Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 13.341
Filter
1.
J Ethnopharmacol ; 336: 118743, 2025 Jan 10.
Article in English | MEDLINE | ID: mdl-39209000

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Non-alcoholic fatty liver disease (NAFLD) is the leading cause of liver-related morbidity and mortality, with hepatic steatosis being the hallmark symptom. Salvia miltiorrhiza Bunge (Smil, Dan-Shen) and Ligusticum striatum DC (Lstr, Chuan-Xiong) are commonly used to treat cardiovascular diseases and have the potential to regulate lipid metabolism. However, whether Smil/Lstr combo can be used to treat NAFLD and the mechanisms underlying its lipid-regulating properties remain unclear. PURPOSE: To assess the feasibility and reliability of a short-term high-fat diet (HFD) induced zebrafish model for evaluating hepatic steatosis phenotype and to investigate the liver lipid-lowering effects of Smil/Lstr, as well as its active components. METHODS: The phenotypic alterations of liver and multiple other organ systems were examined in the HFD zebrafish model using fluorescence imaging and histochemistry. The liver-specific lipid-lowering effects of Smil/Lstr combo were evaluated endogenously. The active molecules and functional mechanisms were further explored in zebrafish, human hepatocytes, and hamster models. RESULTS: In 5-day HFD zebrafish, significant lipid accumulation was detected in the blood vessels and the liver, as evidenced by increased staining with Oil Red O and fluorescent lipid probes. Hepatic hypertrophy was observed in the model, along with macrovesicular steatosis. Smil/Lstr combo administration effectively restored the lipid profile and alleviated hepatic hypertrophy in the HFD zebrafish. In oleic-acid stimulated hepatocytes, Smil/Lstr combo markedly reduced lipid accumulation and cell damage. Subsequently, based on zebrafish phenotypic screening, the natural phthalide senkyunolide I (SEI) was identified as a major molecule mediating the lipid-lowering activities of Smil/Lstr combo in the liver. Moreover, SEI upregulated the expression of the lipid metabolism regulator PPARα and downregulated fatty acid translocase CD36, while a PPARα antagonist sufficiently blocked the regulatory effect of SEI on hepatic steatosis. Finally, the roles of SEI on hepatic lipid accumulation and PPARα signaling were further verified in the hamster model. CONCLUSIONS: We proposed a zebrafish-based screening strategy for modulators of hepatic steatosis and discovered the regulatory roles of Smil/Lstr combo and its component SEI on liver lipid accumulation and PPARα signaling, suggesting their potential value as novel candidates for NAFLD treatment.


Subject(s)
PPAR alpha , Signal Transduction , Zebrafish , Animals , Cricetinae , Humans , Male , Benzofurans/pharmacology , Diet, High-Fat , Disease Models, Animal , Fatty Liver/drug therapy , Hep G2 Cells , Hepatocytes/drug effects , Hepatocytes/metabolism , Lipid Metabolism/drug effects , Liver/drug effects , Liver/metabolism , Liver/pathology , Mesocricetus , Non-alcoholic Fatty Liver Disease/drug therapy , PPAR alpha/metabolism , Signal Transduction/drug effects
2.
Cell Death Dis ; 15(9): 680, 2024 Sep 17.
Article in English | MEDLINE | ID: mdl-39289337

ABSTRACT

Iron overload and cellular senescence have been implicated in liver fibrosis, but their possible mechanistic connection has not been explored. To address this, we have delved into the role of iron and senescence in an experimental model of chronic liver injury, analyzing whether an iron chelator would prevent liver fibrosis by decreasing hepatocyte senescence. The model of carbon tetrachloride (CCl4) in mice was used as an experimental model of liver fibrosis. Results demonstrated that during the progression of liver fibrosis, accumulation of iron occurs, concomitant with the appearance of fibrotic areas and cells undergoing senescence. Isolated parenchymal hepatocytes from CCl4-treated mice present a gene transcriptomic signature compatible with iron accumulation and senescence, which correlates with induction of Reactive Oxygen Species (ROS)-related genes, activation of the Transforming Growth Factor-beta (TGF-ß) pathway and inhibition of oxidative metabolism. Analysis of the iron-related gene signature in a published single-cell RNA-seq dataset from CCl4-treated livers showed iron accumulation correlating with senescence in other non-parenchymal liver cells. Treatment with deferiprone, an iron chelator, attenuated iron accumulation, fibrosis and senescence, concomitant with relevant changes in the senescent-associated secretome (SASP), which switched toward a more anti-inflammatory profile of cytokines. In vitro experiments in human hepatocyte HH4 cells demonstrated that iron accumulates in response to a senescence-inducing reagent, doxorubicin, being deferiprone able to prevent senescence and SASP, attenuating growth arrest and cell death. However, deferiprone did not significantly affect senescence induced by two different agents (doxorubicin and deoxycholic acid) or activation markers in human hepatic stellate LX-2 cells. Transcriptomic data from patients with different etiologies demonstrated the relevance of iron accumulation in the progression of liver chronic damage and fibrosis, correlating with a SASP-related gene signature and pivotal hallmarks of fibrotic changes. Altogether, our study establishes iron accumulation as a clinically exploitable driver to attenuate pathological senescence in hepatocytes.


Subject(s)
Cellular Senescence , Iron Chelating Agents , Liver Cirrhosis , Liver Cirrhosis/pathology , Liver Cirrhosis/metabolism , Liver Cirrhosis/drug therapy , Animals , Cellular Senescence/drug effects , Iron Chelating Agents/pharmacology , Humans , Mice , Male , Disease Progression , Iron/metabolism , Hepatocytes/metabolism , Hepatocytes/drug effects , Hepatocytes/pathology , Mice, Inbred C57BL , Carbon Tetrachloride , Deferiprone/pharmacology , Reactive Oxygen Species/metabolism , Disease Models, Animal
3.
Pestic Biochem Physiol ; 204: 106064, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39277381

ABSTRACT

Environmental pollution caused by arsenic or its compounds is called arsenic pollution. Arsenic pollution mainly comes from people's mining and smelting of arsenic compounds. In addition, the widespread use of arsenic compounds, such as the use and production of arsenic-containing pesticides, is also a source of arsenic contamination. Arsenic contamination leads to an increased risk of arsenic exposure, and the multi-organ toxicity induced by arsenic exposure is a global health problem. As a non-mammalian vertebrate with high nutrient levels, chickens readily absorb and accumulate arsenic from their food. Relevant studies have shown that arsenic exposure induces hepatotoxicity in chickens, and there has been a steady stream of research into the specific mechanisms involved. PANoptosis, a newly discovered and unique mode of programmed cell death (PCD) characterized by both apoptosis, cellular pyroptosis, and necroptosis. There are no studies to indicate whether chicken liver toxicity due to arsenic is associated with PANoptosis. Therefore, we established chicken animal models and chicken primary hepatocyte models exposed to different arsenic concentrations to dissect the role and mechanism of PANoptosis in arsenic exposure-induced hepatotoxicity in chickens. Our histopathological results showed that arsenic treatment caused dose-dependent damage to chicken liver structure. Meanwhile, different doses of arsenic treatment groups caused significant up-regulation of the protein level of ZBP1, a key factor of PANoptosis. And then consequently triggered the abnormal gene and protein expression levels of apoptosis-associated factors (Caspase-8, Caspase-7, Caspase-3), cellular pyroptosis-associated factors (NLRP3, ASC, GSDMD) and necroptosis-associated factors (RIPK1, RIPK3, MLKL). In conclusion, our study revealed that PANoptosis is involved in arsenic-induced chicken hepatotoxicity. Our findings provide a new perspective on the pathogenesis of arsenic exposure-induced hepatotoxicity in chickens.


Subject(s)
Arsenic , Chickens , Liver , Animals , Arsenic/toxicity , Liver/drug effects , Liver/pathology , Liver/metabolism , Hepatocytes/drug effects , Hepatocytes/pathology , Hepatocytes/metabolism , Chemical and Drug Induced Liver Injury/pathology , Necroptosis/drug effects , Apoptosis/drug effects
4.
Pestic Biochem Physiol ; 204: 106098, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39277407

ABSTRACT

Fenitrothion (FNT) is a common organophosphorus pesticide that is widely used in both agricultural and domestic pest control. FNT has been frequently detected in various environmental media, including the human body, and is a notable contaminant. Epidemiological investigations have recently shown the implications of exposure to FNT in the incidence of various metabolic diseases, such as diabetes mellitus in humans, indicating that FNT may be a potential endocrine disruptor. However, the effects of FNT exposure on glucose homeostasis and their underlying mechanisms in model organisms remain largely unknown, which may limit our understanding of the health risks of FNT. In this study, FNT (4 5, 90, 180, and 4 50 µM) exposure model of rat hepatocytes (Buffalo Rat Liver, BRL cells) was established to investigate the effects and potential mechanisms of its toxicity on glucose metabolism. Several key processes of glucose metabolism were detected in this study. The results showed significantly increased glucose levels in the culture medium and decreased glycogen content in the FNT-exposed BRL cells. The results of quantitative real-time PCR and enzymology showed the abnormal expression of genes and activity/content of glucose metabolic enzymes involved in glucose metabolism, which might promote gluconeogenesis and inhibit glucose uptake, glycolysis, and glycogenesis. Furthermore, gluconeogenesis and glycolytic were carried out in the mitochondrial membrane. The abnormal of mitochondrial membrane potential may be a potential mechanism underlying FNT-induced glucose metabolism disorder. In addition, the mRNA and protein expression implicated that FNT may disrupt glucose metabolism by inhibiting the AMPKα and IRS1/PI3K/AKT signaling pathways. In conclusion, results provide in vitro evidence that FNT can cause glucose metabolism disorder, which emphasizes the potential health risks of exposure to FNT in inducing diabetes mellitus.


Subject(s)
AMP-Activated Protein Kinases , Fenitrothion , Glucose , Insulin Receptor Substrate Proteins , Phosphatidylinositol 3-Kinases , Proto-Oncogene Proteins c-akt , Signal Transduction , Animals , Rats , Fenitrothion/toxicity , Proto-Oncogene Proteins c-akt/metabolism , Insulin Receptor Substrate Proteins/metabolism , Signal Transduction/drug effects , AMP-Activated Protein Kinases/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Glucose/metabolism , Liver/drug effects , Liver/metabolism , Hepatocytes/drug effects , Hepatocytes/metabolism , Glucose Metabolism Disorders/chemically induced , Glucose Metabolism Disorders/metabolism , Insecticides/toxicity
5.
Clin Transl Sci ; 17(9): e70028, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39288032

ABSTRACT

A drug-drug interaction (DDI) study was conducted to evaluate the effect of icenticaftor (QBW251) on the pharmacokinetics (PK) of a 5-probe cytochrome P450 (CYP) substrate cocktail, guided by in vitro studies in human hepatocytes and liver microsomes. Another DDI study investigated the effect of icenticaftor on the PK and pharmacodynamics (PD) of a monophasic oral contraceptive (OC) containing ethinyl estradiol (EE) and levonorgestrel (LVG) in premenopausal healthy female subjects. The static-mechanistic DDI assessment indicated that icenticaftor may moderately induce the metabolic clearance of co-medications metabolized by CYP3A4 (area under the concentration-time curve [AUC] ratio: 0.47) and potentially CYP2C; icenticaftor may also weakly inhibit the metabolic clearance of co-medications metabolized by CYP1A2 and CYP3A4 (AUC ratio: 1.35 and 1.86, respectively) and moderately inhibit CYP2B6 (AUC ratio: 2.11). In the CYP substrate cocktail DDI study, icenticaftor 300 mg twice daily (b.i.d.) moderately inhibited CYP1A2 (AUC ratio: 3.35) and CYP2C19 (AUC ratio: 2.70). As expected from the results of the in vitro studies, weak induction was observed for CYP3A4 (AUC ratio: 0.51) and CYP2C8 (AUC ratio: 0.66). In the OC DDI study, co-administration of icenticaftor 450 mg b.i.d. with monophasic OC containing 30-µg EE and 150-µg LVG once daily reduced the plasma exposure of both components by approximately 50% and led to increased levels of follicle-stimulating hormone and luteinizing hormone. These results provide valuable guidance for the use of icenticaftor in patients taking concomitant medications that are substrates of CYP enzymes or patients using OCs.


Subject(s)
Contraceptives, Oral, Combined , Drug Interactions , Ethinyl Estradiol , Humans , Female , Adult , Ethinyl Estradiol/pharmacokinetics , Ethinyl Estradiol/administration & dosage , Ethinyl Estradiol/pharmacology , Young Adult , Contraceptives, Oral, Combined/pharmacokinetics , Contraceptives, Oral, Combined/administration & dosage , Levonorgestrel/pharmacokinetics , Levonorgestrel/administration & dosage , Levonorgestrel/pharmacology , Microsomes, Liver/metabolism , Microsomes, Liver/drug effects , Hepatocytes/metabolism , Hepatocytes/drug effects , Cytochrome P-450 Enzyme System/metabolism , Drug Combinations , Healthy Volunteers , Area Under Curve , Contraceptives, Oral/pharmacokinetics , Contraceptives, Oral/pharmacology , Contraceptives, Oral/administration & dosage , Adolescent
6.
Cells ; 13(17)2024 Aug 29.
Article in English | MEDLINE | ID: mdl-39273020

ABSTRACT

BACKGROUND: Exercise can promote sustainable protection against cold and warm liver ischemia-reperfusion injury (IRI) and tumor metastases. We have shown that this protection is by the induction of hepatic mitochondrial biogenesis pathway. In this study, we hypothesize that ZLN005, a PGC-1α activator, can be utilized as an alternative therapeutic strategy. METHODS: Eight-week-old mice were pretreated with ZLN005 and subjected to liver warm IRI. To establish a liver metastatic model, MC38 cancer cells (1 × 106) were injected into the spleen, followed by splenectomy and liver IRI. RESULTS: ZLN005-pretreated mice showed a significant decrease in IRI-induced tissue injury as measured by serum ALT/AST/LDH levels and tissue necrosis. ZLN005 pretreatment decreased ROS generation and cell apoptosis at the site of injury, with a significant decrease in serum pro-inflammatory cytokines, innate immune cells infiltration, and intrahepatic neutrophil extracellular trap (NET) formation. Moreover, mitochondrial mass was significantly upregulated in hepatocytes and maintained after IRI. This was confirmed in murine and human hepatocytes treated with ZLN005 in vitro under normoxic and hypoxic conditions. Additionally, ZLN005 preconditioning significantly attenuated tumor burden and increased the percentage of intratumoral cytotoxic T cells. CONCLUSIONS: Our study highlights the effective protection of ZLN005 pretreatment as a therapeutic alternative in terms of acute liver injury and tumor metastases.


Subject(s)
Liver Neoplasms , Liver , Mice, Inbred C57BL , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Reperfusion Injury , Animals , Reperfusion Injury/pathology , Reperfusion Injury/prevention & control , Reperfusion Injury/metabolism , Mice , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Liver Neoplasms/secondary , Liver Neoplasms/pathology , Liver/pathology , Liver/drug effects , Liver/metabolism , Humans , Male , Apoptosis/drug effects , Disease Progression , Hepatocytes/metabolism , Hepatocytes/drug effects , Hepatocytes/pathology , Cell Line, Tumor , Mitochondria/metabolism , Mitochondria/drug effects , Reactive Oxygen Species/metabolism , Extracellular Traps/metabolism , Extracellular Traps/drug effects
7.
Molecules ; 29(17)2024 Aug 28.
Article in English | MEDLINE | ID: mdl-39274927

ABSTRACT

Non-alcoholic fatty liver disease (NAFLD) is one of the most common metabolic diseases encountered in clinical practice, which is characterized by the excessive accumulation of triglycerides (steatosis), and a variety of metabolic abnormalities including lipid metabolism and bile acid metabolism are closely related to NAFLD. In China, Gynostemma pentaphyllum is used as functional food and Chinese medicine to treat various diseases, especially NAFLD, for a long time. However, the active components that exert the main therapeutic effects and their mechanisms remain unclear. In this study, Gypensapogenin A was isolated from the total saponins of G. pentaphyllum and prepared as a liposomal delivery system. Gypensapogenin A liposomes could activate FXR, inhibit the expression of CYP7A1 and CYP8B1, increase the expression of CYP27A1, modulate the ratio of CA and CDCA, decrease the content of CA, and increase the content of CDCA, thus forming a virtuous cycle of activating FXR to play a role in lowering blood lipid levels.


Subject(s)
Gynostemma , Lipid Metabolism , Liposomes , Receptors, Cytoplasmic and Nuclear , Receptors, Cytoplasmic and Nuclear/metabolism , Liposomes/chemistry , Lipid Metabolism/drug effects , Humans , Animals , Gynostemma/chemistry , Non-alcoholic Fatty Liver Disease/drug therapy , Non-alcoholic Fatty Liver Disease/metabolism , Saponins/pharmacology , Saponins/chemistry , Hep G2 Cells , Mice , Bile Acids and Salts/metabolism , Hepatocytes/metabolism , Hepatocytes/drug effects
8.
Cell Death Dis ; 15(9): 659, 2024 Sep 09.
Article in English | MEDLINE | ID: mdl-39245717

ABSTRACT

Cell death is a fundamental process in health and disease. Emerging research shows the existence of numerous distinct cell death modalities with similar and intertwined signaling pathways, but resulting in different cellular outcomes, raising the need to understand the decision-making steps during cell death signaling. Paracetamol (Acetaminophen, APAP)-induced hepatocyte death includes several apoptotic processes but eventually is executed by oncotic necrosis without any caspase activation. Here, we studied this paradoxical form of cell death and revealed that APAP not only fails to activate caspases but also strongly impedes their activation upon classical apoptosis induction, thereby shifting apoptosis to necrosis. While APAP intoxication results in massive drop in mitochondrial respiration, low cellular ATP levels could be excluded as an underlying cause of missing apoptosome formation and caspase activation. In contrast, we identified oxidative stress as a key factor in APAP-induced caspase inhibition. Importantly, caspase inhibition and the associated switch from apoptotic to necrotic cell death was reversible through the administration of antioxidants. Thus, exemplified by APAP-induced cell death, our study stresses that cellular redox status is a critical component in the decision-making between apoptotic and necrotic cell death, as it directly affects caspase activity.


Subject(s)
Acetaminophen , Apoptosis , Caspases , Hepatocytes , Oxidative Stress , Oxidative Stress/drug effects , Apoptosis/drug effects , Hepatocytes/drug effects , Hepatocytes/metabolism , Acetaminophen/pharmacology , Caspases/metabolism , Animals , Humans , Necrosis , Mice , Enzyme Activation/drug effects , Mitochondria/drug effects , Mitochondria/metabolism , Antioxidants/pharmacology , Antioxidants/metabolism , Adenosine Triphosphate/metabolism , Male , Signal Transduction/drug effects
9.
Stem Cell Res Ther ; 15(1): 278, 2024 Sep 04.
Article in English | MEDLINE | ID: mdl-39227908

ABSTRACT

BACKGROUND: The immunomodulatory oligodeoxynucleotide (ODN) IMT504 might harbor antifibrotic properties within the liver. METHODS: Fibrosis models were induced in mice through thioacetamide (TAA) administration and bile-duct ligation. Cre-loxP mice were utilized to identify GLAST + Wnt1 + bone marrow stromal progenitors (BMSPs) and to examine their contribution with cells in the liver. In vivo and in vitro assays; flow-cytometry, immunohistochemistry, and qPCR were conducted. RESULTS: IMT504 demonstrated significant inhibition of liver fibrogenesis progression and reversal of established fibrosis. Early responses to IMT504 involved the suppression of profibrogenic and proinflammatory markers, coupled with an augmentation of hepatocyte proliferation. Additionally, this ODN stimulated the proliferation and mobilization of GLAST + Wnt1 + BMSPs, likely amplifying their contribution with endothelial- and hepatocytes-like cells. Moreover, IMT504 significantly modulated the expression levels of Wnt ligands and signaling pathway/target genes specifically within GLAST + Wnt1 + BMSPs, with minimal impact on other BMSPs. Intriguingly, both IMT504 and conditioned media from IMT504-pre-treated GLAST + Wnt1 + BMSPs shifted the phenotype of fibrotic macrophages, hepatic stellate cells, and hepatocytes, consistent with the potent antifibrotic effects observed. CONCLUSION: In summary, our findings identify IMT504 as a promising candidate molecule with potent antifibrotic properties, operating through both direct and indirect mechanisms, including the activation of GLAST + Wnt1 + BMSPs.


Subject(s)
Liver Cirrhosis , Mesenchymal Stem Cells , Wnt1 Protein , Animals , Mice , Liver Cirrhosis/pathology , Liver Cirrhosis/drug therapy , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/cytology , Wnt1 Protein/metabolism , Wnt1 Protein/genetics , Liver/drug effects , Liver/pathology , Liver/metabolism , Oligodeoxyribonucleotides/pharmacology , Male , Mice, Inbred C57BL , Hepatocytes/metabolism , Hepatocytes/drug effects , Thioacetamide
10.
Cell Commun Signal ; 22(1): 431, 2024 Sep 06.
Article in English | MEDLINE | ID: mdl-39243059

ABSTRACT

Recently we have shown that protein disulfide isomerase (PDI or PDIA1) is involved in mediating chemically-induced, glutathione (GSH) depletion-associated ferroptotic cell death through NOS activation (dimerization) and NO accumulation. The present study aims to determine the role of PDI in mediating chemically-induced hepatocyte injury in vitro and in vivo and whether PDI inhibitors can effectively protect against chemically-induced hepatocyte injury. We show that during the development of erastin-induced ferroptotic cell death, accumulation of cellular NO, ROS and lipid-ROS follows a sequential order, i.e., cellular NO accumulation first, followed by accumulation of cellular ROS, and lastly cellular lipid-ROS. Cellular NO, ROS and lipid-ROS each play a crucial role in mediating erastin-induced ferroptosis in cultured hepatocytes. In addition, it is shown that PDI is an important upstream mediator of erastin-induced ferroptosis through PDI-mediated conversion of NOS monomer to its dimer, which then leads to accumulation of cellular NO, ROS and lipid-ROS, and ultimately ferroptotic cell death. Genetic manipulation of PDI expression or pharmacological inhibition of PDI function each can effectively abrogate erastin-induced ferroptosis. Lastly, evidence is presented to show that PDI is also involved in mediating acetaminophen-induced liver injury in vivo using both wild-type C57BL/6J mice and hepatocyte-specific PDI conditional knockout (PDIfl/fl Alb-cre) mice. Together, our work demonstrates that PDI is an important upstream mediator of chemically-induced, GSH depletion-associated hepatocyte ferroptosis, and inhibition of PDI can effectively prevent this injury.


Subject(s)
Glutathione , Hepatocytes , Protein Disulfide-Isomerases , Reactive Oxygen Species , Protein Disulfide-Isomerases/metabolism , Protein Disulfide-Isomerases/genetics , Hepatocytes/metabolism , Hepatocytes/drug effects , Hepatocytes/pathology , Animals , Glutathione/metabolism , Reactive Oxygen Species/metabolism , Mice , Mice, Inbred C57BL , Piperazines/pharmacology , Ferroptosis/drug effects , Nitric Oxide/metabolism , Male , Humans
11.
Cell Mol Biol Lett ; 29(1): 119, 2024 Sep 07.
Article in English | MEDLINE | ID: mdl-39244559

ABSTRACT

BACKGROUND: Drug-induced liver injury (DILI) is gradually becoming a common global problem that causes acute liver failure, especially in acute hepatic damage caused by acetaminophen (APAP). Paeoniflorin (PF) has a wide range of therapeutic effects to alleviate a variety of hepatic diseases. However, the relationship between them is still poorly investigated in current studies. PURPOSE: This work aimed to explore the protective effects of PF on APAP-induced hepatic damage and researched the potential molecular mechanisms. METHODS: C57BL/6J male mice were injected with APAP to establish DILI model and were given PF for five consecutive days for treatment. Aiming to clarify the pharmacological effects, the molecular mechanisms of PF in APAP-induced DILI was elucidated by high-throughput and other techniques. RESULTS: The results demonstrated that serum levels of ALP, γ-GT, AST, TBIL, and ALT were decreased in APAP mice by the preventive effects of PF. Moreover, PF notably alleviated hepatic tissue inflammation and edema. Meanwhile, the results of TUNEL staining and related apoptotic factors coincided with the results of transcriptomics, suggesting that PF inhibited hepatocyte apoptosis by regulated MAPK signaling. Besides, PF also acted on reactive oxygen species (ROS) to regulate the oxidative stress for recovery the damaged mitochondria. More importantly, transmission electron microscopy showed the generation of autophagosomes after PF treatment, and PF was also downregulated mTOR and upregulated the expression of autophagy markers such as ATG5, ATG7, and BECN1 at the mRNA level and LC3, p62, ATG5, and ATG7 at the protein level, implying that the process by which PF exerted its effects was accompanied by the occurrence of autophagy. In addition, combinined with molecular dynamics simulations and western blotting of MAPK, the results suggested p38 as a direct target for PF on APAP. Specifically, PF-activated autophagy through the downregulation of MAPK/mTOR signaling, which in turn reduced APAP injury. CONCLUSIONS: Paeoniflorin mitigated liver injury by activating autophagy to suppress oxidative stress and apoptosis via the MAPK/mTOR signaling pathway. Taken together, our findings elucidate the role and mechanism of paeoniflorin in DILI, which is expected to provide a new therapeutic strategy for the development of paeoniflorin.


Subject(s)
Acetaminophen , Autophagy , Chemical and Drug Induced Liver Injury , Glucosides , Hepatocytes , Mice, Inbred C57BL , Monoterpenes , TOR Serine-Threonine Kinases , Animals , Autophagy/drug effects , Glucosides/pharmacology , TOR Serine-Threonine Kinases/metabolism , Monoterpenes/pharmacology , Male , Hepatocytes/metabolism , Hepatocytes/drug effects , Mice , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/drug therapy , Chemical and Drug Induced Liver Injury/prevention & control , Acetaminophen/adverse effects , Signal Transduction/drug effects , Apoptosis/drug effects , MAP Kinase Signaling System/drug effects , Protective Agents/pharmacology , Reactive Oxygen Species/metabolism , Oxidative Stress/drug effects
12.
Int J Nanomedicine ; 19: 9009-9033, 2024.
Article in English | MEDLINE | ID: mdl-39246425

ABSTRACT

Background: The high infectivity of coronaviruses has led to increased interest in developing new strategies to prevent virus spread. Silver nanoparticles (AgNPs) and graphene oxide (GO) have attracted much attention in the antiviral field. We investigated the potential antiviral activity of GO and AgNPs combined in the nanocomposite GO-Ag against murine betacoronavirus MHV using an in vitro model. Methods: GO, AgNPs, and GO-Ag characterization (size distribution, zeta potential, TEM visualization, FT-IR, and EDX analysis) and XTT assay were performed. The antiviral activity of GO-Ag nanocomposites was evaluated by RT-qPCR and TCID50 assays. The results were compared with free AgNPs and pure GO. Cell growth and morphology of MHV-infected hepatocytes treated with GO-Ag composites were analyzed by JuLI™Br. Immunofluorescence was used to visualize the cell receptor used by MHV. Ultrastructural SEM analysis was performed to examine cell morphology after MHV infection and GO-Ag composite treatment. Results: A significant reduction in virus titer was observed for all nanocomposites tested, ranging from 3.2 to 7.3 log10 TCID50. The highest titer reduction was obtained for GO 5 µg/mL - Ag 25 µg/mL in the post-treatment method. These results were confirmed by RT-qPCR analysis. The results indicate that GO-Ag nanocomposites exhibited better antiviral activity compared to AgNPs and GO. Moreover, the attachment of AgNPs to the GO flake platform reduced their cytotoxicity. In addition, the GO-Ag composite modulates the distribution of the Ceacam1 cell receptor and can modulate cell morphology. Conclusion: Graphene oxide sheets act as a stabilizing agent, inhibiting the accumulation of AgNPs and reducing their cellular toxicity. The GO-Ag composite can physically bind and inhibit murine betacoronavirus from entering cells. Furthermore, the constant presence of GO-Ag can inhibit MHV replication and significantly limit its extracellular release. In conclusion, GO-Ag shows promise as an antiviral coating on solid surfaces to minimize virus transmission and spread.


Subject(s)
Antiviral Agents , Graphite , Metal Nanoparticles , Nanocomposites , Silver , Graphite/pharmacology , Graphite/chemistry , Silver/chemistry , Silver/pharmacology , Animals , Nanocomposites/chemistry , Antiviral Agents/pharmacology , Antiviral Agents/chemistry , Mice , Metal Nanoparticles/chemistry , Murine hepatitis virus/drug effects , Hepatocytes/drug effects , Hepatocytes/virology , Coronavirus Infections/drug therapy , Coronavirus Infections/virology , Cell Line
13.
Redox Rep ; 29(1): 2395779, 2024 Dec.
Article in English | MEDLINE | ID: mdl-39221774

ABSTRACT

OBJECTIVES: Alcohol and its metabolites, such as acetaldehyde, induced hepatic mitochondrial dysfunction play a pathological role in the development of alcohol-related liver disease (ALD). METHODS: In this study, we investigated the potential of nobiletin (NOB), a polymethoxylated flavone, to counter alcohol-induced mitochondrial dysfunction and liver injury. RESULTS: Our findings demonstrate that NOB administration markedly attenuated alcohol-induced hepatic steatosis, endoplasmic reticulum stress, inflammation, and tissue damage in mice. NOB reversed hepatic mitochondrial dysfunction and oxidative stress in both alcohol-fed mice and acetaldehyde-treated hepatocytes. Mechanistically, NOB restored the reduction of hepatic mitochondrial transcription factor A (TFAM) at both mRNA and protein levels. Notably, the protective effects of NOB against acetaldehyde-induced mitochondrial dysfunction and cell death were abolished in hepatocytes lacking Tfam. Furthermore, NOB administration reinstated the levels of hepatocellular NRF1, a key transcriptional regulator of TFAM, which were decreased by alcohol and acetaldehyde exposure. Consistent with these findings, hepatocyte-specific overexpression of Nrf1 protected against alcohol-induced hepatic Tfam reduction, mitochondrial dysfunction, oxidative stress, and liver injury. CONCLUSIONS: Our study elucidates the involvement of the NRF1-TFAM signaling pathway in the protective mechanism of NOB against chronic-plus-binge alcohol consumption-induced mitochondrial dysfunction and liver injury, suggesting NOB supplementation as a potential therapeutic strategy for ALD.


Subject(s)
Flavones , Signal Transduction , Animals , Mice , Flavones/pharmacology , Signal Transduction/drug effects , Male , Transcription Factors/metabolism , Transcription Factors/genetics , Oxidative Stress/drug effects , Mice, Inbred C57BL , Liver/drug effects , Liver/metabolism , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/genetics , Ethanol/toxicity , Ethanol/adverse effects , Mitochondria/drug effects , Mitochondria/metabolism , Liver Diseases, Alcoholic/metabolism , Liver Diseases, Alcoholic/drug therapy , Liver Diseases, Alcoholic/prevention & control , Liver Diseases, Alcoholic/pathology , Hepatocytes/drug effects , Hepatocytes/metabolism , Mitochondrial Proteins/metabolism , Mitochondrial Proteins/genetics , Nuclear Respiratory Factor 1/metabolism , Nuclear Respiratory Factor 1/genetics , Protective Agents/pharmacology , NF-E2-Related Factor 1/metabolism , NF-E2-Related Factor 1/genetics , High Mobility Group Proteins
14.
Cell Signal ; 123: 111379, 2024 Nov.
Article in English | MEDLINE | ID: mdl-39233207

ABSTRACT

BACKGROUND AND AIMS: Acetaminophen (APAP) is the main cause of acute liver injury (ALI) in the Western. Our previous study has shown that fenofibrate activated hepatic expression of fibroblast growth factor 21 (FGF21) can protect the liver form APAP injuries by promoting autophagy. However, the underlying mechanism involved in FGF21-mediated autophagy remains unsolved. METHODS: The ALI mice model was established by intraperitoneal injection of APAP. To investigate the influence of FGF21 on autophagy and Sirt1 expression in APAP-induced ALI, FGF21 knockout (FGF21KO) mice and exogenously supplemented mouse recombinant FGF21 protein were used. In addition, primary isolated hepatocytes and the Sirt1 inhibitor EX527 were used to observe whether FGF21 activated autophagy in APAP injury is regulated by Sirt1 at the cellular level. RESULTS: FGF21, Sirt1, and autophagy levels increased in mice with acute liver injury (ALI) and in primary cultured hepatocytes. Deletion of the FGF21 gene exacerbated APAP-induced liver necrosis and oxidative stress, and decreased mitochondrial potential. It also reduced the mRNA and protein levels of autophagy-related proteins such as Sirt1, LC3-II, and p62, as well as the number of autophagosomes. Replenishment of FGF21 reversed these processes. In addition, EX527 partially counteracted the protective effect of FGF21 by worsening oxidative damage, mitochondrial damage, and reducing autophagy in primary liver cells treated with APAP. CONCLUSION: FGF21 increases autophagy by upregulating Sirt1 to alleviate APAP-induced injuries.


Subject(s)
Acetaminophen , Autophagy , Chemical and Drug Induced Liver Injury , Fibroblast Growth Factors , Hepatocytes , Mice, Inbred C57BL , Sirtuin 1 , Animals , Acetaminophen/adverse effects , Sirtuin 1/metabolism , Sirtuin 1/genetics , Autophagy/drug effects , Fibroblast Growth Factors/metabolism , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/pathology , Mice , Hepatocytes/metabolism , Hepatocytes/drug effects , Male , Mice, Knockout , Oxidative Stress/drug effects , Liver/metabolism , Liver/pathology , Liver/drug effects
15.
Nat Commun ; 15(1): 7991, 2024 Sep 12.
Article in English | MEDLINE | ID: mdl-39266553

ABSTRACT

Interactions between adipose tissue, liver and immune system are at the center of metabolic dysfunction-associated steatotic liver disease and type 2 diabetes. To address the need for an accurate in vitro model, we establish an interconnected microphysiological system (MPS) containing white adipocytes, hepatocytes and proinflammatory macrophages derived from isogenic human induced pluripotent stem cells. Using this MPS, we find that increasing the adipocyte-to-hepatocyte ratio moderately affects hepatocyte function, whereas macrophage-induced adipocyte inflammation causes lipid accumulation in hepatocytes and MPS-wide insulin resistance, corresponding to initiation of metabolic dysfunction-associated steatotic liver disease. We also use our MPS to identify and characterize pharmacological intervention strategies for hepatic steatosis and systemic insulin resistance and find that the glucagon-like peptide-1 receptor agonist semaglutide improves hepatocyte function by acting specifically on adipocytes. These results establish our MPS modeling the adipose tissue-liver axis as an alternative to animal models for mechanistic studies or drug discovery in metabolic diseases.


Subject(s)
Hepatocytes , Induced Pluripotent Stem Cells , Inflammation , Insulin Resistance , Liver , Humans , Induced Pluripotent Stem Cells/metabolism , Hepatocytes/metabolism , Hepatocytes/drug effects , Liver/metabolism , Liver/pathology , Inflammation/metabolism , Inflammation/pathology , Adipocytes/metabolism , Macrophages/metabolism , Macrophages/drug effects , Fatty Liver/metabolism , Fatty Liver/pathology , Glucagon-Like Peptide-1 Receptor/metabolism , Glucagon-Like Peptide-1 Receptor/agonists , Glucagon-Like Peptide-1 Receptor/genetics , Adipose Tissue/metabolism , Microphysiological Systems
16.
PLoS One ; 19(9): e0309976, 2024.
Article in English | MEDLINE | ID: mdl-39236049

ABSTRACT

Sarmentosin (SA) and Quercetin (QC) are two active components of Sedum Sarmentosum Bunge, which is a traditional Chinese herbal medicine. This study aimed to investigate the role and regulatory mechanism of SA and QC in fatty liver of Genetic Improvement of Farmed Tilapia (GIFT) tilapia. GIFT tilapia were randomly divided into two groups with three replicates per treatment (30 fish in each replicate): normal diet group (average weight 3.51±0.31 g) and high-fat diet group (average weight 3.44±0.09 g). After 8 weeks feeding trial, growth index, lipid deposition, and biochemical indexes were measured. Lipid deposition, and lipid and inflammation-related gene expression were detected in a primary hepatocyte model of fatty liver of GIFT tilapia treated with SA or QC. Our results showed that high-fat diet caused lipid deposition and peroxidative damage in the liver of GIFT tilapia. The cell counting kit-8 assay results indicated that 10 µM SA and 10 µM of QC both had the least effect on hepatocyte proliferation. Moreover, both 10 µM of SA and 10 µM of QC showed lipolytic effects and inhibited the expression of lipid-related genes (FAS, Leptin, SREBP-1c, and SREBP2) in fatty liver cells. Interestingly, QC induced autophagosome-like subcellular structure and increased the expression of IL-8 in fatty liver cells. In conclusion, this study confirmed that SA and QC improved fatty liver caused by high-fat diet, providing a novel therapeutic approach for fatty liver of GIFT tilapia.


Subject(s)
Fatty Liver , Hepatocytes , Lipid Metabolism , Quercetin , Animals , Hepatocytes/metabolism , Hepatocytes/drug effects , Quercetin/pharmacology , Lipid Metabolism/drug effects , Fatty Liver/metabolism , Fatty Liver/drug therapy , Fatty Liver/pathology , Cichlids/metabolism , Diet, High-Fat/adverse effects , Liver/metabolism , Liver/drug effects , Liver/pathology , Tilapia/metabolism , Fish Diseases/metabolism , Fish Diseases/drug therapy , Cell Proliferation/drug effects
17.
PLoS One ; 19(9): e0310524, 2024.
Article in English | MEDLINE | ID: mdl-39298444

ABSTRACT

BACKGROUND: Alpha-1 antitrypsin deficiency (AATD) is an inherited disease, the common variant caused by a Pi*Z mutation in the SERPINA1 gene. Pi*Z AAT increases the risk of pulmonary emphysema and liver disease. Berberine (BBR) is a nature dietary supplement and herbal remedy. Emerging evidence revealed that BBR has remarkable liver-protective properties against various liver diseases. In the present study, we investigated the therapeutic effects and toxicities of BBR in Pi*Z hepatocytes and Pi*Z transgenic mice. METHODS: Huh7.5 and Huh7.5Z (which carries the Pi*Z mutation) cells were treated with different concentrations of BBR for 48 hours. MTT was performed for cell viability assay. Intracellular AAT levels were evaluated by western blot. In vivo studies were carried out in wild type, native phenotype AAT (Pi*M), and Pi*Z AAT transgenic mice. Mice were treated with 50 mg/kg/day of BBR or solvent only by oral administration for 30 days. Western blot and liver histopathological examinations were performed to evaluate therapeutic benefits and liver toxicity of BBR. RESULTS: BBR reduced intracellular AAT levels in Huh7.5Z cells, meanwhile, no Pi*Z-specific toxicity was observed. However, BBR did not reduce liver AAT load but significantly potentiated liver inflammation and fibrosis accompanying the activation of unfolded protein response and mTOR in Pi*Z mice, but not in wild type and Pi*M mice. CONCLUSIONS: BBR exacerbated liver inflammation and fibrosis specifically in Pi*Z mice. This adverse effect may be associated with the activation of unfolded protein response and mTOR. This study implicates that BBR should be avoided by AATD patients.


Subject(s)
Berberine , Liver Cirrhosis , Mice, Transgenic , alpha 1-Antitrypsin , Animals , Berberine/pharmacology , Mice , alpha 1-Antitrypsin/genetics , alpha 1-Antitrypsin/metabolism , Humans , Liver Cirrhosis/metabolism , Liver Cirrhosis/pathology , Liver Cirrhosis/drug therapy , Liver Cirrhosis/chemically induced , Disease Models, Animal , TOR Serine-Threonine Kinases/metabolism , Liver/drug effects , Liver/pathology , Liver/metabolism , Hepatocytes/drug effects , Hepatocytes/metabolism , Hepatocytes/pathology , Hepatitis/pathology , Hepatitis/metabolism , Hepatitis/drug therapy , Hepatitis/etiology , Unfolded Protein Response/drug effects
18.
Curr Protoc ; 4(9): e70015, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39283005

ABSTRACT

Primary human hepatocytes (PHHs) are recognized as the "gold standard" for evaluating toxicity of various drugs or chemicals in vitro. However, due to their limited availability, primary hepatocytes isolated from rodents are more commonly used in various experimental studies than PHHs. However, bigger differences in drug metabolism were seen between humans and rats compared to those between human and non-human primates. Here, we describe a method to isolate primary hepatocytes from the liver of rhesus macaques (Macaca mulatta, a species of Old-World monkey) after in situ whole liver perfusion. Techniques for cryopreserving and recovering primary macaque hepatocytes (PMHs) are also described. Given the remarkable physiological and genetic similarity of non-human primates to humans, PMHs isolated using this protocol may serve as a reliable surrogate of PHHs in toxicological research and preclinical studies. Published 2024. This article is a U.S. Government work and is in the public domain in the USA. Basic Protocol 1: In situ whole liver perfusion Basic Protocol 2: Primary macaque hepatocyte isolation and cell plating Basic Protocol 3: Cryopreservation and recovery of primary macaque hepatocytes.


Subject(s)
Cryopreservation , Hepatocytes , Macaca mulatta , Animals , Hepatocytes/cytology , Hepatocytes/drug effects , Cryopreservation/methods , Cell Separation/methods , Liver/cytology , Perfusion/methods , Cells, Cultured
19.
Int J Biol Macromol ; 278(Pt 2): 134717, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39142477

ABSTRACT

Liver sinusoidal endothelial cells (LSECs) are key targets for addressing metabolic dysfunction-associated steatotic liver disease (MASLD). However, isolating and culturing primary LSECs is challenging due to rapid dedifferentiation, resulting in loss of function. The extracellular matrix (ECM) likely plays a crucial role in maintaining the fate and function of LSECs. In this study, we explored the influence of liver-ECM (L-ECM) on liver cells and developed culture conditions that maintain the differentiated function of liver cells in vitro for prolonged periods. Porcine liver-derived L-ECM, containing 34.9 % protein, 0.045 % glycosaminoglycans, and negligible residual DNA (41.2 ng/mg), was utilized to culture primary rat liver cells in generated hydrogels. Proteomic analyses and molecular weight distribution of proteins of solubilized L-ECM revealed the typical diverse ECM core matrisome, with abundant collagens. L-ECM hydrogels showed suitable stiffness and stress relaxation properties. Furthermore, we demonstrated that collagen-rich L-ECM hydrogels enhanced LSECs' and hepatocytes' viability, and reduced the dedifferentiation rate of LSECs. In addition, hepatocyte function was maintained longer by culture on L-ECM hydrogels compared to traditional culturing. These beneficial effects are likely attributed to the bioactive macromolecules including collagens, and mechanical and microarchitectural properties of the L-ECM hydrogels.


Subject(s)
Cell Survival , Collagen , Endothelial Cells , Extracellular Matrix , Hepatocytes , Hydrogels , Liver , Animals , Hydrogels/chemistry , Hydrogels/pharmacology , Hepatocytes/metabolism , Hepatocytes/drug effects , Hepatocytes/cytology , Extracellular Matrix/metabolism , Extracellular Matrix/chemistry , Rats , Endothelial Cells/metabolism , Endothelial Cells/cytology , Endothelial Cells/drug effects , Cell Survival/drug effects , Collagen/chemistry , Collagen/pharmacology , Collagen/metabolism , Liver/metabolism , Liver/cytology , Swine , Cells, Cultured , Male
20.
Food Chem Toxicol ; 192: 114935, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39151875

ABSTRACT

Sodium nitrite (SN), a prevalent food preservative, is known to precipitate hepatotoxicity upon exposure. This study elucidates the hepatoprotective effects of corn oligopeptide (COP) and vitamin E (VE) against SN-induced hepatic injury in canine hepatocytes. Canine liver cells were subjected to SN to induce hepatotoxicity, followed by treatment with COP and VE. Evaluations included assays for cell viability, oxidative stress markers, apoptosis, and inflammatory cytokines. Additionally, transcriptomic and metabolomic analyses were performed to delineate the underlying molecular mechanisms. The findings demonstrated that COP and VE significantly ameliorated SN-induced cytotoxicity, oxidative stress, and apoptosis. It was evidenced by restored cell viability, enhanced antioxidant enzyme activity, reduced cytoplasmic enzyme leakage, and decreased levels of malondialdehyde and inflammatory cytokines, with COP showing superior efficacy. The RNA sequencing revealed that COP treatment suppressed the SN-activated aminoacyl-tRNA biosynthesis pathway and TGF-ß/NF-κB signaling pathways, thereby mitigating amino acid depletion, apoptosis, and inflammation. Moreover, COP treatment upregulated genes associated with protein folding, bile acid synthesis, and DNA repair. Metabolomic analysis corroborated these results, showing that COP restored amino acid levels and enhanced bile acid metabolism, alleviating SN-induced metabolic disruptions. These findings offered significant insights into the protective mechanisms of COP underscoring its prospective application in treating liver injuries.


Subject(s)
Hepatocytes , NF-kappa B , Oligopeptides , Signal Transduction , Sodium Nitrite , Zea mays , Animals , Dogs , Hepatocytes/drug effects , Hepatocytes/metabolism , Signal Transduction/drug effects , Oligopeptides/pharmacology , NF-kappa B/metabolism , NF-kappa B/genetics , Sodium Nitrite/toxicity , RNA, Transfer, Amino Acyl/metabolism , Apoptosis/drug effects , Transforming Growth Factor beta/metabolism , Oxidative Stress/drug effects , Chemical and Drug Induced Liver Injury/metabolism , Cell Survival/drug effects , Metabolic Reprogramming
SELECTION OF CITATIONS
SEARCH DETAIL