Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 188
Filtrer
1.
Biomed Mater ; 2024 Aug 09.
Article de Anglais | MEDLINE | ID: mdl-39121886

RÉSUMÉ

Porous poly (lactic-co-glycolic acid)/ß-tricalcium phosphate/Icaritin (PLGA/ß-TCP/ICT, PTI) scaffold is a tissue engineering scaffold based on PLGA/ß-TCP containing Icaritin, the main active ingredient of the Chinese medicine Epimedium. Due to its excellent mechanical properties and osteogenic effect, PTI scaffold has the potential to promote bone defect repair. However, the release of ICT from the scaffolds is difficult to control. In this study, we constructed Ti3C2Tx@PLGA/ICT microspheres (TIM) and evaluated their characterization as well as ICT release under near-infrared (NIR) irradiation. We utilized TIM to modify the PT scaffold and performed biological experiments. First, we cultured rat bone marrow mesenchymal stem cells on the scaffold to assess biocompatibility and osteogenic potential under on-demand NIR irradiation. Subsequently, to evaluate the osteogenic properties of TIM-modified scaffold in vivo, the scaffold was implanted into a femoral condyle defect model. TIM have excellent drug-loading capacity and encapsulation efficiency for ICT, and the incorporation of Ti3C2Tx endows TIM with photothermal conversion capability. Under 0.90 W cm-2 NIR irradiation, the temperature of TIM maintained at 42.0 ± 0.5°C and the release of ICT was accelerated. Furthermore, while retaining its original properties, the TIM-modified scaffold was biocompatible and could promote cell proliferation, osteogenic differentiation, and biomineralization in vitro, as well as the osteogenesis and osseointegration in vivo, and its effect was further enhanced through the modulation of ICT release under NIR irradiation. In summary, TIM-modified scaffold has the potential to be applied in bone defects repairing.

2.
Biomed Chromatogr ; : e5976, 2024 Aug 10.
Article de Anglais | MEDLINE | ID: mdl-39126342

RÉSUMÉ

A sensitive, rapid, and simple HPLC-MS/MS method was first developed and fully validated to determine the icaritin (ICT) and its novel 3-methylcarbamate prodrug (3N) simultaneously in rat plasma. Analytes were extracted from rat plasma using a liquid-liquid extraction (LLE) method. Chromatographic separation was performed on ACE Excel 2 C18-Amide column. Quantitation of analytes was conducted on an LCMS-8060 triple-quadrupole tandem mass spectrometer. The quantitation mode was the multiple reaction monitoring via positive electrospray ionization. The calibration curve was linear over the concentration range of 1 to 200 ng/ml for ICT with a correlation coefficient of r = 0.9950 and 1 to 400 ng/ml for 3N with a correlation coefficient of r = 0.9956. The intra-precision RSDs were ≤12% for ICT and 3N. The inter-day precision RSDs were ≤10% for ICT and 3N. The accuracy RE was between -2.6% and 7.8% for ICT and 3N. The average ICT, 3N and IS recoveries were 87.9%, 83.6%, and 84.3%. The plasma matrix of ICT and 3N complied with the guidelines. ICT and 3N were stable in rat plasma under various tested conditions. This work has been successfully applied to studying the pharmacokinetics of ICT and 3N.

3.
Front Pharmacol ; 15: 1437515, 2024.
Article de Anglais | MEDLINE | ID: mdl-39055490

RÉSUMÉ

Introduction: Icaritin (ICT), a promising anti-hepatocellular carcinoma (HCC) prenylated flavonoid, is hindered from being applied due to its low water solubility and high lipophilicity in poorly differentiated HCC which is associated with upregulation of CD44 isoforms. Thus, hyaluronic acid (HA), a natural polysaccharide with high binding ability to CD44 receptors, was used to formulate a modified liposome as a novel targeted ICT-delivery system for HCC treatment. Methods: The ICT-Liposomes (Lip-ICT) with and without HA were prepared by a combined method of thin-film dispersion and post-insertion. The particle size, polydispersity (PDI), zeta potential, encapsulation efficacy (%EE), drug loading content (%DLC), and in vitro drug release profiles were investigated for physicochemical properties, whereas MTT assay was used to assess cytotoxic effects on HCC cells, HepG2, and Huh7 cells. Tumor bearing nude mice were used to evaluate the inhibitory effect of HA-Lip-ICT and Lip-ICT in vivo. Results: Lip-ICT and HA-Lip-ICT had an average particle size of 171.2 ± 1.2 nm and 208.0 ± 3.2 nm, with a zeta potential of -13.9 ± 0.83 and -24.8 ± 0.36, respectively. The PDI resulted from Lip-ICT and HA-Lip-ICT was 0.28 ± 0.02 and 0.26 ± 0.02, respectively. HA-Lip-ICT demonstrated higher in vitro drug release when pH was dropped from 7.4 to 5.5, The 12-h release rate of ICT from liposomes increased from 30% at pH7.4 to more than 60% at pH5.5. HA-Lip-ICT displayed higher toxicity than Lip-ICT in both HCC cells, especially Huh7with an IC50 of 34.15 ± 2.11 µM. The in vivo tissue distribution and anti-tumor experiments carried on tumor bearing nude mice indicated that HA-Lip- ICT exhibited higher tumor accumulation and achieved a tumor growth inhibition rate of 63.4%. Discussion: The nano-sized Lip-ICT was able to prolong the drug release time and showed long-term killing HCC cells ability. Following conjugation with HA, HA-Lip-ICT exhibited higher cytotoxicity, stronger tumor targeting, and tumor suppression abilities than Lip-ICT attributed to HA-CD44 ligand-receptor interaction, increasing the potential of ICT to treat HCC.

4.
ACS Appl Mater Interfaces ; 16(29): 37581-37595, 2024 Jul 24.
Article de Anglais | MEDLINE | ID: mdl-38985579

RÉSUMÉ

The effective repair of bone defects has long been a major challenge in clinical practice. Currently, research efforts mostly focus on achieving sufficiently good bone repair, with little attention paid to achieving both good and fast repair. However, achieving highly efficient (H-efficient) bone repair, which is both good and fast, can shorten the treatment cycle and facilitate rapid patient recovery. Therefore, the development of a H-efficient bone repair material is of significant importance. This study incorporated the previously developed osteoinductive photothermal agent (PTA) BPICT into printing paste to prepare a near-infrared (NIR)-responsive BPICT scaffold. Subsequently, the effects of photothermal therapy (PTT) on bone repair and drug release were assessed in vitro. To further validate the H-efficient bone repair properties of the BPICT scaffold, the scaffold was implanted into bone defects and its ability to promote bone repair in vivo was evaluated through radiology and histopathological analysis. The results indicated that compared to scaffolds containing only Icaritin (ICT), the BPICT scaffold can achieve PTT to promote bone repair through NIR irradiation, while also enabling the controlled release of ICT from the scaffold to enhance bone repair. Within the same observation period, the BPICT scaffold achieves more efficient bone repair than the ICT scaffold, significantly shortening the bone repair cycle while ensuring the effectiveness of bone repair. Therefore, the NIR-responsive scaffold based on PTT-mediated controlled release of bone growth factors represents a feasible solution for promoting H-efficient bone repair in the area of bone defects.


Sujet(s)
Régénération osseuse , Rayons infrarouges , Structures d'échafaudage tissulaires , Structures d'échafaudage tissulaires/composition chimique , Animaux , Régénération osseuse/effets des médicaments et des substances chimiques , Thérapie photothermique , Souris , Ostéogenèse/effets des médicaments et des substances chimiques , Humains
5.
World J Clin Cases ; 12(16): 2847-2855, 2024 Jun 06.
Article de Anglais | MEDLINE | ID: mdl-38899296

RÉSUMÉ

BACKGROUND: The prognosis of hepatocellular carcinoma (HCC) combined with portal and hepatic vein cancerous thrombosis is poor, for unresectable patients the combination of targeted therapy and immune therapy was the first-line recommended treatment for advanced HCC, with a median survival time of only about 2.7-6 months. In this case report, we present the case of a patient with portal and hepatic vein cancerous thrombosis who achieved pathologic complete response after conversion therapy. CASE SUMMARY: In our center, a patient with giant HCC combined with portal vein tumor thrombus and hepatic vein tumor thrombus was treated with transcatheter arterial chemoembolization (TACE), radiotherapy, targeted therapy and immunotherapy, and was continuously given icaritin soft capsules for oral regulation. After 7 months of conversion therapy, the patient's tumor shrank and the tumor thrombus subsided significantly. The pathology of surgical resection was in complete remission, and there was no progression in the postoperative follow-up for 7 months, which provided a basis for the future strategy of combined conversion therapy. CONCLUSION: In this case, atezolizumab, bevacizumab, icaritin soft capsules combined with radiotherapy and TACE had a good effect. For patients with hepatocellular carcinoma combined with hepatic vein/inferior vena cava tumor thrombus, adopting a high-intensity, multimodal proactive strategy under the guidance of multidisciplinary team (MDT) is an important attempt to break through the current treatment dilemma.

6.
Biopharm Drug Dispos ; 45(3): 149-158, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38886878

RÉSUMÉ

Icaritin is a prenylflavonoid derivative of the genus Epimedium (Berberidaceae) and has a variety of pharmacological actions. Icaritin is approved by the National Medical Products Administration as an anticancer drug that exhibits efficacy and safety advantages in patients with hepatocellular carcinoma cells. This study aimed to evaluate the inhibitory effects of icaritin on UDP-glucuronosyltransferase (UGT) isoforms. 4-Methylumbelliferone (4-MU) was employed as a probe drug for all the tested UGT isoforms using in vitro human liver microsomes (HLM). The inhibition potentials of UGT1A1 and 1A9 in HLM were further tested by employing 17ß-estradiol (E2) and propofol (PRO) as probe substrates, respectively. The results showed that icaritin inhibits UGT1A1, 1A3, 1A4, 1A7, 1A8, 1A10, 2B7, and 2B15. Furthermore, icaritin exhibited a mixed inhibition of UGT1A1, 1A3, and 1A9, and the inhibition kinetic parameters (Ki) were calculated to be 3.538, 2.117, and 0.306 (µM), respectively. The inhibition of human liver microsomal UGT1A1 and 1A9 both followed mixed mechanism, with Ki values of 2.694 and 1.431 (µM). This study provides supporting information for understanding the drug-drug interaction (DDI) potential of the flavonoid icaritin and other UGT-metabolized drugs in clinical settings. In addition, the findings provide safety evidence for DDI when liver cancer patients receive a combination therapy including icaritin.


Sujet(s)
Interactions médicamenteuses , Flavonoïdes , Glucuronosyltransferase , Microsomes du foie , Glucuronosyltransferase/antagonistes et inhibiteurs , Glucuronosyltransferase/métabolisme , Humains , Flavonoïdes/pharmacologie , Microsomes du foie/métabolisme , Oestradiol/pharmacologie , Hymécromone/pharmacologie , Propofol/pharmacologie , Antienzymes/pharmacologie
7.
J Biotechnol ; 391: 106-116, 2024 Aug 10.
Article de Anglais | MEDLINE | ID: mdl-38871028

RÉSUMÉ

Icaritin, a hydrolysate from total flavonoids of Epimedii (TFE), which has better anti-hepatoma activity than its glycosylated form. In this work, immobilized enzymes 4LP-Tpebgl3@Na-Y and DtRha@ES-107 were used to hydrolyze TFE to prepare icaritin. Five different hydrophobic deep eutectic solvents (HDES) were prepared and the most ideal HDES was successfully selected, which was composed of dodecyl alcohol and thymol with the molar ratio of 2:1. The relative enzyme activity of 4LP-Tpebgl3@Na-Y and DtRha@ES-107 was about 102.4 % and 112.5 %, respectively. In addition, the thermal and binding stability of 4LP-Tpebgl3@Na-Y and DtRha@ES-107 in HDES was not affected negatively. In the biphasic system composed of 50 % (v/v) HDES and Na2HPO4-citric acid buffer (50 mM, pH 5.5), 4LP-Tpebgl3@Na-Y (1.0 U/mL) and TFE (1 g/L) were reacted at 80 °C for 1 h, and then reacted with DtRha@ES-107 (20 U/mL) at 80 °C for 2 h. Finally, TFE was completely converted to 301.8 mg/L icaritin (0.82 mM). After 10 cycles, 4LP-Tpebgl3@Na-Y/DtRha@ES-107 still maintained 84.1 % original activity. In this study, we developed an efficient methodology for icaritin preparation through the integration of enzymatic catalysis and adsorption separation, presenting a viable approach for large-scale, cost-effective production of icaritin.


Sujet(s)
Biotransformation , Enzymes immobilisées , Flavonoïdes , Interactions hydrophobes et hydrophiles , Flavonoïdes/métabolisme , Flavonoïdes/composition chimique , Enzymes immobilisées/métabolisme , Enzymes immobilisées/composition chimique , Solvants eutectiques profonds/composition chimique , Solvants eutectiques profonds/métabolisme , Epimedium/composition chimique , Epimedium/métabolisme , Hydrolyse , Solvants/composition chimique
8.
Int Immunopharmacol ; 138: 112533, 2024 Sep 10.
Article de Anglais | MEDLINE | ID: mdl-38924868

RÉSUMÉ

BACKGROUND: Hepatic ischemia-reperfusion (IR) injury is a major complication of liver transplantation and gravely affects patient prognosis. Icaritin (ICT), the primary plasma metabolite of icariin (ICA), plays a critical role in anti-inflammatory and immunomodulatory processes. However, the role of ICT in hepatic IR injury remains largely undefined. In this study, we aimed to elucidate the role of ICT in hepatic IR injury. METHODS: We established hepatic IR injury models in animals, as well as an oxygen-glucose deprivation/reperfusion (OGD/R) cell model. Liver injury in vivo was assessed by measuring serum alanine aminotransferase (ALT) levels, necrotic areas by liver histology and local hepatic inflammatory responses. For in vitro analyses, we implemented flow-cytometric and western blot analyses, transmission electron microscopy, and an mRFP-GFP-LC3 adenovirus reporter assay to assess the effects of ICT on OGD/R injury in AML12 and THLE-2 cell lines. Signaling pathways were explored in vitro and in vivo to identify possible mechanisms underlying ICT action in hepatic IR injury. RESULTS: Compared to the mouse model group, ICT preconditioning considerably protected the liver against IR stress, and diminished the levels of necrosis/apoptosis and inflammation-related cytokines. In additional studies, ICT treatment dramatically boosted the expression ratios of p-PI3K/PI3K, p-AKT/AKT, and p-mTOR/mTOR proteins in hepatic cells following OGD/R damage. We also applied LY294002 (a PI3K inhibitor) and RAPA (rapamycin, an mTOR inhibitor), which blocked the protective effects of ICT in hepatocytes subjected to OGD/R. CONCLUSION: This study indicates that ICT attenuates ischemia-reperfusion injury by exerting anti-inflammation, anti-oxidative stress, and anti-autophagy effects, as demonstrated in mouse livers. We thus posit that ICT could have therapeutic potential for the treatment of hepatic IR injury.


Sujet(s)
Anti-inflammatoires , Autophagie , Flavonoïdes , Foie , Souris de lignée C57BL , Stress oxydatif , Lésion d'ischémie-reperfusion , Animaux , Lésion d'ischémie-reperfusion/traitement médicamenteux , Lésion d'ischémie-reperfusion/métabolisme , Flavonoïdes/pharmacologie , Flavonoïdes/usage thérapeutique , Foie/anatomopathologie , Foie/effets des médicaments et des substances chimiques , Foie/métabolisme , Stress oxydatif/effets des médicaments et des substances chimiques , Mâle , Souris , Autophagie/effets des médicaments et des substances chimiques , Anti-inflammatoires/pharmacologie , Anti-inflammatoires/usage thérapeutique , Lignée cellulaire , Modèles animaux de maladie humaine , Humains , Sérine-thréonine kinases TOR/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Hépatocytes/effets des médicaments et des substances chimiques , Hépatocytes/métabolisme
9.
Drug Dev Res ; 85(2): e22175, 2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-38567708

RÉSUMÉ

Icaritin is a natural prenylated flavonoid derived from the Chinese herb Epimedium. The compound has shown antitumor effects in various cancers, especially hepatocellular carcinoma (HCC). Icaritin exerts its anticancer activity by modulating multiple signaling pathways, such as IL-6/JAK/STAT3, ER-α36, and NF-κB, affecting the tumor microenvironment and immune system. Several clinical trials have evaluated the safety and efficacy of icaritin in advanced HCC patients with poor prognoses, who are unsuitable for conventional therapies. The results have demonstrated that icaritin can improve survival, delay progression, and produce clinical benefits in these patients, with a favorable safety profile and minimal adverse events. Moreover, icaritin can enhance the antitumor immune response by regulating the function and phenotype of various immune cells, such as CD8+ T cells, MDSCs, neutrophils, and macrophages. These findings suggest that icaritin is a promising candidate for immunotherapy in HCC and other cancers. However, further studies are needed to elucidate the molecular mechanisms and optimal dosing regimens of icaritin and its potential synergistic effects with other agents. Therefore, this comprehensive review of the scientific literature aims to summarize advances in the knowledge of icaritin in preclinical and clinical studies as well as the pharmacokinetic, metabolism, toxicity, and mechanisms action to recognize the main challenge, gaps, and opportunities to develop a medication that cancer patients can use. Thus, our main objective was to clarify the current state of icaritin for use as an anticancer drug.


Sujet(s)
Antinéoplasiques , Carcinome hépatocellulaire , Tumeurs du foie , Humains , Carcinome hépatocellulaire/traitement médicamenteux , Tumeurs du foie/traitement médicamenteux , Antinéoplasiques/pharmacologie , Antinéoplasiques/usage thérapeutique , Flavonoïdes/pharmacologie , Flavonoïdes/usage thérapeutique , Lignée cellulaire tumorale , Microenvironnement tumoral
10.
Nat Prod Res ; : 1-8, 2024 Apr 01.
Article de Anglais | MEDLINE | ID: mdl-38557272

RÉSUMÉ

A series of glycosyl alkyl/triazol-linked icaritin derivatives have been designed and synthesised. The target glycosyl derivatives were evaluated for their anticancer activity against three human cancer cell lines. The results of preliminary anticancer tests in vitro revealed that mannose derivatives 10a-10c (100 µM) with different aliphatic chain lengths exhibited increased cytotoxicity against HepG2 and SK-OV-3 cells compared with the parent compound icaritin. The data indicated that the kind of glycosyl groups and linkers affected the anticancer potency significantly. The ADME analysis of derivatives 10a-10c was also performed.

11.
Chem Biol Interact ; 392: 110926, 2024 Apr 01.
Article de Anglais | MEDLINE | ID: mdl-38431053

RÉSUMÉ

Metabolic reprogramming enables cancer cells to generate energy mainly through aerobic glycolysis, which is achieved by increasing the expression levels of glycolysis-related enzymes. Therefore, the development of drugs targeting aerobic glycolysis could be an effective strategy for cancer treatment. Icaritin (ICT) is an active ingredient from the Chinese herbal plant Epimedium with several biological activities, but its anti-cancer mechanism remains inconclusive. Using normal hepatocytes and hepatoma cells, our results showed that ICT suppressed cell proliferation and clonal formation and decreased glucose consumption and lactate production in liver cancer cells. In consistent, the mRNA and protein levels of several aerobic glycolysis-related genes were decreased upon ICT treatment. Furthermore, our results demonstrated that the expression levels of the aerobic glycolysis-related proteins were correlated with the p53 status in hepatoma cells. Using PFT-α or siRNA-p53, our results confirmed that ICT regulated aerobic glycolysis in a p53-dependent manner. In addition, ICT was found to stabilize p53 at the post-translational level which might be mediated by inhibiting MDM2 expression and affecting its interaction with p53. Finally, our results demonstrated that ICT increased the levels of ROS that activated p53 via the p38 MAPK pathway. In conclusion, ICT increased intracellular ROS levels in liver cancer cells, which promoted the stabilization and activation of p53, inhibiting the expression of aerobic glycolysis-related genes and glycolysis, and ultimately leading to the suppression of liver cancer development.


Sujet(s)
Carcinome hépatocellulaire , Flavonoïdes , Tumeurs du foie , Humains , Carcinome hépatocellulaire/traitement médicamenteux , Carcinome hépatocellulaire/métabolisme , Protéine p53 suppresseur de tumeur/génétique , Protéine p53 suppresseur de tumeur/métabolisme , Espèces réactives de l'oxygène/métabolisme , Tumeurs du foie/traitement médicamenteux , Tumeurs du foie/métabolisme , Glycolyse , Prolifération cellulaire , Lignée cellulaire tumorale
12.
Adv Sci (Weinh) ; 11(16): e2306174, 2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-38368261

RÉSUMÉ

Patients with concurrent intrahepatic cholangiocarcinoma (ICC) and hepatolithiasis generally have poor prognoses. Hepatolithiasis is once considered the primary cause of ICC, although recent insights indicate that bacteria in the occurrence of hepatolithiasis can promote the progression of ICC. By constructing in vitro and in vivo ICC models and patient-derived organoids (PDOs), it is shown that Escherichia coli induces the production of a novel RNA, circGLIS3 (cGLIS3), which promotes tumor growth. cGLIS3 binds to hnRNPA1 and G3BP1, resulting in the assembly of stress granules (SGs) and suppression of hnRNPA1 and G3BP1 ubiquitination. Consequently, the IKKα mRNA is blocked in SGs, decreasing the production of IKKα and activating the NF-κB pathway, which finally results in chemoresistance and produces metastatic phenotypes of ICC. This study shows that a combination of Icaritin (ICA) and gemcitabine plus cisplatin (GP) chemotherapy can be a promising treatment strategy for ICC.


Sujet(s)
Tumeurs des canaux biliaires , Cholangiocarcinome , Évolution de la maladie , Escherichia coli , Facteur de transcription NF-kappa B , Granules de stress , Animaux , Humains , Souris , Tumeurs des canaux biliaires/métabolisme , Tumeurs des canaux biliaires/génétique , Tumeurs des canaux biliaires/anatomopathologie , Cholangiocarcinome/métabolisme , Cholangiocarcinome/génétique , Cholangiocarcinome/anatomopathologie , Modèles animaux de maladie humaine , Helicase , Escherichia coli/génétique , Escherichia coli/métabolisme , , Facteur de transcription NF-kappa B/métabolisme , Facteur de transcription NF-kappa B/génétique , Protéines liant le poly-adp-ribose/métabolisme , Protéines liant le poly-adp-ribose/génétique , RNA helicases , Protéines à motif de reconnaissance de l'ARN/métabolisme , Protéines à motif de reconnaissance de l'ARN/génétique , Transduction du signal/génétique , Granules de stress/métabolisme , Granules de stress/génétique
13.
J Asthma ; : 1-10, 2024 Jan 31.
Article de Anglais | MEDLINE | ID: mdl-38294683

RÉSUMÉ

BACKGROUND: Asthma is a disease characterized by airway hyperresponsiveness and airway inflammation. Icaritin (ICT) is a plant hormone with various pharmacological activities such as anti-inflammatory, immune regulation, and anti-tumor. This study mainly explored the effects of nebulized inhalation of ICT on airway inflammation and airway remodeling in asthmatic mice. METHOD: Different groups of ovalbumin (OVA)-induced asthma mice with acute and chronic airway inflammation received ICT. Asthmatic mice received budesonide (BDND) aerosol inhalation as a positive control, while normal control and asthma model mice received the same volume of saline. Following finishing of the study, analyses were conducted on behavioral tests, biochemical indices, and histological structures of lung tissues. RESULTS: Aerosol inhalation of ICT can notably reduce inflammatory cells infiltration around the airways and pulmonary vessels, and suppressed goblet cell hyperplasia in asthmatic mice. Long-term inhalation of ICT can decrease airway collagen deposition and airway smooth muscle hyperplasia, and alleviate airway hyperresponsiveness, mirroring the effects observed with hormone employed in clinical practice. CONCLUSION: Nebulized inhalation of ICT can effectively inhibit airway inflammation in asthmatic mice, improve airway remodeling, and reduce airway hyperresponsiveness, with effects similar to those of hormones. It may serve as a potential candidate used as a hormone replacement asthma treatment.

14.
Cancer Lett ; 587: 216621, 2024 Apr 10.
Article de Anglais | MEDLINE | ID: mdl-38242198

RÉSUMÉ

Hepatocellular carcinoma (HCC) is among the deadliest malignancies worldwide and still a pressing clinical problem. Icaritin, a natural compound obtained from the Epimedium genus plant, has garnered significant attention as a potential therapeutic drug for HCC therapies. Mitophagy plays a crucial role in mitochondrial quality control through efficiently eliminating damaged mitochondria. However, the specific mechanisms of the interplay between mitophagy and apoptosis in HCC is still unclear. We aimed to explore the cross-talk between icaritin-induced mitophagy and apoptosis in HCC cells and investigate its potential mechanisms. Firstly, we confirmed that icaritin inhibits proliferation and migration while inducing mitochondrial damage and reactive oxygen species (ROS) production in HCC cells. Secondly, based on proteomics analysis, we discovered that icaritin inhibits the growth of tumor cells and disrupts their mitochondrial homeostasis through the regulation of both mitophagy and apoptosis. Thirdly, icaritin causes mitophagy mediated by PINK1-Parkin signaling via regulating feedforward loop. Furthermore, knockdown of PINK1/Parkin leads to inhibition of mitophagy, which promotes cell death induced by icaritin in HCC cells. Finally, autophagy/mitophagy inhibitors remarkably enhance icaritin-induced cell death and anticancer efficacy. Collectively, our findings reveal that icaritin suppresses growth, proliferation and migration of HCC cell through induction of mitophagy and apoptosis, while inhibition of mitophagy significantly increased the anti-cancer and pro-apoptotic effects of icaritin, indicating that targeting autophagy or mitophagy is a novel approach to overcome drug resistance and enhance anticancer therapies.


Sujet(s)
Carcinome hépatocellulaire , Flavonoïdes , Tumeurs du foie , Humains , Mitophagie , Carcinome hépatocellulaire/traitement médicamenteux , Tumeurs du foie/anatomopathologie , Autophagie , Ubiquitin-protein ligases/métabolisme , Protein kinases/métabolisme , Espèces réactives de l'oxygène/métabolisme
15.
J Pharm Biomed Anal ; 241: 115979, 2024 Apr 15.
Article de Anglais | MEDLINE | ID: mdl-38237539

RÉSUMÉ

Osteoporosis is a systemic metabolic bone disease characterized by a reduction in bone mass resulting from multifactorial causes. Icaritin (ICT), a flavonoid glycoside, exhibits a multitude of effects on bone tissue. To examine the influence of ICT on bone trabecular loss in vivo, ovariectomized (OVX) rats were utilized. The ability of ICT to mitigate bone trabecular loss and the underlying anti-osteoporotic pathways were assessed using ovariectomy-induced osteoporosis rats. Furthermore, we gain insights into the osteoprotective mechanisms of ICT on osteoporosis by conducting UPLC-Orbitrap-MS-based metabolomics of rat urine. The results of experiments demonstrated a significant attenuation of bone trabecular loss, as well as improvements in biochemical indices, biomechanical parameters, and microstructure in the ICT administered group compared to the OVX group. Moreover, metabolomics results suggested that the ICT treatment adjusted 33 different metabolites, which associated with the metabolism of amino acids, lipids, and energy. The findings suggest that the anti-osteoporosis effect of ICT may be related to the activation of PI3K/AKT signal and the inhibition of TLR4 pathway regulated by metabonomics. These results contribute to a better understanding of the therapeutic potential of ICT in the treatment of osteoporosis.


Sujet(s)
Ostéoporose , Phosphatidylinositol 3-kinases , Femelle , Rats , Animaux , Rat Sprague-Dawley , Phosphatidylinositol 3-kinases/usage thérapeutique , Ostéoporose/traitement médicamenteux , Ostéoporose/métabolisme , Flavonoïdes/pharmacologie , Flavonoïdes/usage thérapeutique , Métabolomique
16.
CNS Neurosci Ther ; 30(4): e14527, 2024 04.
Article de Anglais | MEDLINE | ID: mdl-37990437

RÉSUMÉ

AIMS: The accumulation and deposition of ß-amyloid (Aß) has always been considered a major pathological feature of Alzheimer's disease (AD). The latest and mainstream amyloid cascade hypothesis indicates that all the main pathological changes in AD are attributed to the accumulation of soluble Aß. However, the exploration of therapeutic drugs for Aß toxicity has progressed slowly. This study aims to investigate the protective effects of Icaritin on the Aß-induced Drosophila AD model and its possible mechanism. METHODS: To identify the effects of Icaritin on AD, we constructed an excellent Drosophila AD model named Aßarc (arctic mutant Aß42) Drosophila. Climbing ability, flight ability, and longevity were used to evaluate the effects of Icaritin on AD phenotypes. Aßarc was determined by immunostaining and ELISA. To identify the effects of Icaritin on oxidative stress, we performed the detection of ROS, hydrogen peroxide, MDA, SOD, catalase, GST, and Caspase-3. To identify the effects of Icaritin on energy metabolism, we performed the detection of ATP and lactate. Transcriptome analysis and qRT-PCR verifications were used to detect the genes directly involved in oxidative stress and energy metabolism. Mitochondrial structure and function were detected by an electron microscopy assay, a mitochondrial membrane potential assay, and a mitochondrial respiration assay. RESULTS: We discovered that Icaritin almost completely rescues the climbing ability, flight ability, and longevity of Aßarc Drosophila. Aßarc was dramatically reduced by Icaritin treatment. We also found that Icaritin significantly reduces oxidative stress and greatly improves impaired energy metabolism. Importantly, transcriptome analysis and qRT-PCR verifications showed that many key genes, directly involved in oxidative stress and energy metabolism, are restored by Icaritin. Next, we found that Icaritin perfectly restores the integrity of mitochondrial structure and function damaged by Aßarc toxicity. CONCLUSION: This study suggested that Icaritin is a potential drug to deal with the toxicity of Aßarc, at least partially realized by restoring the mitochondria/oxidative stress/energy metabolism axis, and holds potential for translation to human AD.


Sujet(s)
Maladie d'Alzheimer , Peptides bêta-amyloïdes , Flavonoïdes , Animaux , Humains , Peptides bêta-amyloïdes/métabolisme , Maladie d'Alzheimer/métabolisme , Stress oxydatif , Drosophila/métabolisme
17.
Nan Fang Yi Ke Da Xue Xue Bao ; 43(10): 1665-1673, 2023 Oct 20.
Article de Chinois | MEDLINE | ID: mdl-37933641

RÉSUMÉ

OBJECTIVE: To explore the radiosensitizing effect of icaritin on nasopharyngeal carcinoma (NPC) cells and the underlying mechanism. METHODS: MTT assay and clonal formation assay were used to evaluate the effect of icaritin on proliferation of human NPC HONE1 and HNE1 cells. The effects of icaritin treatment, γ-ray radiation, or both on production of reactive oxygen species (ROS), cell cycle distribution and apoptosis of the NPC cells were assessed using flow cytometry. The expressions of DNA damage markers γ-H2AX, cycle-related proteins CDC25C, p-CDC25C and cyclin B1, and ferroptosis markers ACSL4 and GXP4 were detected using Western blotting. A nude mouse model bearing subcutaneous HONE1 cell xenograft was used to observe the effect of icaritin and radiation on tumor growth. RESULTS: Icaritin dose-dependently inhibited the viability of the NPC cells and enhanced the inhibitory effect of radiation on cell proliferation. Flow cytometry and Western blotting showed that icaritin treatment prior to radiation significantly promoted ROS production and γ-H2AX expression in the NPC cells (P<0.001). Compared with radiation exposure alone, the combined treatment caused cell cycle arrest in G2 phase, down-regulated CDC25C and cyclin B1 expression, and up-regulated p-CDC25C expression in the cells (P<0.01), resulting also in increased cell apoptosis, enhanced expression of ferroptosis protein ACSL4 and lowered expression of GXP4 (P<0.001). In the tumor-bearing mice, icaritin treatment, compared with radiation alone, significantly reduced the tumor growth rate and decreased tumor weight (P<0.001). CONCLUSION: Icaritin can enhance radiosensitivity of NPC cells both in vitro and in nude mice possibly by enhancing ROS production to promote iron death of the cells.


Sujet(s)
Carcinomes , Tumeurs du rhinopharynx , Humains , Animaux , Souris , Cancer du nasopharynx , Cycline B1 , Carcinomes/métabolisme , Tumeurs du rhinopharynx/génétique , Souris nude , Espèces réactives de l'oxygène , Radiotolérance , Prolifération cellulaire , Lignée cellulaire tumorale , Apoptose
18.
Aging (Albany NY) ; 15(19): 10213-10236, 2023 10 03.
Article de Anglais | MEDLINE | ID: mdl-37793008

RÉSUMÉ

A rapidly aging society and longer life expectancy are causing osteoporosis to become a global epidemic. Over the last five decades, a number of drugs aimed at reducing bone resorption or restoring bone mass have been developed, but their efficacy and safety are limited. Icaritin (ICT) is a natural compound extracted from anti-osteoporosis herb Epimedium spp. and has been shown to inhibit osteoclast differentiation. However, the molecular mechanism by which ICT weaken RANKL-induced osteoclast differentiation has not been completely investigated. Here, we evaluated the anti-osteoclastogenic effect of ICT in vitro and the potential drug candidate for treating osteoporosis in vivo. In vitro study, ICT was found to inhibit osteoclast formation and bone resorption function via downregulating transcription factors activated T cell cytoplasm 1 (NFATc1) and c-fos, which further downregulate osteoclastogenesis-specific gene. In addition, the enhanced mitochondrial mass and function required for osteoclast differentiation was mitigated by ICT. The histomorphological results from an in vivo study showed that ICT attenuated the bone loss associated with ovariectomy (OVX). Based on these results, we propose ICT as a promising new drug strategy for osteoporosis that inhibits osteoclast differentiation.


Sujet(s)
Résorption osseuse , Ostéoporose , Femelle , Humains , Ostéogenèse , Différenciation cellulaire , Ostéoporose/traitement médicamenteux , Ostéoporose/étiologie , Résorption osseuse/traitement médicamenteux , Protéines proto-oncogènes c-fos/génétique , Ovariectomie/effets indésirables
19.
Phytomedicine ; 120: 155062, 2023 Nov.
Article de Anglais | MEDLINE | ID: mdl-37683586

RÉSUMÉ

BACKGROUND: Icaritin has a wide range of pharmacological activities, including significant an-titumor activity. However, the mechanism of action of icaritin in endometrial cancer (UCEC) remains unknown. FOX proteins are a highly conserved transcription factor superfamily that play important roles in epithelial cell differentiation, tumor metastasis, angiogenesis, and cell cycle regulation. FOXC1 is an important member of the FOX protein family. FOXC1 is aberrantly expressed in endometrial cancer and may play a role in the migration and invasion of endometrial cancer; however, its mechanism of action has not yet been reported. O-GlcNAc glycosylation is a common post-translational modification. In endometrial cancer, high levels of O-GlcNAcylation promote cell proliferation, migration, and invasion. Cancer development is often accompanied by O-GlcNAc modification of proteins; however, O-GlcNAc modification of the transcription factor FOXC1 has not been reported to date. PURPOSE: To investigate the inhibitory effects of icaritin on RL95-2 and Ishikawa endometrial cancer cells in vitro and in vivo and to elucidate the possible molecular mechanisms. METHODS/STUDY DESIGN: CCK8, colony formation, migration, and invasion assays were used to determine the inhibitory effects of icaritin on endometrial cancer cells in vitro. Cell cycle regulation was assayed by flow cytometry. Protein levels were measured based on western blotting. The level of FOXC1 expression in endometrial cancer tissues was determined by immunohistochemistry. To assess whether icaritin also has activity in vivo, its effect on tumor xenografts was evaluated. RESULTS: Immunohistochemical analysis of clinical samples revealed that FOXC1 expression was significantly higher in endometrial cancer tissues than in normal tissues. Downregulation of FOXC1 inhibited the proliferative, colony formation, migration, and invasive abilities of RL95-2 and Ishikawa endometrial cancer cells. Icaritin inhibited the proliferation, colony formation, migration, and invasion of endometrial cancer cells and blocked the cell cycle in S phase. Icaritin affected O-GlcNAc modification of FOXC1 and thus the stability of FOXC1, which subsequently triggered the inhibition of endometrial cancer cell proliferation. CONCLUSION: The anti-endometrial cancer effect of icaritin is related to the inhibition of abnormal O-GlcNAc modification of FOXC1, which may provide an important theoretical foundation for the use of icaritin against endometrial cancer.


Sujet(s)
Tumeurs de l'endomètre , Humains , Femelle , Tumeurs de l'endomètre/traitement médicamenteux , Flavonoïdes/pharmacologie , Division cellulaire , Prolifération cellulaire , Facteurs de transcription Forkhead
20.
Phytochemistry ; 215: 113841, 2023 Nov.
Article de Anglais | MEDLINE | ID: mdl-37660725

RÉSUMÉ

As a detoxification and metabolism organ, the liver plays a vital role in human health. However, an excessive consumption of drugs and toxins, exposure to pathogenic viruses, and unhealthy living habits can lead to liver damage, which may even develop into liver cirrhosis and liver cancer. Epimedium brevicornum Maxim. is a traditional Chinese medicine and dietary supplement in which the flavonoid icariin is a main functional component. Although the protective mechanisms of icariin and its metabolites against liver injury are not yet comprehensively understood, an increasing number of studies have confirmed their liver-protective and anticancer effects. Indeed, icaritin, one of the metabolites of icariin, is currently utilized as an active component of an anti-cancer drug. This paper presents a review of the molecular mechanisms through which icariin and its metabolites actively protect against the occurrence and development of liver injury, and, thus, provides a comprehensive reference for further research and their application in liver protection.


Sujet(s)
Epimedium , Extraits de plantes , Humains , Flavonoïdes/pharmacologie , Foie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE