Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Saudi Pharm J ; 32(7): 102124, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38933713

ABSTRACT

Natural products (NPs) play an irreplaceable role in the intervention of various diseases and have been considered a critical source of drug development. Many new pharmacodynamic compounds with potential clinical applications have recently been derived from NPs. These compounds range from small molecules to polysaccharides, polypeptides, proteins, self-assembled nanoparticles, and extracellular vesicles. This review summarizes various active substances found in NPs. The investigation of active substances in NPs can potentiate new drug development and promote the in-depth comprehension of the mechanism of action of NPs that can be beneficial in the prevention and treatment of human diseases.

2.
Chem Biol Interact ; 388: 110829, 2024 Jan 25.
Article in English | MEDLINE | ID: mdl-38101598

ABSTRACT

SUN, a multi-targeted tyrosine kinase inhibitor, exerts cardiotoxicity which hinders its clinical use. It is necessary to elucidate molecular mechanism of SUN-induced cardiotoxicity. To elucidate molecular mechanism of SUN-induced cardiotoxicity and whether it is related to Nrf2-dependent ferroptosis, in vitro model with H9c2 cells derived from rat heart tissue and in vivo model (C57BL/6J male mouse) were used in the present study. In vivo model was established by oral treatment of SUN at dose of 10, 20, 40 mg/kg for 14 days. Body weight, ECG, plasma enzyme activities, histology staining were performed to evaluate heart function. Western-blot was performed to analyze the level of ferroptosis-related proteins. In vitro results indicated that SUN markedly induced ferroptosis embodied as collapsed MMP, accumulated iron and elevated ROS. In vivo results showed that SUN significantly impaired cardiac function. Abnormal electrocardiogram, increased serum CK and lactate LDH levels were significantly observed in SUN groups. Histology staining showed that SUN caused structural injuries and fibrosis deposition. Moreover, SUN increased the level of MDA and Fe2+ content, decreased the level of GSH. Both in vitro and in vivo experiments indicated that SUN reduced the expression of Nrf2, HO-1, NQO1, GPX4 and FTH1, enhanced the TfR expression. This study suggested that oxidative stress and Nrf2-dependent ferroptosis played a vital role in SUN-induced cardiotoxicity.


Subject(s)
Ferroptosis , NF-E2-Related Factor 2 , Male , Mice , Animals , Rats , Mice, Inbred C57BL , Sunitinib , Cardiotoxicity , Oxidative Stress
3.
Saudi Pharm J ; 31(4): 597-604, 2023 Apr.
Article in English | MEDLINE | ID: mdl-37063439

ABSTRACT

A third of the world's population suffers from unexplained fatigue, hugely impacting work learning, efficiency, and health. The fatigue development may be a concomitant state of a disease or the side effect of a drug, or muscle fatigue induced by intense exercise. However, there are no authoritative guides or clinical medication recommendations for various fatigue classifications. Traditional Chinese medicines (TCM) are used as dietary supplements or healthcare products with specific anti-fatigue effects. Thus, TCM may be a potential treatment for fatigue. In this review, we outline the pathogenesis of fatigue, awareness of fatigue in Chinese and western medicine, pharmacodynamics mechanism, and substances. Additionally, we offer a comprehensive summary of fatigue and forecast the potential effect of novel herbal-based medicines against fatigue.

4.
Food Funct ; 14(2): 1087-1098, 2023 Jan 23.
Article in English | MEDLINE | ID: mdl-36594456

ABSTRACT

Imatinib mesylate (IMA) belonging to the selective tyrosine kinase inhibitor family has been proven to induce cardiotoxic effects along with therapeutic strategies. Nrf2-dependent ferroptosis has been implicated in the cardiotoxicity induced by IMA. The present study was designed to investigate the protective effects of berberine hydrochloride (Ber) on cardiac injuries induced by IMA and to explore its potential mechanisms. In H9c2 cells, cell viability, the generation of reactive oxygen species (ROS), mitochondrial membrane potential (MMP) and labile iron pool (LIP) levels were measured. In a mouse model of IMA-induced cardiomyopathy, serum biomarkers and cardiac tissues were examined. A western blot assay was performed to evaluate the expression of ferroptosis-related proteins in vitro and in vivo. Our results indicated that Ber increased cell viability and MMP and decreased cellular ROS and iron levels in comparison to the IMA group of H9c2 cells. In mice, Ber significantly improved cardiac status and attenuated the level of ferroptosis biomarkers including malonaldehyde (MDA) and iron content. Additionally, Ber downregulated the expression of transferrin receptor (TfR) and P53 and upregulated the expression of nuclear factor erythroid 2-related factor 2 (Nrf2), heme oxygenase-1 (HO-1), NAD(P)H quinone oxidoreductase-1 (NQO1), ferritin heavy chain-1 (FTH1), and glutathione peroxidase 4 (GPX4) in H9c2 cells and mice. The present data indicated that Ber has the potential to protect against IMA-induced cardiotoxicity, partly via inhibiting Nrf2-dependent ferroptosis.


Subject(s)
Berberine , Ferroptosis , Animals , Mice , Berberine/pharmacology , Biomarkers , Cardiotoxicity , Imatinib Mesylate/pharmacology , Iron/metabolism , NF-E2-Related Factor 2/metabolism , Reactive Oxygen Species/metabolism , Rats
5.
J Nutr Biochem ; 112: 109241, 2023 02.
Article in English | MEDLINE | ID: mdl-36442718

ABSTRACT

The clinical use of 5-fluorouracil (5-FU), a potent antitumor agent, was limited by severe cardiotoxic effects. The present study was aimed to investigate the protective effects of resveratrol (Res) on 5-FU-induced cardiotoxicity and to explore its potential mechanisms.The cardiotoxicity model was intraperitoneal injection of 5-FU at the dose of 30 mg/kg for 7 consecutive days. Plasma enzymes activities, cardiac tissues were assessed after treatment with Res for 3 weeks. Ferrostatin-1 (Fer-1) was used as ferroptosis inhibitor. In H9c2 cardiomyocyte cells, cell viability, generation of reactive oxygen species (ROS), mitochondrial activity and cellular Fe2+ levels were measured. Western-blot assay was performed to evaluate the protein level of ferroptosis in vitro and in vivo. In the mice model, Res reduced 5-FU-induced cardiomyocyte injury (ferroptosis, myofibrillar loss and vacuolization). In addition, increased serum creatine kinase (CK), lactate dehydrogenase (LDH), malonaldehyde (MDA) and Fe2+ activity and decreased activities of glutathione (GSH) were observed in 5-FU group. These changes were prevented by treatment with Res. In H9c2 cardiomyocyte cells, Res increased the cell viability and attenuated cell ferroptosis as measured by DCFH-DA, TMRE and Calcein AM staining. In addition, 5-FU induced a reduction in GPX4, FTH1, Nrf2 and NQO1 and activation of TfR and P53 compared with the control group. However, Res effectively inhibited the changes in ferroptosis associated proteins in vitro and in vivo. Res possessed the cardioprotective potential against 5-FU induced cardiotoxicity. Moreover, Res attenuates 5-FU-induced cardiotoxicity via inhibiting GPX4 dependent ferroptosis.


Subject(s)
Cardiotoxicity , Ferroptosis , Resveratrol , Animals , Mice , Cardiotoxicity/etiology , Ferroptosis/drug effects , Fluorouracil/toxicity , Glutathione , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Resveratrol/pharmacology , Rats , Phospholipid Hydroperoxide Glutathione Peroxidase/drug effects , Phospholipid Hydroperoxide Glutathione Peroxidase/metabolism
6.
J Pharm Pharmacol ; 75(1): 49-56, 2023 Jan 31.
Article in English | MEDLINE | ID: mdl-36173891

ABSTRACT

OBJECTIVES: Resveratrol (Res) was a naturally occurring polyphenol compound. It has various beneficial effects, including anti-inflammatory, anti-oxidant and anti-cancer effects. However, the anti-cancer activity was hindered by its low targeting and drug release performance. Thus, we synthesized transferrin-cathepsin B cleavable peptide modified mesoporous silica nanoparticle encapsulated Res (Tf-Res-MSN). METHODS: Res was encapsulated in mesoporous silica nanoparticles (MSN), which was a kind of drug carrier complex. Tf was modified to recognize the cancer cells. Cathepsin B cleavable peptide (Pep) was used to combine Res-MSN complex and Tf to construct the final product. Pep was used as linker and trigger for Res release. KEY FINDINGS: The smart nanocarriers were increased the drug release performance of Res in human breast cancer (MCF-7) cells. The physicochemical properties of Tf-Res-MSN were assessed by zeta potential, UV-Prove, diffraction scanning calorimetry (DSC), nitrogen physisorption analysis and transmission electron microscope (TEM). MTT assay, AO and Annexin V-FITC/PI staining were performed to explore the anti-tumour activity of Tf-Res-MSN. The results showed that Tf-Res-MSN significantly decreased cell viability and increased cell apoptosis. The inhibition rate and apoptotic rate of Tf-Res-MSN in MCF-7 cells were 95.75% and 80.8%, respectively. CONCLUSION: Our study demonstrated that Tf-Res-MSN was a valuable technique with potential value in breast cancer applications.


Subject(s)
Breast Neoplasms , Nanoparticles , Humans , Female , Drug Delivery Systems/methods , MCF-7 Cells , Resveratrol/pharmacology , Cathepsin B/pharmacology , Silicon Dioxide , Transferrin/pharmacology , Transferrin/therapeutic use , Drug Carriers/chemistry , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Peptides/pharmacology , Nanoparticles/chemistry , Apoptosis , Porosity
7.
Toxicology ; 468: 153113, 2022 02 28.
Article in English | MEDLINE | ID: mdl-35101590

ABSTRACT

5-Fluorouracil (5-FU) was a key chemotherapeutic agent in the treatment of different solid tumors. However, cardiotoxicity was included among the therapeutic strategies of 5-FU. The molecular mechanism of cardiotoxicity induced by 5-FU remains unclear. The aim of the study was to investigate whether ferroptosis was involved in 5-FU-induced cardiotoxicity in vivo and in vitro. The in vivo cardiotoxicity model was induced by intraperitoneal injection of 5-FU at the dose of 15, 30, 60 mg/kg for 7 days. Body weight, general condition and plasma enzyme activities of the mice were observed to evaluate heart function. In addition, HE staining, MASSON staining and TEM technology was used. Western-blot analysis were performed to evaluate the protein level of iron transport, iron storage and reactive oxygen species (ROS) of ferroptosis. In H9c2 cardiomyocyte cells, cell viability, generation of ROS, mitochondrial activity and cellular Fe2+ levels were measured. The in vivo results showed that 5-FU significantly impaired cardiac function and structure. The serum creatine kinase (CK) and lactate dehydrogenase (LDH) levels were significantly increased in 5-FU group. HE and MASSON staining showed that 5-FU caused structural injuries. In addition, 5-FU increased the level of ferroptosis markers involving malonaldehyde (MDA) and Fe2+ content. Ferrostatin-1 (Fer-1) was an aromatic amine that specifically binds with lipid ROS and protects cells against lipid peroxidation. Furthermore, 5-FU markedly induced ferroptosis in H9c2 cardiomyocyte cells, which mainly embodied as declined cell vitality, accumulated iron, elevated lipid peroxides. Conversely, inhibition of ferroptosis by Fer-1 completely abolished 5-FU-induced effects. Both in vivo and in vitro experiments indicated that 5-FU increased the expression of ferroptosis, mainly by reducing the expression of glutathione peroxidase 4 (GPX4) and ferritin heavy chain 1 (FTH1), but enhancing the expression of transferrin receptor 1 (TfR1). In conclusion, the present study suggested that ROS and iron homeostasis dependent ferroptosis played a vital role in 5-FU induced cardiotoxicity.


Subject(s)
Ferroptosis/physiology , Iron/metabolism , Myocardium/chemistry , Reactive Oxygen Species/metabolism , Animals , Blotting, Western , Body Weight/drug effects , Coloring Agents , Creatine Kinase/blood , Echocardiography , Eosine Yellowish-(YS) , Fluorescent Dyes , Fluorouracil/pharmacology , Heart/anatomy & histology , Heart/drug effects , Heart/physiology , Hematoxylin , Homeostasis , L-Lactate Dehydrogenase/blood , Male , Mice , Mice, Inbred C57BL , Microscopy, Electron, Transmission , Mitochondria/physiology , Myocardium/ultrastructure , Organ Size/drug effects , Plasma/chemistry , Silver Nitrate
8.
Toxicol Appl Pharmacol ; 435: 115852, 2022 01 15.
Article in English | MEDLINE | ID: mdl-34973290

ABSTRACT

Imatinib Mesylate (IMA) has been widely used to treat with chronic myeloid leukemia (CML). However, cardiotoxicity associated with IMA is included among the therapeutic strategies. The present study was aimed to discover whether ferroptosis, a programmed iron-dependent cell death, is involved in IMA-induced cardiotoxicity. In vivo, mouse model was established after treated with 25 mg/kg, 50 mg/kg and 100 mg/kg IMA. Serum CK, LDH, AST activities were determined. Cardiac tissues were examined by H&E and Oil Red O staining. MDA was measured to assess production of lipid peroxide. Tissue iron and GSH content were measured. In vitro, cell viability, mitochondria membrane potential, generation of reactive oxygen species (ROS) and cellular iron levels were performed to explore the mechanism of IMA. The in vivo results revealed that IMA treatment significantly increased serum CK, LDH and AST. H&E staining showed that IMA caused cardiac structural injuries. The dose-dependent decrease of GSH and increase of tissue iron and MDA were observed in IMA-treated groups. Oil Red O staining suggested obvious cardiac lipid accumulation after treated with IMA. In H9c2 cardiomyocytes, IMA significantly inhibited cell proliferation in a dose-dependent manner. Mitochondria membrane potential assay showed that IMA destroyed the mitochondrial function. Additionally, IMA increased the cellular ROS and iron levels. Furthermore, IMA down-regulated the expression of Nrf2 and up-regulated the expression of P53 and TfR. These results provided compelling evidence that ferroptosis participates in IMA-induced cardiotoxicity. Ferroptosis could be regarded as a target to protect against cardiotoxicity in IMA-exposed patients.


Subject(s)
Antineoplastic Agents/toxicity , Cardiotoxicity , Ferroptosis/drug effects , Imatinib Mesylate/toxicity , NF-E2-Related Factor 2/biosynthesis , Protein Kinase Inhibitors/toxicity , Signal Transduction/drug effects , Animals , Cell Line , Dose-Response Relationship, Drug , Down-Regulation/drug effects , Glutathione/metabolism , Iron/metabolism , Lipid Metabolism/drug effects , Lipid Peroxides/metabolism , Male , Membrane Potential, Mitochondrial/drug effects , Mice , Mice, Inbred C57BL , NF-E2-Related Factor 2/drug effects , NF-E2-Related Factor 2/genetics , Reactive Oxygen Species/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...