Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
Add more filters










Publication year range
1.
Int Immunopharmacol ; 136: 112380, 2024 Jul 30.
Article in English | MEDLINE | ID: mdl-38850790

ABSTRACT

BACKGROUND AND AIMS: Impaired intestinal barrier function is key in maintaining intestinal inflammation in Crohn's disease (CD). However, no targeted treatment in clinical practice has been developed. Peiminine (Pm) strongly protects the epithelial barrier, the purpose of this study is to investigate whether Pm affects CD-like colitis and potential mechanisms for its action. METHODS: Trinitro-benzene-sulfonic acid (TNBS)-induced mice and Il-10-/- mice were used as CD animal models. Colitis symptoms, histological analysis, and intestinal barrier permeability were used to assess the Pm's therapeutic effect on CD-like colitis. The colon organoids were induced by TNF-α to evaluate the direct role of Pm in inhibiting apoptosis of the intestinal epithelial cells. Western blotting and small molecule inhibitors were used to investigate further the potential mechanism of Pm in inhibiting apoptosis of intestinal epithelial cells. RESULTS: Pm treatment reduced body weight loss, disease activity index (DAI) score, and inflammatory score, demonstrating that colonic inflammation in mice were alleviated. Pm decreased the intestinal epithelial apoptosis, improved the intestinal barrier function, and prevented the loss of tight junction proteins (ZO1 and claudin-1) in the colon of CD mice and TNF-α-induced colonic organoids. Pm activated Nrf2/HO1 signaling, which may protect intestinal barrier function. CONCLUSIONS: Pm inhibits intestinal epithelial apoptosis in CD mice by activating Nrf2/HO1 pathway. This partially explains the potential mechanism of Pm in ameliorating intestinal barrier function in mice and provides a new approach to treating CD.


Subject(s)
Apoptosis , Colitis , Crohn Disease , Disease Models, Animal , Intestinal Mucosa , Mice, Inbred C57BL , Mice, Knockout , NF-E2-Related Factor 2 , Signal Transduction , Trinitrobenzenesulfonic Acid , Animals , NF-E2-Related Factor 2/metabolism , Crohn Disease/drug therapy , Crohn Disease/pathology , Intestinal Mucosa/drug effects , Intestinal Mucosa/pathology , Intestinal Mucosa/metabolism , Colitis/drug therapy , Colitis/chemically induced , Colitis/pathology , Mice , Signal Transduction/drug effects , Apoptosis/drug effects , Humans , Male , Colon/pathology , Colon/drug effects , Heme Oxygenase-1/metabolism , Heme Oxygenase-1/genetics , Heme Oxygenase (Decyclizing)/metabolism , Heme Oxygenase (Decyclizing)/genetics , Interleukin-10/metabolism , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Membrane Proteins
2.
Chem Biol Drug Des ; 103(6): e14528, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38811358

ABSTRACT

Peimenine (PEI) is a steroid alkaloid substance isolated from Fritillaria thunbergii bulbs. It has various pharmacological activities, such as relief from coughs and asthma, expectorant properties, antibacterial effects, sedative qualities, and anti-inflammatory properties. Notably, PEI can effectively inhibit the proliferation and tumor formation of liver cancer and osteosarcoma cells by inducing autophagic cell death. However, the precise effect and mechanisms of PEI on urothelial bladder cancer (UBC) cells remain uncertain. Thus, this study aims to investigate the impact of PEI on UBC cells both in vivo and in vitro. The IC50 values of BIU-87 and EJ-1 cells after 48 h were 710.3 and 651.1 µg/mL, respectively. Additionally, PEI blocked the cell cycle in BIU-87 and EJ-1 cells during the G1 phase. Furthermore, it hindered the migration of BIU-87 and EJ-1 cells substantially. PEI significantly inhibited the tumor development of EJ-1 cells within the xenograft tumor model in vivo. Mechanically, PEI augmented the protein and mRNA expression of BIM, BAK1, and Cytochrome C (CYCS) in UBC cells. Taken together, PEI suppressed the proliferation of UBC cells both in vitro and in vivo by inducing cell death and cell cycle arrest, suggesting that PEI could be applied in the treatment of UBC.


Subject(s)
Cell Proliferation , Urinary Bladder Neoplasms , Urinary Bladder Neoplasms/drug therapy , Urinary Bladder Neoplasms/pathology , Urinary Bladder Neoplasms/metabolism , Humans , Cell Proliferation/drug effects , Cell Line, Tumor , Animals , Mice , Apoptosis/drug effects , Mice, Nude , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Mice, Inbred BALB C , Xenograft Model Antitumor Assays , Cell Movement/drug effects , Signal Transduction/drug effects
3.
Biomed Pharmacother ; 173: 116353, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38432128

ABSTRACT

Peiminine, the primary biologically active compound from Fritillaria thunbergii Miq., has demonstrated significant pharmacological activities. Doxorubicin is one of the most potent chemotherapeutic agents for breast cancer (BC). This study was designed to investigate the efficacy and underlying mechanisms of Peiminine combined with Doxorubicin in treating BC. Our results demonstrated that the combination of Peiminine and 1 mg/kg Doxorubicin exhibited more significant suppression of tumor growth compared with the monotherapy in MDA-MB-231 xenograft nude mice model, which is comparable to the effect of 3 mg/kg Doxorubicin in vivo. Notably, the 3 mg/kg Doxorubicin monotherapy resulted in organ toxicity, specifically in the liver and heart, whereas no toxicity was observed in the combination group. In vitro, this combined treatment exhibited a synergistic reduction on the viability of BC cells. Peiminine enhanced the cell cycle arrest and DNA damage induced by Doxorubicin. Furthermore, the combination treatment effectively blocked DNA repair by inhibiting the MAPKs signaling pathways. And ZEB1 knockdown attenuated the combined effect of Peiminine and Doxorubicin on cell viability and DNA damage. In conclusion, our study found that the combination of Peiminine and Doxorubicin showed synergistic inhibitory effects on BC both in vivo and in vitro through enhancing Doxorubicin-induced DNA damage. These findings support that their combination is a novel and promising therapeutic strategy for treating BC.


Subject(s)
Breast Neoplasms , Cevanes , Mice , Animals , Humans , Female , Breast Neoplasms/drug therapy , Mice, Nude , Doxorubicin/pharmacology , Doxorubicin/therapeutic use , DNA Adducts/pharmacology , DNA Adducts/therapeutic use , Cell Line, Tumor , Apoptosis , Zinc Finger E-box-Binding Homeobox 1
4.
Int J Environ Health Res ; : 1-13, 2024 Feb 22.
Article in English | MEDLINE | ID: mdl-38385345

ABSTRACT

The deleterious impact of lead (Pb) pollution on human health is evident in both domestic and occupational settings, provoking an inflammatory response across multiple tissue, limited attention has been devoted to its adverse effects on colitis and the underlying mechanisms. Peiminine (PMI) has been recognized for its anti-inflammatory properties, yet its specific anti-inflammatory effects in lead-induced colitis models remain elusive. Through the establishment of both in vivo and in vitro lead exposure models, suggests that lead exposure can induce colitis and that PMI regulates lead exposure-induced colitis by inhibiting the NF-kB signaling pathway, and alleviates the ability of lead to apoptosis and inflammation levels in intestinal epithelial cells. Consequently, these results present a promising avenue for further exploration of the molecular mechanisms underlying lead-induced colitis, evaluation of the associated risks linked to lead exposure, and the development of therapeutic interventions for colitis resulting from lead exposure.

5.
Tissue Cell ; 87: 102323, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38412577

ABSTRACT

BACKGROUND: Peiminine (PMI) is an active alkaloid sourced from Fritillaria thunbergii, which has been shown to suppress the development of a variety of tumors. Whereas, the roles and precise mechanism of PMI in breast cancer (BC) development remain not been clarified. METHODS: The cytotoxic effect of PMI on MCF-10A and BC cell lines (MCF-7 and BT-549) were assessed by MTT and LDH release assay. Cell proliferation was evaluated by EdU staining. Levels of Malondialdehyde (MDA), reactive oxygen species (ROS), glutathione (GSH) activity and iron assay were measured by Enzyme linked immunosorbent assay (ELISA) kits, respectively. Transmission Electron Microscope was performed to observe mitochondrial morphological structure. Immunofluorescence, immunohistochemistry, and western blot were conducted to examine protein levels, respectively. Xenograft model was used to confirm cellular findings. RESULTS: PMI treatment reduced the viability and enhanced LDH level of MCF-7 and BT-549 cells in a time- and concentration-dependent manner, and further suppressed cell proliferation in vitro and tumor growth in vivo. Subsequently, PMI administration resulted in significant increases of ROS, MDA and iron levels, reduction of GSH activity as well as mitochondrial shrinkage and GPX4 reduction, while all these phenomena could be rescued by ferrostatin-1. Mechanistically, PMI treatment led to promoted Nrf2 expression and its nuclear translocation, as well as it's downstream protein HO-1 and NQO1 expressions. Notably, ML-385, a Nrf2 specific inhibitor, greatly reversed the anti-tumor effects and pro-ferroptosis role of PMI in BC cells. CONCLUSION: Taking these finding together, PMI could stimulate ferroptosis to inhibit BC tumor growth by activating Nrf2-HO-1 signaling pathway.


Subject(s)
Breast Neoplasms , Cevanes , Ferroptosis , Humans , Female , Breast Neoplasms/drug therapy , NF-E2-Related Factor 2 , Reactive Oxygen Species , Signal Transduction , Iron
6.
Antioxidants (Basel) ; 13(1)2024 Jan 22.
Article in English | MEDLINE | ID: mdl-38275656

ABSTRACT

Peiminine is the main natural alkaloid compound extracted from the Chinese herb Fritillaria. Although peiminine is known for its antioxidant and anti-inflammatory effects in conditions such as mastitis and arthritis, its impact on inflammation induced by Cutibacterisum acnes (C. acnes) has not been explored. The aim of this study was to investigate the effect of peiminine on C. acnes-induced inflammatory responses in the skin and to identify the underlying mechanism involved. We discovered that peiminine inhibits the C. acnes-induced expression of inflammatory mediators such as pro-interleukin-1ß (pro-IL-1ß), cyclooxygenase-2 (COX-2), tumor necrosis factor-α (TNF-α), and interleukin-6 (IL-6) in mouse bone marrow-derived macrophages (BMDMs). Peiminine suppressed the activation of nuclear factor-kappa B (NF-κB) without affecting the activation of mitogen-activated protein kinase (MAPK) pathways such as JNK, ERK, and p38 MAPK. In addition, we found that peiminine suppressed inflammatory cytokine expression and ameliorated histological symptoms in C. acnes-induced mouse skin. Our study is the first to provide evidence that peiminine has an inhibitory effect on acne, and it points toward the potential of incorporating peiminine into cosmetic and pharmaceutical formulations for acne treatment.

7.
Inflammopharmacology ; 32(1): 657-665, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37855980

ABSTRACT

Ulcerative colitis is a chronic inflammatory disorder of the intestinal mucosa and a prevalent gastrointestinal condition in developed countries. Peiminine, derived from the Fritillaria imperialis plant, exhibits remarkable anti-inflammatory and anti-cancer properties. This study aims to investigate the anti-inflammatory effects of peiminine in an experimental model of ulcerative colitis. Ulcerative colitis was induced intra-rectally in all groups, except the negative control, using 100 µl of 4% acetic acid. Peiminine treatment was initiated after ulcerative colitis induction and symptom manifestation. After the final injection, mice were sacrificed on day 15 for assessment. Various parameters were evaluated, including disease activity index, myeloperoxidase activity, nitric oxide levels, production and expression of IL-1, IL-6, TNF-α cytokines, and expression of IL-1ß, IL-6, TNF-α, iNOS, and COX2 genes. Microscopic pathological evaluation was performed on colon tissue. Peiminine treatment resulted in reduced levels of NO, MPO, IL-1ß, IL-6, and TNF-α. Furthermore, the expression of IL-1ß, IL-6, TNF-α genes, iNOS, and COX2 genes was decreased in response to peiminine treatment in these mice. This study demonstrates the effectiveness of peiminine in alleviating inflammatory manifestations and mitigating intestinal tissue damage in an experimental model of ulcerative colitis, probably by anti-inflammatory procedure. Peiminine holds potential as a therapeutic adjunct for the management of ulcerative colitis.


Subject(s)
Acetic Acid , Cevanes , Colitis, Ulcerative , Animals , Mice , Colitis, Ulcerative/chemically induced , Colitis, Ulcerative/drug therapy , Cyclooxygenase 2 , Interleukin-6 , Tumor Necrosis Factor-alpha , Anti-Inflammatory Agents/pharmacology , Interleukin-1beta , Nitric Oxide
8.
Mol Divers ; 2023 Sep 20.
Article in English | MEDLINE | ID: mdl-37728805

ABSTRACT

ß-secretase 1 (BACE1) is an enzyme that is involved in generating beta-amyloid peptides and is believed to have a significant role in the development of Alzheimer's disease (AD). Therefore, BACE1 has gained attention as a potential therapeutic target for treating AD. Modern drug discovery studies are being conducted to identify potential inhibitors of BACE1, with the goal of reducing the production of beta-amyloid peptides and, thus, slowing the progression of AD. Here, we used a multistep virtual screening methodology to identify phytoconstituents from the IMPPAT library that could inhibit the activity of BACE1. Molecular docking was employed to select initial hits based on their binding affinity toward BACE1. Screening for PAINS patterns, ADMET and PASS properties, was then used to identify potential molecules for BACE1 inhibition. In the end, we discovered two natural compounds, Peiminine and 27-Deoxywithaferin A, which demonstrated a strong affinity, effectiveness, and specific interactions for the BACE1-active site. The elucidated molecules also displayed drug likeliness. A 200 ns molecular dynamics (MD) simulation was conducted to investigate the interaction mechanism, complex stability, and conformational dynamics of BACE1 with Peiminine and 27-Deoxywithaferin A. The MD simulations demonstrated that BACE1 was stable during the simulation with Peiminine and 27-Deoxywithaferin A. Overall, the results suggested that Peiminine and 27-Deoxywithaferin A hold significant potential as scaffolds in drug development efforts targeting BACE1 for the purpose of treating AD.

9.
Cells ; 12(6)2023 03 14.
Article in English | MEDLINE | ID: mdl-36980235

ABSTRACT

Glioblastoma (GBM) is a primary brain tumor of unknown etiology. It is extremely aggressive, incurable and has a short average survival time for patients. Therefore, understanding the precise molecular mechanisms of this diseases is essential to establish effective treatments. In this study, we cloned and sequenced a splice variant of the hydroxysteroid 11-ß dehydrogenase 1 like gene (HSD11B1L) and named it HSD11B1L-181. HSD11 B1L-181 was specifically expressed only in GBM cells. Overexpression of this variant can significantly promote the proliferation, migration and invasion of GBM cells. Knockdown of HSD11B1L-181 expression inhibited the oncogenic potential of GBM cells. Furthermore, we identified the direct interaction of parkin with HSD11B1L-181 by screening the GBM cDNA expression library via yeast two-hybrid. Parkin is an RBR E3 ubiquitin ligase whose mutations are associated with tumorigenesis. Small interfering RNA treatment of parkin enhanced the proliferative, migratory and invasive abilities of GBM. Finally, we found that the alkaloid peiminine from the bulbs of Fritillaria thunbergii Miq blocks the interaction between HSD11B1L-181 and parkin, thereby lessening carcinogenesis of GBM. We further confirmed the potential of peiminine to prevent GBM in cellular, ectopic and orthotopic xenograft mouse models. Taken together, these findings not only provide insight into GBM, but also present an opportunity for future GBM treatment.


Subject(s)
11-beta-Hydroxysteroid Dehydrogenase Type 1 , Brain Neoplasms , Glioblastoma , Ubiquitin-Protein Ligases , Animals , Humans , Mice , 11-beta-Hydroxysteroid Dehydrogenase Type 1/drug effects , 11-beta-Hydroxysteroid Dehydrogenase Type 1/genetics , 11-beta-Hydroxysteroid Dehydrogenase Type 1/metabolism , Brain Neoplasms/drug therapy , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Carcinogenesis/genetics , Cevanes/pharmacology , Glioblastoma/drug therapy , Glioblastoma/genetics , Glioblastoma/metabolism , Protein Isoforms/drug effects , Protein Isoforms/genetics , Protein Isoforms/metabolism , Ubiquitin-Protein Ligases/drug effects , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
10.
Phytother Res ; 37(7): 2841-2853, 2023 Jul.
Article in English | MEDLINE | ID: mdl-36799485

ABSTRACT

Peiminine is a major biologically active component of Fritillaria thunbergii Miq that exhibits good anticancer, antiinflammatory, and anti-osteoclast effects. However, its effects on osteoporosis (OP) remain unknown. This study aimed to explore whether Peiminine was able to regulate osteogenesis and adipogenesis in ovariectomized (OVX) rat. The effects on the differentiation of bone marrow stem cells (BMSCs), function of Wnt/ß-catenin pathway, ALP activity, calcium nodule deposition, as well as adipocyte formation in vitro by Peiminine at different concentrations, were detected. The curative effects of Peiminine on the ovariectomy-induced osteoporosis model by micro-CT and bone histomorphology assays were analyzed. The promotion of osteogenic differentiation and inhibition of adipogenic differentiation by Peiminine (5-40 µg/mL) was detected and the optimum concentration was 20 µg/mL. Mechanistically, Peiminine regulated the fate of BMSCs in vitro, and activated Wnt/ß-catenin signaling pathway by restraining phosphorylation of ß-catenin and promoting the nuclear translocation of ß-catenin. Moreover, Peiminine prevented ovariectomy-induced osteoporosis by alleviating trabecular bone loss and inhibiting adipose formation. Our data suggested that Peiminine could attenuate ovariectomy-induced osteoporosis by alleviating trabecular bone loss and inhibiting adipose formation. These encouraging discoveries could lay the foundation for Peiminine to be a promising preventive treatment strategy for skeletal diseases, such as osteoporosis.


Subject(s)
Osteoporosis , Wnt Signaling Pathway , Female , Rats , Animals , Osteogenesis , beta Catenin/metabolism , Cell Differentiation , Osteoporosis/drug therapy , Cells, Cultured
11.
Front Pharmacol ; 13: 1090599, 2022.
Article in English | MEDLINE | ID: mdl-36582529

ABSTRACT

Introduction: The clinical efficacy of Yiqi Sanjie (YQSJ) formula in the treatment of stage III colorectal cancer (CRC) has been demonstrated. However, the underlying antitumor mechanisms remain poorly understood. Materials and methods: The aim of the present study was to comprehensively characterize the molecular and microbiota changes in colon tissues and fecal samples from CRC mice and in CRC cell lines treated with YQSJ or its main active component, peiminine. Integrative tandem mass tag-based proteomics and ultra-performance liquid chromatography coupled with time-of-flight tandem mass spectrometry metabolomics were used to analyze azoxymethane/dextran sulfate sodium-induced CRC mouse colon tissues. Results: The results showed that 0.8% (57/7568) of all detected tissue proteins and 3.2% (37/1141) of all detected tissue metabolites were significantly changed by YQSJ treatment, with enrichment in ten and six pathways associated with colon proteins and metabolites, respectively. The enriched pathways were related to inflammation, sphingolipid metabolism, and cholesterol metabolism. Metabolomics analysis of fecal samples from YQSJ-treated mice identified 121 altered fecal metabolites and seven enriched pathways including protein digestion and absorption pathway. 16S rRNA sequencing analysis of fecal samples indicated that YQSJ restored the CRC mouse microbiota structure by increasing the levels of beneficial bacteria such as Ruminococcus_1 and Prevotellaceae_UCG_001. In HCT-116 cells treated with peiminine, data-independent acquisition-based proteomics analysis showed that 1073 of the 7152 identified proteins were significantly altered and involved in 33 pathways including DNA damage repair, ferroptosis, and TGF-ß signaling. Conclusion: The present study identified key regulatory elements (proteins/metabolites/bacteria) and pathways involved in the antitumor mechanisms of YQSJ, suggesting new potential therapeutic targets in CRC.

12.
Korean J Physiol Pharmacol ; 26(2): 87-94, 2022 Mar 01.
Article in English | MEDLINE | ID: mdl-35203059

ABSTRACT

Myocardial infarction promotes cardiac remodeling and myocardial fibrosis, thus leading to cardiac dysfunction or heart failure. Peiminine has been regarded as a traditional anti-fibrotic Chinese medicine in pulmonary fibrosis. However, the role of peiminine in myocardial infarction-induced myocardial injury and fibrosis remained elusive. Firstly, rat model of myocardial infarction was established using ligation of the left coronary artery, which were then intraperitoneally injected with 2 or 5 mg/kg peiminine once a day for 4 weeks. Echocardiography and haemodynamic evaluation results showed that peiminine treatment reduced left ventricular end-diastolic pressure, and enhanced maximum rate of increase/decrease of left ventricle pressure (± dP/dt max) and left ventricular systolic pressure, which ameliorate the cardiac function. Secondly, myocardial infarction-induced myocardial injury and infarct size were also attenuated by peiminine. Moreover, peiminine inhibited myocardial infarction-induced increase of interleukin (IL)-1ß, IL-6 and tumor necrosis factor-α production, as well as the myocardial cell apoptosis, in the rats. Thirdly, peiminine also decreased the myocardial fibrosis related protein expression including collagen I and collagen III. Lastly, peiminine reduced the expression of p38 and phosphorylation of extracellular signal-regulated kinase 1/2 in rat model of myocardial infarction. In conclusion, peiminine has a cardioprotective effect against myocardial infarction-induced myocardial injury and fibrosis, which can be attributed to the inactivation of mitogen-activated protein kinase pathway.

13.
Front Pharmacol ; 12: 770846, 2021.
Article in English | MEDLINE | ID: mdl-34867399

ABSTRACT

Aims: Peiminine has been reported to have various pharmacological properties, including anticancer activity. In this study, we investigated the effect of this alkaloid on osteosarcoma and explored the underlying mechanisms. Methods: To evaluate the antiosteosarcoma effects of peiminine in vitro, cell viability was assessed by CCK-8 and live/dead assays; the effects of the drug on apoptosis and the cell cycle were examined by flow cytometry; the effects on cell migration and invasion were detected by wound healing and Transwell assays, respectively, while its effects on autophagy were observed by transmission electron microscopy and an LC3 fluorescent puncta formation assay. The role of autophagy in the peiminine-mediated effects in osteosarcoma cells was evaluated by CCK-8 assay and western blotting after the application of the autophagy inhibitor chloroquine. The effect of peiminine on reactive oxygen species (ROS) production was analyzed using fluorescence confocal microscopy and spectrophotometry. Additionally, peiminine-treated osteosarcoma cells were exposed to SP600125, a JNK inhibitor, and N-acetylcysteine, a ROS scavenger, after which the contribution of the ROS/JNK signaling pathway to osteosarcoma was assessed using cell viability and LC3 fluorescent puncta formation assays, flow cytometry, and western blotting. A xenograft mouse model of osteosarcoma was generated to determine the antitumor effects of peiminine in vivo. Results: Peiminine suppressed proliferation and metastasis and induced cell cycle arrest, apoptosis, and autophagy in osteosarcoma cells. These anticancer effects of peiminine were found to be dependent on intracellular ROS generation and activation of the JNK pathway. In line with these results, peiminine significantly inhibited xenograft tumor growth in vivo. Conclusions: Peiminine induced G0/G1-phase arrest, apoptosis, and autophagy in human osteosarcoma cells via the ROS/JNK signaling pathway both in vitro and in vivo. Our study may provide an experimental basis for the evaluation of peiminine as an alternative drug for the treatment of osteosarcoma.

14.
Front Endocrinol (Lausanne) ; 12: 736863, 2021.
Article in English | MEDLINE | ID: mdl-34630331

ABSTRACT

Osteoclasts (OCs) play an important role in osteoporosis, a disease that is mainly characterized by bone loss. In our research, we aimed to identify novel approach for regulating osteoclastogenesis and thereby treating osteoporosis. Previous studies have set a precedent for screening traditional Chinese herbal extracts for effective inhibitors. Peiminine is an alkaloid extracted from the bulb of Fritillaria thunbergii Miq that reportedly has anticancer and anti-inflammatory effects. Thus, the potential inhibitory effect of peiminine on OC differentiation was investigated via a series of experiments. According to the results, peiminine downregulated the levels of specific genes and proteins in vitro and consequently suppressed OC differentiation and function. Based on these findings, we further investigated the underlying molecular mechanisms and identified the NF-κB and ERK1/2 signaling pathways as potential targets of peiminine. In vivo, peiminine alleviated bone loss in an ovariectomized mouse model.


Subject(s)
Cevanes/pharmacology , Osteoclasts/drug effects , Osteogenesis/drug effects , RANK Ligand/pharmacology , Signal Transduction/drug effects , Animals , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Femur/drug effects , Femur/metabolism , Mice , NF-kappa B/metabolism , NFATC Transcription Factors/metabolism , Osteoclasts/metabolism , Ovariectomy
15.
Int J Mol Sci ; 22(19)2021 Sep 23.
Article in English | MEDLINE | ID: mdl-34638579

ABSTRACT

Parkinson's disease (PD) is a degenerative disease that can cause motor, cognitive, and behavioral disorders. The treatment strategies being developed are based on the typical pathologic features of PD, including the death of dopaminergic (DA) neurons in the substantia nigra of the midbrain and the accumulation of α-synuclein in neurons. Peiminine (PMN) is an extract of Fritillaria thunbergii Miq that has antioxidant and anti-neuroinflammatory effects. We used Caenorhabditis elegans and SH-SY5Y cell models of PD to evaluate the neuroprotective potential of PMN and address its corresponding mechanism of action. We found that pretreatment with PMN reduced reactive oxygen species production and DA neuron degeneration caused by exposure to 6-hydroxydopamine (6-OHDA), and therefore significantly improved the DA-mediated food-sensing behavior of 6-OHDA-exposed worms and prolonged their lifespan. PMN also diminished the accumulation of α-synuclein in transgenic worms and transfected cells. In our study of the mechanism of action, we found that PMN lessened ARTS-mediated degradation of X-linked inhibitor of apoptosis (XIAP) by enhancing the expression of PINK1/parkin. This led to reduced 6-OHDA-induced apoptosis, enhanced activity of the ubiquitin-proteasome system, and increased autophagy, which diminished the accumulation of α-synuclein. The use of small interfering RNA to down-regulate parkin reversed the benefits of PMN in the PD models. Our findings suggest PMN as a candidate compound worthy of further evaluation for the treatment of PD.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Caenorhabditis elegans Proteins/metabolism , Cevanes/pharmacology , Parkinson Disease/metabolism , Protein Serine-Threonine Kinases/metabolism , Ubiquitin-Protein Ligases/metabolism , X-Linked Inhibitor of Apoptosis Protein/metabolism , alpha-Synuclein/metabolism , Animals , Animals, Genetically Modified , Caenorhabditis elegans/metabolism , Dopamine/metabolism , Dopaminergic Neurons/metabolism , Nerve Degeneration/metabolism , Proteasome Endopeptidase Complex/metabolism , Substantia Nigra/metabolism , Ubiquitin/metabolism
16.
J Ethnopharmacol ; 257: 112796, 2020 Jul 15.
Article in English | MEDLINE | ID: mdl-32344236

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: The effective-component compatibility of Bufei Yishen formula I (ECC-BYF I), a combination of 10 compounds, including total ginsenosides, astragaloside IV, icariin, and paeonol, etc., is derived from Bufei Yishen formula (BYF). The efficacy and safety of ECC-BYF I is equal to BYF. However, the composition of ECC-BYF I needs to be further optimized. Based on the beneficial effects of BYF and ECC-BYF I on chronic obstructive pulmonary disease (COPD), this study aimed to optimize the composition of ECC-BYF I and to explore the effects and mechanisms of optimized ECC-BYF I (ECC-BYF II) on mucus hypersecretion in COPD rats. MATERIALS AND METHODS: ECC-BYF I was initially optimized to six groups: optimized ECC-BYF I (OECC-BYF I)-A~F. Based on a COPD rat model, the effects of OECC-BYF I-A~F on COPD rats were evaluated. R-value comprehensive evaluation was used to evaluate the optimal formula, which was named ECC-BYF II. The changes in goblet cells and expression of mucins and the mRNA and proteins involved in the epidermal growth factor receptor/phosphoinositide-3-kinase/mammalian target of rapamycin (EGFR/PI3K/mTOR) pathway were evaluated to explore the effects and mechanisms of ECC-BYF II on mucus hypersecretion. RESULTS: ECC-BYF I and its six optimized groups, OECC-BYF I-A~F, had beneficial effects on COPD rats in improving pulmonary function and lung tissue pathology, reducing inflammation and oxidative stress, and improving the protease/anti-protease imbalance and collagen deposition. R-value comprehensive evaluation found that OECC-BYF I-E (paeonol, icariin, nobiletin, total ginsenoside, astragaloside IV) was the optimal formula for improving the comprehensive effects (lung function: VT, MV, PEF, EF50, FVC, FEV 0.1, FEV 0.1/FVC; histological changes: MLI, MAN; IL-1ß, IL-6, TNF-α, MMP-9, TIMP-1, T-AOC, LPO, MUC5AC, Collagen I and Collagen III). OECC-BYF I-E was named ECC-BYF II. Importantly, the effect of ECC-BYF II showed no significant difference from BYF and ECC-BYF I. ECC-BYF II inhibited mucus hypersecretion in COPD rats, which manifested as reducing the expression of MUC5AC and MUC5B and the hyperplasia rate of goblet cells. The mRNA and protein expression levels of EGFR, PI3K, Akt, and mTOR were increased in COPD rats and were obviously downregulated after ECC-BYF II administration. CONCLUSION: ECC-BYF II, which consists of paeonol, icariin, nobiletin, total ginsenoside and astragaloside IV, has beneficial effects equivalent to BYF and ECC-BYF I on COPD rats. ECC-BYF II significantly inhibited mucus hypersecretion, which may be related to the regulation of the EGFR/PI3K/mTOR pathway.


Subject(s)
Drugs, Chinese Herbal/pharmacology , ErbB Receptors/metabolism , Mucus/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Pulmonary Disease, Chronic Obstructive/metabolism , TOR Serine-Threonine Kinases/metabolism , Animals , Bronchi/pathology , Cytokines/metabolism , Inflammation/metabolism , Lung/drug effects , Lung/metabolism , Lung/pathology , Rats, Sprague-Dawley , Signal Transduction/drug effects
17.
Inflammation ; 43(3): 1110-1119, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32152924

ABSTRACT

Acute lung injury (ALI) is a kind of lung serious disease which leads to the damage of alveolar epithelial cells and capillary endothelial. Lipopolysaccharide (LPS) is one of the common factors inducing ALI. The previous study has reported that the anti-inflammatory effect of peiminine, but little is known about its effect on the ALI induced by LPS. The aim of this study is to investigate the therapeutic effect of peiminine on LPS-induced acute lung injury and potential mechanisms. Mice were given LPS through nasal cavity to establish ALI model, and then the peiminine (1, 3, or 5 mg/kg) was injected into the mice as the experimental group. In the present study, we would measure the W/D ratio, activity of MPO, the histopathological changes, and the levels of cytokines. The results showed that peiminine could reduce the W/D ratio and the MPO activity significantly. Furthermore, the histopathological changes and the expression of TNF-α, IL-1ß, and IL-6 were inhibited after the peiminine treatment. In vitro, peiminine significantly inhibited LPS-induced IL-8 production in A549 lung epithelial cells. Meanwhile, the activity of NF-κB signaling pathway was suppressed obviously by peiminine with the western blot analysis. Also, peiminine significantly attenuated LPS-induced AKT and PI3K phosphorylation. In addition, peiminine was found to disrupt lipid rafts formation by attenuating the cholesterol content. In conclusion, peiminine could attenuate LPS-induced ALI in mice and it may become a new approach to treat ALI.


Subject(s)
Acute Lung Injury/drug therapy , Acute Lung Injury/metabolism , Cevanes/therapeutic use , Lipopolysaccharides/toxicity , Membrane Microdomains/drug effects , Membrane Microdomains/metabolism , A549 Cells , Acute Lung Injury/chemically induced , Alkaloids/pharmacology , Alkaloids/therapeutic use , Animals , Cell Survival/drug effects , Cell Survival/physiology , Cevanes/pharmacology , Dose-Response Relationship, Drug , Humans , Male , Mice , Mice, Inbred BALB C
18.
Front Pharmacol ; 10: 737, 2019.
Article in English | MEDLINE | ID: mdl-31333459

ABSTRACT

Chronic obstructive pulmonary disease (COPD) has been a major public health problem and is still a formidable challenge for clinicians. It is urgent to find new compounds for minimizing the risk of disease progression and exacerbation especially in the early phase of COPD. A traditional Chinese medicine (TCM) formula, Chuan Bei Pi Pa dropping pills (CBPP), was tested in this study to investigate its potential mechanisms in preventing the exacerbation of COPD. Phosphoproteomics analysis for a smog stimulated early stage COPD mice model was employed to detect the underlying molecular mechanisms of CBPP. In addition, protein-protein interaction (PPI) and bioinformatics analyses were included to analyze the key proteins and predict the key bioactive compounds. The results indicated that peiminine (PEI) target epidermal growth factor receptor (EGFR) prevented the exacerbation of COPD by inhibiting the EGFR signaling pathway, and ursolic acid (UA) can alleviate inflammation disorders via inhibition of CASP3 on mitogen-activated protein kinase (MAPK) signaling pathway. After in vivo and in vitro evaluations, we revealed that PEI from CBPP, as a lead compound, can improve lung function and alleviate pulmonary fibrosis by acting on the EGFR and MLC2 signaling pathways. Furthermore, the approach described here is an effective way to analyze and identify the bioactive ingredients from a mixture by functional proteomics analysis.

19.
J Chromatogr A ; 1593: 156-163, 2019 May 24.
Article in English | MEDLINE | ID: mdl-30718059

ABSTRACT

Nonaqueous capillary electrophoresis (CE) and organic-aqueous CE using high content of organic solvents are common techniques in capillary electrophoresis-mass spectrometry (CE-MS) due to various advantages. However, the long-term stability of commonly used polyimide coated fused silica capillary remains a major problem that polyimide coating is prone to swelling in organic solvents and aminolysis in alkaline buffer, which cause serious problems like irregular electrospray, current drop or clogging event. In this work, robust PEEK capillary was used as separation capillary for tolerance to high content of organic solvents in CE-MS system. It is interesting that the high mechanical and chemical stability of PEEK capillary ensured stable CE separation and electrospray when used in CE-MS. All quantitative experiments were accomplished by single PEEK capillary. In addition, the using of organic solvents as running buffer reduced adsorption of hydrophobic compounds on PEEK capillary wall and improved the symmetry of peak shape. The PEEK-based CE-MS method showed good separation performance in analysis of several groups of active alkaloids in traditional Chinese medicines and was applied for quantitative analysis of peimine and peiminine in Bulbus Fritillariae thunbergii herb with high sensitivity. Based on the advantages like simple pretreatment, easy to cut, stable and good separation performance of PEEK column in CE-MS, this study provides an effective way for improving the stability and employment of high content of organic solvents in CE-MS analysis.


Subject(s)
Electrophoresis, Capillary/methods , Ketones/chemistry , Mass Spectrometry/methods , Polyethylene Glycols/chemistry , Alkaloids/analysis , Benzophenones , Drugs, Chinese Herbal/chemistry , Polymers , Sensitivity and Specificity , Solvents/chemistry
20.
Article in Chinese | WPRIM (Western Pacific) | ID: wpr-851268

ABSTRACT

Objective: To detect the influence of peiminine on the invasion and migration of human lung cancer A549 cells. Methods A549 cells were treated with peiminine at the final concentrations of 0, 50, 100, and 200 μmol/L, respectively. The influence of peiminine on the invasion and migration of A549 cells and its underlying mechanisms were investigated by cell invasion experiment, cell scratch experiment, real-time quantitative PCR, ELISA, and Western blotting. Results Compared with 0 μmol/L peiminine group, the cell transmembrane number and scratch wound healing rate and expressions of MMP-9 and MMP-2 in the group treated with different concentrations of peiminine significantly decreased (P < 0.01). Compared with 0 μmol/L peiminine group, the mRNA expression of E-cadherin increased significantly (P < 0.01), while the mRNA expressions of N-cadherin and vimentin decreased significantly (P < 0.01). Compared with 0 μmol/L peiminine group, FN protein expression was significantly decreased in all the groups with different concentrations of peiminine group except treatment with 50 μmol/L peiminine after 24 h (P < 0.05, 0.01). Compared with 0 μmol/L peiminine group, the ratio of p-PI3K/PI3K and p-mTOR/mTOR in all concentrations of peiminine groups and p-Akt/Akt in 100 and 200 peiminine groups were significantly decreased (P < 0.05, 0.01). Conclusion Peiminine can inhibit the invasion and migration of A549 cells, which may be related to the activation of PI3K/Akt/mTOR pathway and decreasing the epithelial- mesenchymal transition process.

SELECTION OF CITATIONS
SEARCH DETAIL
...