Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 160
Filter
1.
Mol Med ; 30(1): 59, 2024 May 14.
Article in English | MEDLINE | ID: mdl-38745316

ABSTRACT

Microglial activation and polarization play a central role in poststroke inflammation and neuronal damage. Modulating microglial polarization from pro-inflammatory to anti-inflammatory phenotype is a promising therapeutic strategy for the treatment of cerebral ischemia. Polyphyllin I (PPI), a steroidal saponin, shows multiple bioactivities in various diseases, but the potential function of PPI in cerebral ischemia is not elucidated yet. In our study, the influence of PPI on cerebral ischemia-reperfusion injury was evaluated. Mouse middle cerebral artery occlusion (MCAO) model and oxygen-glucose deprivation and reoxygenation (OGD/R) model were constructed to mimic cerebral ischemia-reperfusion injury in vivo and in vitro. TTC staining, TUNEL staining, RT-qPCR, ELISA, flow cytometry, western blot, immunofluorescence, hanging wire test, rotarod test and foot-fault test, open-field test and Morris water maze test were performed in our study. We found that PPI alleviated cerebral ischemia-reperfusion injury and neuroinflammation, and improved functional recovery of mice after MCAO. PPI modulated microglial polarization towards anti-inflammatory M2 phenotype in MCAO mice in vivo and post OGD/R in vitro. Besides, PPI promoted autophagy via suppressing Akt/mTOR signaling in microglia, while inhibition of autophagy abrogated the effect of PPI on M2 microglial polarization after OGD/R. Furthermore, PPI facilitated autophagy-mediated ROS clearance to inhibit NLRP3 inflammasome activation in microglia, and NLRP3 inflammasome reactivation by nigericin abolished the effect of PPI on M2 microglia polarization. In conclusion, PPI alleviated post-stroke neuroinflammation and tissue damage via increasing autophagy-mediated M2 microglial polarization. Our data suggested that PPI had potential for ischemic stroke treatment.


Subject(s)
Autophagy , Disease Models, Animal , Microglia , Neuroinflammatory Diseases , Reperfusion Injury , Animals , Microglia/drug effects , Microglia/metabolism , Mice , Reperfusion Injury/drug therapy , Reperfusion Injury/metabolism , Reperfusion Injury/etiology , Autophagy/drug effects , Male , Neuroinflammatory Diseases/etiology , Neuroinflammatory Diseases/drug therapy , Neuroinflammatory Diseases/metabolism , Diosgenin/analogs & derivatives , Diosgenin/pharmacology , Diosgenin/therapeutic use , Brain Ischemia/drug therapy , Brain Ischemia/metabolism , Signal Transduction/drug effects , Infarction, Middle Cerebral Artery/drug therapy , TOR Serine-Threonine Kinases/metabolism , Mice, Inbred C57BL , Cell Polarity/drug effects
2.
Biochem Biophys Res Commun ; 712-713: 149941, 2024 Jun 18.
Article in English | MEDLINE | ID: mdl-38643718

ABSTRACT

While diosgenin has been demonstrated effective in various cardiovascular diseases, its specific impact on treating heart attacks remains unclear. Our research revealed that diosgenin significantly improved cardiac function in a myocardial infarction (MI) mouse model, reducing cardiac fibrosis and cell apoptosis while promoting angiogenesis. Mechanistically, diosgenin upregulated the Hand2 expression, promoting the proliferation and migration of endothelial cells under hypoxic conditions. Acting as a transcription factor, HAND2 activated the angiogenesis-related gene Aggf1. Conversely, silencing Hand2 inhibited the diosgenin-induced migration of hypoxic endothelial cells and angiogenesis. In summary, these findings provide new insights into the protective role of diosgenin in MI, validating its effect on angiogenic activity and providing a theoretical basis for clinical treatment strategies.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors , Diosgenin , Mice, Inbred C57BL , Myocardial Infarction , Neovascularization, Physiologic , Animals , Diosgenin/pharmacology , Diosgenin/therapeutic use , Myocardial Infarction/drug therapy , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Neovascularization, Physiologic/drug effects , Basic Helix-Loop-Helix Transcription Factors/metabolism , Basic Helix-Loop-Helix Transcription Factors/genetics , Male , Mice , Cell Proliferation/drug effects , Cell Movement/drug effects , Apoptosis/drug effects , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Endothelial Cells/pathology , Humans , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Angiogenesis
3.
Mol Psychiatry ; 28(6): 2398-2411, 2023 06.
Article in English | MEDLINE | ID: mdl-37085711

ABSTRACT

Central nervous system axons have minimal capacity to regenerate in adult brains, hindering memory recovery in Alzheimer's disease (AD). Although recent studies have shown that damaged axons sprouted in adult and AD mouse brains, long-distance axonal re-innervation to their targets has not been achieved. We selectively visualized axon-growing neurons in the neural circuit for memory formation, from the hippocampus to the prefrontal cortex, and showed that damaged axons successfully extended to their native projecting area in mouse models of AD (5XFAD) by administration of an axonal regenerative agent, diosgenin. In vivo transcriptome analysis detected the expression profile of axon-growing neurons directly isolated from the hippocampus of 5XFAD mice. Secreted protein acidic and rich in cysteine (SPARC) was the most expressed gene in axon-growing neurons. Neuron-specific overexpression of SPARC via adeno-associated virus serotype 9 delivery in the hippocampus recovered memory deficits and axonal projection to the prefrontal cortex in 5XFAD mice. DREADDs (Designer receptors exclusively activated by designer drugs) analyses revealed that SPARC overexpression-induced axonal growth in the 5XFAD mouse brain directly contributes to memory recovery. Elevated levels of SPARC on axonal membranes interact with extracellular rail-like collagen type I to promote axonal remodeling along their original tracings in primary cultured hippocampal neurons. These findings suggest that SPARC-driven axonal growth in the brain may be a promising therapeutic strategy for AD and other neurodegenerative diseases.


Subject(s)
Alzheimer Disease , Diosgenin , Mice , Animals , Alzheimer Disease/genetics , Diosgenin/metabolism , Diosgenin/pharmacology , Diosgenin/therapeutic use , Osteonectin/metabolism , Osteonectin/therapeutic use , Axons/metabolism , Hippocampus/metabolism , Disease Models, Animal , Mice, Transgenic
4.
Pharmacol Res ; 175: 105983, 2022 01.
Article in English | MEDLINE | ID: mdl-34822972

ABSTRACT

Angiogenesis plays an important role in the growth and metastasis of solid tumors including melanoma. Inhibiting tumor-associated angiogenesis is a tactic in treating melanoma. Dioscin restrains angiogenesis in colon tumor and has anti-melanoma effects in cell and animal models. In a previous study, we found that dioscin inhibits Src/STAT3 signaling in melanoma cells. Activation of the Src/STAT3 pathway has been shown to promote tumor angiogenesis. This study aimed to determine whether dioscin's anti-melanoma effects is related to inhibiting Src/STAT3 signaling-mediated angiogenesis. In a B16F10 allograft mouse model, we found that dioscin inhibited melanoma growth and angiogenesis. To exclude the impact of tumor growth on angiogenesis, a chicken chorioallantoic membrane (CAM) model was used to verify the anti-angiogenic effect of dioscin. Results showed that dioscin suppressed vessel formation in CAM. To determine if tumor secreted pro-angiogenic cytokines are involved in the anti-angiogenic effect of dioscin, conditioned media from dioscin-treated A375 melanoma cells were used to culture human umbilical vein endothelial cells (HUVECs), and tube formation was monitored. It was observed that the tube formation of HUVECs was inhibited. Mechanistic studies revealed that dioscin inhibited the activation of Src and STAT3, and lowered mRNA and protein levels of STAT3 transcriptionally-regulated genes, in B16F10 melanomas. ELISA assays showed that dioscin decreased the secretion of MMP-2, MMP-9 and VEGF from A375 cells. Over-activation of STAT3 lessened the effects of dioscin in decreasing the secretion of pro-angiogenic cytokines from melanoma cells, and in inhibiting tube formation of HUVECs cultured with conditioned media from melanoma cell cultures. In summary, we for the first time demonstrated that inhibiting Src/STAT3 signaling-mediated angiogenesis is involved in the anti-melanoma effects of dioscin. This study provides further pharmacological groundwork for developing dioscin as an anti-melanoma agent.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Diosgenin/analogs & derivatives , Melanoma, Experimental/drug therapy , Neovascularization, Pathologic/drug therapy , STAT3 Transcription Factor/antagonists & inhibitors , src-Family Kinases/antagonists & inhibitors , Angiogenesis Inhibitors/pharmacology , Animals , Cell Line, Tumor , Diosgenin/pharmacology , Diosgenin/therapeutic use , Human Umbilical Vein Endothelial Cells/drug effects , Humans , Male , Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Mice, Inbred C57BL , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , STAT3 Transcription Factor/metabolism , Tumor Burden/drug effects , src-Family Kinases/metabolism
5.
Drug Chem Toxicol ; 45(3): 1264-1275, 2022 May.
Article in English | MEDLINE | ID: mdl-32924642

ABSTRACT

Idiopathic Pulmonary Fibrosis (IPF) is a grave disease characterized by abnormal wound healing associated with chronic, progressive, irreversible fatal lung disease, leading to persistent injuries to the alveolar epithelium. A consequent disturbance of fibroblast proliferation and apoptosis results in subsequent release of pro-inflammatory and pro-fibrotic mediators coupled with accumulation of extracellular matrix within the interstitium. Inexorable distortion of lung alveolar architecture leads to respiratory failure with a median survival rate of 3-5 years. Currently available drugs can only slowdown the progression of fibrosis and novel drugs are warranted to treat this disease. In this study, we demonstrate the fibro-protective effect of diosgenin in experimental lung fibrosis through regulation of Epithelial Mesenchymal Transition (EMT). A single dose of 3 U/kg body weight (b.wt) Bleomycin (BLM) was administered intratracheally in Wistar male albino rats and fibrotic animals were treated with diosgenin (100 mg/kg b.wt) orally for 28 days. BLM administered rat show histological alteration with increased mast cell and collagen accumulation. BLM induced abnormalities were significantly reduced upon treatment with diosgenin. Western blot analysis revealed an increased level of pro-inflammatory and pro-fibrotic molecules such as IL-1ß and TGF-ß in BLM induced rats. Rats supplemented with diosgenin showed a decreased expression of inflammatory and pro-fibrotic mediators. Markers of EMT molecules were evaluated by immunoblot. The results of immunoblot demonstrate that diosgenin regulated the expression of TGF-ß mediated EMT. Hence, from the overall study, administration of diosgenin prevents pulmonary fibrosis by restraint inflammation and EMT.


Subject(s)
Diosgenin , Pulmonary Fibrosis , Animals , Bleomycin/toxicity , Diosgenin/metabolism , Diosgenin/pharmacology , Diosgenin/therapeutic use , Epithelial-Mesenchymal Transition , Lung , Male , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/drug therapy , Pulmonary Fibrosis/prevention & control , Rats , Rats, Wistar , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta/pharmacology , Transforming Growth Factor beta/therapeutic use , Transforming Growth Factor beta1/metabolism
6.
Drug Dev Res ; 83(8): 1725-1738, 2022 12.
Article in English | MEDLINE | ID: mdl-36126194

ABSTRACT

Diosgenin, a steroidal saponin, is a natural product found in many plants. Diosgenin has a wide range of pharmacological activities, and has been used to treat cancer, nervous system diseases, inflammation, and infections. Numerous studies have shown that diosgenin has potential therapeutic value for lipid metabolism diseases via various pathways and mechanisms, such as controlling lipid synthesis, absorption, and inhibition of oxidative stress. These mechanisms and pathways have provided ideas for researchers to develop related drugs. In this review, we focus on data from animal and clinical studies, summarizing the toxicity of diosgenin, its pharmacological mechanism, recent research advances, and the related mechanisms of diosgenin as a drug for the treatment of lipid metabolism, especially in obesity, hyperlipidemia, nonalcoholic fatty liver disease, atherosclerosis, and diabetes. This systematic review will briefly describe the advantages of diosgenin as a potential therapeutic drug and seek to enhance our understanding of the pharmacological mechanism, recipe-construction, and the development of novel therapeutics against lipid metabolism diseases.


Subject(s)
Diosgenin , Animals , Diosgenin/pharmacology , Diosgenin/therapeutic use , Lipid Metabolism , Oxidative Stress , Antioxidants/pharmacology , Inflammation/drug therapy
7.
Pharmacol Res ; 172: 105796, 2021 10.
Article in English | MEDLINE | ID: mdl-34343656

ABSTRACT

Restoring immune balance by targeting macrophage polarization is a potentially valuable therapeutic strategy for ulcerative colitis (UC). Dioscin is a steroidal saponin with potent anti-inflammatory, immunoregulatory, and hypolipidemic effects. This study examined the protective effect of Dioscin on UC in mice and explored the underlying mechanisms. Mice were induced colitis by dextran sulfate sodium (DSS) and concurrently treated with Dioscin oral administration. RAW264.7 cells were skewed to M1 macrophage polarization by lipopolysaccharide (LPS) and interferon-γ (INF-γ) in vitro, and received Dioscin treatment. The results showed that Dioscin ameliorated colitis in mice, reduced macrophage M1 polarization, but markedly promoted M2 polarization in mice colon. Dioscin inhibited mammalian target rapamycin complex 1 (mTORC1)/hypoxia-inducible factor-1α (HIF-1α) signaling and restrained glycolysis in RAW264.7; however, it activated mammalian target rapamycin complex 2 (mTORC2)/peroxisome proliferator-activated receptor-γ (PPAR-γ) signal and facilitated fatty acid oxidation (FAO). The modulation of mTORs signaling may inhibit M1, but promote M2 polarization. Furthermore, the effect of Dioscin on M2 polarization was neutralized by the FAO inhibitor Etomoxir and the mTORC2 inhibitor JR-AB2-011. In parallel, the inhibitory effect of Dioscin on M1 polarization was mitigated by the mTORC1 agonist L-leucine. Both JR-AB2-011 and L-leucine blocked the therapeutic effect of Dioscin in mice with UC. Therefore, Dioscin ameliorated UC in mice, possibly by restraining M1, while skewing M2 polarization of macrophages. Regulation of mTORC1/HIF-1α and mTORC2/PPAR-γ signals is a possible mechanism by which Dioscin inhibited aerobic glycolysis and promoted FAO of macrophages. In summary, Dioscin protected mice against DSS-induced UC by regulating mTOR signaling, thereby adjusting macrophage metabolism and polarization.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Colitis, Ulcerative/drug therapy , Diosgenin/analogs & derivatives , Macrophages/drug effects , Animals , Anti-Inflammatory Agents/pharmacology , Colitis, Ulcerative/chemically induced , Colitis, Ulcerative/immunology , Colitis, Ulcerative/pathology , Colon/drug effects , Colon/immunology , Colon/pathology , Cytokines/genetics , Dextran Sulfate , Diosgenin/pharmacology , Diosgenin/therapeutic use , Disease Models, Animal , Hypoxia-Inducible Factor 1, alpha Subunit/antagonists & inhibitors , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Macrophages/immunology , Male , Mechanistic Target of Rapamycin Complex 1/antagonists & inhibitors , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice , Mice, Inbred BALB C , PPAR gamma/metabolism , RAW 264.7 Cells
8.
Bioorg Med Chem ; 37: 116109, 2021 05 01.
Article in English | MEDLINE | ID: mdl-33780813

ABSTRACT

A novel series of multitargeted molecules were designed and synthesized by combining the pharmacological role of cholinesterase inhibitor and antioxidant of steroid as potential ligands for the treatment of Vascular Dementia (VD). The oxygen-glucose deprivation (OGD) model was used to evaluate these molecules, among which the most potent compound ML5 showed the highest activity. Firstly, ML5 showed appropriate inhibition of cholinesterases (ChEs) at orally 15 mg/kg in vivo. The further test revealed that ML5 promoted the nuclear translocation of Nrf2. Furthermore, ML5 has significant neuroprotective effect in vivo model of bilateral common carotid artery occlusion (BCCAO), significantly increasing the expression of Nrf2 protein in the cerebral cortex. In the molecular docking research, we predicted the ML5 combined with hAChE and Keap1. Finally, compound ML5 displayed normal oral absorption and it was nontoxic at 500 mg/kg, po, dose. We can draw the conclusion that ML5 could be considered as a new potential compound for VD treatment.


Subject(s)
Central Nervous System Agents/therapeutic use , Cholinesterase Inhibitors/therapeutic use , Dementia, Vascular/drug therapy , Diosgenin/analogs & derivatives , Diosgenin/therapeutic use , Protective Agents/therapeutic use , Acetylcholinesterase/metabolism , Animals , Cell Survival/drug effects , Central Nervous System Agents/chemical synthesis , Central Nervous System Agents/metabolism , Central Nervous System Agents/toxicity , Cholinesterase Inhibitors/chemical synthesis , Cholinesterase Inhibitors/metabolism , Cholinesterase Inhibitors/toxicity , Diosgenin/metabolism , Diosgenin/toxicity , Humans , Kelch-Like ECH-Associated Protein 1/metabolism , Learning/drug effects , Male , Memory/drug effects , Mice, Inbred ICR , Molecular Docking Simulation , NF-E2-Related Factor 2/metabolism , Neuroprotection/drug effects , Protective Agents/chemical synthesis , Protective Agents/metabolism , Protective Agents/toxicity , Protein Binding , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism
9.
Molecules ; 26(9)2021 Apr 23.
Article in English | MEDLINE | ID: mdl-33922675

ABSTRACT

Although the anti-allergic and prebiotic activities of diosgenin have been reported, the influence of diosgenin on intestinal immune and epithelial cells remains unclear. As the gut microbiota plays an important role in allergic disorders, this study aimed to investigate whether the anti-allergic diarrhea effect of diosgenin occurs via improving gut dysbiosis. In a murine food allergy model, the density of fecal bacterial growth on de Man, Rogossa and Sharpe (MRS) plates was diminished, and growth on reinforced clostridial medium (RCM) and lysogeny broth (LB) agar plates was elevated. However, the oral administration of diosgenin reduced the density of fecal bacteria and ameliorated diarrhea severity. Concordantly, reshaped diversity and an abundance of fecal microbes were observed in some of the diosgenin-treated mice, which showed a milder severity of diarrhea. The relevant fecal strains from the diosgenin-treated mice were defined and cultured with Caco-2 cells and allergen-primed mesenteric lymph node (MLN) cells. These strains exhibited protective effects against the cytokine/chemokine network and allergen-induced T-cell responses to varying degrees. By contrast, diosgenin limitedly regulated cytokine production and even reduced cell viability. Taken together, these findings show that diosgenin per se could not directly modulate the functionality of intestinal epithelial cells and immune cells, and its anti-allergic effect is most likely exerted via improving gut dysbiosis.


Subject(s)
Anti-Allergic Agents/therapeutic use , Diosgenin/therapeutic use , Dysbiosis/drug therapy , Food Hypersensitivity/drug therapy , Animals , Caco-2 Cells , Disease Models, Animal , Gastrointestinal Microbiome , Humans , Mice
10.
Arch Biochem Biophys ; 687: 108363, 2020 07 15.
Article in English | MEDLINE | ID: mdl-32335049

ABSTRACT

Polyphyllin I (PPI), an extract from Paris polyphylla, has been demonstrated to possess antitumor activity against multiple cancers. However, whether PPI can inhibit bladder cancer (BCa) and the underlying mechanisms have never been researched. In this study, we initially found that PPI could induce BCa cell apoptosis and cell cycle arrest, as well as inhibit cell proliferation in vitro. Additionally, PPI could effectively suppress the in vivo growth of BCa in the xenograft mice model. Furthermore, we found that forkhead box O3 (FOXO3) and its targets including BIM or NOXA were significantly upregulated in BCa cells following PPI treatment. Interestingly, we observed that FOXO3 knockdown partly reversed the effects of PPI on BCa cells. Taken together, our findings suggested that PPI exerted a cytotoxic effect in vitro and an antitumor activity in vivo against BCa partly by activating FOXO3 signaling pathway. Therefore, PPI may serve as a promising chemotherapy agent for BCa treatment.


Subject(s)
Apoptosis/drug effects , Diosgenin/analogs & derivatives , Forkhead Box Protein O3/metabolism , Signal Transduction/drug effects , Urinary Bladder Neoplasms/drug therapy , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Diosgenin/therapeutic use , Female , Forkhead Box Protein O3/genetics , G2 Phase Cell Cycle Checkpoints/drug effects , Gene Knockdown Techniques , Humans , Mice, Inbred BALB C , S Phase Cell Cycle Checkpoints/drug effects , Xenograft Model Antitumor Assays
11.
Arch Biochem Biophys ; 687: 108285, 2020 07 15.
Article in English | MEDLINE | ID: mdl-32074500

ABSTRACT

Polyphyllin I (PPI), a bioactive constituent extracted from the rhizomes of Paris polyphylla, is cytotoxic to several cancer types. This study was designed to explore whether PPI prevents non-small-cell lung cancer (NSCLC) growth and to investigate the molecular mechanism. AMP-activated protein kinase (AMPK) has been implicated in the activation of autophagy in distinct tissues. In cultured human NSCLC cell lines, PPI induces autophagy by activating AMPK and then inhibiting mTOR signaling in a concentration-dependent manner. Furthermore, the activation of autophagy induced by PPI was reversed by the AMPK inhibitor compound C. Computational docking showed that PPI directly interacted with the allosteric drug and metabolite site of AMPK to stabilize its activation. Microscale thermophoresis and Drug Affinity Responsive Targeting Stability (DARTS) assay further confirmed the high affinity between PPI and AMPK. In vivo studies indicated that PPI suppressed the growth of NSCLC and increased the levels of LC3-II and phosphorylated AMPK in tumors isolated from a xenograft model of NSCLC in mice. Moreover, PPI exhibited favorable pharmacokinetics in rats. In summary, PPI conclusively acts as a direct AMPK activator to induce cell autophagy which inhibits the growth of NSCLC cells. In the future, PPI therapy should be applied to treat patients with NSCLC.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Antineoplastic Agents/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Diosgenin/analogs & derivatives , Enzyme Activators/therapeutic use , Lung Neoplasms/drug therapy , AMP-Activated Protein Kinases/chemistry , Allosteric Site , Animals , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacokinetics , Apoptosis/drug effects , Autophagy/drug effects , Cell Line, Tumor , Diosgenin/metabolism , Diosgenin/pharmacokinetics , Diosgenin/therapeutic use , Enzyme Activators/metabolism , Enzyme Activators/pharmacokinetics , Female , Humans , Male , Mice, Nude , Molecular Docking Simulation , Protein Binding , Rats, Sprague-Dawley , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism , Xenograft Model Antitumor Assays
12.
J Biochem Mol Toxicol ; 34(2): e22422, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31729780

ABSTRACT

M1 macrophages serve one edge as proinflammatory and M2 macrophages serve the other edge as an anti-inflammatory macrophage. It appears that a related "switch" in macrophage morphology may also happen in the course of atherosclerosis, which has not yet been elucidated. An atherogenic diet (AD) was given to rats, and induction of macrophage differentiation and the nuclear localization of nuclear factor-kappa B (NFκB) were investigated by Western blot and immunofluorescence. Chemokines were analyzed using an antibody array with 32 target proteins. M2 macrophage transformation was confirmed in diosgenin-treated aorta by immunofluorescence and was validated in vitro using THP-1 cells. MAC387 (macrophage marker) and NFκBp65 (inflammatory hub) were upregulated in oxidatively-modified low-density lipoprotein (OxyLDL) and AD-induced condition. Macrophage differentiation, which induced the formation of inflammatory mediators, was not significantly suppressed by the inhibition of NFκB using dexamethasone. M1 macrophage polarization was identified in OxyLDL-induced monocytes, which are proinflammatory in nature, whereas M2 macrophage polarization was noticed in diosgenin-treated monocytes, which exhibit anti-inflammatory properties. M1-and M2-specific chemokines were analyzed using chemokine antibody array. Furthermore, the expression of proinflammatory macrophage (M1) was noticed in AD-induced aorta and anti-inflammatory macrophage (M2) was observed in diosgenin-treated aorta. This is the first report where, unifying the mechanism of diosgenin as aan nti-atherosclerotic and the expression of M1 and M2 specific chemokines is shown by downregulating NFκB and not by preventing the differentiation of monocyte into a macrophage, but by allowing macrophage to differentiate into M2, which aids in preventing the atherosclerotic progression.


Subject(s)
Aorta/metabolism , Atherosclerosis/metabolism , Cell Polarity , Cytokines/metabolism , Diosgenin/pharmacology , Macrophages/metabolism , Plant Extracts/pharmacology , Transcription Factor RelA/metabolism , Animals , Antigens, CD/genetics , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/genetics , Antigens, Differentiation, Myelomonocytic/metabolism , Atherosclerosis/drug therapy , Atherosclerosis/prevention & control , CD36 Antigens/genetics , CD36 Antigens/metabolism , Cell Differentiation/drug effects , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Dexamethasone/pharmacology , Diet, Atherogenic/adverse effects , Dioscorea/chemistry , Diosgenin/therapeutic use , Humans , Lipoproteins, LDL/pharmacology , Male , Monocytes/metabolism , Plant Extracts/therapeutic use , Rats , Signal Transduction/drug effects , THP-1 Cells , Transcription Factor RelA/antagonists & inhibitors , Transcription Factor RelA/genetics
13.
Pharmacol Res ; 139: 431-439, 2019 01.
Article in English | MEDLINE | ID: mdl-30503840

ABSTRACT

Inflammatory reaction and cell apoptosis are two important processes in intestinal ischemia/reperfusion (II/R) injury, and exploration of effective lead compounds against II/R injury via regulating inflammation and apoptosis is critical important. In this paper, the results indicated that dioscin significantly increased cell viability, and inhibited inflammation and apoptosis caused by hypoxia-reoxygenation (H/R) injury in IEC-6 cells. in vivo II/R injury, dioscin markedly suppressed inflamma- tion and apoptosis, improved pathological changes, and depressed chiu' score in rats. Mechanistic studies indicated that dioscin notably up-regulated the expression level of MAPK13 through decreasing miR-351-5p level, and thereby decreased the expression levels of p-PKD1, NF-κB, Apaf-1, cleaved Caspase-3 and cleaved Caspase-9. Furthermore, miR-351-5p mimic and inhibitor experiments in IEC-6 cells further proved that dioscin up-regulated MAPK13 expression by decreasing miR-351-5p level to inhibit inflammation and apoptosis. Therefore, dioscin showed protective effect against II/R injury via adjusting miR-351-5/MAPK13-mediated inflammation and apoptosis. Dioscin should be considered as one potent candidate and miR-351-5/ MAPK13 should be one effective drug target for the treatment of II/R injury.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Diosgenin/analogs & derivatives , Protective Agents/therapeutic use , Reperfusion Injury/drug therapy , Animals , Anti-Inflammatory Agents/pharmacology , Apoptosis/drug effects , Cell Hypoxia/drug effects , Cell Line , Diosgenin/pharmacology , Diosgenin/therapeutic use , Intestinal Mucosa/metabolism , Male , MicroRNAs/metabolism , Mitogen-Activated Protein Kinase 13/metabolism , Protective Agents/pharmacology , Rats, Sprague-Dawley , Reperfusion Injury/metabolism
14.
Pharmacol Res ; 139: 273-285, 2019 01.
Article in English | MEDLINE | ID: mdl-30471409

ABSTRACT

Colorectal cancer (CRC) is one of the leading causes of cancer-related morbidity and mortality worldwide. Accumulating evidence suggests that mitochondrial dynamics are closely implicated in carcinogenesis including CRC. Paris Saponin II (PSII), a major steroidal saponin extracted from Rhizoma Paris polyphylla, has emerged as a potential anticancer agent. However, the effects of PSII on CRC and its underlying mechanisms remain unknown. In the present study, we found PSII induced apoptosis and inhibited colony formation in HT 29 and HCT 116 cells, and cell cycle arrest in G1 phase. PSII inhibited the phosphorylation of ERK1/2 and mitochondrial translocation of dynamin-related protein 1 (Drp1) by dephosphorylating Drp1 at Ser616, leading to the suppression of mitochondrial fission. PSII also suppressed NF-κB activation as a result of the inhibition of IKKß and p65 translocation. Drp1 knockdown remarkably downregulated the nuclear expression of p65 and its target genes cyclin D1 and c-Myc in HCT 116 cell, confirming the link between mitochondrial fission and NF-κB pathway. Silencing of Drp 1 enhanced the inhibitory effects of PSII on p65 phosphorylation and the expressions of cyclin D1 and c-Myc, revealing that the inhibitory effects of PSII on cyclin D1 and c-Myc were relevant in the suppression of Drp1 and NF-κB activation. An in vivo study demonstrated PSII remarkably decreased the xenograft tumor size and suppressed the phosphorylation of ERK1/2 and Drp1 at Ser616. Taken together, our results suggested that PSII could inhibit colorectal carcinogenesis, at least in part, by regulating mitochondrial fission and NF-κB pathway.


Subject(s)
Antineoplastic Agents, Phytogenic/therapeutic use , Colorectal Neoplasms/drug therapy , Diosgenin/analogs & derivatives , Saponins/therapeutic use , Animals , Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Proliferation/drug effects , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Diosgenin/pharmacology , Diosgenin/therapeutic use , Female , HCT116 Cells , HT29 Cells , Humans , Mice, Nude , Mitochondrial Dynamics/drug effects , NF-kappa B/antagonists & inhibitors , NF-kappa B/metabolism , Saponins/pharmacology
15.
Mediators Inflamm ; 2019: 3041438, 2019.
Article in English | MEDLINE | ID: mdl-31263381

ABSTRACT

Despite the usefulness of glucocorticoids, they may cause hazardous side effects that limit their use. Searching for compounds that are as equally efficient as glucocorticoids, but with less side effects, the current study compared plant steroids, namely, glycyrrhetinic acid, guggulsterone, boswellic acid, withaferin A, and diosgenin with the classical glucocorticoid, fluticasone. This was approached both in silico using molecular docking against glucocorticoid receptor (GR) and in vivo in two different animal models. All tested compounds interacted with GR, but only boswellic acid and withaferin A showed docking results comparable to fluticasone, as well as similar in vivo anti-inflammatory effects, by significantly decreasing serum levels of interleukin-6 and tumor necrosis factor-α in cotton pellet-induced granuloma in rats. In addition, both compounds significantly decreased the percent of change in ear weight in croton oil-induced ear edema in mice and the granuloma weight in cotton pellet-induced granuloma in rats, to levels comparable to that of fluticasone. Both boswellic acid and withaferin A had no effect on adrenal index, but only withaferin A significantly increased the thymus index. In conclusion, boswellic acid may have comparable anti-inflammatory effects to fluticasone with fewer side effects.


Subject(s)
Ear Diseases/drug therapy , Ear Diseases/metabolism , Inflammation/drug therapy , Inflammation/metabolism , Phytosterols/therapeutic use , Receptors, Glucocorticoid/metabolism , Animals , Anti-Inflammatory Agents/therapeutic use , Croton Oil/toxicity , Diosgenin/therapeutic use , Ear Diseases/blood , Ear Diseases/chemically induced , Edema/blood , Edema/chemically induced , Edema/drug therapy , Edema/metabolism , Enzyme-Linked Immunosorbent Assay , Glycyrrhetinic Acid/therapeutic use , Inflammation/chemically induced , Inflammation/immunology , Interleukin-6/blood , Mice , Molecular Docking Simulation , Pregnenediones/therapeutic use , Rats , Software , Thymus Gland/drug effects , Thymus Gland/metabolism , Triterpenes/therapeutic use , Tumor Necrosis Factor-alpha/blood , Withanolides/therapeutic use
16.
Cell Physiol Biochem ; 47(2): 759-773, 2018.
Article in English | MEDLINE | ID: mdl-29807357

ABSTRACT

BACKGROUND/AIMS: Polyphyllin I (PPI), one of the steroidal saponins in Paris polyphylla, reportedly exhibits antitumor effects. However, the detailed mechanism underlying PPI, particularly in enhancing the effect of the androgen receptor inhibitor enzalutamide in controlling castration-resistant prostate cancer (CRPC) has not been explored. METHODS: Cell viability and cell cycle distribution were measured using 3-(4, 5-dimethylthiazol-2-yl)-2, 5-diphenyltetrazolium bromide (MTT) and flow cytometry assays, respectively. Long non-coding RNA (lncRNA) HOX transcript antisense RNA (HOTAIR) expression was measured by quantitative real time-PCR (qRT-PCR). Western blot analysis was performed to determine the protein expression levels of MUC1, p65, and p50. Silencing of HOTAIR was evaluated using the siRNA procedure. The promoter activity of the MUC1 gene was determined using Secrete-Pair Dual Luminescence Assay Kit. Exogenous expression of HOTAIR, p65, and MUC1 was conducted by transient transfection assay. A xenograft tumor model in nude mice was used to further evaluate the effect of the combination of PPI and enzalutamide in vivo. RESULTS: We showed that PPI significantly inhibited growth and induced cell cycle arrest in CRPC cells. PPI also decreased p65 and MUC1 protein expression and reduced HOTAIR expression. Exogenously expressed p65 resisted the PPI-inhibited expression of HOTAIR, whereas silenced HOTAIR reduced MUC1 protein but exerted no effect on the expression of p65 and p50 proteins. Conversely, exogenously expressed HOTAIR resisted the PPI-inhibited MUC1 protein expression, and excessive expression of MUC1 antagonized the PPI-inhibited cell growth. Notably, PPI combined with enzalutamide exerted a synergistic effect. Consistent with this finding, PPI inhibited tumor growth, HOTAIR expression, as well as p65 and MUC1 protein expressions in vivo. CONCLUSIONS: Our results indicate that PPI inhibits the growth of CRPC cells by inhibiting p65 protein and concomitantly reducing HOTAIR expression, thereby suppressing MUC1 gene expression. The novel regulatory interaction of p65 and HOTAIR converge in the inhibition of MUC1 expression and overall PPI response. The combination of PPI and enzalutamide exhibits synergy. This study reveals a novel mechanism underlying the synergistic inhibitory effect of PPI and enzalutamide on the growth of CRPC cells.


Subject(s)
Apoptosis/drug effects , Diosgenin/analogs & derivatives , Mucin-1/metabolism , Phenylthiohydantoin/analogs & derivatives , RNA, Long Noncoding/metabolism , Transcription Factor RelA/metabolism , Animals , Benzamides , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Diosgenin/pharmacology , Diosgenin/therapeutic use , Drug Synergism , Drug Therapy, Combination , Female , Humans , Male , Mice , Mice, Nude , Mucin-1/genetics , NF-kappa B p50 Subunit/metabolism , Nitriles , Phenylthiohydantoin/pharmacology , Phenylthiohydantoin/therapeutic use , Prostatic Neoplasms, Castration-Resistant/drug therapy , Prostatic Neoplasms, Castration-Resistant/metabolism , Prostatic Neoplasms, Castration-Resistant/pathology , RNA Interference , RNA, Long Noncoding/antagonists & inhibitors , RNA, Long Noncoding/genetics , RNA, Small Interfering/metabolism , Transcription Factor RelA/genetics
17.
Pharmacol Res ; 137: 259-269, 2018 11.
Article in English | MEDLINE | ID: mdl-30315966

ABSTRACT

Currently, the numbers of patients with cancer, fibrosis, diabetes, chronic kidney disease, stroke and osteoporosis are increasing fast and fast. It's critical necessary to discovery lead compounds for new drug development. Dioscin, one active compound in some medicinal plants, has anti-inflammation, immunoregulation, hypolipidemic, anti-viral, anti-fungal and anti-allergic effects. In recent years, dioscin has reached more and more attention with its potent effects to treat liver, kidney, brain, stomach and intestine damages, and metabolic diseases including diabetes, osteoporosis, obesity, hyperuricemia as well as its anti-cancer activities through adjusting multiple targets and multiple signals. Therefore, dioscin is a promising multi-target candidate to treat various diseases. This review paper summarized the progress on pharmacological activities and mechanisms of dioscin, which may provide useful data for development and exploration of this natural product in the further.


Subject(s)
Anti-Infective Agents/therapeutic use , Anti-Inflammatory Agents/therapeutic use , Antineoplastic Agents/therapeutic use , Diosgenin/analogs & derivatives , Metabolic Diseases/drug therapy , Neoplasms/drug therapy , Animals , Diosgenin/therapeutic use , Humans
18.
Pharmacol Res ; 131: 51-60, 2018 05.
Article in English | MEDLINE | ID: mdl-29574225

ABSTRACT

Dioscin, one natural product, has active effect against non-alcoholic fatty liver disease (NAFLD) in our previous work. However, the pharmacological data are insufficient and the mechanisms have not been reported. Thus, this study aims to comprehensively investigate the effects and molecular mechanisms of dioscin against NAFLD. The primary cultured hepatocytes, AML-12 and HepG-2 cells were treated with palmic acid (PA) after dioscin treatment. The mice and rats were induced by high fat diet to establish the in vivo models of NAFLD. Dioscin obviously alleviated liver lipid accumulation symptoms and improved the levels of serum and hepatic biochemical parameters in vitro and in vivo. Further investigations revealed that dioscin significantly attenuated lipid metabolism via adjusting SIRT1/AMPK signal pathway to regulate the expression levels of SREBP-1c, CPT, FAS, SCD, FoxO1 and ATGL. In addition, suppression of SIRT1 by Nicotinamide or abrogation of AMPK by Compound C eliminated the inhibitory effects of dioscin on lipid metabolism. Therefore, our findings further demonstrated that dioscin markedly prevented NAFLD through adjusting lipid metabolism via SIRT1/AMPK signal pathway, which should be developed as a new candidate for NAFLD.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Diosgenin/analogs & derivatives , Lipid Metabolism/drug effects , Non-alcoholic Fatty Liver Disease/drug therapy , Signal Transduction/drug effects , Sirtuin 1/metabolism , Animals , Cell Line , Diosgenin/therapeutic use , Hep G2 Cells , Humans , Liver/drug effects , Liver/metabolism , Liver/pathology , Male , Mice , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/pathology , Rats, Wistar
19.
Med Sci Monit ; 24: 246-253, 2018 Jan 12.
Article in English | MEDLINE | ID: mdl-29329279

ABSTRACT

BACKGROUND Diosgenin, a phytosteroid sapogenin, has anti-inflammatory properties shown to reduce myocardial ischemia-reperfusion injury (MIRI). However, the specific mechanism by which this is achieved is not clear. This study investigated the protective effects of diosgenin on myocardial ischemia/reperfusion (I/R) and the potential anti-inflammatory mechanisms. MATERIAL AND METHODS Healthy adult male SD rats, body weight (b.w.) 250-280 g, were used to model ischemia-reperfusion injury (IRI) and were administered diosgenin (50 mg/kg and 100 mg/kg b.w.) intragastrically for 4 consecutive weeks before surgery. The left anterior descending artery (LAD) was ligated to induce myocardial ischemia for 30 min and reperfusion for 30 min, 60 min, and 120 min while relevant indicators were detected. RESULTS Both 50 mg and 100 mg diosgenin oral administration increased left ventricular developed pressure (LVDP) and maximum changing rate of ventricular pressure (±dp/dtmax), decreased left ventricular end-diastolic pressure (LVEDP), and myocardial enzyme markers. TTC staining suggested that diosgenin reduced myocardial infarct size in the rat model. Pathological results showed that myocardial ischemia and inflammation were alleviated by diosgenin. In addition, the increased expression of tumor necrosis factor-alpha (TNF-α), interleukin-1 beta (IL-1ß) in serum, and myeloperoxidase (MPO) in myocardium were significantly suppressed by diosgenin administration. Diosgenin further inhibited the phosphorylation of transcription factor NF-κB and modulated the expression of downstream inflammatory cytokines by regulating the activation of p38-MAPK and JNK pathways. CONCLUSIONS Results demonstrate diosgenin plays an anti-inflammatory role in the protection of MIRI through regulation of p38-MAPK and JNK pathways and phosphorylation of NF-κB.


Subject(s)
Cardiotonic Agents/therapeutic use , Diosgenin/therapeutic use , Inflammation/drug therapy , Inflammation/etiology , Myocardial Reperfusion Injury/complications , Myocardial Reperfusion Injury/drug therapy , Animals , Cardiotonic Agents/pharmacology , Diosgenin/pharmacology , Heart Function Tests/drug effects , Inflammation/blood , Inflammation/physiopathology , Interleukin-1beta/blood , MAP Kinase Signaling System/drug effects , Male , Myocardial Reperfusion Injury/blood , Myocardial Reperfusion Injury/physiopathology , Myocardium/enzymology , Myocardium/pathology , NF-kappa B/metabolism , Peroxidase/blood , Phosphorylation/drug effects , Rats, Sprague-Dawley , Tumor Necrosis Factor-alpha/blood
20.
Int J Mol Sci ; 19(3)2018 Mar 10.
Article in English | MEDLINE | ID: mdl-29534453

ABSTRACT

Fenugreek is a common herb possessing several bioactive components including diosgenin. Here, dietary fenugreek seed flour and diosgenin were evaluated on a model of endothelium-dependent vasorelaxation by abdominal aortas isolated from rats receiving high-fat, high-sugar diet (HFHSD). 60 male Wistar rats were randomized into six groups: (i) negative control getting conventional rat feed regimen; (ii) positive control receiving HFHSD; (iii) a test group fed 2 g/kg bw/day fenugreek seed flour (containing 10 mg/kg bw/day diosgenin) + HFHSD; (iv) three test groups fed 1, 10 and 50 mg/kg bw/day diosgenin + HFHSD. Alimentary treatments were carried out for six weeks. The abdominal aortas were isolated, and 2 mm wide rings were sectioned off and mounted at a resting tension of 10 mN in organ baths containing Krebs solution (36 °C) exposed to 95% O2 and 5% CO2. After 60-min incubation, a norepinephrine concentration-response (E/c) curve was generated to determine their half-maximal effective concentration (EC50) value. After 60-min wash-out, a pre-contraction with norepinephrine EC50 was made, followed by an acetylcholine E/c curve. Plasma glutathione levels, glutathione-handling enzyme activities and blood antioxidant capacities were also determined. HFHSD significantly decreased the dilatory response to acetylcholine and increased plasma glutathione levels and these effects were significantly reversed by fenugreek seed flour, 10 and 50 mg/kg bw/day diosgenin. Both fenugreek and diosgenin treatments prevent HFHSD-induced endothelial dysfunction and redox changes. As fenugreek treatment was more effective at lower acetylcholine concentrations than diosgenin treatments, components of fenugreek other than diosgenin may contribute to the beneficial effects of dietary fenugreek seed flour.


Subject(s)
Diosgenin/pharmacology , Endothelium, Vascular/drug effects , Metabolic Syndrome/drug therapy , Plant Extracts/pharmacology , Vasodilation , Animals , Arteries/drug effects , Arteries/physiology , Diosgenin/administration & dosage , Diosgenin/therapeutic use , Endothelium, Vascular/physiology , Male , Metabolic Syndrome/prevention & control , Plant Extracts/administration & dosage , Plant Extracts/therapeutic use , Rats , Rats, Wistar , Trigonella
SELECTION OF CITATIONS
SEARCH DETAIL