de Anglais | MEDLINE | ID: mdl-39147926

RÉSUMÉ

The simultaneous abuse of alcohol-cocaine is known to cause stronger and more unpredictable cellular damage in the liver, heart, and brain. However, the mechanistic crosstalk between cocaine and alcohol in liver injury remains unclear. The findings revealed cocaine-induced liver injury and inflammation in both marmosets and mice. Of note, co-administration of cocaine and ethanol in mice causes more severe liver damage than individual treatment. The metabolomic analysis confirmed that hippuric acid (HA) is the most abundant metabolite in marmoset serum after cocaine consumption and that is formed in primary marmoset hepatocytes. HA, a metabolite of cocaine, increases mitochondrial DNA leakage and subsequently increases the production of proinflammatory factors via STING signaling in Kupffer cells (KCs). In addition, conditioned media of cocaine-treated KC induced hepatocellular necrosis via alcohol-induced TNFR1. Finally, disruption of STING signaling in vivo ameliorated co-administration of alcohol- and cocaine-induced liver damage and inflammation. These findings postulate intervention of HA-STING-TNFR1 axis as a novel strategy for treatment of alcohol- and cocaine-induced excessive liver damage.


Sujet(s)
Cocaïne , ADN mitochondrial , Hippurates , Maladies alcooliques du foie , Protéines membranaires , Transduction du signal , Animaux , Cocaïne/pharmacologie , Cocaïne/toxicité , Transduction du signal/effets des médicaments et des substances chimiques , Maladies alcooliques du foie/métabolisme , Maladies alcooliques du foie/anatomopathologie , ADN mitochondrial/métabolisme , ADN mitochondrial/effets des médicaments et des substances chimiques , Souris , Hippurates/métabolisme , Mâle , Protéines membranaires/métabolisme , Hépatocytes/métabolisme , Hépatocytes/effets des médicaments et des substances chimiques , Cellules de Küpffer/effets des médicaments et des substances chimiques , Cellules de Küpffer/métabolisme , Foie/effets des médicaments et des substances chimiques , Foie/métabolisme , Foie/anatomopathologie , Éthanol/toxicité , Souris de lignée C57BL , Troubles liés à la cocaïne/métabolisme , Récepteur au facteur de nécrose tumorale de type I/métabolisme
19.
Clin Transl Med ; 14(8): e1812, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-39152680

RÉSUMÉ

The liver possesses a distinctive capacity for regeneration within the human body. Under normal circumstances, liver cells replicate themselves to maintain liver function. Compensatory replication of healthy hepatocytes is sufficient for the regeneration after acute liver injuries. In the late stage of chronic liver damage, a large number of hepatocytes die and hepatocyte replication is blocked. Liver regeneration has more complex mechanisms, such as the transdifferentiation between cell types or hepatic progenitor cells mediated. Dysregulation of liver regeneration causes severe chronic liver disease. Gaining a more comprehensive understanding of liver regeneration mechanisms would facilitate the advancement of efficient therapeutic approaches. This review provides an overview of the signalling pathways linked to different aspects of liver regeneration in various liver diseases. Moreover, new knowledge on cellular interactions during the regenerative process is also presented. Finally, this paper explores the potential applications of new technologies, such as nanotechnology, stem cell transplantation and organoids, in liver regeneration after injury, offering fresh perspectives on treating liver disease.


Sujet(s)
Régénération hépatique , Régénération hépatique/physiologie , Humains , Maladies du foie/thérapie , Maladies du foie/physiopathologie , Communication cellulaire/physiologie , Foie/traumatismes , Hépatocytes/métabolisme , Transduction du signal , Animaux
20.
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
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE