Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
Regen Ther ; 26: 132-144, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38872979

ABSTRACT

Compared to bioactive glass 45S5, bioactive glass 1393 has shown greater potential in activating tissue cells and promoting angiogenesis for bone repair. Nevertheless, the effect of bioactive glass 1393 in the context of wound healing remains extensively unexplored, and its mechanism in wound healing remains unclear. Considering that angiogenesis is a critical stage in wound healing, we hypothesize that bioactive glass 1393 may facilitate wound healing through the stimulation of angiogenesis. To validate this hypothesis and further explore the mechanisms underlying its pro-angiogenic effects, we investigated the impact of bioactive glass 1393 on wound healing angiogenesis through both in vivo and in vitro studies. The research demonstrated that bioactive glass 1393 accelerated wound healing by promoting the formation of granulation, deposition of collagen, and angiogenesis. The results of Western blot analysis and immunofluorescence staining revealed that bioactive glass 1393 up-regulated the expression of angiogenesis-related factors. Additionally, bioactive glass 1393 inhibited the expression of ROS and P53 to promote angiogenesis. Furthermore, bioactive glass 1393 stimulated angiogenesis through the P53 signaling pathway, as evidenced by P53 activation assays. Collectively, these findings indicate that bioactive glass 1393 accelerates wound healing by promoting angiogenesis via the ROS/P53/MMP9 signaling pathway.

2.
Aging (Albany NY) ; 16(4): 3612-3630, 2024 02 14.
Article in English | MEDLINE | ID: mdl-38364249

ABSTRACT

PURPOSE: The age-induced imbalance in ecological niches leads to the loss of GSCs, which is the main reason for ovarian germline senescence. Ginsenoside Rg1 can delay ovarian senescence. Here, we shed light on new insights of ginsenoside Rg1 in regulating the niche to maintain GSCs self-renewal and discussing related molecular mechanisms. METHODS: The differences among GSC number, reproductive capacity of naturally aging female Drosophila after ginsenoside Rg1 feeding were analyzed by immunofluorescence and behavior monitoring. The expressions of the active factors in the niche and the BMP signaling were analyzed through Western blot and RT-qPCR. The target effect was verified in the ECR mutant and combined with the molecular docking. RESULTS: Ginsenoside Rg1 inhibited the age-induced reduction of the GSCs number and restored offspring production and development. Ginsenoside Rg1 promoted the expression of anchor proteins E-cadherin, stemness maintenance factor Nos and differentiation promoting factor Bam, thereby GSCs niche homeostasis was regulated. In addition, ginsenoside Rg1 was bound to the LBD region of the hormone receptor ECR. Ginsenoside Rg1 promotes the regeneration of GSCs by targeting the ECR to increase pSmad1/5/8 expression and thereby activating the BMP signaling pathway. In addition, ginsenoside Rg1 maintenance of niche homeostasis to promote GSCs regeneration is dependent on ECR as demonstrated in ECR mutants. CONCLUSIONS: Ginsenoside Rg1 regulated the ecological niche homeostasis of GSCs and promoted the regeneration of GSCs by targeting the ECR/BMP signaling pathway in hormone-deficient states in aging ovaries. It is of great significance for prolonging fertility potential and delaying ovarian senescence.


Subject(s)
Drosophila Proteins , Drosophila , Ginsenosides , Animals , Female , Drosophila/physiology , Drosophila Proteins/metabolism , Molecular Docking Simulation , Stem Cells/metabolism , Signal Transduction , Hormones/metabolism , Germ Cells
3.
J Anim Sci ; 1022024 Jan 03.
Article in English | MEDLINE | ID: mdl-38189595

ABSTRACT

The objective was to determine the effects of maternal inflammation on offspring muscle development and postnatal innate immune response. Sixteen first-parity gilts were randomly allotted to repeated intravenous injections with lipopolysaccharide (LPS; n = 8, treatment code INFLAM) or comparable volume of phosphate buffered saline (CON, n = 8). Injections took place every other day from gestational day (GD) 70 to GD 84 with an initial dose of 10 µg LPS/kg body weight (BW) increasing by 12% each time to prevent endotoxin tolerance. On GD 70, 76, and 84, blood was collected at 0 and 4 h postinjection via jugular or ear venipuncture to determine tumor necrosis factor (TNF)-α, interleukin (IL)-6, and IL-1ß concentrations. After farrowing, litter mortality was recorded, and the pig closest to litter BW average was used for dissection and muscle fiber characterization. On weaning (postnatal day [PND] 21), pigs were weighed individually and 2 barrows closest to litter BW average were selected for another study. The third barrow closest to litter BW average was selected for the postnatal LPS challenge. On PND 52, pigs were given 5 µg LPS/kg BW via intraperitoneal injection, and blood was collected at 0, 4, and 8 h postinjection to determine TNF-α concentration. INFLAM gilt TNF-α concentration increased (P < 0.01) 4 h postinjection compared to 0 h postinjection, while CON gilt TNF-α concentration did not differ between time points. INFLAM gilt IL-6 and IL-1ß concentrations increased (P = 0.03) 4 h postinjection compared to 0 h postinjection on GD 70, but did not differ between time points on GD 76 and 84. There were no differences between INFLAM and CON gilts litter mortality outcomes (P ≥ 0.13), but INFLAM pigs were smaller (P = 0.04) at birth and tended (P = 0.09) to be smaller at weaning. Muscle and organ weights did not differ (P ≥ 0.17) between treatments, with the exception of semitendinosus, which was smaller (P < 0.01) in INFLAM pigs. INFLAM pigs tended (P = 0.06) to have larger type I fibers. INFLAM pig TNF-α concentration did not differ across time, while CON pig TNF-α concentration peaked (P = 0.01) 4 h postinjection. TNF-α concentration did not differ between treatments at 0 and 8 h postinjection, but CON pigs had increased (P = 0.01) TNF-α compared to INFLAM pigs 4 h postinjection. Overall, maternal immune activation did not alter pig muscle development, but resulted in suppressed innate immune activation.


Maternal inflammation or immune activation impacts fetal development and subsequently the offspring's postnatal performance. In particular, maternal immune activation may be detrimental to fetal muscle development and alter postnatal immune responses, both of which are vital in determining livestock efficiency. However, understanding the relationship between maternal immune activation and offspring development is difficult as many models use a live pathogen. This introduces many confounding factors, including increased mortality, persistent postnatal infection, and potential copathogens. Therefore, the objective of this study was to determine the effect of maternal inflammation on offspring muscle development and postnatal inflammatory response using repeated injections of a nonpathogenic immune stimulant. Each injection successfully induced an inflammatory response as indicated by increased rectal temperature and circulating inflammatory markers. The gestational challenge did not result in increased litter mortality. Further, muscle development was not altered in piglets exposed to gestational inflammation. However, when challenged with the same immune stimulant given to the dams, pigs exposed to maternal inflammation had a remarkably suppressed immune response compared to controls. Overall, maternal inflammation independent of infection affected offspring immune function, but not muscle development.


Subject(s)
Lipopolysaccharides , Tumor Necrosis Factor-alpha , Pregnancy , Swine , Animals , Female , Lipopolysaccharides/pharmacology , Sus scrofa/physiology , Weaning , Muscle Fibers, Skeletal , Interleukin-6
4.
Biomed Pharmacother ; 167: 115442, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37699318

ABSTRACT

Alzheimer's disease (AD) is a neurological condition that progresses with age. Amyloid-ß (Aß) aggregation has been suggested to be a key pathogenic process in Alzheimer's disease. Ginseng polysaccharides (GP), the main biologically active components isolated from Panax ginseng C. A. Meyer (ginseng), may act as neuroprotective agents with potential benefits for AD patients. However, GP effects on Aß pathology and AD symptoms are still unclear. Here, a 4.7-kDa GP termed GP4 was purified and subjected to basic physicochemical characterization. The biological effects of GP4 to prevent Aß aggregation were then assessed with cross-species AD models, including Aftin-5-treated SH-SY5Y cells and cerebral organoids, and transgenic C. elegans overexpressing the full-length human Aß42 peptide. These analyses ultimately demonstrated that GP4 was capable of inhibiting Aß accumulation both in vivo and vitro, and with early intervention of GP4 being sufficient to alleviate Aß42-associated aging phenotypes and memory loss in C. elegans model of AD. Furthermore, neuroinflammation was significantly down-regulated in human cells and cerebral organoids. From a mechanistic perspective, the ability of GP4 to inhibit Aß aggregation was found to be related to its ability to promote neuronal mitophagic activity. This finding offers a robust theoretical foundation for the further development of GP4 as a candidate drugs with the potential to treat AD.

5.
J Ginseng Res ; 46(6): 759-770, 2022 Nov.
Article in English | MEDLINE | ID: mdl-36312736

ABSTRACT

Background: Aerobic cellular respiration provides chemical energy, adenosine triphosphate (ATP), to maintain multiple cellular functions. Sirtuin 1 (SIRT1) can deacetylate peroxisome proliferator-activated receptor gamma coactivator 1 alpha (PGC-1α) to promote mitochondrial biosynthesis. Targeting energy metabolism is a potential strategy for the prevention and treatment of various diseases, such as cardiac and neurological disorders. Ginsenosides, one of the major bioactive constituents of Panax ginseng, have been extensively used due to their diverse beneficial effects on healthy subjects and patients with different diseases. However, the underlying molecular mechanisms of total ginsenosides (GS) on energy metabolism remain unclear. Methods: In this study, oxygen consumption rate, ATP production, mitochondrial biosynthesis, glucose metabolism, and SIRT1-PGC-1α pathways in untreated and GS-treated different cells, fly, and mouse models were investigated. Results: GS pretreatment enhanced mitochondrial respiration capacity and ATP production in aerobic respiration-dominated cardiomyocytes and neurons, and promoted tricarboxylic acid metabolism in cardiomyocytes. Moreover, GS clearly enhanced NAD+-dependent SIRT1 activation to increase mitochondrial biosynthesis in cardiomyocytes and neurons, which was completely abrogated by nicotinamide. Importantly, ginsenoside monomers, such as Rg1, Re, Rf, Rb1, Rc, Rh1, Rb2, and Rb3, were found to activate SIRT1 and promote energy metabolism. Conclusion: This study may provide new insights into the extensive application of ginseng for cardiac and neurological protection in healthy subjects and patients.

6.
Front Endocrinol (Lausanne) ; 13: 964069, 2022.
Article in English | MEDLINE | ID: mdl-36017314

ABSTRACT

Aging ovaries caused diminished fertility and depleted steroid hormone level. Ginsenosides, the active ingredient in ginseng, had estrogen-like hormonal effects. Although ginsenosides were well known for their ability to alleviate many age-related degenerative diseases, the effect of ginsenosides on the decline in reproductive capability caused by aging, as well as the mechanism, are unknown. We found that ginsenosides improved the quantity and quality of the offspring, prolonged life and restored muscle ability in aged female Drosophila. In addition, ginsenosides inhibited ovarian atrophy and maintained steroid hormone 20-Hydroxyecdysone (20E) and juvenile-preserving hormone (JH)) levels. Ginsenosides activated ecdysteroid receptor (ECR) and increased the expression of the early transcription genes E74 and Broad (Br), which triggered steroid signaling pathway. Meanwhile, ginsenosides promoted JH biosynthesis by increasing the expression of Hydroxyl-methylglutaryl-CoA reductase (HMGR) and juvenile hormone acid O-methyltransferase (JHAMT). Subsequently, JH was bound to Methoprene Tolerant (Met) and activated the transcription of the responsive gene Kruppel Homolog 1 (Kr-h1), which coordinated with 20E signaling to promote the reproduction of aged female Drosophila. The reproductive capacity and steroid hormone levels were not improved and the steroid signaling pathway was not activated in ginsenoside-treated ECR knockout Drosophila. This suggested that ginsenosides played a role dependent on targeted ECR. Furthermore, 17 kinds of ginsenoside monomers were identified from the total ginsenosides. Among them, Rg1, Re and Rb1 improved the reproductive capacity and steroid hormone levels of aged female Drosophila, which has similar effects to the total ginsenoside. These results indicated that ginsenosides could enhance the reproductive capacity of aged female Drosophila by activating steroid signals dependent on nuclear receptor ECR. In addition, ginsenoside monomers Rg1, Rb1 and Re are the main active components of total ginsenosides to improve reproductive ability. This will provide strong evidence that ginsenosides had the potential to alleviate age-induced reproductive degradation.


Subject(s)
Drosophila Proteins , Ginsenosides , Animals , Drosophila/genetics , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Ecdysterone/pharmacology , Female , Ginsenosides/metabolism , Ginsenosides/pharmacology , Juvenile Hormones/pharmacology , Receptors, Steroid , Reproduction
7.
Food Funct ; 13(16): 8605-8615, 2022 Aug 15.
Article in English | MEDLINE | ID: mdl-35894549

ABSTRACT

The effects of ginseng oligosaccharides (GSOs) on neuronal oxidative injury induced by glutamate (GLU) and the molecular mechanisms involved were investigated. Cell damage was assessed using MTT assays, and the lactate dehydrogenase (LDH) release rate and flow cytometry were used to detect the accumulation of reactive oxygen species (ROS) and mitochondrial membrane potential respectively. The levels of catalase (CAT) and glutathione (GSH) were measured in PC12 cells and Drosophila brain tissue. The climbing ability of Drosophila was observed. Levels of proteins, including Cyt C, Bcl-2/BAX, and Nrf2/HO-1-associated proteins, were determined by western blotting and immunofluorescence. It was found that GSOs reversed GLU-induced reductions in cell viability and the LDH release rate, and rescued ROS accumulation. GSOs also mitigated the deleterious effects of GLU on the mitochondrial membrane potential and Cyt C release, thus alleviating mitochondrial dysfunction, and increased GSH levels and CAT activity in both cells and Drosophila brain tissue. The climbing index in GSO-treated Drosophila was significantly higher than that in the tert-butyl-hydroperoxide-treated flies. Furthermore, GSOs protected cells against GLU-induced apoptosis by reducing the expression of the mitochondrial apoptosis-associated Bcl-2 family effector proteins and protected cells from GLU-induced oxidative damage by increasing the nuclear translocation of Nrf2 and HO-1 expression. These findings indicate that GSOs protect against GLU-induced neuronal oxidative damage through Nrf2/HO-1 activation.


Subject(s)
NF-E2-Related Factor 2 , Panax , Animals , Apoptosis , Drosophila/metabolism , Glutamic Acid/metabolism , Glutathione/metabolism , Heme Oxygenase-1/genetics , Heme Oxygenase-1/metabolism , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Neurons/metabolism , Oligosaccharides/pharmacology , Oxidative Stress , Panax/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Rats , Reactive Oxygen Species/metabolism , Signal Transduction
8.
J Anim Sci ; 100(5)2022 May 01.
Article in English | MEDLINE | ID: mdl-35426431

ABSTRACT

The goals of this study were to determine the impact of maternal PRRSV infection on offspring muscle and immune development and the potential of dietary soy isoflavones to mitigate those effects. Thirteen first-parity gilts ("gilts") were randomly allotted into one of three treatments: not infected and fed a diet devoid of isoflavones (CON), infected with porcine reproductive and respiratory syndrome virus (PRRSV) and fed the control diet (POS) or that supplemented with 1,500 mg/kg soy-derived isoflavones (ISF). Gilts were inoculated with PRRSV intranasally on gestational day (GD) 70. After farrowing (GD 114 ± 2), 1-2 offspring ("pigs") closest to the average litter weight were selected either at birth (3 ± 2 d of age) or weaning (21 ± 2 d of age) to determine body, muscle, and organ weights as well as muscle cell number and size. Four weaned pigs of average body weight within each litter were selected for postnatal immune challenge. At PND 52, pigs were injected with 5 µg/kg BW lipopolysaccharide (LPS) intraperitoneally. Serum was collected at 0, 4, and 8 h following LPS administration to analyze tumor necrosis factor alpha (TNF-α). At PND 59, pigs were administered a novel vaccine to elicit an adaptive immune response. At PND 59, 66, and 73, peripheral blood mononuclear cells were isolated and T-cell populations determined by flow cytometry. Both POS and ISF pigs exhibited persistent PRRSV infections throughout the study (PND 1-73). At PND 3, whole body, muscle, and organ weights were not different (P > 0.22) between groups, with the exception of relative liver weight, which was increased (P < 0.05) in POS compared with CON pigs. At PND 21, ISF pigs had reduced (P ≤ 0.05) whole body and muscle weights, but greater (P < 0.05) kidney weight compared with CON, and greater (P < 0.05) relative liver weight compared with CON and POS. Muscle fiber number and size were not different (P > 0.39) between groups at birth or weaning. After LPS administration, TNF-α was greatest in ISF pigs (P < 0.05) at both 0 and 8 h post-challenge. At the peak time-point of 4 h post-challenge, ISF pigs had the greatest concentration of TNF-α and CON pigs had the lowest, with POS pigs being intermediate (P = 0.01). After vaccination, ISF offspring had shifts in T-cell populations indicating an impaired immune response. These data indicate that maternal PRRSV infection may impact offspring organ growth and immune function, particularly when the dam is supplemented with isoflavones.


Gestational health challenges may influence growth performance and immunity of offspring pigs during postnatal life. In particular, porcine reproductive and respiratory syndrome virus (PRRSV) is endemic in the U.S. herd, but its effects on surviving offspring pigs are largely unknown. Further, dietary supplementation with soy isoflavones lessened the severity of PRRSV infections in weaning and growing pigs. Therefore, the goals of this study were to determine the impact of maternal PRRSV infection on offspring muscle and immune development and the potential of isoflavones to mitigate those effects. Isoflavone supplementation reduced viral load in dams 21 d after infection, but did not alter clinical illness indicators. Pig mortality was increased by PRRSV infection in dams, and surviving pigs were infected with PRRSV throughout the study. Interestingly, muscle and organ weights were not different among treatments at birth, but infected litters were lighter at weaning, likely due to postnatal infection. Muscle fiber number and size did not differ between treatments. Pigs born to infected dams had slower responses during innate immune stimulation and then failed to mount a proper vaccine response during adaptive immune stimulation. Overall, maternal infection altered offspring immune responses but not muscle fiber development. Isoflavone supplementation did not mitigate these effects.


Subject(s)
Isoflavones , Porcine Reproductive and Respiratory Syndrome , Porcine respiratory and reproductive syndrome virus , Swine Diseases , Adaptive Immunity , Animal Feed/analysis , Animals , Diet/veterinary , Dietary Supplements , Female , Isoflavones/pharmacology , Leukocytes, Mononuclear , Lipopolysaccharides/pharmacology , Muscle Fibers, Skeletal , Pregnancy , Sus scrofa , Swine , Tumor Necrosis Factor-alpha
9.
Eur J Pharmacol ; 922: 174887, 2022 May 05.
Article in English | MEDLINE | ID: mdl-35306001

ABSTRACT

Under pathological conditions, human tau (htau) hyperphosphorylation promotes formation of proteotoxic intracellular amyloid aggregates that may underlie neurodegenerative diseases known as tauopathies, prompting researchers to develop treatments that inhibit htau aggregation as a promising therapeutic strategy. Ginsenosides, the main active constituents of Panax ginseng C. A. Meyer (ginseng), appear to inhibit tau aggregation and disassociation in tauopathy models, although their active components and molecular mechanisms are unknown. Here, we used a novel Caenorhabditis elegans (C. elegans) tauopathy model to identify ginsenoside monomers which may repress htau proteotoxicity. Our findings indicated that ginsenoside Rf prevented tau aggregation and reversed abnormal tau aggregation-induced phenotypes and alleviated neurodegeneration in worms. Notably, deep RNA-seq analysis of ginsenoside Rf-treated and untreated worms with tauopathy revealed that ginsenoside Rf altered expression levels of 24 up- and 36 down-regulated lncRNA transcripts, 32 up- and 22 down-regulated miRNAs and 65 up- and 30 down-regulated mRNA transcripts. Based on GO and KEGG pathway annotation analyses, identified mRNAs, miRNAs and lncRNAs-associated gene targets were functionally related to neuron-related terms (e.g., neuron development, axon and motor neuron axon guidance) and longevity regulating pathways. Importantly, RT-qRCR results suggested that 6 miRNAs (miR-786, miR-2208b, miR-34, miR-241, miR-247 and miR-4805), 8 lncRNAs (MSTRG.20812.2, MSTRG.22617.2, MSTRG.28210.13, MSTRG.5728.12, MSTRG.29708.1, MSTRG.3342.25, MSTRG.3342.31 and MSTRG.8841.8) and 7 mRNAs (nas-33, math-28, T14B4.19, col-17, rol-6, sqt-1 and irg-4) were potential targets of ginsenoside Rf inhibition of tauopathy. These results partially explain mechanisms underlying ginsenoside Rf-associated alleviation of htau proteotoxicity and will guide future strategies to discover potential therapeutic targets for preventing and alleviating tauopathies.


Subject(s)
Ginsenosides , MicroRNAs , Panax , RNA, Long Noncoding , Tauopathies , Animals , Caenorhabditis elegans/genetics , Ginsenosides/pharmacology , Humans , MicroRNAs/genetics , RNA, Long Noncoding/genetics , RNA, Messenger/genetics , Tauopathies/drug therapy , Tauopathies/genetics , tau Proteins/metabolism
10.
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
11.
Biochem Pharmacol ; 180: 114145, 2020 10.
Article in English | MEDLINE | ID: mdl-32653593

ABSTRACT

We previously found that 20(S)-ginsenoside Rg3 (S-Rg3) promotes myoblast differentiation via an unknown mechanism. Here we measured levels of myosin heavy chain (MHC) and myogenin, markers of myoblast differentiation, using Western blot analysis and immunofluorescence staining. Notably, S-Rg3 treatment of C2C12 myoblasts led to increased muscle differentiation and protection from muscle atrophy in a dexamethasone (DEX)-treated C2C12 myotube-based muscle atrophy model. This effect was likely caused by S-Rg3 treatment-induced promotion of Akt/mTOR phosphorylation and inhibition of FoxO3 nuclear transcription. Additionally, S-Rg3 treatment also led to increased fruit fly climbing distances (Drosophila melanogaster) and prevented muscle atrophy in aged fruit flies. Our study provides a mechanistic framework for understanding how S-Rg3 enhances myoblast differentiation and inhibits myotube atrophy through activation of the Akt/mTOR/FoxO3 signaling pathway, as demonstrated in vitro in C2C12 cells and in vivo in fruit flies.


Subject(s)
Drosophila Proteins/metabolism , Forkhead Box Protein O3/metabolism , Ginsenosides/pharmacology , Muscle Fibers, Skeletal/metabolism , Muscular Atrophy/metabolism , Myoblasts/metabolism , Nuclear Matrix-Associated Proteins/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Animals , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cell Line , Dose-Response Relationship, Drug , Drosophila melanogaster , Mice , Muscle Fibers, Skeletal/drug effects , Muscular Atrophy/pathology , Muscular Atrophy/prevention & control , Myoblasts/drug effects , Signal Transduction/drug effects , Signal Transduction/physiology
12.
J Agric Food Chem ; 68(12): 3742-3749, 2020 Mar 25.
Article in English | MEDLINE | ID: mdl-32129993

ABSTRACT

The protective effect and mechanism of action of p-coumaric acid for alleviating palmitic acid (PA)-induced hepatocyte injury were investigated using a PA-induced human hepatoma cell (HepG2)-based hepatocellular injury model and MTT cell viability determinations. Additionally, reduced glutathione content and catalase activity were detected using commercial kits, while intracellular lipid accumulation and total triglyceride content were measured using Oil Red O staining and a triglyceride quantification kit, respectively. Meanwhile, levels of proteins (fatty acid synthase, sterol regulatory element-binding protein-1, stearoyl-CoA desaturase-1) and proliferator-activated receptor-α mRNA were determined using western blotting and real-time quantitative polymerase chain reaction, respectively. After p-coumaric acid targets were identified using network pharmacological analysis, cyclooxygenase-2 (COX-2) expression was assessed via western blotting, while prostaglandin E2 accumulation was measured via an enzyme-linked immunosorbent assay. Notably, PA-treated hepatocytes exhibited increased viability (87.3 ± 2.2% vs 65.5 ± 2.5% for untreated cells), with reduced intracellular lipid accumulation reflecting promotion of lipolysis and fatty acid ß-oxidation; this protective effect may depend on inhibition of both PA-induced HepG2 cell COX-2 expression and PGE2 accumulation.


Subject(s)
Coumaric Acids/pharmacology , Hepatocytes/drug effects , Lipid Metabolism/drug effects , Palmitic Acid/metabolism , Protective Agents/pharmacology , Cell Survival/drug effects , Cytoprotection/drug effects , Hep G2 Cells , Hepatocytes/metabolism , Hepatocytes/pathology , Humans
13.
J Food Sci ; 85(3): 576-581, 2020 Mar.
Article in English | MEDLINE | ID: mdl-32078759

ABSTRACT

Elevated free fatty acids may impair insulin-mediated signaling to eNOS that contributes to the pathophysiology of endothelial dysfunction. Previous studies have indicated the protective effect of ginseng and the regulatory potential of phenolic acid components from other plants on endothelial function. Therefore, this study investigated the protective effects of phenolic acid extract from ginseng (PG2) on endothelial cells against palmitate-induced damage. We found that PG2 increases cell viability, inhibits the palmitate-induced intracellular accumulation of lipids, and the overexpression of endothelin-1 (ET-1) through enhancing the phosphorylation of the phosphatidylinositol 3-kinase/Akt/endothelial nitric oxide synthase (PI3K/Akt/eNOS) signaling pathway. The results of this study may be valuable for the development of PG2 to combat the endothelial cell damage caused by hyperlipidemia. PRACTICAL APPLICATION: We proved that phenolic acid extract from ginseng has a protective effect on free fatty acid-induced endothelial dysfunction in vitro. This study provides experimental data for the application of ginseng-derived phenolic acids in treating cardiovascular disease.


Subject(s)
Endothelial Cells/drug effects , Hydroxybenzoates/pharmacology , Nitric Oxide Synthase Type III/metabolism , Panax/chemistry , Phosphatidylinositol 3-Kinase/metabolism , Plant Extracts/pharmacology , Protective Agents/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Cell Survival/drug effects , Endothelial Cells/enzymology , Endothelin-1/metabolism , Humans , Insulin/metabolism , Palmitates/toxicity , Phosphorylation/drug effects , Signal Transduction/drug effects
15.
Biomed Pharmacother ; 121: 109676, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31810119

ABSTRACT

OBJECTIVE: Oxidative stress is increasingly recognized as a risk factor associated with the development and progression of osteoporosis. Fufang Lurong Jiangu Capsule (FLJC) has a known anti-osteoporotic effect, but its pharmacological effect on osteoblasts is not clearly understood. This study was designed to investigate FLJC effects/mechanisms on in vitro hydrogen peroxide (H2O2)-induced oxidative damage of osteoblasts and on in vivo lipopolysaccharide (LPS)-induced mice bone loss. FLJC alleviates osteoporosis via unknown pharmacological mechanisms. METHODS: Chemical compositions of FLJC preparations were analyzed using high-performance liquid chromatographic fingerprinting. After rat bone marrow mesenchymal stem cell differentiation induction, resulting osteoblasts received various 48 h FLJC pretreatments before H2O2-based (200 µM) oxidative stress exposure. FLJC effects were measured on osteoblast cell viability, morphological changes, levels of intracellular reactive oxygen species (ROS), localization of mitochondria, activity of antioxidant enzymes, alkaline phosphatase (ALP) and mineralization, the secretion of Col I and expression of osteogenic markers. The percentages of apoptosis were determined by flow cytometric analysis; apoptosis-related protein levels, including nuclear factor (erythroid-derived 2)-like 2 (Nrf2) and heme oxygenase-1 (HO-1) with or without Nrf2 inhibitor were analyzed via western blot. Hematoxylin and eosin (H&E) and ALP staining revealed in vivo FLJC effect on mice LPS-induced bone loss. RESULTS: Five chemical components in FLJC were identified, and fingerprint analysis showed good reproducibility. FLJC pretreatment significantly reduced H2O2-induced ROS levels in osteoblasts and increased antioxidant enzyme activities to reduce oxidative damage. With regard to osteoblast differentiation, FLJC pretreatment increased ALP expression, as well as levels of mineralization and osteoblast markers. Additionally, FLJC protected against H2O2-induced apoptosis by inhibiting changes in expression of major Bcl-2 family effector proteins of the mitochondrial apoptosis pathway. Furthermore, FLJC protected cells from H2O2-induced oxidative damage by up-regulating Nrf2 and HO-1 protein levels. Finally, we confirmed that FLJC administration could reverse the bone loss in LPS-induced mice. CONCLUSION: These results indicate that FLJC may significantly attenuate oxidative damage of osteoblasts induced by H2O2 via the Nrf2/HO-1 signaling pathway, providing new insights to guide development of treatments for osteoporosis induced by oxidative injury.


Subject(s)
Cytoprotection/drug effects , Drugs, Chinese Herbal/pharmacology , Heme Oxygenase-1/metabolism , Hydrogen Peroxide/toxicity , Mesenchymal Stem Cells/pathology , NF-E2-Related Factor 2/metabolism , Osteoblasts/pathology , Oxidative Stress/drug effects , Alkaline Phosphatase/metabolism , Animals , Apoptosis/drug effects , Biomarkers/metabolism , Bone Resorption/pathology , Calcification, Physiologic/drug effects , Cell Differentiation/drug effects , Cell Shape/drug effects , Cell Survival/drug effects , Collagen Type I/metabolism , Down-Regulation/drug effects , Lipopolysaccharides , Male , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Mice, Inbred C57BL , Osteoblasts/drug effects , Osteoblasts/metabolism , Protective Agents/pharmacology , Rats, Sprague-Dawley , Signal Transduction/drug effects
16.
Funct Plant Biol ; 46(12): 1123-1133, 2019 11.
Article in English | MEDLINE | ID: mdl-31581976

ABSTRACT

Red-skin disorder (RSD), a non-infectious disorder in Panax ginseng, impairs the quality and yield of ginseng and impedes continuous cropping. Since the mechanism of this disorder is unknown, there are no effective prevention measures for RSD. The proteomic changes in RSD ginseng were analysed in this study by two-dimensional electrophoresis (2-DE) and isobaric tags for relative and absolute quantification (iTRAQ). The differential expression of 137 proteins (60 from 2-DE and 77 from iTRAQ) was identified in RSD ginseng as compared with healthy ginseng. Most changes are related to carbon- and nitrogen- metabolism, redox homeostasis, and stress resistance. We also found that the concentration of metal elements, such as iron (Fe), aluminium (Al), and manganese (Mn), was significantly increased in RSD ginseng. These increased metals would be chelated with phenols to form red spots on the ginseng epidermis. Moreover, RSD disturbed the carbon and nitrogen metabolism and affected the biosynthesis of nutrients (sugar, proteins, amino acids) and active components (ginsenosides), which reduced the survival rate and medicinal value of ginseng. These differences between RSD and healthy ginseng will contribute to the understanding of RSD mechanism.


Subject(s)
Ginsenosides , Panax , Carbon , Nitrogen , Proteomics
17.
Biomed Res Int ; 2019: 3719643, 2019.
Article in English | MEDLINE | ID: mdl-31467885

ABSTRACT

BACKGROUND: The clinical anti-inflammatory drug dexamethasone (DEX) can cause many side effects such as muscle atrophy for long-term use. Muscle atrophy induced by DEX may be caused by decrease of glucose consumption. Panax ginseng C.A. Meyer was previously considered to be an antiatrophic agent for glucocorticoid- (GC-) treated therapies. As one of the main components, it remains unclear whether ginseng total protein (GP) facilitates recovery from muscle atrophy induced by DEX. METHODS: In this study, GP was extracted and purified with Sephadex-G50. C2C12 myoblasts was induced with 2% horse serum to differentiate into C2C12 myotubes. Cell viability was analyzed by the MTT assay, and Ca2+ concentration was analyzed by a flow cytometer. The release of lactic dehydrogenase (LDH) and the glucose consumption were analyzed by spectrophotometry. The phosphorylation of AMP-activated protein kinase (AMPK), phosphoinositide 3-kinase (PI3K), and protein kinase B (Akt) and the expression of glucose transporter 4 (GLUT4) were analyzed by Western blotting. The phosphorylation of AS160 was quantified by Immunofluorescence staining. RESULTS: We found that GP increased cell viability and increased myotube diameter in high-dose DEX-treated C2C12 myotubes for 24 h, but this activity was not found in the enzymatic hydrolyzed GP group. GP reduced muscle atrophy by decreasing the expression of key proteins such as muscle RING-finger protein-1 and muscle atrophy F-box, reducing the Ca2+ concentration, and decreasing the release of LDH in DEX-injured C2C12 myotubes. Moreover, GP improved glucose consumption and increased the phosphorylation of AMPK, PI3K, Akt, and AS160 and the expression of GLUT4 in DEX-treated C2C12 myotubes. CONCLUSION: The results of this study suggest that GP has effects on recovering DEX-induced muscle atrophy and cell injury, which may improve glucose consumption via the AMPK and PI3K/Akt pathways in high-dose DEX-treated C2C12 myotubes. This study provides in vitro mechanistic insights into the recovery of muscle atrophy with GP treatment.


Subject(s)
Glucose/metabolism , Muscular Atrophy/drug therapy , Panax/chemistry , Plant Extracts/pharmacology , Animals , Dexamethasone/toxicity , Gene Expression/drug effects , Glucose Transporter Type 4 , Humans , Mice , Muscle Fibers, Skeletal/drug effects , Muscle, Skeletal/drug effects , Muscle, Skeletal/pathology , Muscular Atrophy/chemically induced , Muscular Atrophy/pathology , Myoblasts/drug effects , Myoblasts/pathology , Phosphorylation/drug effects , Plant Extracts/chemistry
18.
Biomed Pharmacother ; 115: 108840, 2019 Jul.
Article in English | MEDLINE | ID: mdl-31048189

ABSTRACT

BACKGROUND AND AIMS: Hirudo is an important Chinese medicine that has been widely used in patients with thrombosis-related diseases. We aimed to evaluate the protective effect and potential mechanism of Hirudo extract (HE) on the process of atherosclerosis (AS) as well as identify its active components in the lipopolysaccharide (LPS) - or oxidized low-density lipoprotein (ox-LDL)-induced cell models. METHODS: After treatment, adhesion molecules and pro-inflammatory cytokines induced by LPS were examined by qPCR and ELISA. ROS production, cell apoptosis, and lipid accumulation in ox-LDL-induced cells were analyzed by flow cytometry, qPCR, western blotting, and immunofluorescence staining. In addition, the main active components of HE were identified and analyzed for preventing the progression of AS. RESULTS: In this study, we found that HE pretreatment for 48 h significantly inhibited monocyte adhesion and reduced the levels of adhesion factors (ICAM-1 and VCAM-1) and pro-inflammatory factors (IL-6 and TNF-α) in LPS-induced endothelial cells. Moreover, HE attenuated ox-LDL-induced ROS accumulation and apoptosis in macrophage cells via mitochondrial apoptotic pathways. Additionally, HE pretreatment effectively inhibited cholesterol uptake and increased cholesterol efflux by regulating the LOX-1/LXR-α/ABCA1 pathway. Importantly, the polypeptides from HE (PP) with a molecular weight < 10,000 Da accounted for about 62.9% of the total amount of polypeptides, which in turn may be active components of HE that are responsible for inhibiting inflammation, foam cell formation and apoptosis. CONCLUSION: PP from HE potently inhibits endothelial cell inflammatory injury and macrophage foam cell formation and apoptosis by regulating the LOX-1/LXR-α/ABCA1 pathway, thereby providing additional support to the beneficial effects of HE in preventing AS.


Subject(s)
Foam Cells/drug effects , Human Umbilical Vein Endothelial Cells/drug effects , Leeches/chemistry , Macrophages/drug effects , Peptides/pharmacology , Scavenger Receptors, Class E/metabolism , ATP Binding Cassette Transporter 1/genetics , ATP Binding Cassette Transporter 1/metabolism , Animals , Cell Adhesion/drug effects , Cholesterol/metabolism , Dose-Response Relationship, Drug , Gene Expression Regulation/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Inflammation/drug therapy , Inflammation/metabolism , Lipopolysaccharides/administration & dosage , Lipopolysaccharides/toxicity , Lipoproteins, LDL/administration & dosage , Lipoproteins, LDL/toxicity , Liver X Receptors/genetics , Liver X Receptors/metabolism , Mice , Peptides/chemistry , RAW 264.7 Cells , Reactive Oxygen Species , Scavenger Receptors, Class E/genetics , THP-1 Cells
19.
Biomed Pharmacother ; 115: 108890, 2019 Jul.
Article in English | MEDLINE | ID: mdl-31022597

ABSTRACT

OBJECTIVE: Pig brain polypeptides (PBP), active polypeptides hydrolysate extracted from fresh porcine brain tissue, has been shown to have neuroprotective effects in both in vitro and in vivo studies. The present study aimed to explore the molecular mechanisms underlying the neuroprotective effects of PBP in corticosterone (CORT)-induced rat adrenal pheochromocytoma PC12 cells. METHODS: Cell viability and lactate dehydrogenase (LDH) release were measured in PC12 cells induced with 200 µM CORT in the presence or absence of various concentrations of PBP for 48 h. Intracellular reactive oxygen species (ROS) generation, the activities of superoxide dismutase (SOD) and glutathione peroxidase (GSH-Px) and glutathione (GSH) content were examined to analyze the effect of PBP on CORT-induced oxidative stress. The levels of pro-inflammatory factors, the percentage of apoptotic cells, and apoptosis-related protein expression in PC12 cells were determined. RESULTS: PBP is mainly composed of protein subunits with molecular weights ranging from 1000 to 10,000 Da. PBP treatment increased cell viability and decreased the release of LDH in CORT-stimulated PC12 cells. Moreover, PBP reduced the level of CORT-induced oxidative stress by decreasing ROS levels and increasing SOD, GSH-Px activities and GSH content. PBP had an inhibitory effect on the CORT-induced inflammatory response through inhibition of the NF-κB signaling pathway. PBP also inhibited CORT-induced apoptosis by downregulating the mitochondrial apoptotic signaling pathway. CONCLUSION: These results suggest that PBP exerts a neuroprotective effect against CORT-induced cell injury by inhibiting oxidative stress, inflammation, and apoptosis. PBP could act as a neuroprotective agent against nerve injury induced by CORT.


Subject(s)
Apoptosis/drug effects , Brain Chemistry , Corticosterone/toxicity , Inflammation/chemically induced , Oxidative Stress/drug effects , Peptides/pharmacology , Animals , Cell Survival/drug effects , Cytokines/genetics , Cytokines/metabolism , Gene Expression Regulation, Enzymologic/drug effects , Glutathione/metabolism , Glutathione Peroxidase/genetics , Glutathione Peroxidase/metabolism , Inflammation/prevention & control , L-Lactate Dehydrogenase/metabolism , PC12 Cells , Peptides/chemistry , Rats , Reactive Oxygen Species , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism , Swine
20.
Biomed Pharmacother ; 108: 1751-1758, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30372878

ABSTRACT

Cigarette smoke causes many adverse effects such as inflammation, oxidative stress, and excessive accumulation of the extracellular matrix (ECM). Ginsenoside Rb3 has anti-inflammatory and anti-oxidative effects, which may contribute to delaying the injury caused by cigarette smoke. In this study, we used cigarette smoke extract (CSE) to establish cell injury models in WI-38 human fetal lung fibroblasts and 16HBE human bronchial epithelial cells. Our results showed that Rb3 protected against CSE-induced cytotoxicity in both cell lines. In addition, it significantly inhibited the secretion of inflammatory factors, such as interleukin-8 and tumor necrosis factor alpha, by inhibiting the phosphorylation of p38 mitogen-activated protein kinase (MAPK) and nuclear factor kappa B (NF-κB). Moreover, Rb3 pre-treatment led to an increase in the levels of glutathione (GSH) and activities of superoxide dismutase, glutathione peroxidase (GSH-Px), and catalase to reduce the oxidative stress induced by CSE. Additionally, Rb3 decreased the levels of ECM proteins including collagen I (Col I), Col III, and elastin after CSE treatment by inhibiting the expression of transforming growth factor beta 1 (TGF-ß1)-induced vascular endothelial growth factor (VEGF). Our findings suggest that Rb3 prevented CSE-induced inflammation and oxidative stress as well as the excessive accumulation of ECM in WI-38 and 16HBE cells to protect against cell injury by inhibiting the p38 MAPK/NF-κB and TGF-ß1/VEGF pathways. The results of this study may be valuable for the development of Rb3 to combat the damage caused by cigarette smoke.


Subject(s)
Cigarette Smoking , Epithelial Cells/metabolism , Fibroblasts/metabolism , Ginsenosides/pharmacology , NF-kappa B/metabolism , Transforming Growth Factor beta1/metabolism , Vascular Endothelial Growth Factor A/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Antioxidants/pharmacology , Cell Line , Cell Survival/drug effects , Epithelial Cells/drug effects , Extracellular Matrix/drug effects , Extracellular Matrix/metabolism , Fibroblasts/drug effects , Ginsenosides/chemistry , Humans , Inflammation/pathology , Protective Agents/chemistry , Protective Agents/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...