Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 452
Filter
Add more filters

Complementary Medicines
Country/Region as subject
Publication year range
1.
Cell Mol Life Sci ; 79(4): 193, 2022 Mar 17.
Article in English | MEDLINE | ID: mdl-35298717

ABSTRACT

Aberrant insulin-like growth factor 1 (IGF-1) signaling has been proposed as a contributing factor to the development of neurodegenerative disorders including diabetic neuropathy, and delivery of exogenous IGF-1 has been explored as a treatment for Alzheimer's disease and amyotrophic lateral sclerosis. However, the role of autocrine/paracrine IGF-1 in neuroprotection has not been well established. We therefore used in vitro cell culture systems and animal models of diabetic neuropathy to characterize endogenous IGF-1 in sensory neurons and determine the factors regulating IGF-1 expression and/or affecting neuronal health. Single-cell RNA sequencing (scRNA-Seq) and in situ hybridization analyses revealed high expression of endogenous IGF-1 in non-peptidergic neurons and satellite glial cells (SGCs) of dorsal root ganglia (DRG). Brain cortex and DRG had higher IGF-1 gene expression than sciatic nerve. Bidirectional transport of IGF-1 along sensory nerves was observed. Despite no difference in IGF-1 receptor levels, IGF-1 gene expression was significantly (P < 0.05) reduced in liver and DRG from streptozotocin (STZ)-induced type 1 diabetic rats, Zucker diabetic fatty (ZDF) rats, mice on a high-fat/ high-sugar diet and db/db type 2 diabetic mice. Hyperglycemia suppressed IGF-1 gene expression in cultured DRG neurons and this was reversed by exogenous IGF-1 or the aldose reductase inhibitor sorbinil. Transcription factors, such as NFAT1 and CEBPß, were also less enriched at the IGF-1 promoter in DRG from diabetic rats vs control rats. CEBPß overexpression promoted neurite outgrowth and mitochondrial respiration, both of which were blunted by knocking down or blocking IGF-1. Suppression of endogenous IGF-1 in diabetes may contribute to neuropathy and its upregulation at the transcriptional level by CEBPß can be a promising therapeutic approach.


Subject(s)
Aging/metabolism , Axons/pathology , CCAAT-Enhancer-Binding Protein-beta/metabolism , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Energy Metabolism , Insulin-Like Growth Factor I/metabolism , Sensory Receptor Cells/metabolism , Animals , Antibodies, Neutralizing/pharmacology , Axons/drug effects , Axons/metabolism , Base Sequence , CCAAT-Enhancer-Binding Protein-beta/genetics , Cell Respiration/drug effects , Cells, Cultured , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/pathology , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/pathology , Energy Metabolism/drug effects , Ganglia, Spinal/drug effects , Ganglia, Spinal/metabolism , Gene Expression Regulation/drug effects , Glycolysis/drug effects , HEK293 Cells , Humans , Insulin-Like Growth Factor I/genetics , Liver/metabolism , Male , Mitochondria/drug effects , Mitochondria/metabolism , NFATC Transcription Factors/metabolism , Neuronal Outgrowth/drug effects , Polymers/metabolism , Promoter Regions, Genetic/genetics , Protein Transport/drug effects , Rats, Sprague-Dawley , Sensory Receptor Cells/pathology , Signal Transduction/drug effects
2.
Eur J Pharmacol ; 910: 174458, 2021 Nov 05.
Article in English | MEDLINE | ID: mdl-34480884

ABSTRACT

Abnormal melanogenesis and melanosome transport can cause skin pigmentation disorders that are often treated using ginseng-based formulation. We previously found that phenolic acid compounds in ginseng root could inhibit melanin production and as a skin-whitening agents. However, mechanisms of action underlying effects of ginseng phenolic acid monomers on melanogenesis remain unclear. This study was conducted to investigate effects of salicylic acid, a main ginseng root phenolic acid component, on melanogenesis and melanosome functions in melanocytes of zebrafish and other species. Salicylic acid exhibited no cytotoxicity and reduced melanin levels and tyrosinase activity in B16F10 murine melanoma cells and normal human epidermal melanocytes regardless of prior cell stimulation with α-melanocyte stimulating hormone. Additionally, salicylic acid treatment reduced expression of melanogenic enzymes tyrosinase, tyrosinase-related protein 1 and tyrosinase-related protein 2, while reducing expression of their master transcriptional regulator, microphthalmia-associated transcription factor. Moreover, reduced phosphorylation of cAMP response-element binding protein was observed due to reduced cAMP levels resulting from salicylic acid inhibition of upstream signal regulators (adenylyl cyclase and protein kinase A). Furthermore, salicylic acid treatment suppressed expression of transport complex-associated proteins melanophilin and myosin Va in two UVB-treated melanocytic cell lines, suppressed phagocytosis of fluorescent microspheres by UVB-stimulated human keratinocytes (HaCaT), inhibited protease-activated receptor 2 activation by reducing both Ca2+ release and activation of phosphoinositide 3 kinase/AKT and mitogen-activated protein kinases and induced anti-melanogenic effects in zebrafish. Collectively, these results indicate that salicylic acid within ginseng root can inhibit melanocyte melanogenesis and melanin transport, while also suppressing keratinocyte phagocytic function.


Subject(s)
Hyperpigmentation/drug therapy , Melanins/metabolism , Melanosomes/metabolism , Panax/chemistry , Salicylic Acid/pharmacology , Animals , Calcium/metabolism , Cell Line , Cyclic AMP/antagonists & inhibitors , Cyclic AMP-Dependent Protein Kinases/metabolism , Humans , Intramolecular Oxidoreductases/metabolism , Keratinocytes/drug effects , Melanins/antagonists & inhibitors , Melanocytes/drug effects , Melanosomes/drug effects , Mice , Microphthalmia-Associated Transcription Factor/metabolism , Monophenol Monooxygenase/antagonists & inhibitors , Monophenol Monooxygenase/metabolism , Oxidoreductases/metabolism , Phagocytosis/drug effects , Protein Transport/drug effects , Receptor, PAR-2/metabolism , Signal Transduction/drug effects , Ultraviolet Rays , Zebrafish , alpha-MSH/pharmacology
3.
Molecules ; 26(14)2021 Jul 18.
Article in English | MEDLINE | ID: mdl-34299620

ABSTRACT

Type 2 diabetes mellitus (T2DM) is linked to insulin resistance and a loss of insulin sensitivity, leading to millions of deaths worldwide each year. T2DM is caused by reduced uptake of glucose facilitated by glucose transporter 4 (GLUT4) in muscle and adipose tissue due to decreased intracellular translocation of GLUT4-containing vesicles to the plasma membrane. To treat T2DM, novel medications are required. Through a fluorescence microscopy-based high-content screen, we tested more than 600 plant extracts for their potential to induce GLUT4 translocation in the absence of insulin. The primary screen in CHO-K1 cells resulted in 30 positive hits, which were further investigated in HeLa and 3T3-L1 cells. In addition, full plasma membrane insertion was examined by immunostaining of the first extracellular loop of GLUT4. The application of appropriate inhibitors identified PI3 kinase as the most important signal transduction target relevant for GLUT4 translocation. Finally, from the most effective hits in vitro, four extracts effectively reduced blood glucose levels in chicken embryos (in ovo), indicating their applicability as antidiabetic pharmaceuticals or nutraceuticals.


Subject(s)
Blood Glucose/drug effects , Glucose/metabolism , Hypoglycemic Agents/pharmacology , Insulin/pharmacology , Plant Extracts/pharmacology , 3T3-L1 Cells , Adipocytes/drug effects , Adipocytes/metabolism , Animals , CHO Cells , Cell Line , Cell Line, Tumor , Cell Membrane/drug effects , Cell Membrane/metabolism , Cricetulus , Diabetes Mellitus, Type 2 , Glucose Transporter Type 4/metabolism , HeLa Cells , Humans , Insulin Resistance/physiology , Mice , Protein Transport/drug effects , Signal Transduction/drug effects
4.
Biochem Pharmacol ; 190: 114622, 2021 08.
Article in English | MEDLINE | ID: mdl-34043967

ABSTRACT

Lung cancer is the leading cause of cancer deaths in the world. Non-small cell lung cancer (NSCLC), with poor prognosis and resistance to chemoradiotherapy, is the most common histological type of lung cancer. Therefore, it is necessary to develop new and more effective treatment strategy for NSCLC. Nur77, an orphan member of the nuclear receptor superfamily, induces apoptosis in cancer cells including NSCLC cells, by high expression and translocation to mitochondria. Small molecules trigger expression and mitochondrial localization of Nur77 may be an ideal anti-cancer drug candidate. Here, we report malayoside, a cardiac glycoside in the extract of Antiaris toxicaria Lesch., had different sensitivities to NSCLC cells. Malayoside induced apoptosis in NCI-H460 cells. Meanwhile, malayoside induced Nur77 expression and mitochondrial localization, and its induction of apoptosis was Nur77-dependent. To investigate the molecular mechanism of malayoside inducing Nur77 and apoptosis, we found that malayoside activated MAPK signaling pathway, including both ERK and p38 phosphorylation. The suppression of MAPK signaling activation inhibited the expression of Nur77 and apoptosis induced by malayoside. Our studies in nude mice showed that malayside potently inhibited the growth of tumor cells in vivo. Furthermore, the anti-cancer effect of malayosidwas in vivo was also related to the elevated expression of Nur77, p-ERK, and p-p38 proteins. Our results suggest that malayoside possesses an anti-NSCLC activity in vitro and in vivo mainly via activation of MAPK-Nur77 signaling pathway, indicating that malayoside is a promising chemotherapeutic candidate for NSCLC.


Subject(s)
Antiaris/chemistry , Apoptosis/drug effects , Cardiac Glycosides/pharmacology , Mitogen-Activated Protein Kinase Kinases/metabolism , Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism , Animals , Carcinoma, Non-Small-Cell Lung , Cardiac Glycosides/chemistry , Cell Line, Tumor , Cell Proliferation , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Mice , Mice, Nude , Nuclear Receptor Subfamily 4, Group A, Member 1/genetics , Phytotherapy , Protein Transport/drug effects
5.
Oxid Med Cell Longev ; 2021: 8839479, 2021.
Article in English | MEDLINE | ID: mdl-33747350

ABSTRACT

Black berry (Syzygium cumini) fruit is useful in curing diabetic complications; however, its role in diabetes-induced cardiomyopathy is not yet known. In this study, we investigated the regulation of gelatinase-B (MMP-9) by S. cumini methanol seed extract (MSE) in diabetic cardiomyopathy using real-time PCR, RT-PCR, immunocytochemistry, gel diffusion assay, and substrate zymography. The regulatory effects of MSE on NF-κB, TNF-α, and IL-6 were also examined. Identification and estimation of polyphenol constituents present in S. cumini extract were carried out using reverse-phase HPLC. Further, in silico docking studies of identified polyphenols with gelatinase-B were performed to elucidate molecular level interaction in the active site of gelatinase-B. Docking studies showed strong interaction of S. cumini polyphenols with gelatinase-B. Our findings indicate that MSE significantly suppresses gelatinase-B expression and activity in high-glucose- (HG-) stimulated cardiomyopathy. Further, HG-induced activation of NF-κB, TNF-α, and IL-6 was also remarkably reduced by MSE. Our results suggest that S. cumini MSE may be useful as an effective functional food and dietary supplement to regulate HG-induced cardiac stress through gelatinase.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Hyperglycemia/pathology , Matrix Metalloproteinase 9/metabolism , Myocytes, Cardiac/pathology , Oxidative Stress , Plant Extracts/pharmacology , Seeds/chemistry , Syzygium/chemistry , Animals , Cell Line , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Chromatography, High Pressure Liquid , Gene Expression Regulation, Enzymologic/drug effects , Glucose , Hyperglycemia/genetics , Inflammation/pathology , Interleukin-6/metabolism , Matrix Metalloproteinase 9/genetics , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/enzymology , NF-kappa B/metabolism , Oxidative Stress/drug effects , Phenols/pharmacology , Protein Transport/drug effects , Rats , Substrate Specificity/drug effects , Thermodynamics , Tumor Necrosis Factor-alpha/metabolism
6.
J Biochem Mol Toxicol ; 35(6): 1-10, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33720461

ABSTRACT

Lung cancer is a noxious disease with substandard overall survival. Despite this, there are several treatment strategies for lung cancer include chemotherapy, radiotherapy, surgery; however, the overall survival remains poor. Punicalagin has been documented as a potential phytomedicine to selectively inhibit the progression and expansion of numerous cancers. In the present study, we evaluated the antiproliferative ability of punicalagin against lung cancer A549 cells by inducing apoptosis by inhibiting STAT-3 activation. Punicalagin induces toxic effects of A549 cells in a dose-associated manner after 24 h treatment. And we also observed that punicalagin (10, 20, and 30 µM) induced reactive oxygen species generation, alters the mitochondrion membrane potential and apoptotic morphological changes in A549 cells. The STAT-3 overexpression regulates apoptosis, proliferation, and angiogenesis. Here, the punicalagin inhibited STAT-3 translocation and thereby induces apoptosis by inhibiting expression Bcl-2 and enhanced expression of Bax, cytochrome-c, caspase-9, and caspase-3 in A549 cells. Hence, we stated that the punicalagin is a possible therapy for non-small cell lung, malignancies. Altogether, the punicalagin is a promising phytomedicine in malignancy treatment and further endeavors are needed to unveil the complete potential.


Subject(s)
Apoptosis/drug effects , Hydrolyzable Tannins/pharmacology , Lung Neoplasms , Neoplasm Proteins/metabolism , Reactive Oxygen Species/metabolism , STAT3 Transcription Factor/metabolism , A549 Cells , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Protein Transport/drug effects
7.
Oxid Med Cell Longev ; 2021: 1246491, 2021.
Article in English | MEDLINE | ID: mdl-33613822

ABSTRACT

Anthocyanins from the petals of Hibiscus syriacus L. (PS) possess anti-inflammatory, antioxidant, and antimelanogenic activities. However, it remains unclear whether PS inhibit the NLR family pyrin domain-containing 3 (NLRP3) inflammasome activation and assembly. This study is aimed at investigating whether PS downregulate NLRP3-mediated inflammasome by inhibiting nuclear factor-κB (NF-κB) and endoplasmic reticulum (ER) stress. BV2 microglia cells were treated with PS in the presence of lipopolysaccharide and adenosine triphosphate (LPS/ATP), and the NLRP3-related signaling pathway was investigated. In this study, we found that LPS/ATP treatment activated the NLRP3 inflammasome, which resulted in the release of interleukin-1ß (IL-1ß) and IL-18. Meanwhile, PS reduced LPS/ATP-mediated NLRP3 inflammasome at 12 h by inhibiting ER stress-mediated Ca2+ accumulation and subsequent mitochondrial reactive oxygen species (mtROS) production, which, in turn, decreased IL-1ß and IL-18 release. Furthermore, PS inhibited the NLRP3 inflammasome 1 h after LPS/ATP treatment by suppressing the NF-κB pathway, which downregulated Ca2+ accumulation and mtROS production. These data showed that PS negatively regulated activation of the NLRP3 inflammasome in a time-different manner by inhibiting the NF-κB signaling pathway in the early stage and the ER stress response in the late stage. The pathways shared Ca2+ accumulation-mediated mtROS production, which was significantly inhibited in the presence of PS. In conclusion, our results suggested that PS has potential as a supplement against NLRP3 inflammasome-related inflammatory disorders; nevertheless, further studies are needed to determine the effect of PS in the noncanonical NLRP3 inflammasome pathways and pathological conditions in vivo.


Subject(s)
Anthocyanins/pharmacology , Calcium/metabolism , Endoplasmic Reticulum Stress , Hibiscus/chemistry , Microglia/metabolism , Mitochondria/metabolism , NF-kappa B/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Adenosine Triphosphate/pharmacology , Animals , Apoptosis/drug effects , Cell Line , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Down-Regulation/drug effects , Endoplasmic Reticulum Stress/drug effects , Flowers/chemistry , Inflammasomes/metabolism , Interleukin-18/metabolism , Interleukin-1beta/metabolism , Lipopolysaccharides/pharmacology , Membrane Potential, Mitochondrial/drug effects , Mice , Mitochondria/drug effects , Models, Biological , Protein Transport/drug effects , Reactive Oxygen Species/metabolism
8.
Nutrients ; 13(2)2021 Jan 29.
Article in English | MEDLINE | ID: mdl-33572926

ABSTRACT

Rosa canina L. is a natural polyphenol-rich medicinal plant that exhibits antioxidant and anti-inflammatory activities. Recent in vivo studies have demonstrated that a methanol extract of Rosa canina L. (RCME) has reversed an inflammatory bowel disease (IBD)-like phenotype that has been triggered by dextran sulfate sodium (DSS) in mice. In the current study, we investigated the effects of RCME on perturbations of cellular mechanisms induced by DSS-treatment of intestinal Caco-2 cells, including stress response in the endoplasmic reticulum (ER), protein trafficking and sorting as well as lipid rafts integrity and functional capacities of an intestinal enzyme. 6 days post-confluent cells were treated for 24 h with DSS (3%) or simultaneously with DSS (3%) and RCME (100 µg/mL) or exclusively with RCME (100 µg/mL) or not treated. The results obtained demonstrate the ability of RCME to counteract the substantial increase in the expression levels of several ER stress markers in DSS-treated cells. Concomitantly, the delayed trafficking of intestinal membrane glycoproteins sucrase-isomaltase (SI) and dipeptidyl peptidase 4 (DPP4) induced by DSS between the ER and the Golgi has been compromised by RCME. Furthermore, RCME restored the partially impaired polarized sorting of SI and DPP4 to the brush border membrane. An efficient sorting mechanism of SI and DPP4 is tightly associated with intact lipid rafts structures in the trans-Golgi network (TGN), which have been distorted by DSS and normalized by RCME. Finally, the enzymatic activities of SI are enhanced in the presence of RCME. Altogether, DSS treatment has triggered ER stress, impaired trafficking and function of membrane glycoproteins and distorted lipid rafts, all of which can be compromised by RCME. These findings indicate that the antioxidants in RCME act at two major sites in Caco-2 cells, the ER and the TGN and are thus capable of maintaining the membrane integrity by correcting the sorting of membrane-associated proteins.


Subject(s)
Endoplasmic Reticulum/drug effects , Inflammatory Bowel Diseases/therapy , Methanol/pharmacology , Plant Extracts/pharmacology , Protein Transport/drug effects , Rosa/chemistry , Animals , Caco-2 Cells , Dextran Sulfate , Dipeptidyl Peptidase 4/metabolism , Disease Models, Animal , Humans , Inflammatory Bowel Diseases/chemically induced , Intestinal Mucosa/drug effects , Membrane Microdomains/metabolism , Mice , Microvilli/metabolism , Phenotype , Sucrase-Isomaltase Complex/metabolism
9.
Sci Rep ; 11(1): 4537, 2021 02 25.
Article in English | MEDLINE | ID: mdl-33633156

ABSTRACT

Vasopressin (AVP) increases water permeability in the renal collecting duct through the regulation of aquaporin-2 (AQP2) trafficking. Several disorders, including hypertension and inappropriate antidiuretic hormone secretion (SIADH), are associated with abnormalities in water homeostasis. It has been shown that certain phytocompounds are beneficial to human health. Here, the effects of the Olive Leaf Extract (OLE) have been evaluated using in vitro and in vivo models. Confocal studies showed that OLE prevents the vasopressin induced AQP2 translocation to the plasma membrane in MCD4 cells and rat kidneys. Incubation with OLE decreases the AVP-dependent increase of the osmotic water permeability coefficient (Pf). To elucidate the possible effectors of OLE, intracellular calcium was evaluated. OLE increases the intracellular calcium through the activation of the Calcium Sensing Receptor (CaSR). NPS2143, a selective CaSR inhibitor, abolished the inhibitory effect of OLE on AVP-dependent water permeability. In vivo experiments revealed that treatment with OLE increases the expression of the CaSR mRNA and decreases AQP2 mRNA paralleled by an increase of the AQP2-targeting miRNA-137. Together, these findings suggest that OLE antagonizes vasopressin action through stimulation of the CaSR indicating that this extract may be beneficial to attenuate disorders characterized by abnormal CaSR signaling and affecting renal water reabsorption.


Subject(s)
Aquaporin 2/metabolism , Olea/chemistry , Plant Extracts/pharmacology , Plant Leaves/chemistry , Receptors, Calcium-Sensing/agonists , Vasopressins/pharmacology , Animals , Cell Line , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Humans , Kidney Tubules, Collecting/cytology , Kidney Tubules, Collecting/metabolism , Plant Extracts/chemistry , Protein Transport/drug effects , Rats , Receptors, Calcium-Sensing/metabolism
10.
Neuropeptides ; 87: 102134, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33639357

ABSTRACT

Parkinson's disease (PD) is the second most common neurodegenerative disorder, characterized by the loss of dopaminergic neurons in the substantia nigra and the deposition of Lewy bodies. Mitochondrial dysfunction, oxidative stress, and autophagy dysfunction are involved in the pathogenesis of PD. Ghrelin is a brain-gut peptide that has been reported that protected against 1-methyl-4-phenyl-1,2,3,6- tetrahydropyran (MPTP)/MPP+-induced toxic effects. In the present work, human neuroblastoma SH-SY5Y cells were exposed to rotenone as a PD model to explore the underlying mechanism of ghrelin. We found that ghrelin inhibited rotenone-induced cytotoxicity, mitochondrial dysfunction, and apoptosis by improving cell viability, increasing the ratio of red/green of JC-1, inhibiting the production of reactive oxidative species (ROS), and regulating Bcl-2, Bax, Cytochrome c, caspase-9, and caspase-3 expression. Besides, ghrelin promoted mitophagy accompanied by up-regulating microtubule-associated protein 1 Light Chain 3B-II/I(LC3B-II/I) and Beclin1 but decreasing the expression of p62. Moreover, ghrelin promoted PINK1/Parkin mitochondrial translocation. Additionally, we investigated that ghrelin activated the AMPK/SIRT1/PGC1α pathway and pharmacological inhibition of AMPK and SIRT1 abolished the cytoprotection of ghrelin, decreased the level of mitophagy, and PINK1/Parkin mitochondrial translocation. Taken together, our findings suggested that mitophagy and AMPK/SIRT1/PGC1α pathways were related to the cytoprotection of ghrelin. These findings provided novel insights into the underlying mechanisms of ghrelin, further mechanistic studies on preclinical and clinical levels are required to be conducted with ghrelin to avail and foresee it as a potential agent in the treatment and management of PD.


Subject(s)
Ghrelin/physiology , Mitochondria/drug effects , Mitophagy/physiology , Nerve Tissue Proteins/physiology , Rotenone/toxicity , Signal Transduction/drug effects , AMP-Activated Protein Kinases/antagonists & inhibitors , AMP-Activated Protein Kinases/physiology , Apoptosis/drug effects , Cell Line, Tumor , Drug Evaluation, Preclinical , Gene Expression Regulation/drug effects , Ghrelin/pharmacology , Humans , Membrane Potential, Mitochondrial/drug effects , Mitochondria/metabolism , Nerve Tissue Proteins/antagonists & inhibitors , Neuroblastoma , Oxidative Stress/drug effects , Parkinson Disease , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/physiology , Protein Kinases/metabolism , Protein Transport/drug effects , Reactive Oxygen Species , Rotenone/antagonists & inhibitors , Sirtuin 1/antagonists & inhibitors , Sirtuin 1/physiology , Ubiquitin-Protein Ligases/metabolism , alpha-Synuclein/biosynthesis , alpha-Synuclein/genetics
11.
Mol Neurobiol ; 58(6): 2494-2507, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33443681

ABSTRACT

Oxidative stress is believed to be one of the primary causes in ischemic stroke injury, and the nuclear factor erythroid 2-related factor 2 (Nrf2) signaling pathway is the most important endogenous antioxidative stress damage pathway. Cottonseed oil (CSO), which is used mostly as a solvent for lipid-soluble drugs, has been shown to exert antioxidative effects against peripheral tissue injury. However, the effects and mechanisms of CSO on ischemic stroke-induced oxidative stress injury and the Nrf2 signaling pathway remain largely unknown. In this study, we investigated the potential of CSO in regulating oxidative stress injury induced by middle cerebral artery occlusion and reperfusion (MCAO-R), or oxygen and glucose deprivation and reperfusion (OGD-R). We found that 1.3 mL/kg CSO treatment of male rats with a subcutaneous injection once every other day for 3 weeks significantly improved neurological deficit; reduced infarction volume; alleviated neuronal injuries; reduced the content of ROS and MDA; increased the activity of SOD, GSH, and GSH-PX; and markedly increased the expression of Nrf2. Furthermore, treatment with 10-9 µL/mL CSO to a neuron cell line (HT-22) for 24 h significantly increased cell viability and decreased cell apoptosis after OGD-R injury; significantly reduced the levels of ROS and MDA; increased the activity of SOD, GSH, and GSH-PX; and induced an increase in Nrf2 nuclear translocation. Based on our findings, we conclude that CSO treatment alleviates ischemic stroke injury-induced oxidative stress via activating the Nrf2 signaling pathway, highlighting the potential that CSO has as a therapeutic for ischemic strokes.


Subject(s)
Cottonseed Oil/therapeutic use , Ischemic Stroke/drug therapy , NF-E2-Related Factor 2/metabolism , Oxidative Stress , Signal Transduction , Animals , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cottonseed Oil/pharmacology , Glucose/deficiency , Male , Mice , Neurons/drug effects , Neurons/pathology , Oxidative Stress/drug effects , Oxygen , Protein Transport/drug effects , Rats, Sprague-Dawley , Reperfusion Injury/pathology
12.
Biomed Pharmacother ; 133: 111032, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33378945

ABSTRACT

Insulin resistance is associated with obesity and can lead to several metabolic disorders including type II diabetes, nonalcoholic fatty liver disease and cardiovascular problems. Search for the small molecules which can either induce or mimic the insulin action are of great interest and can be utilized to manage insulin resistance. There are several dietary phytochemicals which can potentially have insulinomimetic action. Nevertheless, high throughput screening methods to test efficiency of small molecules to act as an insulinomimetic are not fully established. In this paper we have performed chemical screen analysis based on GLUT4 translocation using a cell line CHO-HIRC-myc-GLUT4 eGFP that expresses GLUT4-GFP in association with human Insulin receptor. We have established a high content screening-based method which can track and quantify the GLUT4 translocation from perinuclear area to the cell membrane. The assay involves measuring fluorescence intensity in a defined perinuclear area and a defined area along the cell membrane; and the results are expressed as the ratio of fluorescence intensity in the perinuclear to membrane area. The assay could collect real time data of GLUT4 translocation from thousand of cells/ sample and from many such samples in one experiment. We validated the assay using Insulin, insulin mimics/sensitizers and insulin inhibitors. The agonist or antagonists were analyzed for their ability to enhance or block the GLUT4 translocation independent of insulin. The outcome of the assay was correlated by performing glucose uptake assay using differentiated 3T3L1 cells. Using this platform we further identified several plant extracts which had the insulin mimetic action. We confirmed that these plant extracts were non-toxic to the beta cells using RIN mf5cells and 3T3L1 cells. We have identified plant extracts with the potential insulinomimetic action using novel high-content screening approach; these can be further tested for their efficiency in-vivo in pre-clinical trials.


Subject(s)
Adipocytes/metabolism , Biological Assay , Glucose Transporter Type 4/metabolism , Hepatocytes/metabolism , High-Throughput Screening Assays , 3T3 Cells , Adipocytes/drug effects , Animals , CHO Cells , Cricetulus , Glucose Transporter Type 4/genetics , Hep G2 Cells , Hepatocytes/drug effects , Humans , Hypoglycemic Agents/pharmacology , Insulin/pharmacology , Insulin Resistance , Mice , Protein Transport/drug effects , Reproducibility of Results , Time Factors
13.
Mol Biol Cell ; 32(2): 98-108, 2021 01 15.
Article in English | MEDLINE | ID: mdl-33236955

ABSTRACT

The posttranslational regulation of transferrin receptor (TfR1) is largely unknown. We investigated whether iron availability affects TfR1 endocytic cycle and protein stability in HepG2 hepatoma cells exposed to ferric ammonium citrate (FAC). NH4Cl and bafilomycin A1, but not the proteasomal inhibitor MG132, prevented the FAC-mediated decrease in TfR1 protein levels, thus indicating lysosomal involvement. Knockdown experiments showed that TfR1 lysosomal degradation is independent of 1) endocytosis mediated by the clathrin adaptor AP2; 2) Tf, which was suggested to facilitate TfR1 internalization; 3) H-ferritin; and 4) MARCH8, previously implicated in TfR1 degradation. Notably, FAC decreased the number of TfR1 molecules at the cell surface and increased the Tf endocytic rate. Colocalization experiments confirmed that, upon FAC treatment, TfR1 was endocytosed in an AP2- and Tf-independent pathway and trafficked to the lysosome for degradation. This unconventional endocytic regulatory mechanism aimed at reducing surface TfR1 may represent an additional posttranslational control to prevent iron overload. Our results show that iron is a key regulator of the trafficking of TfR1, which has been widely used to study endocytosis, often not considering its function in iron homeostasis.


Subject(s)
Endocytosis , Iron/pharmacology , Receptors, Transferrin/metabolism , Adaptor Protein Complex 2/metabolism , Adaptor Protein Complex mu Subunits/metabolism , Apoferritins/metabolism , Down-Regulation/drug effects , Endocytosis/drug effects , HeLa Cells , Hep G2 Cells , Humans , Lysosomes/drug effects , Lysosomes/metabolism , Protein Transport/drug effects , Transferrin/metabolism
14.
Neuromolecular Med ; 23(1): 199-210, 2021 03.
Article in English | MEDLINE | ID: mdl-33025396

ABSTRACT

Neuroinflammation has been shown to exacerbate ischemic brain injury, and is considered as a prime target for the development of stroke therapies. Clinacanthus nutans Lindau (C. nutans) is widely used in traditional medicine for treating insect bites, viral infection and cancer, due largely to its anti-oxidative and anti-inflammatory properties. Recently, we reported that an ethanol extract from the leaf of C. nutans could protect the brain against ischemia-triggered neuronal death and infarction. In order to further understand the molecular mechanism(s) for its beneficial effects, two experimental paradigms, namely, in vitro primary cortical neurons subjected to oxygen-glucose deprivation (OGD) and in vivo rat middle cerebral artery (MCA) occlusion, were used to dissect the anti-inflammatory effects of C. nutans extract. Using promoter assays, immunofluorescence staining, and loss-of-function (siRNA) approaches, we demonstrated that transient OGD led to marked induction of IL-1ß, IL-6 and TNFα, while pretreatment with C. nutans suppressed production of inflammatory cytokines in primary neurons. C. nutans inhibited IL-1ß transcription via preventing NF-κB/p65 nuclear translocation, and siRNA knockdown of either p65 or IL-1ß mitigated OGD-mediated neuronal death. Correspondingly, post-ischemic treatment of C. nutans attenuated IκBα degradation and decreased IL-1ß, IL-6 and TNFα production in the ischemic brain. Furthermore, IL-1ß siRNA post-ischemic treatment reduced cerebral infarct, thus mimicking the beneficial effects of C. nutans. In summary, our findings demonstrated the ability for C. nutans to suppress NF-κB nuclear translocation and inhibit IL-1ß transcription in ischemic models. Results further suggest the possibility for using C. nutans to prevent and treat stroke patients.


Subject(s)
Acanthaceae/chemistry , Anti-Inflammatory Agents/therapeutic use , Brain Ischemia/drug therapy , Infarction, Middle Cerebral Artery/drug therapy , Interleukin-1beta/biosynthesis , NF-kappa B/metabolism , Neurons/drug effects , Plant Extracts/pharmacology , Plant Leaves/chemistry , Plants, Medicinal/chemistry , Animals , Anti-Inflammatory Agents/pharmacology , Cell Death/drug effects , Cells, Cultured , Cerebral Infarction/pathology , Drug Evaluation, Preclinical , Glucose/pharmacology , Interleukin-1beta/genetics , Male , NF-KappaB Inhibitor alpha/metabolism , Oxygen/pharmacology , Phytotherapy , Promoter Regions, Genetic , Protein Transport/drug effects , RNA Interference , RNA, Small Interfering/genetics , Rats , Rats, Long-Evans , Transcription Factor RelA/antagonists & inhibitors , Transcription Factor RelA/genetics , Transcription, Genetic/drug effects , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/genetics
15.
Nutrients ; 12(10)2020 Oct 06.
Article in English | MEDLINE | ID: mdl-33036197

ABSTRACT

In age-related macular degeneration (AMD), both systemic and local zinc levels decline. Elevation of zinc in clinical studies delayed the progression to end-stage AMD. However, the molecular pathways underpinning this beneficial effect are not yet identified. In this study, we used differentiated primary human fetal retinal pigment epithelium (RPE) cultures and long-term zinc supplementation to carry out a combined transcriptome, proteome and secretome analysis from three genetically different human donors. After combining significant differences, we identified the complex molecular networks using Database for Annotation, Visualization and Integrated Discovery (DAVID) and Ingenuity Pathway Analysis (IPA). The cell cultures from the three donors showed extensive pigmentation, development of microvilli and basal infoldings and responded to zinc supplementation with an increase in transepithelial electrical resistance (TEER) (apical supplementation: 443.2 ± 79.3%, basal supplementation: 424.9 ± 116.8%, compared to control: 317.5 ± 98.2%). Significant changes were observed in the expression of 1044 genes, 151 cellular proteins and 124 secreted proteins. Gene set enrichment analysis revealed changes in specific molecular pathways related to cell adhesion/polarity, extracellular matrix organization, protein processing/transport, and oxidative stress response by zinc and identified a key upstream regulator effect similar to that of TGFB1.


Subject(s)
Micronutrients , Proteome , Retinal Pigment Epithelium/cytology , Retinal Pigment Epithelium/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Transcriptome , Transforming Growth Factor beta1/physiology , Zinc/pharmacology , Cell Adhesion/drug effects , Cell Adhesion/genetics , Cell Polarity/drug effects , Cell Polarity/genetics , Cells, Cultured , Electric Impedance , Extracellular Matrix/metabolism , Humans , Macular Degeneration/genetics , Macular Degeneration/metabolism , Macular Degeneration/prevention & control , Microvilli/drug effects , Oxidative Stress/drug effects , Oxidative Stress/genetics , Pigmentation/drug effects , Protein Transport/drug effects , Retinal Pigment Epithelium/embryology , Retinal Pigment Epithelium/physiology , Zinc/metabolism
16.
Plant Cell ; 32(8): 2639-2659, 2020 08.
Article in English | MEDLINE | ID: mdl-32434855

ABSTRACT

Karrikins (KARs) are butenolides found in smoke that can influence germination and seedling development of many plants. The KAR signaling mechanism is hypothesized to be very similar to that of the plant hormone strigolactone (SL). Both pathways require the F-box protein MORE AXILLARY GROWTH2 (MAX2), and other core signaling components have shared ancestry. Putatively, KAR activates the receptor KARRIKIN INSENSITIVE2 (KAI2), triggering its association with the E3 ubiquitin ligase complex SCFMAX2 and downstream targets SUPPRESSOR OF MAX2 1 (SMAX1) and SMAX1-LIKE2 (SMXL2). Polyubiquitination and proteolysis of SMAX1 and SMXL2 then enable growth responses to KAR. However, many of the assumptions of this model have not been demonstrated. Therefore, we investigated the posttranslational regulation of SMAX1 from the model plant Arabidopsis (Arabidopsis thaliana). We find evidence that SMAX1 is degraded by KAI2-SCFMAX2 but is also subject to MAX2-independent turnover. We identify SMAX1 domains that are responsible for its nuclear localization, KAR-induced degradation, association with KAI2, and ability to interact with other SMXL proteins. KAI2 undergoes MAX2-independent degradation after KAR treatment, which we propose results from its association with SMAX1 and SMXL2. Finally, we discover an SMXL domain that mediates receptor-target interaction preferences in KAR and SL signaling, laying the foundation for understanding how these highly similar pathways evolved to fulfill different roles.


Subject(s)
Arabidopsis Proteins/chemistry , Arabidopsis Proteins/metabolism , Furans/pharmacology , Hydrolases/metabolism , Intracellular Signaling Peptides and Proteins/chemistry , Intracellular Signaling Peptides and Proteins/metabolism , Proteolysis , Pyrans/pharmacology , Amino Acid Motifs , Carrier Proteins/metabolism , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Conserved Sequence , Green Fluorescent Proteins/metabolism , Heterocyclic Compounds, 3-Ring/pharmacology , Hydrolases/chemistry , Lactones/pharmacology , Plant Extracts , Protein Binding/drug effects , Protein Domains , Protein Transport/drug effects , Proteolysis/drug effects , Sequence Deletion , Structure-Activity Relationship , Nicotiana/drug effects
17.
Clin Sci (Lond) ; 134(10): 1167-1180, 2020 05 29.
Article in English | MEDLINE | ID: mdl-32458968

ABSTRACT

In the present study, we evaluated the metabolic effects of green tea polyphenols (GTPs) in high-fat diet (HFD) fed Zucker fatty (ZF) rats, in particular the effects of GTP on skeletal muscle insulin sensitivity. Body weight, visceral fat, glucose tolerance, lipid profiles and whole-body insulin sensitivity were measured in HFD-fed ZF rats after 8-week-treatment with GTP (200 mg/kg of body weight) or saline (5 ml/kg of body weight). Zucker lean rats were studied as controls. Ex vivo insulin-mediated muscle glucose uptake was assessed. Immunoblotting was used to evaluate the expression of key insulin signalling proteins in skeletal muscle. GTP treatment attenuated weight gain (P<0.05) and visceral fat accumulation (27.6%, P<0.05), and significantly reduced fasting serum glucose (P<0.05) and insulin (P<0.01) levels. Homoeostasis model assessment of insulin resistance (HOMA-IR), a measure of insulin resistance, was lower (P<0.01) in GTP-treated animals compared with ZF controls. Moreover, insulin-stimulated glucose uptake by isolated soleus muscle was increased (P<0.05) in GTP-ZF rats compared with ZF-controls. GTP treatment attenuated the accumulation of ectopic lipids (triacyl- and diacyl-glycerols), enhanced the expression and translocation of glucose transporter-4, and decreased pSer612IRS-1 and increased pSer473Akt2 expression in skeletal muscle. These molecular changes were also associated with significantly decreased activation of the inhibitory (muscle-specific) protein kinase (PKC) isoform, PKC-θ. Taken together, the present study has shown that regular ingestion of GTP exerts a number of favourable metabolic and molecular effects in an established animal model of obesity and insulin resistance. The benefits of GTP are mediated in part by inhibiting PKC-θ and improving muscle insulin sensitivity.


Subject(s)
Insulin Resistance , Insulin/metabolism , Muscle, Skeletal/metabolism , Polyphenols/pharmacology , Signal Transduction , Tea/chemistry , Adiposity/drug effects , Animals , Body Weight/drug effects , Glucose/metabolism , Glucose Tolerance Test , Glucose Transporter Type 4/metabolism , Intra-Abdominal Fat/drug effects , Intra-Abdominal Fat/metabolism , Isoenzymes/metabolism , Male , Muscle Proteins/metabolism , Muscle, Skeletal/drug effects , Muscle, Skeletal/enzymology , Phosphorylation/drug effects , Protein Kinase C/metabolism , Protein Transport/drug effects , Rats, Zucker
18.
Sci Rep ; 10(1): 6427, 2020 04 14.
Article in English | MEDLINE | ID: mdl-32286474

ABSTRACT

Diabetic cardiomyopathy (DCM) is the principal cause of death in people with diabetes. However, there is currently no effective strategy to prevent the development of DCM. Although cyclovirobuxine D (CVB-D) has been widely used to treat multiple cardiovascular diseases, the possible beneficial effects of CVB-D on DCM remained unknown. The present aim was to explore the potential effects and underlying mechanisms of CVB-D on DCM. We explored the effects of CVB-D in DCM by using high fat high sucrose diet and streptozotocin-induced rat DCM model. Cardiac function and survival in rats with DCM were improved via the amelioration of oxidative damage after CVB-D treatment. Our data also demonstrated that pre-treatment with CVB-D exerted a remarkable cytoprotective effect against high glucose -or H2O2 -induced neonatal rat cardiomyocyte damage via the suppression of reactive oxygen species accumulation and restoration of mitochondrial membrane potential; this effect was associated with promotion of Nrf2 nuclear translocation and its downstream antioxidative stress signals (NQO-1, Prdx1). Overall, the present data has provided the first evidence that CVB-D has potential therapeutic in DCM, mainly by activation of the Nrf2 signalling pathway to suppress oxidative stress. Our findings also have positive implications on the novel promising clinical applications of CVB-D.


Subject(s)
Antioxidants/metabolism , Diabetic Cardiomyopathies/drug therapy , Diabetic Cardiomyopathies/metabolism , Drugs, Chinese Herbal/therapeutic use , NF-E2-Related Factor 2/metabolism , Animals , Animals, Newborn , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cells, Cultured , Diabetic Cardiomyopathies/pathology , Diabetic Cardiomyopathies/physiopathology , Drugs, Chinese Herbal/chemistry , Drugs, Chinese Herbal/pharmacology , Female , Glucose/toxicity , Heart Function Tests , Hydrogen Peroxide/toxicity , Models, Biological , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Oxidation-Reduction , Oxidative Stress/drug effects , Protein Transport/drug effects , Rats, Sprague-Dawley
19.
Phytomedicine ; 68: 153178, 2020 Mar.
Article in English | MEDLINE | ID: mdl-32126492

ABSTRACT

BACKGROUND: Lowering blood glucose levels by increasing glucose uptake in insulin target tissues, such as skeletal muscle and adipose tissue, is one strategy to discover and develop antidiabetic drugs from natural products used as traditional medicines. PURPOSE: Our goal was to reveal the mechanism and activity of acacetin (5,7-dihydroxy-4'-methoxyflavone), one of the major compounds in Agastache rugose, in stimulating glucose uptake in muscle cells. METHODS: To determine whether acacetin promotes GLUT4-dependent glucose uptake in cultured L6 skeletal muscle cells, we performed a [14C] 2-deoxy-D-glucose (2-DG) uptake assay after treating differentiated L6-GLUT4myc cells with acacetin. RESULTS: Acacetin dose-dependently increased 2-DG uptake by enhancing GLUT4 translocation to the plasma membrane. Our results revealed that acacetin activated the CaMKII-AMPK pathway by increasing intracellular calcium concentrations. We also found that aPKCλ/ζ phosphorylation and intracellular reactive oxygen species (ROS) production were involved in acacetin-induced GLUT4 translocation. Moreover, acacetin-activated AMPK inhibited intracellular lipid accumulation and increased 2-DG uptake in HepG2 cells. CONCLUSION: Taken together, these results suggest that acacetin might be useful as an antidiabetic functional ingredient. Subsequent experiments using disease model animals are needed to verify our results.


Subject(s)
DNA-Binding Proteins/metabolism , Flavones/pharmacology , Glucose/metabolism , Insulin/metabolism , Muscle Fibers, Skeletal/drug effects , Transcription Factors/metabolism , Animals , Cell Membrane/drug effects , Cell Membrane/metabolism , Cells, Cultured , Deoxyglucose/pharmacokinetics , Dose-Response Relationship, Drug , Glucose/pharmacokinetics , Glucose Transporter Type 4/metabolism , Hep G2 Cells , Humans , Hypoglycemic Agents/pharmacology , Muscle Fibers, Skeletal/metabolism , Phosphorylation , Protein Transport/drug effects , Reactive Oxygen Species/metabolism
20.
Cells ; 9(1)2019 12 20.
Article in English | MEDLINE | ID: mdl-31861943

ABSTRACT

Hepatic fibrosis is characterized by the abnormal deposition of extracellular matrix (ECM) proteins. During hepatic fibrogenesis, hepatic stellate cell (HSC) activation followed by chronic injuries is considered a key event in fibrogenesis, and activated HSCs are known to comprise approximately 90% of ECM-producing myofibroblasts. Here, we demonstrated that (-)-catechin-7-O-ß-d-apiofuranoside (C7A) significantly inhibited HSC activation via blocking the signal transducer and activator of transcription 3 (STAT3) signaling pathway. This is the first study to show the hepatic protective effects of C7A with possible mechanisms in vitro and in vivo. In our bioactivity screening, we figured out that the EtOH extract of Ulmusdavidiana var. japonica root barks, which have been used as a Korean traditional medicine, inhibited collagen synthesis in HSCs. Four catechins isolated from the EtOAc fraction of the EtOH extract were compared with each other in terms of reduction in collagen, which is considered as a marker of hepatic protective effects, and C7A showed the strongest inhibitory effects on HSC activation in protein and qPCR analyses. As a possible mechanism, we investigated the effects of C7A on the STAT3 signaling pathway, which is known to activate HSCs. We found that C7A inhibited phosphorylation of STAT3 and translocation of STAT3 to nucleus. C7A also inhibited expressions of MMP-2 and MMP-9, which are downstream genes of STAT3 signaling. Anti-fibrotic effects of C7A were evaluated in a thioacetamide (TAA)-induced liver fibrosis model, which indicated that C7A significantly inhibited ECM deposition through inhibiting STAT3 signaling. C7A decreased serum levels of aspartate amino transferase and alanine transaminase, which were markedly increased by TAA injection. Moreover, ECM-associated proteins and mRNA expression were strongly suppressed by C7A. Our study provides the experimental evidence that C7A has inhibitory effects on HSC activation after live injury and has preventive and therapeutic potentials for the management of hepatic fibrosis.


Subject(s)
Catechin/administration & dosage , Hepatic Stellate Cells/cytology , STAT3 Transcription Factor/metabolism , Ulmus/chemistry , Animals , Catechin/chemistry , Catechin/pharmacology , Cell Line , Cell Proliferation/drug effects , Cell Survival , Disease Models, Animal , Gene Expression Regulation/drug effects , Hepatic Stellate Cells/drug effects , Hepatic Stellate Cells/metabolism , Humans , Male , Phosphorylation , Plant Bark/chemistry , Plant Extracts/chemistry , Protein Transport/drug effects , Signal Transduction/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL