Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 483
Filter
1.
Drug Des Devel Ther ; 18: 4241-4256, 2024.
Article in English | MEDLINE | ID: mdl-39323973

ABSTRACT

Objective: This study investigates the targets, pathways, and mechanisms of Schisandrin B (Sch B) in alleviating renal ischemia-reperfusion injury (RIRI) using RNA sequencing and network pharmacology. Methods: The effects of Sch B on RIRI were assessed using hematoxylin-eosin (HE) and periodic acid-Schiff (PAS) staining, along with measurements of blood creatinine and urea nitrogen (BUN). Differential gene expression in mouse models treated with RIRI and Sch B+RIRI was analyzed through RNA-Seq. Key processes, targets, and pathways were examined using network pharmacology techniques. The antioxidant capacity of Sch B was evaluated using assays for reactive oxygen species (ROS), mitochondrial superoxide, and JC-1 membrane potential. Molecular docking was employed to verify the interactions between key targets and Sch B, and the expression of these targets and pathway was confirmed using qRT-PCR, Western blot, and immunofluorescence. Results: Sch B pre-treatment significantly reduced renal pathological damage, inflammatory response, and apoptosis in a mouse RIRI model. Pathological damage scores dropped from 4.33 ± 0.33 in the I/R group to 2.17 ± 0.17 and 1.5 ± 0.22 in Sch B-treated groups (p < 0.01). Creatinine and BUN levels were also reduced (from 144.6 ± 21.05 µmol/L and 53.51 ± 2.34 mg/dL to 50.44 ± 5.61 µmol/L and 17.18 ± 0.96 mg/dL, p < 0.05). Transcriptomic analysis identified four key targets (AKT1, ALB, ACE, CCL5) and the PI3K/AKT pathway. Experimental validation confirmed Sch B modulated these targets, reducing apoptosis and oxidative stress, and enhancing renal recovery. Conclusion: Sch B reduces oxidative stress, inflammation, and apoptosis by modulating key targets such as AKT1, ALB, ACE, and CCL5, while activating the PI3K/AKT pathway, leading to improved renal recovery in RIRI.


Subject(s)
Cyclooctanes , Lignans , Polycyclic Compounds , Reperfusion Injury , Lignans/pharmacology , Lignans/chemistry , Animals , Cyclooctanes/pharmacology , Cyclooctanes/chemistry , Polycyclic Compounds/pharmacology , Polycyclic Compounds/chemistry , Mice , Reperfusion Injury/drug therapy , Reperfusion Injury/metabolism , Reperfusion Injury/pathology , Male , Transcriptome/drug effects , Mice, Inbred C57BL , Molecular Docking Simulation , Protective Agents/pharmacology , Protective Agents/chemistry , Disease Models, Animal , Apoptosis/drug effects , Network Pharmacology
2.
Fitoterapia ; 178: 106184, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39142529

ABSTRACT

The phytochemical investigations on the fruits of Kadsura coccinea led to the isolation of six undescribed dibenzocyclooctadiene lignans named kadcolignans B-G, together with eleven previously described analogues. The structures of these compounds were established by spectroscopic methods including NMR, HRESIMS, and CD experiments. All isolated compounds were evaluated for their hepatoprotective activity by measuring the levels of triglyceride (TG), total cholesterol (TC), and reactive oxygen species (ROS) in FFA-induced HepG2 cells. As a result, compounds 4, 5, 9, 13, and 15 showed potent inhibitory effects on hepatocyte lipid accumulation at a concentration of 100 µM. Our research not only broadens the understanding on the chemical composition of K. coccinea but also provides experimental and theoretical evidences supporting the fruit's active ingredients in alleviating nonalcoholic fatty liver disease (NAFLD).


Subject(s)
Cyclooctanes , Fruit , Kadsura , Lignans , Phytochemicals , Lignans/pharmacology , Lignans/isolation & purification , Fruit/chemistry , Humans , Kadsura/chemistry , Hep G2 Cells , Cyclooctanes/pharmacology , Cyclooctanes/isolation & purification , Molecular Structure , Phytochemicals/pharmacology , Phytochemicals/isolation & purification , Protective Agents/pharmacology , Protective Agents/isolation & purification , Reactive Oxygen Species/metabolism , Triglycerides , China , Cholesterol , Non-alcoholic Fatty Liver Disease/drug therapy
3.
Biomed Pharmacother ; 178: 117241, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39111082

ABSTRACT

Calcific aortic valve disease (CAVD) primarily involves osteogenic differentiation in human aortic valve interstitial cells (hVICs). Schisandrol B (SolB), a natural bioactive constituent, has known therapeutic effects on inflammatory and fibrotic disorders. However, its impact on valve calcification has not been reported. We investigated the effect of SolB on osteogenic differentiation of hVICs. Transcriptome sequencing was used to analyze potential molecular pathways affected by SolB treatment. The study also included an in vivo murine model using aortic valve wire injury surgery to observe SolB's effect on valve calcification. SolB inhibited the osteogenic differentiation of hVICs, reversing the increase in calcified nodule formation and osteogenic proteins. In the murine model, SolB significantly decreased the peak velocity of the aortic valve post-injury and reduced valve fibrosis and calcification. Transcriptome sequencing identified the p53 signaling pathway as a key molecular target of SolB, demonstrating its role as a molecular glue in the mouse double minute 2 (MDM2)-p53 interaction, thereby promoting p53 ubiquitination and degradation, which further inhibited p53-related inflammatory and senescence response. These results highlighted therapeutic potential of SolB for CAVD via inhibiting p53 signaling pathway and revealed a new molecular mechanism of SolB which provided a new insight of theraputic mechanism for CAVD.


Subject(s)
Aortic Valve Stenosis , Aortic Valve , Calcinosis , Cyclooctanes , Lignans , Tumor Suppressor Protein p53 , Animals , Humans , Male , Mice , Aortic Valve/pathology , Aortic Valve/drug effects , Aortic Valve/metabolism , Aortic Valve Stenosis/drug therapy , Aortic Valve Stenosis/pathology , Calcinosis/drug therapy , Calcinosis/pathology , Calcinosis/metabolism , Cell Differentiation/drug effects , Cells, Cultured , Cellular Senescence/drug effects , Cyclooctanes/pharmacology , Disease Models, Animal , Inflammation/drug therapy , Inflammation/pathology , Inflammation/metabolism , Lignans/pharmacology , Mice, Inbred C57BL , Osteogenesis/drug effects , Proto-Oncogene Proteins c-mdm2/metabolism , Signal Transduction/drug effects , Tumor Suppressor Protein p53/metabolism
4.
J Nanobiotechnology ; 22(1): 498, 2024 Aug 20.
Article in English | MEDLINE | ID: mdl-39164657

ABSTRACT

Microcarrier is a promising drug delivery system demonstrating significant value in treating cancers. One of the main goals is to devise microcarriers with ingenious structures and functions to achieve better therapeutic efficacy in tumors. Here, inspired by the nucleus-cytoplasm structure of cells and the material exchange reaction between them, we develop a type of biorthogonal compartmental microparticles (BCMs) from microfluidics that can separately load and sequentially release cyclooctene-modified doxorubicin prodrug (TCO-DOX) and tetrazine-modified indocyanine green (Tz-ICG) for tumor therapy. The Tz-ICG works not only as an activator for TCO-DOX but also as a photothermal agent, allowing for the combination of bioorthogonal chemotherapy and photothermal therapy (PTT). Besides, the modification of DOX with cyclooctene significantly decreases the systemic toxicity of DOX. As a result, the developed BCMs demonstrate efficient in vitro tumor cell eradication and exhibit notable tumor growth inhibition with favorable safety. These findings illustrate that the formulated BCMs establish a platform for bioorthogonal prodrug activation and localized delivery, holding significant potential for cancer therapy and related applications.


Subject(s)
Doxorubicin , Drug Delivery Systems , Indocyanine Green , Photothermal Therapy , Prodrugs , Doxorubicin/pharmacology , Doxorubicin/chemistry , Photothermal Therapy/methods , Humans , Prodrugs/pharmacology , Prodrugs/chemistry , Animals , Indocyanine Green/chemistry , Indocyanine Green/pharmacology , Mice , Drug Delivery Systems/methods , Cell Line, Tumor , Neoplasms/therapy , Neoplasms/drug therapy , Cyclooctanes/chemistry , Cyclooctanes/pharmacology , Mice, Inbred BALB C , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Female
5.
Sci Rep ; 14(1): 18418, 2024 08 08.
Article in English | MEDLINE | ID: mdl-39117695

ABSTRACT

Bacterial testicular inflammation is one of the important causes of male infertility. Using plant-derived compounds to overcome the side effects of antibiotics is an alternative treatment strategy for many diseases. Schizandrin B (SchB) is a bioactive compound of herbal medicine Schisandra chinensis which has multiple pharmacological effects. However its effect and the mechanism against testicular inflammation are unknown. Here we tackled these questions using models of lipopolysaccharide (LPS)-induced mice and -Sertoli cells (SCs). Histologically, SchB ameliorated the LPS-induced damages of the seminiferous epithelium and blood-testicular barrier, and reduced the production of pro-inflammatory mediators in mouse testes. Furthermore, SchB decreased the levels of pro-inflammatory mediators and inhibited the nuclear factor kB (NF-κB) and MAPK (especially JNK) signaling pathway phosphorylation in LPS-induced mSCs. The bioinformatics analysis based on receptor prediction and the molecular docking was further conducted. We targeted androgen receptor (AR) and illustrated that AR might bind with SchB in its function. Further experiments indicate that the AR expression was upregulated by LPS stimulation, while SchB treatment reversed this phenomenon; similarly, the expression of the JNK-related proteins and apoptotic-related protein were also reversed after AR activator treatment. Together, SchB mitigates LPS-induced inflammation and apoptosis by inhibiting the AR-JNK pathway.


Subject(s)
Apoptosis , Cyclooctanes , Lignans , Lipopolysaccharides , Polycyclic Compounds , Sertoli Cells , Animals , Male , Cyclooctanes/pharmacology , Polycyclic Compounds/pharmacology , Polycyclic Compounds/therapeutic use , Lignans/pharmacology , Lignans/therapeutic use , Apoptosis/drug effects , Mice , Sertoli Cells/drug effects , Sertoli Cells/metabolism , Receptors, Androgen/metabolism , MAP Kinase Signaling System/drug effects , Inflammation/drug therapy , Inflammation/metabolism , Inflammation/pathology , Molecular Docking Simulation , Testis/drug effects , Testis/metabolism , Testis/pathology , NF-kappa B/metabolism
6.
Eur J Pharmacol ; 982: 176938, 2024 Nov 05.
Article in English | MEDLINE | ID: mdl-39181224

ABSTRACT

There has been significant global interest in respiratory health driven by the coronavirus disease (COVID-19) and severe environmental pollution. This study explored the potential of schisantherin A (SchA), a compound derived from Schisandra chinensis, to protect against acute pneumoconiosis. We assessed the effects of SchA on phorbol 12-myristate 13-acetate (PMA)-stimulated A549 alveolar epithelial cells and SiO2/TiO2-induced pulmonary injury in mice. In A549 cells, SchA significantly decreased pro-inflammatory mediators such as inducible nitric oxide synthase (iNOS), cyclooxygenase-2 (COX-2), and interleukin (IL)-8 levels. SchA-mediated reduction in inflammatory mediators was associated with the downregulation of PMA-stimulated nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) and mitogen-activated protein kinase (MAPK) signaling activation. In SiO2/TiO2-induced lung-injured mice, SchA administration significantly reduced MUC5AC production in lung tissue. SchA administration significantly downregulated the overexpression of NK-κB and the subsequent production of COX-2, iNOS, and NOD-like receptor pyrin domain-containing protein 3 (NLRP3) inflammasomes. It significantly suppressed expected increases in total cell numbers and pro-inflammatory cytokines, including tumor necrosis factor-alpha (TNF-α) and IL-1ß in the bronchoalveolar lavage fluid (BALF) in SiO2/TiO2-stimulated mice. In contrast, the SiO2/TiO2-mediated decrease in IL-10 levels was significantly improved by SchA treatment. These fundamental results can be used to develop potential treatments involving SchA for acute pneumoconiosis.


Subject(s)
Acute Lung Injury , Cyclooctanes , Nanoparticles , Silicon Dioxide , Titanium , Animals , Silicon Dioxide/toxicity , Titanium/toxicity , Humans , Cyclooctanes/pharmacology , Cyclooctanes/therapeutic use , Mice , Acute Lung Injury/chemically induced , Acute Lung Injury/drug therapy , Acute Lung Injury/pathology , Acute Lung Injury/metabolism , A549 Cells , Male , Nanoparticles/chemistry , Lignans/pharmacology , Lignans/therapeutic use , Mucin 5AC/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , NF-kappa B/metabolism , Nitric Oxide Synthase Type II/metabolism , Silicosis/pathology , Silicosis/drug therapy , Silicosis/metabolism , Cyclooxygenase 2/metabolism
7.
Int Immunopharmacol ; 139: 112660, 2024 Sep 30.
Article in English | MEDLINE | ID: mdl-39018688

ABSTRACT

Cardiac tissue remodeling is characterized by altered heart tissue architecture and dysfunction, leading to heart failure. Sustained activation of the renin-angiotensin-aldosterone system (RAAS) greatly promotes the development of myocardial remodeling. Angiotensin II (Ang II), which is the major component of RAAS, can directly lead to cardiac remodeling by inducing an inflammatory response. Schisandrin B (Sch B), the active component extracted from the fruit of Schisandra chinensis (Turcz.) Baill has been shown to exhibit anti-inflammatory activity through its ability to target TLR4 and its adaptor protein, MyD88. In this study, we explored whether Sch B alleviates Ang II-induced myocardial inflammation and remodeling via targeting MyD88. Sch B significantly suppressed Ang II-induced inflammation as well as increased the expression of several genes of tissue remodeling (ß-Mhc, Tgfb, Anp, α-Ska) both in vivo and in vitro. These protective effects of Sch B were due to the inhibition of recruitment of MyD88 to TLR2 and TLR4, suppressing the Ang II-induced NF-κB activation and reducing the following inflammatory responses. Moreover, the knockdown of Myd88 in cardiomyocytes abrogated the Ang II-induced increases in the production of inflammatory cytokines and expression of remodeling genes. These findings provide new evidence that the mechanism of Sch B protection was attributed to selective inhibition of MyD88 signaling. This finding could pave the way for novel therapeutic strategies for myocardial inflammatory diseases.


Subject(s)
Angiotensin II , Cyclooctanes , Lignans , Mice, Inbred C57BL , Myeloid Differentiation Factor 88 , Myocytes, Cardiac , Polycyclic Compounds , Toll-Like Receptor 4 , Animals , Cyclooctanes/pharmacology , Cyclooctanes/therapeutic use , Lignans/pharmacology , Lignans/therapeutic use , Myeloid Differentiation Factor 88/metabolism , Polycyclic Compounds/pharmacology , Polycyclic Compounds/therapeutic use , Angiotensin II/metabolism , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Toll-Like Receptor 4/metabolism , Toll-Like Receptor 4/genetics , Mice , Male , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 2/genetics , Ventricular Remodeling/drug effects , Signal Transduction/drug effects , Cells, Cultured , NF-kappa B/metabolism
8.
Tissue Cell ; 89: 102440, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39002288

ABSTRACT

Abnormal proliferation, migration, and foam cell formation of Vascular smooth muscle cells (VSMCs) each play a role in the development of atherosclerosis (AS). Schisandrin (Sch) is the active lignan ingredient with broad-spectrum pharmacological effects. However, the role of Sch in the AS process is not clear. Therefore, this study was proposed to explore the therapeutic effect and potential mechanism of Sch on VSMCs. Ox-LDL was selected to create an atherosclerosis injury environment for VSMCs and macrophages. The MTT assay, Oil red O staining, wound healing, transwell experiments and ELISA were used to investigate the phenotype effects of Sch. Network pharmacology, molecular docking, flow cytometry, and western blot were used to investigate the underlying mechanisms of Sch on AS progression. Our findings implied that Sch treatment inhibited the proliferation and migration of VSMCs, and suppressed the ROS production and inflammatory cytokines up-regulation of VSMCs and macrophages. Moreover, Sch reduced lipid uptake and foam cell formation through downregulating LOX-1. Mechanistically, we found that Sch can inhibit the activation of JAK2/STAT3 signaling by targeting JAK2, and arrest cell cycle in GO/G1 phase. In summary, Sch can inhibit VSMCs proliferation and migration by arresting cell cycle and targeting JAK2 to regulating the JAK2/STAT3 pathway. Sch may serve as a potential drug for patients with AS.


Subject(s)
Cell Movement , Cell Proliferation , Cyclooctanes , Janus Kinase 2 , Lignans , Muscle, Smooth, Vascular , Polycyclic Compounds , STAT3 Transcription Factor , Signal Transduction , Janus Kinase 2/metabolism , STAT3 Transcription Factor/metabolism , Cell Movement/drug effects , Cell Proliferation/drug effects , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/cytology , Lignans/pharmacology , Signal Transduction/drug effects , Cyclooctanes/pharmacology , Polycyclic Compounds/pharmacology , Humans , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Cell Cycle Checkpoints/drug effects , Animals , Atherosclerosis/pathology , Atherosclerosis/metabolism , Atherosclerosis/drug therapy
9.
J Control Release ; 373: 749-765, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39084465

ABSTRACT

Convincing evidence suggests that aberrant gut microbiota changes play a critical role in the progression and pathogenesis of inflammatory bowel disease (IBD). Probiotic therapeutic interventions targeting the microbiota may provide alternative avenues to treat IBD, but currently available probiotics often suffer from low intestinal colonization and limited targeting capability. Here, we developed azido (N3)-modified Prussian blue nanozyme (PB@N3) spatio-temporal guidance enhances the targeted colonization of probiotics to alleviate intestinal inflammation. First, clickable PB@N3 targets intestinal inflammation, simultaneously, it scavenges reactive oxygen species (ROS). Subsequently, utilizing "click" chemistry to spatio-temporally guide targeted colonization of dibenzocyclooctyne (DBCO)-modified Lactobacillus reuteri DSM 17938 (LR@DBCO). The "click" reaction between PB@N3 and LR@DBCO has excellent specificity and efficacy both in vivo and in vitro. Despite the complex physiological environment of IBD, "click" reaction can prolong the retention time of probiotics in the intestine. Dextran sulfate sodium (DSS)-induced colitis mice model, demonstrates that the combination of PB@N3 and LR@DBCO effectively mitigates levels of ROS, enhances the colonization of probiotics, modulates intestinal flora composition and function, regulates immune profiles, restores intestinal barrier function, and alleviates intestinal inflammation. Hence, PB@N3 spatio-temporal guidance enhances targeted colonization of LR@DBCO provides a promising medical treatment strategy for IBD.


Subject(s)
Dextran Sulfate , Inflammatory Bowel Diseases , Limosilactobacillus reuteri , Mice, Inbred C57BL , Probiotics , Animals , Probiotics/administration & dosage , Probiotics/pharmacology , Probiotics/therapeutic use , Inflammatory Bowel Diseases/therapy , Inflammatory Bowel Diseases/drug therapy , Male , Gastrointestinal Microbiome/drug effects , Colitis/chemically induced , Colitis/drug therapy , Colitis/microbiology , Colitis/therapy , Reactive Oxygen Species/metabolism , Mice , Humans , Cyclooctanes/administration & dosage , Cyclooctanes/chemistry , Cyclooctanes/pharmacology , Cyclooctanes/therapeutic use
10.
Int Immunopharmacol ; 137: 112430, 2024 Aug 20.
Article in English | MEDLINE | ID: mdl-38852519

ABSTRACT

Mastitis, one of the most significant problems in women, is commonly caused by pathogens, especially Staphylococcus aureus (S.aureus). Schisandrin B (SCB), the main abundant derivatives from Schisandra chinensis, has been proven to have the ability to inhibiting inflammation and bacteria. However, few relevant researches systematically illustrate the role SCB in the treatment of mastitis. The aim of the present study is to demonstrate the mechanism that SCB functions in reducing pathological injury to the mammary gland in treating S.aureus-induced mastitis. H&E staining was used to identify pathological changes and injuries in mastitis. The levels of cytokines associated with inflammation were detected by ELISA. Key signals relevant to ferroptosis and Nrf2 signaling pathway were tested by western blot analysis and iron assay kit. Compared with the control group, inflammation-associated factors, such as IL-1ß, TNF-α, MPO activity, increased significantly in S. aureus-treated mice. However, these changes were inhibited by SCB. Ferroptosis-associated factors Fe2+ and MDA increased significantly, and GSH, GPX4 and ferritin expression decreased markedly in S. aureus-treated mice. SCB treatment could attenuate S.aureus-induced ferroptosis. Furthermore, SCB increase SIRT1 and SLC7A11 expression and down-regulated p53 expression and NF-κB activation. In conclusion, SCB alleviates S.aureus-induced mastitis via up-regulating SIRT1/p53/SLC7A11 signaling pathway, attenuating the activation of inflammation-associated cytokines and ferroptosis in the mammary gland tissues.


Subject(s)
Cyclooctanes , Ferroptosis , Lignans , Mastitis , Polycyclic Compounds , Signal Transduction , Sirtuin 1 , Staphylococcal Infections , Staphylococcus aureus , Tumor Suppressor Protein p53 , Animals , Lignans/pharmacology , Lignans/therapeutic use , Cyclooctanes/pharmacology , Cyclooctanes/therapeutic use , Ferroptosis/drug effects , Mastitis/drug therapy , Mastitis/chemically induced , Mastitis/immunology , Mastitis/metabolism , Polycyclic Compounds/pharmacology , Polycyclic Compounds/therapeutic use , Female , Sirtuin 1/metabolism , Signal Transduction/drug effects , Mice , Staphylococcus aureus/drug effects , Tumor Suppressor Protein p53/metabolism , Staphylococcal Infections/drug therapy , Staphylococcal Infections/immunology , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Mammary Glands, Animal/drug effects , Mammary Glands, Animal/pathology , Mammary Glands, Animal/immunology , Cytokines/metabolism , Inflammation/drug therapy , Humans
11.
Phytomedicine ; 132: 155811, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38924927

ABSTRACT

Oxidative stress is one of the earlier events causing neuronal dysfunction in Alzheimer's disease (AD). Gomisin N (GN), a lignin isolated from Schisandra chinensis, has anti-oxidative stress effects. There are currently no studies on the neuroprotective potential of GN in AD. In this study, two AD models were treated with GN for 8 weeks. The cognitive functions, amyloid deposition, and neuronal death were assessed. Additionally, the expressions of critical proteins in the GSK3ß/Nrf2 signaling pathway were determined in vivo and in vitro. We showed that GN significantly upregulated the expressions of Nrf2, p-GSK3ßSer9/GSK3ß, NQO1 and HO-1 proteins in SHSY-5Y/APPswe cells after H2O2 injury, whereas the PI3K inhibitor LY294002 reversed the increase in the expressions of Nrf2, p-GSK3ßSer9/GSK3ß, NQO1 and HO-1 proteins induced by GN administration. In a further study, GN could significantly improve the learning and memory dysfunctions of the rat and mouse AD models, reduce the area of Aß plaques in the hippocampus and cortex, and increase the number and function of neurons. Here, we first demonstrate the neuroprotective effects of GN on AD in vivo and in vitro. A possible mechanism by which GN prevents AD is proposed: GN significantly increased the expressions of Nrf2, p-GSK3Ser9/GSK3ß and NQO1 proteins in the brain of AD animal models and promoted Nrf2 nuclear translocation, then activated Nrf2 downstream genes to combat oxidative stress in AD pathogenesis. GN might be a promising therapeutic agent for AD.


Subject(s)
Alzheimer Disease , Cognitive Dysfunction , Glycogen Synthase Kinase 3 beta , Lignans , NF-E2-Related Factor 2 , Neuroprotective Agents , Oxidative Stress , Signal Transduction , NF-E2-Related Factor 2/metabolism , Alzheimer Disease/drug therapy , Animals , Glycogen Synthase Kinase 3 beta/metabolism , Signal Transduction/drug effects , Lignans/pharmacology , Male , Oxidative Stress/drug effects , Cognitive Dysfunction/drug therapy , Neuroprotective Agents/pharmacology , Humans , Mice , Rats, Sprague-Dawley , Rats , Disease Models, Animal , Schisandra/chemistry , Hippocampus/drug effects , Hippocampus/metabolism , Cyclooctanes/pharmacology , Cell Line, Tumor , Chromones/pharmacology , Neurons/drug effects , Neurons/metabolism
12.
Toxicol In Vitro ; 99: 105852, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38789064

ABSTRACT

Cisplatin is an effective chemotherapeutic agent; however, ototoxicity is one of its negative effects that greatly limits the use of cisplatin in clinical settings. Previous research has shown that the most important process cisplatin damage to inner ear cells, such as hair cells (HCs), is the excessive production and accumulation of ROS. Schisandrin B (SchB), is a low-toxicity, inexpensive, naturally occurring antioxidant with a variety of pharmacological effects. Therefore, the potential antioxidant effects of SchB may be useful for cisplatin ototoxicity treatment. In this study, the effects of SchB on cochlear hair cell viability, ROS levels, and expression of apoptosis-related molecules were evaluated by CCK-8, immunofluorescence, flow cytometry, and qRT-PCR, as well as auditory brainstem response (ABR) and dysmorphic product otoacoustic emission (DPOAE) tests to assess the effects on inner ear function. The results showed that SchB treatment increased cell survival, prevented apoptosis, and reduced cisplatin-induced ROS formation. SchB treatment reduced the loss of cochlear HCs caused by cisplatin in exosome culture. In addition, SchB treatment attenuated cisplatin-induced hearing loss and HC loss in mice. This study demonstrates the ability of SchB to inhibit cochlear hair cell apoptosis and ROS generation and shows its potential therapeutic effect on cisplatin ototoxicity.


Subject(s)
Antineoplastic Agents , Apoptosis , Cell Survival , Cisplatin , Cyclooctanes , Hair Cells, Auditory, Inner , Lignans , Oxidative Stress , Polycyclic Compounds , Reactive Oxygen Species , Cisplatin/toxicity , Cyclooctanes/pharmacology , Polycyclic Compounds/pharmacology , Polycyclic Compounds/toxicity , Animals , Apoptosis/drug effects , Lignans/pharmacology , Oxidative Stress/drug effects , Antineoplastic Agents/toxicity , Cell Survival/drug effects , Reactive Oxygen Species/metabolism , Hair Cells, Auditory, Inner/drug effects , Mice , Mice, Inbred C57BL , Protective Agents/pharmacology , Antioxidants/pharmacology , Evoked Potentials, Auditory, Brain Stem/drug effects , Male , Ototoxicity/prevention & control
13.
J Asian Nat Prod Res ; 26(5): 604-615, 2024 May.
Article in English | MEDLINE | ID: mdl-38634612

ABSTRACT

We established myocardial injury models in vivo and in vitro to investigate the cardioprotective effect of gomisin D obtained from Schisandra chinensis. Gomisin D significantly inhibited isoproterenol-induced apoptosis and hypertrophy in H9C2 cells. Gomisin D decreased serum BNP, ANP, CK-MB, cTn-T levels and histopathological alterations, and inhibited myocardial hypertrophy in mice. In mechanisms research, gomisin D reversed ISO-induced accumulation of intracellular ROS and Ca2+. Gomisin D further improved mitochondrial energy metabolism disorders by regulating the TCA cycle. These results demonstrated that gomisin D had a significant effect on isoproterenol-induced myocardial injury by inhibiting oxidative stress, calcium overload and improving mitochondrial energy metabolism.


Subject(s)
Apoptosis , Isoproterenol , Oxidative Stress , Polycyclic Compounds , Schisandra , Animals , Isoproterenol/pharmacology , Mice , Molecular Structure , Schisandra/chemistry , Oxidative Stress/drug effects , Apoptosis/drug effects , Calcium/metabolism , Male , Reactive Oxygen Species/metabolism , Lignans/pharmacology , Lignans/chemistry , Cardiotonic Agents/pharmacology , Cell Line , Myocytes, Cardiac/drug effects , Cyclooctanes/pharmacology , Cyclooctanes/chemistry
14.
Radiol Oncol ; 58(2): 221-233, 2024 06 01.
Article in English | MEDLINE | ID: mdl-38452391

ABSTRACT

BACKGROUND: The aim of the study was to explore the therapeutic effect of ultrasound targeted destruction of schisandrin A contrast microbubbles on liver cancer and its related mechanism. MATERIALS AND METHODS: The Span-PEG microbubbles loaded with schisandrin A were prepared using Span60, NaCl, PEG-1500, and schisandrin A. The loading rate of schisandrin A in Span-PEG composite microbubbles was determined by ultraviolet spectrophotometry method. The Walker-256 cell survival rate of schisandrin A was determined by 3-(4,5)-dimethylthiahiazo (-z-y1)-3,5-di-phenytetrazoliumromide (MTT) assay. The content of schisandrin A in the cells was determined by high performance liquid chromatography. Ultrasound imaging was used to evaluate the therapeutic effect in situ. Enzyme linked immunosorbent assay (ELISA) was used to measure the content of inflammatory factors in serum. Hematoxylin-eosin (HE) staining was used to observe the pathological changes of experimental animals in each group. Immunohistochemistry was used to detect the expression of hypoxia inducible factor-1α (HIF-1α), vascular endothlial growth factor (VEGF) and vascular endothelial growth factor receptor 2 (VEGFR-2) in tumor tissues, and western blot was used to detect the protein expression of phosphoinositide 3-kinase (PI3K)/AKT/mammalian target of rapamycin (mTOR) signaling pathway in tumor tissues. RESULTS: The composite microbubbles were uniform in size, and the particle size distribution was unimodal and stable, which met the requirements of ultrasound contrast agents. The loading rate of schisandrin A in Span-PEG microbubbles was 8.84 ± 0.14%, the encapsulation efficiency was 82.24±1.21%. The IC50 value of schisandrin A was 2.87 µg/mL. The drug + microbubbles + ultrasound (D+M+U) group had the most obvious inhibitory effect on Walker-256 cancer cells, the highest intracellular drug concentration, the largest reduction in tumor volume, the most obvious reduction in serum inflammatory factors, and the most obvious improvement in pathological results. The results of immunohistochemistry showed that HIF-1α, VEGF and VEGFR-2 protein decreased most significantly in D+M+U group (P < 0.01). WB results showed that D+M+U group inhibited the PI3K/AKT/mTOR signaling pathway most significantly (P < 0.01). CONCLUSIONS: Schisandrin A had an anti-tumor effect, and its mechanism might be related to the inhibition of the PI3K/AKT/mTOR signaling pathway. The schisandrin A microbubbles could promote the intake of schisandrin A in tumor cells after being destroyed at the site of tumor under ultrasound irradiation, thus playing the best anti-tumor effect.


Subject(s)
Contrast Media , Cyclooctanes , Lignans , Liver Neoplasms , Microbubbles , Polycyclic Compounds , Ultrasonic Therapy , Animals , Male , Rats , Cell Line, Tumor , Cell Survival/drug effects , Cyclooctanes/pharmacology , Lignans/pharmacology , Liver Neoplasms/drug therapy , Liver Neoplasms/diagnostic imaging , Polycyclic Compounds/pharmacology , Polycyclic Compounds/therapeutic use , Polyethylene Glycols , Ultrasonic Therapy/methods , Ultrasonography/methods , Rats, Sprague-Dawley
15.
Int J Antimicrob Agents ; 63(5): 107150, 2024 May.
Article in English | MEDLINE | ID: mdl-38513748

ABSTRACT

OBJECTIVES: To analyse the impact of the most clinically relevant ß-lactamases and their interplay with low outer membrane permeability on the activity of cefiderocol, ceftazidime/avibactam, aztreonam/avibactam, cefepime/enmetazobactam, cefepime/taniborbactam, cefepime/zidebactam, imipenem/relebactam, meropenem/vaborbactam, meropenem/xeruborbactam and meropenem/nacubactam against recombinant Escherichia coli strains. METHODS: We constructed 82 E. coli laboratory transformants expressing the main ß-lactamases circulating in Enterobacterales (70 expressing single ß-lactamase and 12 producing double carbapenemase) under high (E. coli TG1) and low (E. coli HB4) permeability conditions. Antimicrobial susceptibility testing was determined by reference broth microdilution. RESULTS: Aztreonam/avibactam, cefepime/zidebactam, cefiderocol, meropenem/xeruborbactam and meropenem/nacubactam were active against all E. coli TG1 transformants. Imipenem/relebactam, meropenem/vaborbactam, cefepime/taniborbactam and cefepime/enmetazobactam were also highly active, but unstable against most of MBL-producing transformants. Combination of ß-lactamases with porin deficiency (E. coli HB4) did not significantly affect the activity of aztreonam/avibactam, cefepime/zidebactam, cefiderocol or meropenem/nacubactam, but limited the effectiveness of the rest of carbapenem- and cefepime-based combinations. Double-carbapenemase production resulted in the loss of activity of most of the compounds tested, an effect particularly evident for those E. coli HB4 transformants in which MBLs were present. CONCLUSIONS: Our findings highlight the promising activity that cefiderocol and new ß-lactam/ß-lactamase inhibitors have against recombinant E. coli strains expressing widespread ß-lactamases, including when these are combined with low permeability or other enzymes. Aztreonam/avibactam, cefiderocol, cefepime/zidebactam and meropenem/nacubactam will help to mitigate to some extent the urgency of new compounds able to resist MBL action, although NDM enzymes represent a growing challenge against which drug development efforts are still needed.


Subject(s)
Anti-Bacterial Agents , Azabicyclo Compounds , Borinic Acids , Carboxylic Acids , Cefepime , Cefiderocol , Ceftazidime , Cephalosporins , Cyclooctanes , Drug Combinations , Escherichia coli , Lactams , Microbial Sensitivity Tests , Triazoles , beta-Lactamase Inhibitors , beta-Lactamases , Escherichia coli/drug effects , Escherichia coli/genetics , beta-Lactamases/genetics , beta-Lactamases/metabolism , Cephalosporins/pharmacology , beta-Lactamase Inhibitors/pharmacology , Azabicyclo Compounds/pharmacology , Anti-Bacterial Agents/pharmacology , Cyclooctanes/pharmacology , Ceftazidime/pharmacology , Cefepime/pharmacology , Boronic Acids/pharmacology , Meropenem/pharmacology , Aztreonam/pharmacology , Imipenem/pharmacology , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Heterocyclic Compounds, 1-Ring/pharmacology , Cell Membrane Permeability/drug effects
16.
Int J Mol Sci ; 25(6)2024 Mar 19.
Article in English | MEDLINE | ID: mdl-38542438

ABSTRACT

Schisandra chinensis (Schisandraceae) is a medicinal plant widely used in traditional Chinese medicine. Under the name Wu Wei Zi, it is used to treat many diseases, especially as a stimulant, adaptogen, and hepatoprotective. Dibenzocyclooctadiene lignans are the main compounds responsible for the effect of S. chinensis. As a part of ongoing studies to identify and evaluate anti-inflammatory natural compounds, we isolated a series of dibenzocyclooctadiene lignans and evaluated their biological activity. Furthermore, we isolated new sesquiterpene 7,7-dimethyl-11-methylidenespiro[5.5]undec-2-ene-3-carboxylic acid. Selected dibenzocyclooctadiene lignans were tested to assess their anti-inflammatory potential in LPS-stimulated monocytes by monitoring their anti-NF-κB activity, antioxidant activity in CAA assay, and their effect on gap junction intercellular communication in WB-ras cells. Some S. chinensis lignans showed antioxidant activity in CAA mode and affected the gap junction intercellular communication. The anti-inflammatory activity was proven for (-)-gomisin N, (+)-γ-schisandrin, rubrisandrin A, and (-)-gomisin J.


Subject(s)
Lignans , Polycyclic Compounds , Schisandra , Lignans/pharmacology , Cyclooctanes/pharmacology , Anti-Inflammatory Agents/pharmacology
17.
J Pharm Pharmacol ; 76(6): 681-690, 2024 Jun 06.
Article in English | MEDLINE | ID: mdl-38422325

ABSTRACT

OBJECTIVES: Schisandrin B (Sch B) has been shown to possess anti-inflammatory and antioxidant properties, however, its antirheumatoid arthritis properties and potential mechanism remain unexplored. This study evaluated the potential of Sch B in adjuvant-induced arthritic (AIA) rats. METHODS: AIA was induced by injecting 0.1 ml of CFA into the paw of rats and the animals were administered with Sch B (50 mg/kg) for 28 days. The effects of Sch B were evaluated using arthritis severity, serum levels of oxido-inflammatory, and metabolic index parameters. KEY FINDINGS: Sch B eased arthritic symptoms by significantly reducing paw swelling and arthritic score and increased body weight gain. Moreover, Sch B alleviated the levels of oxido-inflammatory markers including interleukin-1 beta, interleukin-6, tumor necrosis factor alpha, nuclear factor kappa B, transforming growth factor ß1, inducible nitric oxide synthase and malonaldehyde, as well as increased the levels of superoxide dismutase, glutathione, and Nrf2. Sch B also remarkably restored the altered levels of triglyceride, aspartate aminotransferase, lactic acid, pyruvate, phosphoenolpyruvate carboxylase, glucose, hypoxia inducible factor-1 alpha, and vascular endothelial growth factor. In addition, Sch B markedly alleviated p65 expression in the treated AIA rats. CONCLUSION: This study suggests that Sch B alleviated AIA by reducing oxidative stress, inflammation, and angiogenesis.


Subject(s)
Anti-Inflammatory Agents , Arthritis, Experimental , Cyclooctanes , Hypoxia-Inducible Factor 1, alpha Subunit , Inflammation Mediators , Lignans , Oxidative Stress , Polycyclic Compounds , Vascular Endothelial Growth Factor A , Animals , Cyclooctanes/pharmacology , Cyclooctanes/therapeutic use , Lignans/pharmacology , Lignans/therapeutic use , Oxidative Stress/drug effects , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Arthritis, Experimental/drug therapy , Arthritis, Experimental/metabolism , Polycyclic Compounds/pharmacology , Polycyclic Compounds/therapeutic use , Vascular Endothelial Growth Factor A/metabolism , Rats , Anti-Inflammatory Agents/pharmacology , Male , Inflammation Mediators/metabolism , Antioxidants/pharmacology , Signal Transduction/drug effects , Rats, Sprague-Dawley , Inflammation/drug therapy , Inflammation/metabolism
18.
Cancer Med ; 13(2): e6942, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38376003

ABSTRACT

OBJECTIVE: The purpose of this study is to explore the biological mechanism of Schizandrin A (SchA) inducing non-small cell lung cancer (NSCLC) apoptosis. METHODS: The reverse molecular docking tool "Swiss Target Prediction" was used to predict the targets of SchA. Protein-protein interaction analysis was performed on potential targets using the String database. Functional enrichment analyses of potential targets were performed with Gene Ontology and Kyoto Encyclopedia of Genes and Genomes. The conformation of SchA binding to target was simulated by chemical-protein interactomics and molecular docking. The effect of SchA on the expression and phosphorylation level of EGFR was detected by Western blot. Lipofectamine 3000 and EGFR plasmids were used to overexpress EGFR. Apoptosis was tested with Annexin V-FITC and propidium iodide staining, and cell cycle was detected by propidium iodide staining. RESULTS: The "Swiss Target Prediction" database predicted 112 and 111 targets based on the 2D and 3D structures of SchA, respectively, of which kinases accounted for the most, accounting for 24%. Protein interaction network analyses showed that molecular targets such as ERBB family and SRC were at the center of the network. Functional enrichment analyses indicated that ERBB-related signaling pathways were enriched. Compound-protein interactomics and molecular docking revealed that SchA could bind to the ATP-active pocket of the EGFR tyrosine kinase domain. Laboratory results showed that SchA inhibited the phosphorylation of EGFR. Insulin could counteract the cytotoxic effect of SchA. EGFR overexpression and excess EGF or IGF-1 had limited impacts on the cytotoxicity of SchA. CONCLUSIONS: Network pharmacology analyses suggested that ERBB family members may be the targets of SchA. SchA can inhibit NSCLC at least in part by inhibiting EGFR phosphorylation, and activating the EGFR bypass can neutralize the cytotoxicity of SchA.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Cyclooctanes , Lignans , Lung Neoplasms , Polycyclic Compounds , Humans , Apoptosis , Carcinoma, Non-Small-Cell Lung/genetics , Cell Line, Tumor , Cyclooctanes/pharmacology , ErbB Receptors/genetics , Lignans/pharmacology , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Molecular Docking Simulation , Polycyclic Compounds/pharmacology
19.
Naunyn Schmiedebergs Arch Pharmacol ; 397(2): 783-794, 2024 02.
Article in English | MEDLINE | ID: mdl-37658213

ABSTRACT

Schisandrin stands as one of the primary active compounds within the widely used traditional medicinal plant Schisandra chinensis (Turcz.) Baill. This compound exhibits sedative, hypnotic, anti-aging, antioxidant, and immunomodulatory properties, showcasing its effectiveness across various liver diseases while maintaining a favorable safety profile. However, the bioavailability of schisandrin is largely affected by hepatic and intestinal first-pass metabolism, which limits the clinical efficacy of schisandrin. In this paper, we review the various pharmacological effects and related mechanisms of schisandrin, in order to provide reference for subsequent drug research and promote its medicinal value.


Subject(s)
Drugs, Chinese Herbal , Lignans , Polycyclic Compounds , Drugs, Chinese Herbal/pharmacology , Lignans/pharmacology , Cyclooctanes/pharmacology , Polycyclic Compounds/pharmacology
20.
Chem Biodivers ; 20(12): e202301298, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37990607

ABSTRACT

Since ancient times, China has used natural medicine as the primary way to combat diseases and has a rich arsenal of natural medicines. With the progress of the times, the extraction of bioactive molecules from natural drugs has become the new development direction for natural medicines. Among the numerous natural drugs, Schisandrin C (Sch C), derived from Schisandra Chinensis (Turcz.) Baill. It has excellent potential for development and has been shown to possess various pharmacological properties, including hepatoprotective, antitumor and anti-inflammatory activities. Based on the biological properties of hepatoprotection, scholars have explored Sch C and its synthetic products in depth; some studies have shown that pentosidine has the effect of improving the symptoms of liver fibrosis and reducing the concentration of alanine transaminase (ALT) and aspartate aminotransferase (AST) in the serum of rats, which is an essential inspiration for the development of anti-liver fibrosis drugs. But more in vivo and ex vivo studies still need to be included. This paper focuses on Sch C's extraction and synthesis, biological activities and drug development progress. The future application prospects of Sch C are discussed to perfect its development work further.


Subject(s)
Lignans , Polycyclic Compounds , Schisandra , Rats , Animals , Lignans/pharmacology , Polycyclic Compounds/pharmacology , Cyclooctanes/pharmacology , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL