Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 92
Filter
1.
Neurochem Int ; 176: 105725, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38561151

ABSTRACT

Epilepsy constitutes a global health concern, affecting millions of individuals and approximately one-third of patients exhibit drug resistance. Recent investigations have revealed alterations in cerebral iron content in both epilepsy patients and animal models. However, the extant literature lacks a comprehensive exploration into the ramifications of modulating iron homeostasis as an intervention in epilepsy. This study investigated the impact of deferasirox, a iron ion chelator, on epilepsy. This study unequivocally substantiated the antiepileptic efficacy of deferasirox in a kainic acid-induced epilepsy model. Furthermore, deferasirox administration mitigated seizure susceptibility in a pentylenetetrazol-induced kindling model. Conversely, the augmentation of iron levels through supplementation has emerged as a potential exacerbating factor in the precipitating onset of epilepsy. Intriguingly, our investigation revealed a hitherto unreported discovery: ITPRIP was identified as a pivotal modulator of excitatory synaptic transmission, regulating seizures in response to deferasirox treatment. In summary, our findings indicate that deferasirox exerts its antiepileptic effects through the precise targeting of ITPRIP and amelioration of cerebral iron homeostasis, suggesting that deferasirox is a promising and novel therapeutic avenue for interventions in epilepsy.


Subject(s)
Anticonvulsants , Brain , Deferasirox , Epilepsy , Iron Chelating Agents , Iron , Membrane Proteins , Animals , Male , Mice , Anticonvulsants/pharmacology , Anticonvulsants/therapeutic use , Brain/drug effects , Brain/metabolism , Deferasirox/pharmacology , Epilepsy/drug therapy , Epilepsy/metabolism , Homeostasis/drug effects , Homeostasis/physiology , Iron/metabolism , Iron Chelating Agents/pharmacology , Iron Chelating Agents/therapeutic use , Kindling, Neurologic/drug effects , Pentylenetetrazole/toxicity , Rats, Sprague-Dawley , Membrane Proteins/drug effects , Membrane Proteins/metabolism
2.
Adv Mater ; 36(15): e2304328, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38229577

ABSTRACT

Currently, certain cancer patients exhibit resistance to radiotherapy due to reduced DNA damage under hypoxic conditions and acquired immune tolerance triggered by transforming growth factor-ß1 (TGF-ß1) and membrane-localized programmed death ligand-1 (PD-L1). Meanwhile, cytoplasm-distributed PD-L1 induces radiotherapy resistance through accelerating DNA damage repair (DDR). However, the disability of clinically used PD-L1 antibodies in inhibiting cytoplasm-distributed PD-L1 limits their effectiveness. Therefore, a nanoadjuvant is developed to sensitize cancer to radiotherapy via multi-level immunity activation through depressing PD-L1 and TGF-ß1 by triphenylphosphine-derived metformin, and activating the cGAS-STING pathway by generating Mn2+ from MnO2 and producing more dsDNA via reversing tumor hypoxia and impairing DDR. Thus, Tpp-Met@MnO2@Alb effectively enhances the efficiency of radiotherapy to inhibit the progression of irradiated local and abscopal tumors and tumor lung metastases, offering a long-term memory of antitumor immunity without discernible side effects. Overall, Tpp-Met@MnO2@Alb has the potential to be clinically applied for overcoming radio-immunotherapy resistance.


Subject(s)
Adjuvants, Pharmaceutic , Lung Neoplasms , Neoplasms , Humans , B7-H1 Antigen/antagonists & inhibitors , Immunotherapy , Lung Neoplasms/radiotherapy , Lung Neoplasms/therapy , Manganese Compounds/pharmacology , Neoplasms/radiotherapy , Neoplasms/therapy , Oxides , Transforming Growth Factor beta1/antagonists & inhibitors , Adjuvants, Pharmaceutic/pharmacology , Adjuvants, Pharmaceutic/therapeutic use , Nucleotidyltransferases/drug effects , Membrane Proteins/drug effects
3.
Nutrients ; 13(10)2021 Oct 08.
Article in English | MEDLINE | ID: mdl-34684530

ABSTRACT

In hypertensive individuals, platelet morphology and function have been discovered to be altered, and this has been linked to the development of vascular disease, including erectile dysfunction (ED). The impact of nutritional supplementation with Cyperus esculentus (tiger nut, TN) and Tetracarpidium conophorum (walnut, WN) on androgen levels, ectonucleotidases, and adenosine deaminase (ADA) activities in platelets from L-NAME (Nω-nitro-L-arginine methyl ester hydrochloride) challenged rats were investigated. We hypothesized that these nuts may show a protective effect on platelets aggregation and possibly enhance the sex hormones, thereby reverting vasoconstriction. Wistar rats (male; 250-300 g; n = 10) were grouped into seven groups as follows: basal diet control group (I); basal diet/L-NAME/Viagra (5 mg/kg/day) as positive control group (II); ED-induced group (basal diet/L-NAME) (III); diet supplemented processed TN (20%)/L-NAME (IV); diet supplemented raw TN (20%)/L-NAME (V); diet supplemented processed WN (20%)/L-NAME (VI); and diet supplemented raw WN (20%)/L-NAME (VII). The rats were given their regular diet for 2 weeks prior to actually receiving L-NAME (40 mg/kg/day) for ten days to induce hypertension. Platelet androgen levels, ectonucleotidases, and ADA were all measured. L-NAME considerably lowers testosterone levels (54.5 ± 2.2; p < 0.05). Supplementing the TN and WN diets revealed improved testosterone levels as compared to the control (306.7 ± 5.7), but luteinizing hormone levels remained unchanged. Compared to control groups, the L-NAME-treated group showed a rise in ATP (127.5%) hydrolysis and ADA (116.7%) activity, and also a decrease in ADP (76%) and AMP (45%) hydrolysis. Both TN and WN supplemented diets resulted in substantial (p < 0.05) reversal effects. Enhanced testosterone levels and modulation of the purinergic system in platelets by TN and WN could be one of the mechanisms by which they aid in vasoconstriction control.


Subject(s)
Blood Platelets/drug effects , Cyperus , Dietary Supplements , Hypertension/therapy , Juglans , NG-Nitroarginine Methyl Ester/pharmacology , Adenosine Deaminase/drug effects , Adenosine Diphosphate/metabolism , Adenosine Monophosphate/metabolism , Adenosine Triphosphatases/metabolism , Adenosine Triphosphate/metabolism , Animals , Diet/methods , Hydrolysis/drug effects , Hypertension/blood , Hypertension/chemically induced , Male , Membrane Proteins/drug effects , Platelet Aggregation/drug effects , Purinergic Agents/pharmacology , Rats , Rats, Wistar , Testosterone/blood , Vasoconstriction/drug effects
4.
Sci Rep ; 11(1): 16580, 2021 08 16.
Article in English | MEDLINE | ID: mdl-34400719

ABSTRACT

Membrane proteins constitute a substantial fraction of the human proteome, thus representing a vast source of therapeutic drug targets. Indeed, newly devised technologies now allow targeting "undruggable" regions of membrane proteins to modulate protein function in the cell. Despite the advances in technology, the rapid translation of basic science discoveries into potential drug candidates targeting transmembrane protein domains remains challenging. We address this issue by harmonizing single molecule-based and ensemble-based atomistic simulations of ligand-membrane interactions with patient-derived induced pluripotent stem cell (iPSC)-based experiments to gain insights into drug delivery, cellular efficacy, and safety of molecules directed at membrane proteins. In this study, we interrogated the pharmacological activation of the cardiac Ca2+ pump (Sarcoplasmic reticulum Ca2+-ATPase, SERCA2a) in human iPSC-derived cardiac cells as a proof-of-concept model. The combined computational-experimental approach serves as a platform to explain the differences in the cell-based activity of candidates with similar functional profiles, thus streamlining the identification of drug-like candidates that directly target SERCA2a activation in human cardiac cells. Systematic cell-based studies further showed that a direct SERCA2a activator does not induce cardiotoxic pro-arrhythmogenic events in human cardiac cells, demonstrating that pharmacological stimulation of SERCA2a activity is a safe therapeutic approach targeting the heart. Overall, this novel multiscale platform encompasses organ-specific drug potency, efficacy, and safety, and opens new avenues to accelerate the bench-to-patient research aimed at designing effective therapies directed at membrane protein domains.


Subject(s)
Membrane Proteins/drug effects , Molecular Targeted Therapy/methods , Sarcoplasmic Reticulum Calcium-Transporting ATPases/drug effects , Small Molecule Libraries/therapeutic use , Animals , Enzyme Activation/drug effects , Giant Cells/enzymology , Humans , Induced Pluripotent Stem Cells/enzymology , Microsomes/enzymology , Molecular Dynamics Simulation , Molecular Structure , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/enzymology , Phosphatidylcholines , Protein Domains/drug effects , Sarcoplasmic Reticulum/enzymology , Small Molecule Libraries/adverse effects , Small Molecule Libraries/pharmacology , Swine , Water
5.
Pak J Pharm Sci ; 34(2): 493-498, 2021 Mar.
Article in English | MEDLINE | ID: mdl-34275821

ABSTRACT

P. petiolosa as a typical Chinese herbal medicine has been generally utilized as Chinese native medicine formulation for treatment of chronic bronchitis, bronchial asthma and pneumoconiosis. The objective of this study was to evaluate the anti-inflammatory and antibacterial activities of P. petiolosa ethyl acetate extract (PPEAE) against S. aureusin mice. In our study, mice were infected pneumonia by S. aureus, colonization of S. aureus in lung tissue was calculated and the number of white blood cells (WBC) in blood was measured. Meanwhile, the hematoxylin-eosin staining (H&E) was observed and the Real-time PCR was employed to determine the relative mRNA expression. The results showed that, after treated with PPEAE the wet/dry (W/D) weight ratio and the number of WBC decreased dramatically, the number of S. aureus was significantly reduced. Furthermore, H&E staining showed that PPEAE obviously relieved the inflammation of infected mice and real-time PCR results indicated that PPEAE significantly down regulated the inflammatory iNOS, TNF-α and up regulated the anti-inflammatory HO-1 mRNA. In summary, our study revealed that application of crude product PPEAE had prominent antibacterial activity against S. aureus. PPEAE significantly reduced the biomass of S. aureus and effectively relieved the inflammation of S. aureus-induced pneumonia.


Subject(s)
Anti-Bacterial Agents/pharmacology , Anti-Inflammatory Agents/pharmacology , Drugs, Chinese Herbal/pharmacology , Lung/drug effects , Plant Extracts/pharmacology , Pneumonia, Staphylococcal/genetics , Polypodiaceae , Staphylococcus aureus/drug effects , Animals , Heme Oxygenase-1/drug effects , Heme Oxygenase-1/genetics , Inflammation/genetics , Inflammation/metabolism , Lung/metabolism , Lung/microbiology , Membrane Proteins/drug effects , Membrane Proteins/genetics , Mice , Nitric Oxide Synthase Type II/drug effects , Nitric Oxide Synthase Type II/genetics , Pneumonia, Staphylococcal/metabolism , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Tumor Necrosis Factor-alpha/drug effects , Tumor Necrosis Factor-alpha/genetics
6.
Biomed Pharmacother ; 141: 111846, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34225018

ABSTRACT

In this study, untargeted lipidomics based on UPLC-Q/TOF-MS, network pharmacology and atomic force microscopy were used to explore the common biomarkers of hyperlipidemia and coronary heart disease, the therapeutic mechanism of the main components of Salvia miltiorrhiza as well as the action mechanism of key lipids. Firstly, the serum samples of 30 healthy people, 30 patients with coronary heart disease and 30 patients with hyperlipidemia were analyzed by using lipidomics technology to obtain biomarkers which can be used to link hyperlipidemia and coronary heart disease and to find potential targets; then, the key components and core targets of Salvia miltiorrhiza intervention in hyperlipidemia and coronary heart disease were analyzed by network pharmacology, the results were verified by atomic force microscopy. It showed that SMS2 might be the key target. And through network pharmacology and atomic force microscope analysis, it can be inferred that salvianolic acid A can combine with SMS2 to play a therapeutic role.


Subject(s)
Coronary Disease/prevention & control , Hyperlipidemias/prevention & control , Lipidomics , Metabolic Networks and Pathways/drug effects , Network Pharmacology , Salvia miltiorrhiza/chemistry , Caffeic Acids/pharmacology , Chromatography, High Pressure Liquid , Female , Healthy Volunteers , Humans , Lactates/pharmacology , Male , Membrane Proteins/drug effects , Microscopy, Atomic Force , Middle Aged , Nerve Tissue Proteins/drug effects , Transferases (Other Substituted Phosphate Groups)/drug effects
7.
Nutrients ; 13(3)2021 Mar 09.
Article in English | MEDLINE | ID: mdl-33803357

ABSTRACT

Benign prostatic hyperplasia (BPH) is the most common symptomatic abnormality of the human prostate characterized by uncontrolled proliferation of the prostate gland. In this study, we investigated the effect of bamboo, Phyllostachys pubescens, leaves extract (PPE) on human 5α-reductase type 2 (SRD5A2) gene promoter activity in human prostate cell lines and the protective effect of PPE on a testosterone-induced BPH rat model. PPE repressed human SRD5A2 promoter activity and its mRNA expression. The rats treated with PPE for 4 weeks showed a significantly attenuated prostate weight compared to vehicle control. PPE-treated rats also showed reduced serum dihydrotestosterone, testosterone, prostate-specific antigen, and SRD5A2 levels by testosterone injection. Quantitative real-time polymerase chain reaction showed that PPE treatment significantly decreased mRNA expression of SRD5A2, androgen receptor (AR), proliferating cell nuclear antigen (PCNA), and fibroblast growth factor 2 compared with the vehicle-treated, testosterone-injected rats in the prostate. Furthermore, PPE treatment showed reduced AR, PCNA, and tumor necrosis factor alpha expression in the prostate via immunohistofluorescence staining. In conclusion, oral administration of PPE prevented and inhibited the development and progression of enlarged prostate lesions in testosterone-induced animal models through various anti-proliferative and anti-inflammatory pharmacological effects and induced suppression of SRD5A2 gene expression.


Subject(s)
3-Oxo-5-alpha-Steroid 4-Dehydrogenase/drug effects , Membrane Proteins/drug effects , Plant Extracts/pharmacology , Plant Leaves/chemistry , Prostatic Hyperplasia/drug therapy , Sasa/chemistry , Animals , Anti-Inflammatory Agents/pharmacology , Cell Line , Cell Proliferation/drug effects , Disease Models, Animal , Humans , Male , Prostate/drug effects , Prostatic Hyperplasia/chemically induced , Prostatic Hyperplasia/genetics , Rats , Testosterone/adverse effects
8.
Med Sci Monit ; 26: e923163, 2020 Jun 18.
Article in English | MEDLINE | ID: mdl-32555127

ABSTRACT

BACKGROUND Many bioactive ingredients of medicinal plants are known to produce vaso-protective benefits. Puerarin is one of the major isoflavone glucosides found in the root of kudzu vine and it exerts an anti-inflammatory effect and many other pharmacological actions. However, the mechanism underlying the vascular effect of puerarin is incompletely understood. Therefore, the present study aims to examine how puerarin reduces endothelium-dependent contractions (EDCs) in mouse arteries. MATERIAL AND METHODS EDCs were evoked by acetylcholine (ACh) in isolated mouse carotid arteries with intact endothelium pretreated with Nω-NO2-L-Arg-OMe (L-NAME). The arteries were pretreated with puerarin and other pharmacological inhibitors before the addition of cumulative concentrations of ACh. The concentration of several prostaglandins (PGs) was measured by high performance liquid chromatography-coupled spectrometry (HPLC-MS). RESULTS EDCs induced by ACh only presented in endothelium-intact arteries pretreated by L-NAME and EDCs were prevented by the treatment with cyclooxygenase (COX) inhibitor indomethacin (3 µmol/L) or thromboxane prostanoid receptor (TP receptor) antagonist S18886 (30 nmol/L). Acute 40-minute treatment with puerarin reduced EDCs in a concentration-dependent manner without affecting U46619-induced contraction. However, treatment with puerarin did not inhibit ACh-induced production of prostaglandins (PGs) in endothelium-intact arteries. CONCLUSIONS The present results show that puerarin is able to suppress EDCs in mouse carotid arteries, independent of inhibition of TP receptor or COX2-derived PGs.


Subject(s)
Carotid Arteries/drug effects , Endothelium, Vascular/drug effects , Isoflavones/pharmacology , Muscle Contraction/drug effects , Muscle, Smooth, Vascular/drug effects , Vasoconstriction/drug effects , Vasodilator Agents/pharmacology , Acetylcholine/pharmacology , Animals , Cyclooxygenase 1/drug effects , Cyclooxygenase 1/metabolism , Cyclooxygenase 2/drug effects , Cyclooxygenase 2/metabolism , Cyclooxygenase Inhibitors/pharmacology , Endothelium, Vascular/metabolism , Enzyme Inhibitors/pharmacology , Membrane Proteins/drug effects , Membrane Proteins/metabolism , Mice , Myography , NG-Nitroarginine Methyl Ester/pharmacology
9.
Drug Deliv ; 27(1): 15-25, 2020 Dec.
Article in English | MEDLINE | ID: mdl-31830840

ABSTRACT

Small interfering RNA (siRNA) exhibits great potential as a novel therapeutic option due to its highly sequence-specific ability to silence genes. However, efficient and safe delivery carriers are required for developing novel therapeutic paradigms. Thus, the successful development of efficient delivery platforms for siRNA is a crucial issue for the development of siRNA-based drugs in cancer treatments. In this study, biocompatible selenium nanoparticles (SeNPs) were loaded with RGDfC peptide to fabricate tumor-targeting gene delivery vehicle RGDfC-SeNPs. Subsequently, RGDfC-SeNPs were loaded with Derlin1-siRNA to fabricate RGDfC-Se@siRNA, which are functionalized selenium nanoparticles. RGDfC-Se@siRNA showed greater uptake in HeLa cervical cancer cells in comparison with that in human umbilical vein endothelial cells (HUVECs), verifying the RGDfC-mediated specific uptake of RGDfC-Se@siRNA. RGDfC-Se@siRNA was capable of entering HeLa cells via clathrin-associated endocytosis, and showed faster siRNA release in a cancer cell microenvironment in comparison with a normal physiological environment. qPCR and western blotting assays both indicated that RGDfC-Se@siRNA exhibited an obvious gene silencing efficacy in HeLa cells. RGDfC-Se@siRNA suppressed the invasion, migration and the proliferation of HeLa cells, and triggered HeLa cell apoptosis. Moreover, RGDfC-Se@siRNA induced the disruption of mitochondrial membrane potentials. Meanwhile, RGDfC-Se@siRNA enhanced the generation of reactive oxygen species (ROS) in HeLa cell, suggesting that mitochondrial dysfunction mediated by ROS might play a significant role in RGDfC-Se@siRNA-induced apoptosis. Interestingly, RGDfC-SeNPs@siRNA exhibited significant antitumor activity in a HeLa tumor-bearing mouse model. Additionally, RGDfC-SeNPs@siRNA is nontoxic to main organ of mouse. The above results indicate that RGDfC-Se@siRNA provides a promising potential for cervical cancer therapy.


Subject(s)
Membrane Proteins/drug effects , Nanoparticles/chemistry , Oligopeptides/pharmacology , RNA, Small Interfering/administration & dosage , Selenium/chemistry , Apoptosis/drug effects , Blotting, Western , Cell Migration Inhibition , Cell Proliferation/drug effects , Female , Gene Silencing/drug effects , HeLa Cells , Human Umbilical Vein Endothelial Cells , Humans , Membrane Potential, Mitochondrial/drug effects , Oligopeptides/administration & dosage , Oligopeptides/pharmacokinetics , Reactive Oxygen Species/metabolism , Tumor Microenvironment , Uterine Cervical Neoplasms/drug therapy
10.
Gynecol Oncol ; 155(2): 349-358, 2019 11.
Article in English | MEDLINE | ID: mdl-31477281

ABSTRACT

OBJECTIVE: Platinum compounds have been widely used as a primary treatment for many types of cancer. However, resistance is the major cause of therapeutic failure for patients with metastatic or recurrent disease, thus highlighting the need to identify novel factors driving resistance to Platinum compounds. Metadherin (MTDH, also known as AEG-1 and LYRIC), located in a frequently amplified region of chromosome 8, has been consistently associated with resistance to chemotherapeutic agents, though the precise mechanisms remain incompletely defined. METHODS: The mRNA of FANCD2 and FANCI was pulled down by RNA-binding protein immunoprecipitation. Pristimerin-loaded nanoparticles were prepared using the nanoprecipitation method. Immunocompromised mice bearing patient-derived xenograft tumors were treated with pristimerin-loaded nanoparticles, cisplatin and a combination of the two. RESULTS: MTDH, through its recently discovered role as an RNA binding protein, regulates expression of FANCD2 and FANCI, two components of the Fanconi anemia complementation group (FA) that play critical roles in interstrand crosslink damage induced by platinum compounds. Pristimerin, a quinonemethide triterpenoid extract from members of the Celastraceae family used to treat inflammation in traditional Chinese medicine, significantly decreased MTDH, FANCD2 and FANCI levels in cancer cells, thereby restoring sensitivity to platinum-based chemotherapy. Using a patient-derived xenograft model of endometrial cancer, we discovered that treatment with pristimerin in a novel nanoparticle formulation markedly inhibited tumor growth when combined with cisplatin. CONCLUSIONS: MTDH is involved in post-transcriptional regulation of FANCD2 and FANCI. Pristimerin can increase sensitivity to platinum-based agents in tumors with MTDH overexpression by inhibiting the FA pathway.


Subject(s)
Antineoplastic Agents/pharmacology , Fanconi Anemia Complementation Group D2 Protein/antagonists & inhibitors , Fanconi Anemia Complementation Group Proteins/antagonists & inhibitors , Membrane Proteins/drug effects , Triterpenes/pharmacology , Animals , Cisplatin/pharmacology , Cystadenocarcinoma, Serous/drug therapy , Down-Regulation , Drug Resistance, Neoplasm/drug effects , Female , Male , Mice, Knockout , Nanoparticles , Pentacyclic Triterpenes , RNA-Binding Proteins , Uterine Neoplasms/drug therapy
11.
Phytother Res ; 33(5): 1562-1569, 2019 May.
Article in English | MEDLINE | ID: mdl-30932261

ABSTRACT

In view of the recognized anti-tumor properties of eugenol against non-small cell lung cancer (NSCLC) in cell culture, here we further set out to investigate the potential therapeutic effect of eugenol in vivo and elucidate the underlying molecular mechanism. The relative expression levels of TRIM59 and p65 in NSCLC were quantified by real-time polymerase chain reaction. Xenograft tumor model was established with TRIM59-deficient H1975 cells, and tumor progression was monitored. Kaplan-Meier's analysis was performed to measure overall survival. Protein levels of TRIM59 and p65 in xenograft tumor were determined by western blot. Direct binding of p65 on the TRIM59 promoter was analyzed by chromatin immunoprecipitation assay, and the regulatory effect was interrogated with luciferase reporter assay. Both TRIM59 and p65 were up-regulated in NSCLC. Eugenol treatment significantly inhibited xenograft tumor progression and prolonged the overall survival of tumor-bearing mice. Mechanistically, eugenol suppressed p65 expression, which subsequently decreased TRIM59 expression. TRIM59 deficiency fully recapitulated the anti-tumoral phenotype elicited by eugenol. Ectopic expression of TRIM59 completely abolished the tumor suppressive effect of eugenol, which underlined the predominant role of TRIM59 in mediating the signaling downstream of eugenol treatment. Eugenol inhibited NSCLC via repression NF-κB-TRIM59 pathway.


Subject(s)
Carcinoma, Non-Small-Cell Lung/drug therapy , Eugenol/chemistry , Lung Neoplasms/drug therapy , Membrane Proteins/drug effects , Metalloproteins/drug effects , NF-kappa B/drug effects , Animals , Carcinoma, Non-Small-Cell Lung/mortality , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cell Proliferation , Disease Models, Animal , Humans , Intracellular Signaling Peptides and Proteins , Lung Neoplasms/mortality , Lung Neoplasms/pathology , Mice , NF-kappa B/metabolism , Survival Rate , Tripartite Motif Proteins , Xenograft Model Antitumor Assays
12.
Life Sci ; 204: 71-77, 2018 Jul 01.
Article in English | MEDLINE | ID: mdl-29738778

ABSTRACT

AIMS: Mutant EGFR Non-small cell lung cancer has benefit from gefitinib, but it has limited effect for wild-type EGFR tumors. Shikonin, a natural naphthoquinone isolated from a traditional Chinese medicine, the plant Lithospermum erythrorhizon (zicao), not only can inhibit the tumor growth, but also overcome cancer drug resistance. Our aim is to investigate whether shikonin can enhance antitumor effect of gefitinib in EGFR wild-type lung cancer cells in vitro and in vivo. MATERIALS AND METHODS: CCK-8 was used to determine the proliferation of EGFR wild-type non-small cell lung cancer. Apoptosis and cell cycle were detected by flow cytometry. PKM2, STAT3, p-STAT3 and cyclinD1 were detected by Western blot. A549 tumor model was established to observe the antitumor effect of shikonin combination with gefitinib in vivo. KEY FINDINGS: The results showed that combination of shikonin with gefitinib exhibited synergistic antitumor effect in vitro and in vivo. Its potential molecular mechanisms may be associated with inhibition of PKM2/STAT3/cyclinD1. SIGNIFICANCE: These results provide a promising therapeutic approach for the treatment of wild-type EGFR non-small cell lung cancer.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Antineoplastic Agents/pharmacology , Carcinoma, Non-Small-Cell Lung/drug therapy , ErbB Receptors/drug effects , Lung Neoplasms/drug therapy , Naphthoquinones/pharmacology , Quinazolines/pharmacology , Signal Transduction/drug effects , A549 Cells , Animals , Carrier Proteins/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Cyclin D1/drug effects , Drug Synergism , Gefitinib , Humans , Immunohistochemistry , Membrane Proteins/drug effects , Mice , Mice, Nude , STAT3 Transcription Factor/drug effects , Sincalide/drug effects , Thyroid Hormones , Thyroid Hormone-Binding Proteins
13.
PLoS Pathog ; 14(2): e1006876, 2018 02.
Article in English | MEDLINE | ID: mdl-29451901

ABSTRACT

The acylphloroglucinol rhodomyrtone is a promising new antibiotic isolated from the rose myrtle Rhodomyrtus tomentosa, a plant used in Asian traditional medicine. While many studies have demonstrated its antibacterial potential in a variety of clinical applications, very little is known about the mechanism of action of rhodomyrtone. Preceding studies have been focused on intracellular targets, but no specific intracellular protein could be confirmed as main target. Using live cell, high-resolution, and electron microscopy we demonstrate that rhodomyrtone causes large membrane invaginations with a dramatic increase in fluidity, which attract a broad range of membrane proteins. Invaginations then form intracellular vesicles, thereby trapping these proteins. Aberrant protein localization impairs several cellular functions, including the respiratory chain and the ATP synthase complex. Being uncharged and devoid of a particular amphipathic structure, rhodomyrtone did not seem to be a typical membrane-inserting molecule. In fact, molecular dynamics simulations showed that instead of inserting into the bilayer, rhodomyrtone transiently binds to phospholipid head groups and causes distortion of lipid packing, providing explanations for membrane fluidization and induction of membrane curvature. Both its transient binding mode and its ability to form protein-trapping membrane vesicles are unique, making it an attractive new antibiotic candidate with a novel mechanism of action.


Subject(s)
Anti-Bacterial Agents/pharmacology , Membrane Fluidity/drug effects , Membrane Proteins/drug effects , Transport Vesicles/drug effects , Xanthones/pharmacology , Anti-Bacterial Agents/pharmacokinetics , Bacillus subtilis/drug effects , Bacillus subtilis/physiology , Bacillus subtilis/ultrastructure , Cell Membrane Permeability/drug effects , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Microbial Sensitivity Tests , Molecular Docking Simulation , Transport Vesicles/metabolism , Xanthones/pharmacokinetics
14.
Clin Toxicol (Phila) ; 56(5): 313-326, 2018 05.
Article in English | MEDLINE | ID: mdl-29124976

ABSTRACT

INTRODUCTION: There is increasing evidence that the pathophysiological target of mercury is in fact selenium, rather than the covalent binding of mercury to sulfur in the body's ubiquitous sulfhydryl groups. The role of selenium in mercury poisoning is multifaceted, bidirectional, and central to understanding the target organ toxicity of mercury. METHODS: An initial search was performed using Medline/PubMed, Toxline, Google Scholar, and Google for published work on mercury and selenium. These searches yielded 2018 citations. Publications that did not evaluate selenium status or evaluated environmental status (e.g., lake or ocean sediment) were excluded, leaving approximately 500 citations. This initial selection was scrutinized carefully and 117 of the most relevant and representative references were selected for use in this review. Binding of mercury to thiol/sulfhydryl groups: Mercury has a lower affinity for thiol groups and higher affinity for selenium containing groups by several orders of magnitude, allowing for binding in a multifaceted way. The established binding of mercury to thiol moieties appears to primarily involve the transport across membranes, tissue distribution, and enhanced excretion, but does not explain the oxidative stress, calcium dyshomeostasis, or specific organ injury seen with mercury. Effects of mercury on selenium and the role this plays in the pathophysiology of mercury toxicity: Mercury impairs control of intracellular redox homeostasis with subsequent increased intracellular oxidative stress. Recent work has provided convincing evidence that the primary cellular targets are the selenoproteins of the thioredoxin system (thioredoxin reductase 1 and thioredoxin reductase 2) and the glutathione-glutaredoxin system (glutathione peroxidase). Mercury binds to the selenium site on these proteins and permanently inhibits their function, disrupting the intracellular redox environment. A number of other important possible target selenoproteins have been identified, including selenoprotein P, K, and T. Impairment of the thioredoxin and glutaredoxin systems allows for proliferation intracellular reactive oxygen species which leads to glutamate excitosis, calcium dyshomeostasis, mitochondrial injury/loss, lipid peroxidation, impairment of protein repair, and apoptosis. Methylmercury is a more potent inhibitor of the thioredoxin system, partially explaining its increased neurotoxicity. A second important mechanism is due to the high affinity of mercury for selenium and the subsequent depletion of selenium stores needed for insertion into de novo generation of replacement selenoproteins. This mercury-induced selenium deficiency state inhibits regeneration of the selenoproteins to restore the cellular redox environment. The effects of selenium on mercury and the role this plays in biological response to mercury: Early research suggested selenium may provide a protective role in mercury poisoning, and with limitations this is true. The roles selenium plays in this reduction of mercury toxicity partially depends on the form of mercury and may be multifaceted including: 1) facilitating demethylation of organic mercury to inorganic mercury; 2) redistribution of mercury to less sensitive target organs; 3) binding to inorganic mercury and forming an insoluble, stable and inert Hg:Se complex; 4) reduction of mercury absorption from the GI tract; 5) repletion of selenium stores (reverse selenium deficiency); and 6) restoration of target selenoprotein activity and restoring the intracellular redox environment. There is conflicting evidence as to whether selenium increases or hinders mercury elimination, but increased mercury elimination does not appear to be a major role of selenium. Selenium supplementation has been shown to restore selenoprotein function and reduce the toxicity of mercury, with several significant limitations including: the form of mercury (methylmercury toxicity is less responsive to amelioration) and mercury dose. CONCLUSIONS: The interaction with selenium is a central feature in mercury toxicity. This interaction is complex depending on a number of features such as the form of mercury, the form of selenium, the organ and dose. The previously suggested "protective effect" of selenium against mercury toxicity may in fact be backwards. The effect of mercury is to produce a selenium deficiency state and a direct inhibition of selenium's role in controlling the intracellular redox environment in organisms. Selenium supplementation, with limitations, may have a beneficial role in restoring adequate selenium status from the deficiency state and mitigating the toxicity of mercury.


Subject(s)
Mercury Poisoning/physiopathology , Selenium/metabolism , Acute Kidney Injury/chemically induced , Humans , Membrane Proteins/drug effects , Mercury/metabolism , Mercury/toxicity , Mercury Poisoning/metabolism , Mitochondria/drug effects , Mitochondria/metabolism , Nerve Tissue Proteins/drug effects , Oxidation-Reduction/drug effects , Selenoproteins/drug effects , Selenoproteins/metabolism
15.
Am J Physiol Endocrinol Metab ; 314(6): E605-E619, 2018 06 01.
Article in English | MEDLINE | ID: mdl-28655718

ABSTRACT

In striated muscle, eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) have differential effects on the metabolism of glucose and differential effects on the metabolism of protein. We have shown that, despite similar incorporation, treatment of C2C12 myotubes (CM) with EPA but not DHA improves glucose uptake and protein accretion. We hypothesized that these differential effects of EPA and DHA may be due to divergent shifts in lipidomic profiles leading to altered proteomic profiles. We therefore carried out an assessment of the impact of treating CM with EPA and DHA on lipidomic and proteomic profiles. Fatty acid methyl esters (FAME) analysis revealed that both EPA and DHA led to similar but substantials changes in fatty acid profiles with the exception of arachidonic acid, which was decreased only by DHA, and docosapentanoic acid (DPA), which was increased only by EPA treatment. Global lipidomic analysis showed that EPA and DHA induced large alterations in the cellular lipid profiles and in particular, the phospholipid classes. Subsequent targeted analysis confirmed that the most differentially regulated species were phosphatidylcholines and phosphatidylethanolamines containing long-chain fatty acids with five (EPA treatment) or six (DHA treatment) double bonds. As these are typically membrane-associated lipid species we hypothesized that these treatments differentially altered the membrane-associated proteome. Stable isotope labeling by amino acids in cell culture (SILAC)-based proteomics of the membrane fraction revealed significant divergence in the effects of EPA and DHA on the membrane-associated proteome. We conclude that the EPA-specific increase in polyunsaturated long-chain fatty acids in the phospholipid fraction is associated with an altered membrane-associated proteome and these may be critical events in the metabolic remodeling induced by EPA treatment.


Subject(s)
Docosahexaenoic Acids/pharmacology , Eicosapentaenoic Acid/pharmacology , Glucose/metabolism , Lipid Metabolism/drug effects , Membrane Proteins/drug effects , Muscle, Skeletal/drug effects , Proteome/drug effects , Animals , Carbohydrate Metabolism/drug effects , Cell Membrane/drug effects , Cell Membrane/metabolism , Cells, Cultured , Eicosapentaenoic Acid/analogs & derivatives , Fatty Acids/metabolism , Membrane Proteins/metabolism , Mice , Muscle, Skeletal/metabolism , Proteome/metabolism , Triglycerides/metabolism
16.
Kidney Blood Press Res ; 42(3): 530-540, 2017.
Article in English | MEDLINE | ID: mdl-28854437

ABSTRACT

BACKGROUND/AIMS: Renal damage and dysfunction is attributed to sustained hyperglycaemia in overt diabetes. Subcutaneous insulin injections are beneficial in delaying the progression of renal dysfunction and damage in diabetics. However, the current mode of administration is associated with severe undesirable effects. In this study, we evaluated the ameliorative effects of pectin-insulin dermal patches on renal dysfunction in diabetes. METHODS: Pectin-insulin patches (20.0, 40.8 and 82.9 µg/kg) were applied on the skin of streptozotocin-induced diabetic rats, thrice daily for 5 weeks. Blood glucose concentration, blood pressure and urine output volume were recorded on week 5 after which the animals were sacrificed after which the kidneys and plasma were collected. Kidney nephrin expression and urinary nephrin concentration, albumin excretion rate (AER), creatinine clearance (CC) and albumin creatinine ratio (ACR) were assessed. RESULTS: Patch application resulted in reduced blood glucose concentration and blood pressure. Furthermore, pectin-insulin patch treatment resulted in increased kidney nephrin expression and reduced urinary nephrin concentration. AER, CC ACR were also reduced post patch application. CONCLUSIONS: The application of pectin-insulin patch limited diabetes associated kidney damaged and improved kidney function. These observations suggest that pectin-insulin patches may ameliorate kidney dysfunction that is associated with chronic subcutaneous insulin administration.


Subject(s)
Diabetes Mellitus, Experimental/chemically induced , Diabetic Nephropathies/drug therapy , Insulin/administration & dosage , Pectins/administration & dosage , Animals , Blood Glucose/drug effects , Blood Pressure/drug effects , Membrane Proteins/drug effects , Membrane Proteins/metabolism , Rats , Streptozocin , Transdermal Patch
17.
Am J Chin Med ; 45(1): 85-104, 2017.
Article in English | MEDLINE | ID: mdl-28081630

ABSTRACT

Notch signaling has been demonstrated to be involved in ductular reactions and fibrosis. Previous studies have shown that Huang Qi Decoction (HQD) can prevent the progression of cholestatic liver fibrosis (CLF). However, whether HQD affects the Notch signaling pathway is unclear. In this study, CLF was established by common bile duct ligation (BDL) in rats. At the end of the first week, the rats were randomly divided into a model group (i.e., BDL), an HQD group, and a sorafenib positive control group (SORA) and were treated for 3 weeks. Bile duct proliferation and liver fibrosis were determined by tissue staining. Activation of the Notch signaling pathway was evaluated by analyzing expressions of Notch-1, -2, -3, and -4, Jagged (JAG) 1, and Delta like (DLL)-1, -3, and -4. The results showed that HQD significantly reduced the deposition of collagen and the Hyp content of liver tissue and inhibited the activation of HSCs compared with the BDL group. In addition, HQD significantly decreased the protein and mRNA expressions of TGF-[Formula: see text]1 and [Formula: see text]-SMA. In contrast, HQD significantly enhanced expression of the Smad 7 protein. HQD also reduced biliary epithelial cell proliferation, and reduced the mRNA levels of CK7, CK8, CK18, SRY-related high mobility group-box gene (SOX) 9, epithelial cell adhesion molecule (EpCAM) and the positive areas of CK19 and OV6. In addition, the mRNA and protein expressions of Notch-3, -4, JAG1, and DLL-1, -3 were significantly reduced in the HQD compared to the BDL group. These results demonstrated that HQD may prevent biliary liver fibrosis through inhibition of the Notch signaling pathway, and it may be a potential treatment for cholestatic liver disease.


Subject(s)
Drugs, Chinese Herbal/pharmacology , Liver/drug effects , RNA, Messenger/drug effects , Receptors, Notch/drug effects , Actins/drug effects , Actins/genetics , Actins/metabolism , Animals , Astragalus propinquus , Biliary Tract/drug effects , Cell Proliferation/drug effects , Cholestasis/etiology , Cholestasis/prevention & control , Collagen/drug effects , Collagen/metabolism , Common Bile Duct/surgery , Disease Models, Animal , Epithelial Cell Adhesion Molecule/drug effects , Epithelial Cell Adhesion Molecule/genetics , Epithelial Cells/drug effects , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Jagged-1 Protein/drug effects , Jagged-1 Protein/genetics , Jagged-1 Protein/metabolism , Keratins/drug effects , Keratins/genetics , Ligation , Liver/metabolism , Liver Cirrhosis/etiology , Liver Cirrhosis/prevention & control , Membrane Proteins/drug effects , Membrane Proteins/genetics , Membrane Proteins/metabolism , Rats , Receptors, Notch/genetics , Receptors, Notch/metabolism , Signal Transduction/drug effects , Smad7 Protein/drug effects , Transforming Growth Factor beta1/drug effects , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/metabolism
18.
Int J Pediatr Otorhinolaryngol ; 92: 61-66, 2017 Jan.
Article in English | MEDLINE | ID: mdl-28012535

ABSTRACT

OBJECTIVE: Cisplatin is commonly used to treat solid tumors. However, permanent hearing loss is a major side effect of cisplatin chemotherapy and often results in dose reduction of the cisplatin chemotherapy. Peanut sprouts show cytoprotective properties owing to their antioxidant activities. This study was designed to investigate the effect of peanut sprout extract (PSE) on cisplatin-induced ototoxicity in an auditory cell line, HEI-OC1 cells. METHODS: Cells were exposed to cisplatin for 24 h, with or without pre-treatment with PSE, cell viability was examined using the MTT assay. Apoptotic cells were identified by double staining with Hoechst 33258 and propidium iodide. Western blot analysis was performed to examine apoptotic proteins including C-PARP and C-caspase, anti-apoptotic protein Bcl-2, and Nrf2 redox system activation. Mitochondrial reactive oxygen species (ROS) were investigated to examine whether PSE could scavenge cisplatin-induced ROS. Real-time PCR analyses were performed to investigate the mRNA levels of antioxidant enzymes including NQO1, HO-1, GPx2, Gclc, and catalase. RESULTS: The cisplatin-treated group showed reduced cell viability, increased apoptotic properties and markers, and increased ROS levels. PSE pre-treatment before cisplatin exposure significantly increased cell viability and reduced apoptotic properties and ROS production. These effects resulted from the up-regulation of antioxidant genes, including NQO1, HO-1, GPx2, Gclc, and catalase through Akt phosphorylation and Nrf2 activation. CONCLUSION: Our results demonstrate that PSE protects from cisplatin-induced cytotoxicity by activating the antioxidant effects via the Akt/Nrf-2 pathway in this auditory cell line, and indicate that PSE may provide novel treatment to prevent cisplatin-induced ototoxicity.


Subject(s)
Antineoplastic Agents/toxicity , Apoptosis/drug effects , Arachis , Cisplatin/toxicity , Hair Cells, Auditory/drug effects , Labyrinth Supporting Cells/drug effects , Plant Extracts/pharmacology , Seedlings , Animals , Blotting, Western , Caspases/drug effects , Caspases/metabolism , Catalase/drug effects , Catalase/genetics , Cell Line , Cell Survival/drug effects , Glutamate-Cysteine Ligase/drug effects , Glutamate-Cysteine Ligase/genetics , Glutathione Peroxidase/drug effects , Glutathione Peroxidase/genetics , Heme Oxygenase-1/drug effects , Heme Oxygenase-1/genetics , In Vitro Techniques , Membrane Proteins/drug effects , Membrane Proteins/genetics , Mice , Mitochondria/drug effects , Mitochondria/metabolism , NAD(P)H Dehydrogenase (Quinone)/drug effects , NAD(P)H Dehydrogenase (Quinone)/genetics , NF-E2-Related Factor 2/drug effects , NF-E2-Related Factor 2/metabolism , Oxidation-Reduction/drug effects , Poly(ADP-ribose) Polymerases/drug effects , Poly(ADP-ribose) Polymerases/metabolism , Proto-Oncogene Proteins c-bcl-2/drug effects , Proto-Oncogene Proteins c-bcl-2/metabolism , Reactive Oxygen Species/metabolism , Real-Time Polymerase Chain Reaction , Up-Regulation
19.
Endocrinology ; 157(2): 799-809, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26646203

ABSTRACT

The WNT pathway was shown to play an important role in the adult central nervous system. We previously identified the WNT pathway as a novel integration site of the adipokine leptin in mediating its neuroendocrine control of metabolism in obese mice. Here we investigated the implication of WNT signaling in seasonal body weight regulation exhibited by the Djungarian hamster (Phodopus sungorus), a seasonal mammal that exhibits profound annual changes in leptin sensitivity. We furthermore investigated whether crucial components of the WNT pathway are regulated in a diurnal manner. Gene expression of key components of the WNT pathway in the hypothalamus of hamsters acclimated to either long day (LD) or short day (SD) photoperiod was analyzed by in situ hybridization. We detected elevated expression of the genes WNT-4, Axin-2, Cyclin-D1, and SFRP-2, in the hypothalamic arcuate nucleus, a key energy balance integration site, during LD compared with SD as well as a diurnal regulation of Axin-2, Cyclin-D1, and DKK-3. Investigating the effect of photoperiod as well as leptin on the activation (phosphorylation) of the WNT coreceptor LRP-6-(Ser1490) by immunohistochemistry, we found elevated activity in the arcuate nucleus during LD relative to SD as well as after leptin treatment (2 mg/kg body weight). These findings indicate that differential WNT signaling may be associated with seasonal body weight regulation and is partially regulated in a diurnal manner in the adult brain. Furthermore, they suggest that this pathway plays a key role in the neuroendocrine regulation of body weight and integration of the leptin signal.


Subject(s)
Arcuate Nucleus of Hypothalamus/metabolism , Axin Protein/genetics , Body Weight/genetics , Circadian Rhythm/genetics , Cyclin D1/genetics , Photoperiod , Wnt Signaling Pathway/genetics , Wnt4 Protein/genetics , Animals , Arcuate Nucleus of Hypothalamus/drug effects , Axin Protein/drug effects , Axin Protein/metabolism , Body Weight/drug effects , Circadian Rhythm/drug effects , Cricetinae , Cyclin D1/drug effects , Cyclin D1/metabolism , Energy Metabolism/drug effects , Energy Metabolism/genetics , Female , Gene Expression Profiling , Hypothalamus/drug effects , Hypothalamus/metabolism , Immunohistochemistry , In Situ Hybridization , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Leptin/pharmacology , Membrane Proteins/drug effects , Membrane Proteins/genetics , Membrane Proteins/metabolism , Phodopus , Seasons , Wnt Signaling Pathway/drug effects , Wnt4 Protein/drug effects , Wnt4 Protein/metabolism
20.
PLoS One ; 10(11): e0142401, 2015.
Article in English | MEDLINE | ID: mdl-26551613

ABSTRACT

Because of the perpetual development of resistance to current therapies for malaria, the Medicines for Malaria Venture developed the Malaria Box to facilitate the drug development process. We tested the 80 most potent compounds from the box for bilayer-mediated effects on membrane protein conformational changes (a measure of likely toxicity) in a gramicidin-based stopped flow fluorescence assay. Among the Malaria Box compounds tested, four compounds altered membrane properties (p< 0.05); MMV007384 stood out as a potent bilayer-perturbing compound that is toxic in many cell-based assays, suggesting that testing for membrane perturbation could help identify toxic compounds. In any case, MMV007384 should be approached with caution, if at all.


Subject(s)
Antimalarials/pharmacology , Antimalarials/toxicity , Lipid Bilayers/chemistry , Biophysical Phenomena , Cell Line , Cell Membrane/chemistry , Cell Membrane/drug effects , Drug Discovery , Drug Evaluation, Preclinical , Humans , Membrane Proteins/chemistry , Membrane Proteins/drug effects , Protein Conformation/drug effects , Spectrometry, Fluorescence
SELECTION OF CITATIONS
SEARCH DETAIL