Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 161
Filter
1.
Sci Rep ; 13(1): 13475, 2023 08 18.
Article in English | MEDLINE | ID: mdl-37596361

ABSTRACT

Pulmonary fibrosis (PF) is a serious progressive fibrotic disease that is characterized by excessive accumulation of extracellular matrix (ECM), thus resulting in stiff lung tissues. Lysyl oxidase (LOX) is an enzyme involved in fibrosis by catalyzing collagen cross-linking. Studies found that the ingredients in schisandra ameliorated bleomycin (BLM)-induced PF, but it is unknown whether the anti-PF of schisandra is related to LOX. In this study, we established models of PF including a mouse model stimulated by BLM and a HFL1 cell model induced by transforming growth factor (TGF)-ß1 to evaluate the inhibition effects of Schisandrin C (Sch C) on PF. We observed that Sch C treatment decreased pulmonary indexes compared to control group. Treatment of Sch C showed a significant reduction in the accumulation of ECM as evidenced by decreased expressions of α-SMA, FN, MMP2, MMP9, TIMP1 and collagen proteins such as Col 1A1, and Col 3A1. In addition, the expression of LOX in the lung tissue of mice after Sch C treatment was effectively decreased compared with the MOD group. The inhibition effects in vitro were consistent with those in vivo. Mechanistic studies revealed that Sch C significantly inhibited TGF-ß1/Smad2/3 and TNF-α/JNK signaling pathways. In conclusion, our data demonstrated that Sch C significantly ameliorated PF in vivo and vitro, which may play an important role by reducing ECM deposition and inhibiting the production of LOX.


Subject(s)
Lignans , Polycyclic Compounds , Pulmonary Fibrosis , Animals , Mice , Pulmonary Fibrosis/drug therapy , Collagen , Lignans/pharmacology , Lignans/therapeutic use , Polycyclic Compounds/pharmacology , Polycyclic Compounds/therapeutic use
2.
Molecules ; 27(3)2022 Jan 29.
Article in English | MEDLINE | ID: mdl-35164203

ABSTRACT

A series of novel pleuromutilin derivatives containing nitrogen groups on the side chain of C14 were synthesized under mild conditions. Most of the synthesized derivatives displayed potent antibacterial activities. Compound 9 was found to be the most active antibacterial derivative against MRSA (MIC = 0.06 µg/mL). Furthermore, the result of time-kill curves showed that compound 9 had a certain inhibitory effect against MRSA in vitro. Moreover, according to a surface plasmon resonance (SPR) study, compound 9 (KD = 1.77 × 10-8 M) showed stronger affinity to the 50S ribosome than tiamulin (KD = 2.50 × 10-8 M). The antibacterial activity of compound 9 was further evaluated in an MRSA-infected murine thigh model. Compared to the negative control group, tiamulin reduced MRSA load (~0.7 log10 CFU/mL), and compound 9 performed a treatment effect (~1.3 log10 CFU/mL). In addition, compound 9 was evaluated in CYP450 inhibition assay and showed only moderate in vitro CYP3A4 inhibition (IC50 = 2.92 µg/mL).


Subject(s)
Anti-Bacterial Agents/therapeutic use , Diterpenes/therapeutic use , Drug Discovery , Methicillin-Resistant Staphylococcus aureus/isolation & purification , Polycyclic Compounds/therapeutic use , Staphylococcal Infections/drug therapy , Animals , Humans , Mice , Surface Plasmon Resonance , Pleuromutilins
3.
J Pharmacol Sci ; 148(2): 255-261, 2022 Feb.
Article in English | MEDLINE | ID: mdl-35063141

ABSTRACT

Mechanical stimulation of cultured keratinocytes and a living epidermis increases intracellular calcium ion concentrations ([Ca2+]i) in stimulated cells. This action propagates a Ca2+ wave to neighboring keratinocytes via ATP/P2Y2 receptors. Recent behavioral, pharmacological studies revealed that exogenous ATP induces itching via P2X3 receptors in mice. We previously showed that alloknesis occurs when an external stimulus is applied to the skin with increased epidermal histamine in the absence of spontaneous pruritus. Based on these results, we investigated the effects of histamine at a concentration that does not cause itching on ATP-induced itching. The mean number of scratching events induced by the mixture of ATP and histamine increased by 28% over the sum of that induced by histamine alone or ATP alone. A317491, a P2X3 receptor antagonist, suppressed the mixture-induced scratching more often than the ATP-induced scratching. Next, we examined the ATP-induced [Ca2+]i change before and after histamine stimulation using normal human epidermal keratinocytes. Some cells did not respond to ATP before histamine stimulation but responded to ATP afterward, the phenomenon suppressed by chlorpheniramine maleate. These findings suggest that histamine enhances ATP-induced itching and that a potential mechanism could involve increased responsiveness to ATP in keratinocytes.


Subject(s)
Adenosine Triphosphate/metabolism , Adenosine Triphosphate/pharmacology , Histamine/pharmacology , Keratinocytes/metabolism , Pruritus/chemically induced , Pruritus/drug therapy , Animals , Calcium/metabolism , Cells, Cultured , Dose-Response Relationship, Drug , Male , Mice, Inbred ICR , Phenols/pharmacology , Phenols/therapeutic use , Physical Stimulation , Polycyclic Compounds/pharmacology , Polycyclic Compounds/therapeutic use , Purinergic P2X Receptor Agonists , Receptors, Purinergic P2X3/metabolism , Receptors, Purinergic P2Y2/metabolism
4.
J Intern Med ; 291(1): 51-63, 2022 01.
Article in English | MEDLINE | ID: mdl-34425035

ABSTRACT

Lefamulin is a novel antibiotic agent within the pleuromutilin derivative class approved for the treatment of community-acquired bacterial pneumonia (CABP) by the United States Food and Drug Administration and the European Commission in 2019 and 2020, respectively. The objective of this article is to provide a summary of clinically relevant data underlying lefamulin and to provide recommendations for its place in therapy. In vitro data establish lefamulin's activity against a number of Gram-positive, Gram-negative and atypical organisms relevant in the treatment of CABP, including Streptococcus pneumoniae, Haemophilus influenzae, Moraxella catarrhalis, Legionella pneumophila, Mycoplasma pneumoniae and Chlamydophila pneumoniae. Two phase-3 studies, the Lefamulin Evaluation Against Pneumonia trials, established non-inferiority of lefamulin against moxifloxacin in the treatment of CABP, including the sequential transition from intravenous to oral therapy and across a broad set of patient demographics and severities. Pooled and post hoc analyses have confirmed these effects for a variety of subgroups and secondary endpoints. Real-world study data post-approval have largely not yet emerged for lefamulin, and there is a need for further investigation into safety/efficacy for off-label indications such as acute bacterial skin and skin structure infections and sexually transmitted infections. Further data regarding tolerability, particularly with long-term use, as well as the emergence of resistance over time, are still undefined.


Subject(s)
Anti-Bacterial Agents , Community-Acquired Infections , Diterpenes/therapeutic use , Pneumonia, Bacterial , Polycyclic Compounds/therapeutic use , Thioglycolates/therapeutic use , Anti-Bacterial Agents/therapeutic use , Community-Acquired Infections/drug therapy , Humans , Pneumonia, Bacterial/drug therapy , United States , Pleuromutilins
5.
Int J Parasitol ; 52(5): 275-284, 2022 04.
Article in English | MEDLINE | ID: mdl-34875254

ABSTRACT

Schistosomiasis is a tropical parasitic disease, in which the major clinical manifestation includes hepatosplenomegaly, portal hypertension, and organs fibrosis. Clinically, treatment of schistosomiasis involves the use of praziquantel (PZQ) and supportive care, which does not improve the patient's outcome as liver injuries persist. Here we show the beneficial effects of using PZQ in combination with Schisandrin B (Sch B). Concomitant treatment with PZQ and Sch B resulted in a significant improvement of hepatosplenomegaly and fibrosis, compared with single-agent treatment. We also demonstrated that PZQ-Sch B treatment ameliorates injuries in the lungs and intestine better than the sole use of PZQ or Sch B. In addition, PZQ-Sch B treatment improves the survival of S. mansoni-infected mice, and the treatment combination yields better therapeutic outcomes, as indicated by a partial improvement in neurological function. These results were accompanied by a reduction in neurological injuries. Collectively, we suggest that PZQ-Sch B concomitant therapy may be useful to alleviate schistosomiasis-associated liver injuries and prevent systemic complications.


Subject(s)
Anthelmintics , Polycyclic Compounds , Schistosomiasis mansoni , Animals , Anthelmintics/pharmacology , Cyclooctanes , Lignans , Mice , Polycyclic Compounds/pharmacology , Polycyclic Compounds/therapeutic use , Praziquantel , Schistosoma mansoni , Schistosomiasis mansoni/complications , Schistosomiasis mansoni/drug therapy , Schistosomiasis mansoni/parasitology
6.
J Med Chem ; 64(23): 17346-17365, 2021 12 09.
Article in English | MEDLINE | ID: mdl-34844412

ABSTRACT

Evodiamine (Evo) is a quinazolinocarboline alkaloid found in Evodia rutaecarpa and exhibits moderate antiproliferative activity. Herein, we report using a scaffold-hopping approach to identify a series of novel polycyclic heterocyclic derivatives based on Evo as the topoisomerase I (Top1) inhibitor for the treatment of triple-negative breast cancer (TNBC), which is an aggressive subtype of breast cancer with limited treatment options. The most potent compound 7f inhibited cell growth in a human breast carcinoma cell line (MDA-MB-231) with an IC50 value of 0.36 µM. Further studies revealed that Top1 was the target of 7f, which directly induced irreversible Top1-DNA covalent complex formation or induced an oxidative DNA lesion through an indirect mechanism mediated by reactive oxygen species. More importantly, in vivo studies showed that 7f exhibited potent antitumor activity in a TNBC-patient-derived tumor xenograft model. These results suggest that compound 7f deserves further investigation as a promising candidate for the treatment of TNBC.


Subject(s)
Antineoplastic Agents/therapeutic use , Heterocyclic Compounds/chemistry , Heterocyclic Compounds/therapeutic use , Polycyclic Compounds/chemistry , Polycyclic Compounds/therapeutic use , Quinazolines/chemistry , Triple Negative Breast Neoplasms/drug therapy , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Heterocyclic Compounds/pharmacology , Humans , Polycyclic Compounds/pharmacology , Triple Negative Breast Neoplasms/pathology
7.
Cell Mol Biol (Noisy-le-grand) ; 67(2): 25-32, 2021 Sep 29.
Article in English | MEDLINE | ID: mdl-34817341

ABSTRACT

There has been an exponential growth in the field of molecular oncology and cutting-edge research has enabled us to develop a better understanding of therapeutically challenging nature of cancer. Based on the mechanistic insights garnered from decades of research, puzzling mysteries of multifaceted nature of cancer have been solved to a greater extent. Our rapidly evolving knowledge about deregulated oncogenic cell signaling pathways has allowed us to dissect different oncogenic transduction cascades which play critical role in cancer onset, progression and metastasis. Pharmacological targeting of deregulated pathways has attracted greater than ever attention in the recent years. Henceforth, discovery and identification of high-quality biologically active chemicals and products is gaining considerable momentum. There has been an explosion in the dimension of natural product research because of tremendous potential of chemopreventive and pharmaceutical significance of natural products. Schisandrin is mainly obtained from Schisandra chinensis. Schisandrin has been shown to be effective against different cancers because of its ability to inhibit/prevent cancer via modulation of different cell signaling pathways. Importantly, regulation of non-coding RNAs by schisandrin is an exciting area of research that still needs detailed and comprehensive research.   However, we still have unresolved questions about pharmacological properties of schisandrin mainly in context of its regulatory role in TGF/SMAD, SHH/GLI, NOTCH and Hippo pathways.


Subject(s)
Cyclooctanes/therapeutic use , Lignans/therapeutic use , Neoplasms/prevention & control , Polycyclic Compounds/therapeutic use , Schisandra/chemistry , Signal Transduction/drug effects , Animals , Apoptosis/drug effects , Apoptosis/genetics , Carcinogenesis/drug effects , Carcinogenesis/genetics , Cell Movement/drug effects , Cell Movement/genetics , Clinical Trials as Topic , Cyclooctanes/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Humans , Lignans/pharmacology , Neoplasms/genetics , Neoplasms/metabolism , Plant Extracts/pharmacology , Plant Extracts/therapeutic use , Polycyclic Compounds/pharmacology , Protein Interaction Maps/drug effects , Protein Interaction Maps/genetics , Treatment Outcome
8.
J Immunol Res ; 2021: 8029963, 2021.
Article in English | MEDLINE | ID: mdl-34258300

ABSTRACT

BACKGROUND: Asthma is a complex inflammatory disorder that plagues a large number of people. Schisandrin B is an active ingredient of the traditional Chinese herbal medicine Schisandra with various proven physiological activities such as anti-inflammatory and antioxidant activities. In this study, we explored the anti-inflammatory and antioxidant effects and provided the mechanistic insights into the activity of schisandrin B in a mouse model of ovalbumin- (OVA-) induced allergic asthma. METHODS: Male BALB/c mice were sensitized and challenged with OVA to induce asthma and treated with various doses (15 mg/kg, 30 mg/kg, and 60 mg/kg) of SCH to alleviate the features of allergic asthma, airway hyperresponsiveness, inflammatory response, OVA-specific immunoglobulin (Ig)E level, and pathological injury. RESULTS: Schisandrin B significantly attenuated the airway hyperresponsiveness induced by OVA. Moreover, schisandrin B administration suppressed inflammatory responses, reduced the level of IgE, and attenuated pathological injury. Mechanistically, schisandrin B treatment promoted the activation of nuclear erythroid 2-related factor 2 (Nrf2), but suppressed the stimulation of the NF-κB pathway caused by OVA. CONCLUSION: Taken together, our study suggests that schisandrin B attenuates the features of asthmatic lungs by inhibiting the NF-κB pathway and activating the Nrf2 signaling pathway.


Subject(s)
Asthma/drug therapy , Lignans/pharmacology , NF-E2-Related Factor 2/agonists , NF-kappa B/antagonists & inhibitors , Polycyclic Compounds/pharmacology , Animals , Asthma/diagnosis , Asthma/immunology , Asthma/pathology , Bronchoalveolar Lavage Fluid/cytology , Bronchoalveolar Lavage Fluid/immunology , Cyclooctanes/pharmacology , Cyclooctanes/therapeutic use , Disease Models, Animal , Humans , Lignans/therapeutic use , Lung/drug effects , Lung/immunology , Lung/pathology , Male , Mice , NF-E2-Related Factor 2/metabolism , NF-kappa B/metabolism , Ovalbumin/administration & dosage , Ovalbumin/immunology , Oxidative Stress/drug effects , Oxidative Stress/immunology , Polycyclic Compounds/therapeutic use , Signal Transduction/drug effects , Signal Transduction/immunology , Specific Pathogen-Free Organisms
9.
Eur J Med Chem ; 223: 113624, 2021 Nov 05.
Article in English | MEDLINE | ID: mdl-34153574

ABSTRACT

A series of pleuromutilin derivatives with 1,2,4-triazole-3-substituted Schiff base structure were designed and synthesized under mild conditions. The in vitro antibacterial activities of the synthesized derivatives against 4 strains of Staphylococcus aureus (MRSA ATCC 43300, S.aureus ATCC 29213, S.aureus 144 and S.aureus AD3) and 1 strain of E. coli (ATCC 25922) were evaluated by the broth dilution method. Among these derivatives, compound 60 exhibited superior in vitro antibacterial effect against MRSA (MIC = 0.25 µg/mL) than tiamulin (MIC = 0.5 µg/mL), and compound 60 (-2.28 log10 CFU/mL) also displayed superior in vivo antibacterial efficacy than tiamulin (-1.40 log10 CFU/mL) in reducing MRSA load in the mouse thigh infection model. The time-kill study and the post-antibiotic effect study indicated that compound 60 showed a faster bactericidal kinetic and longer PAE time (exposure to 2 × MIC and 4 × MIC for 2 h, the PAE was 4.06 and 4.27 h) against MRSA compared with tiamulin (exposure to 2 × MIC and 4 × MIC for 2 h, the PAE was 1.72 and 2.14 h). Meanwhile, most of these compounds had no significant inhibitory effect on RAW 264.7 cells and HepG2 cells at the concentration of 4 µg/mL. Additionally, the development of resistance study showed that MRSA did not easily develop resistance against compound 60 compared with tiamulin after induction for 8 passages.


Subject(s)
Anti-Bacterial Agents/therapeutic use , Diterpenes/therapeutic use , Methicillin-Resistant Staphylococcus aureus/drug effects , Polycyclic Compounds/therapeutic use , Schiff Bases/therapeutic use , Staphylococcal Infections/drug therapy , Animals , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/pharmacology , Diterpenes/chemical synthesis , Diterpenes/pharmacology , Drug Design , Female , Mice, Inbred ICR , Microbial Sensitivity Tests , Polycyclic Compounds/chemical synthesis , Polycyclic Compounds/pharmacology , Schiff Bases/chemical synthesis , Schiff Bases/pharmacology , Pleuromutilins
10.
Eur J Pharmacol ; 906: 174220, 2021 Sep 05.
Article in English | MEDLINE | ID: mdl-34081905

ABSTRACT

Schisandrin C (Sch C) is one of the main components of Schisandra chinensis (Schisandra). Since the olden times, Schisandra has been used as a traditional herbal medicine in Asia. Recent studies have shown that Schisandra is effective against irritable bowel syndrome (IBS) in an animal model and affects IBS through the 5-HT3A pathway in the IBS rat model. However, there lacks fundamental research on the interaction of specific components of Schisandra with the 5-HT3A receptor for the treatment of IBS. We hypothesized that a component of Schisandra binds to the 5-HT3A receptor and identified Sch C via a screening work using two electrode-voltage clamps (TEVC). Thus, we aimed to elucidate the neuropharmacological actions between Sch C and the 5-HT3A receptor at molecular and cellular levels. Co-treatment of Sch C with 5-HT inhibited I5-HT in a reversible, concentrate-dependent, like-competition, and voltage-independent manner, and IC50 values of Sch C. Besides, the main binding positions of Sch C were identified through 3D modeling and point mutation were V225A and V288Y on 5-HT3A receptor. Thus, we suggest the potential of Sch C in treating IBS in a manner that suppresses excessive neuronal serotonin signaling in the synapse of sensory neurons and enterochromaffin (EC) cells. In conclusion, the results demonstrate the mechanism of interaction between Sch C and 5-HT3A receptor and reveal Sch C as a novel antagonist.


Subject(s)
Lignans/pharmacology , Polycyclic Compounds/pharmacology , Receptors, Serotonin, 5-HT3/metabolism , Serotonin 5-HT3 Receptor Antagonists/pharmacology , Animals , Cyclooctanes/pharmacology , Cyclooctanes/therapeutic use , Enterochromaffin Cells/drug effects , Enterochromaffin Cells/metabolism , Humans , Inhibitory Concentration 50 , Intestinal Mucosa/drug effects , Intestinal Mucosa/innervation , Intestinal Mucosa/pathology , Irritable Bowel Syndrome/drug therapy , Irritable Bowel Syndrome/pathology , Lignans/therapeutic use , Molecular Docking Simulation , Oocytes , Patch-Clamp Techniques , Polycyclic Compounds/therapeutic use , Receptors, Serotonin, 5-HT3/genetics , Receptors, Serotonin, 5-HT3/isolation & purification , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Sensory Receptor Cells/drug effects , Sensory Receptor Cells/metabolism , Serotonin 5-HT3 Receptor Antagonists/therapeutic use , Xenopus laevis
11.
J Med Chem ; 64(11): 7404-7421, 2021 06 10.
Article in English | MEDLINE | ID: mdl-34038111

ABSTRACT

Imbalance miRNAs contribute to tumor formation; therefore, the development of small-molecule compounds that regulate miRNA biogenesis is an important strategy in oncotherapy. Here, (-)-Gomisin M1 (GM) was found to modulate miRNA biogenesis to inhibit the proliferation, migration, and invasion of hepatocellular carcinoma (HCC) cells. GM modulated expression profiles of miRNA and protein in HCC cells and suppressed tumor growth in a mouse model. Mechanistically, GM affected miRNA maturation by targeting TAR RNA-binding protein 2 (TRBP), with an efficacy higher than that of enoxacin, and promoted the binding of TRBP with Dicer. Structural simplification and a preliminary structure-activity relationship study via the synthesis of 20 GM derivatives showed that compound 9 exhibited more potent inhibitory activity in HCC cell proliferation and affinity for TRBP than did GM. These results suggest that TRBP may be a novel potential therapeutic target in HCC and compound 9 may be a potential drug candidate for the treatment of HCC.


Subject(s)
Polycyclic Compounds/chemistry , RNA-Binding Proteins/metabolism , Animals , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , DEAD-box RNA Helicases/chemistry , DEAD-box RNA Helicases/metabolism , Enoxacin/chemistry , Enoxacin/metabolism , Enoxacin/pharmacology , Enoxacin/therapeutic use , Gene Expression Regulation, Neoplastic/drug effects , Humans , Liver Neoplasms/drug therapy , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Mice , Mice, Nude , MicroRNAs/metabolism , Polycyclic Compounds/metabolism , Polycyclic Compounds/pharmacology , Polycyclic Compounds/therapeutic use , Proteome/drug effects , Proteome/metabolism , RNA-Binding Proteins/antagonists & inhibitors , Ribonuclease III/chemistry , Ribonuclease III/metabolism , Structure-Activity Relationship , Transcriptome/drug effects , Transplantation, Heterologous
12.
Oncol Rep ; 45(6)2021 Jun.
Article in English | MEDLINE | ID: mdl-33907830

ABSTRACT

Lung cancer is one of the most common types of cancer in the world, resulting in numerous cancer­associated deaths. The properties of cancer stem cells (CSCs) are important for the initiation and deterioration of lung cancer. Schisandrin B (SchB), an active compound extracted from Schisandra chinensis, exerts anticancer effects in various malignancies, including lung cancer. Nevertheless, the potential of SchB in epithelial­mesenchymal transition (EMT) and CSC features of large­cell lung cancer remains unclear. The present study established cancer stem­like cells derived from large­cell lung cancer cells, NCI­H460 and H661, and revealed that SchB inhibited the viability of cancer stem­like cells at concentrations of ≥40 µmol/l. Moreover, SchB prominently inhibited cell migration, invasion and EMT. Sphere­forming assays and western blotting demonstrated that the stemness of cancer stem­like cells was alleviated by SchB treatment. Mechanistically, the current findings revealed that SchB contributed to the suppression of the NF­κB and p38 MAPK signaling pathways. Notably, further results revealed that the malignant behaviors of NCI­H460­CSCs induced by the activation of the NF­κB and p38 MAPK signaling pathways were suppressed by SchB treatment. Consistently, the inhibitory role of SchB in EMT and CSC activities, as well as in the activation of the NF­κB and p38 MAPK signaling pathways, was confirmed in vivo. In conclusion, the present study demonstrated that SchB exerted inhibitory effects on large­cell lung cancer cells via targeting the NF­κB and p38 MAPK signaling pathways, suggesting that SchB may act as a potential therapeutic drug for large­cell lung cancer.


Subject(s)
Carcinoma, Non-Small-Cell Lung/drug therapy , Cell Self Renewal/drug effects , Epithelial-Mesenchymal Transition/drug effects , Lignans/pharmacology , Lung Neoplasms/drug therapy , Polycyclic Compounds/pharmacology , Animals , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cyclooctanes/pharmacology , Cyclooctanes/therapeutic use , Humans , Lignans/therapeutic use , Lung Neoplasms/pathology , MAP Kinase Signaling System/drug effects , Male , Mice , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/pathology , Polycyclic Compounds/therapeutic use , Transcription Factor RelA/metabolism , Xenograft Model Antitumor Assays , p38 Mitogen-Activated Protein Kinases/metabolism
13.
Cells ; 10(2)2021 02 21.
Article in English | MEDLINE | ID: mdl-33669953

ABSTRACT

Multidrug resistance (MDR) is one of the major clinical challenges in cancer treatment and compromises the effectiveness of conventional anticancer chemotherapeutics. Among known mechanisms of drug resistance, drug efflux via ATP binding cassette (ABC) transporters, namely P-glycoprotein (P-gp) has been characterized as a major mechanism of MDR. The primary function of ABC transporters is to regulate the transport of endogenous and exogenous small molecules across the membrane barrier in various tissues. P-gp and similar efflux pumps are associated with MDR because of their overexpression in many cancer types. One of the intensively studied approaches to overcome this mode of MDR involves development of small molecules to modulate P-gp activity. This strategy improves the sensitivity of cancer cells to anticancer drugs that are otherwise ineffective. Although multiple generations of P-gp inhibitors have been identified to date, reported compounds have demonstrated low clinical efficacy and adverse effects. More recently, natural polyphenols have emerged as a promising class of compounds to address P-gp linked MDR. This review highlights the chemical structure and anticancer activities of selected members of a structurally unique class of 'biaryl' polyphenols. The discussion focuses on the anticancer properties of ellagic acid, ellagic acid derivatives, and schisandrins. Research reports regarding their inherent anticancer activities and their ability to sensitize MDR cell lines towards conventional anticancer drugs are highlighted here. Additionally, a brief discussion about the axial chirality (i.e., atropisomerism) that may be introduced into these natural products for medicinal chemistry studies is also provided.


Subject(s)
Cyclooctanes/therapeutic use , Drug Resistance, Multiple , Drug Resistance, Neoplasm , Ellagic Acid/therapeutic use , Lignans/therapeutic use , Neoplasms/drug therapy , Polycyclic Compounds/therapeutic use , Polyphenols/therapeutic use , Animals , Cyclooctanes/chemistry , Drug Discovery , Ellagic Acid/chemistry , Humans , Lignans/chemistry , Polycyclic Compounds/chemistry , Polyphenols/chemistry
15.
Immunopharmacol Immunotoxicol ; 43(2): 212-222, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33588680

ABSTRACT

Aim: The present study was aimed to evaluate the anxiolytic and antidepressant-like effects of schizandrin (from Schisandra chinensis (Turcz.) Baill. which is a functional food) against chronic liver injury in mice.Methods: Chronic liver injury was induced by the treatment of d-galactose (d-GaIN, 200 mg/kg, s.c.) for 8 weeks.Results: Administration of schizandrin (30 mg/kg, i.g.) significantly ameliorated d-GaIN-induced anxiety and depression-like behavior as evident from the results of open field test (OFT), sucrose preference test (SPT), tail suspension test (TST), forced swimming test (FST), novelty-suppressed feeding test (NSFT), and elevated plus maze (EPM) test. In addition, schizandrin remarkably reduced the oxidative stress due to its potential to enhance the levels of decreased CAT, GSH/GSSG, SOD, and increased MDA in peripheral and brain, the antioxidant activities might be related with the Nrf2/HO-1 pathway. Furthermore, schizandrin could dramatically inhibit the neuroinflammation in mice by reducing pro-inflammatory cytokines (TNF-α, IL-1ß, and IL-6) through regulating NF-κB/NLRP3/Iba-1 signaling. Besides, the elevated levels of ammonia, AST, and ALT were significantly reduced by schizandrin.Conclusion: The present data revealed that hyperammonemia produced due to liver injury-induced oxidative stress and neuroinflammation in the hippocampus and prefrontal cortex resulting in anxiety and depression were improved by schizandrin.


Subject(s)
Anxiety/drug therapy , Chemical and Drug Induced Liver Injury/drug therapy , Cyclooctanes/therapeutic use , Depression/drug therapy , Inflammation Mediators/antagonists & inhibitors , Lignans/therapeutic use , Oxidative Stress/drug effects , Polycyclic Compounds/therapeutic use , Animals , Anxiety/chemically induced , Anxiety/metabolism , Chemical and Drug Induced Liver Injury/metabolism , Cyclooctanes/pharmacology , Depression/chemically induced , Depression/metabolism , Galactose/toxicity , Inflammation Mediators/metabolism , Lignans/pharmacology , Locomotion/drug effects , Locomotion/physiology , Male , Maze Learning/drug effects , Maze Learning/physiology , Mice , Mice, Inbred C57BL , Oxidative Stress/physiology , Polycyclic Compounds/pharmacology , Schisandra
16.
Pharmacol Res ; 166: 105459, 2021 04.
Article in English | MEDLINE | ID: mdl-33545313

ABSTRACT

Schisandrin B (Sch B) is the major active constituent of the traditional Chinese medicine Schisandra chinensis and has anti-inflammatory activity, but the target of Sch B remains unclear. T helper 17 (TH17) cells have been involved in the pathogenesis of many autoimmune and inflammatory diseases. Here, we showed that Sch B could decrease IL-17A production of CD4+ T cells by targeting STAT3 in vitro. Importantly, Sch B has therapeutic effects on DSS-induced acute and chronic colitis, CD4+CD45RBhigh T cell-induced colitis. Furthermore, we identified TH17 cells as the direct target of Sch B for mediating its anti-inflammatory activity. Sch B could serve as a lead for developing new therapeutics against TH17 cells or IL-17A cytokine-driven diseases.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Inflammatory Bowel Diseases/drug therapy , Lignans/therapeutic use , Polycyclic Compounds/therapeutic use , Th17 Cells/drug effects , Animals , Anti-Inflammatory Agents/pharmacology , Cell Differentiation/drug effects , Cells, Cultured , Cyclooctanes/pharmacology , Cyclooctanes/therapeutic use , Female , Humans , Inflammatory Bowel Diseases/pathology , Lignans/pharmacology , Mice, Inbred C57BL , Polycyclic Compounds/pharmacology , Th17 Cells/pathology
17.
Pharmacology ; 106(3-4): 177-188, 2021.
Article in English | MEDLINE | ID: mdl-33486482

ABSTRACT

INTRODUCTION: Although oxidative stress has been demonstrated to mediate acute ethanol-induced changes in autophagy in the heart, the precise mechanism behind redox regulation in acute ethanol heart disease remains largely unknown. METHODS: Wild-type C57BL/6 mice were intraperitoneally injected with ethanol (3 g/kg/day) for 3 consecutive days. The effects of ethanol on cultured primary cardiomyocytes and H9c2 myoblasts were also studied in vitro. Levels of autophagic flux, cardiac apoptosis and function, reactive oxygen species (ROS) accumulation, NOX4, and NOX2 were examined. The NOX4 gene was knocked down with NOX4 siRNA. RESULTS: In this study, we demonstrated that schisandrin B inhibited acute ethanol-induced autophagy and sequent apoptosis. In addition, schisandrin B treatment improved cardiac function in ethanol-treated mice. Furthermore, NOX4 protein expression was increased during acute ethanol exposure, and the upregulation of NOX4 was significantly inhibited by schisandrin B treatment. The knockdown of NOX4 prevented ROS accumulation, cell autophagy, and apoptosis. CONCLUSION: These results highlight that NOX4 is a critical mediator of ROS and elaborate the role of the NOX4/ROS axis in the effect of schisandrin B on autophagy and autophagy-mediated apoptosis in acute ethanol exposure, which suggests a therapeutic strategy for acute alcoholic cardiomyopathy.


Subject(s)
Autophagy/drug effects , Cardiomyopathy, Alcoholic/prevention & control , Heart Injuries/prevention & control , Lignans/pharmacology , NADPH Oxidase 4/metabolism , Polycyclic Compounds/pharmacology , Protective Agents/pharmacology , Animals , Apoptosis/drug effects , Autophagy/genetics , Cyclooctanes/pharmacology , Cyclooctanes/therapeutic use , Down-Regulation , Ethanol/toxicity , Gene Knockdown Techniques , Heart Injuries/chemically induced , Heart Injuries/metabolism , Lignans/therapeutic use , Male , Mice, Inbred C57BL , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , NADPH Oxidase 4/antagonists & inhibitors , NADPH Oxidase 4/genetics , Polycyclic Compounds/therapeutic use , Primary Cell Culture , Protective Agents/therapeutic use , Reactive Oxygen Species/antagonists & inhibitors , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects
18.
Drugs ; 81(2): 233-256, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33247830

ABSTRACT

Lefamulin is a novel oral and intravenous (IV) pleuromutilin developed as a twice-daily treatment for community-acquired bacterial pneumonia (CABP). It is a semi-synthetic pleuromutilin with a chemical structure that contains a tricyclic core of five-, six-, and eight-membered rings and a 2-(4-amino-2-hydroxycyclohexyl)sulfanylacetate side chain extending from C14 of the tricyclic core. Lefamulin inhibits bacterial protein synthesis by binding to the 50S bacterial ribosomal subunit in the peptidyl transferase center (PTC). The pleuromutilin tricyclic core binds to a pocket close to the A site, while the C14 side chain extends to the P site causing a tightening of the rotational movement in the binding pocket referred to as an induced-fit mechanism. Lefamulin displays broad-spectrum antibacterial activity against Gram-positive and Gram-negative aerobic and anaerobic bacteria as well as against atypical bacteria that commonly cause CABP. Pleuromutilin antibiotics exhibit low rates of resistance development and lack cross-resistance to other antimicrobial classes due to their unique mechanism of action. However, pleuromutilin activity is affected by mutations in 23S rRNA, 50S ribosomal subunit proteins rplC and rplD, ATP-binding cassette (ABC)-F transporter proteins such as vga(A), and the methyltransferase cfr. The pharmacokinetic properties of lefamulin include: volume of distribution (Vd) ranging from 82.9 to 202.8 L, total clearance (CLT) of 19.5 to 21.4 L/h, and terminal elimination half-life (t1/2) of 6.9-13.2 h; protein binding of lefamulin is high and non-linear. The oral bioavailability of lefamulin has been estimated as 24% in fasted subjects and 19% in fed subjects. A single oral dose of lefamulin 600 mg administered in fasted patients achieved a maximum plasma concentration (Cmax) of 1.2-1.5 mg/L with a time of maximum concentration (Tmax) ranging from 0.8 to 1.8 h, and an area under the plasma concentration-time curve from 0 to infinity (AUC0-∞) of 8.5-8.8 mg h/L. The pharmacodynamic parameter predictive of lefamulin efficacy is the free plasma area under the concentration-time curve divided by the minimum inhibitory concentration (fAUC24h/MIC). Lefamulin efficacy has been demonstrated using various animal models including neutropenic murine thigh infection, pneumonia, lung infection, and bacteremia. Lefamulin clinical safety and efficacy was investigated through a Phase II clinical trial of acute bacterial skin and skin structure infection (ABSSSI), as well as two Phase III clinical trials of CABP. The Phase III trials, LEAP 1 and LEAP 2 established non-inferiority of lefamulin to moxifloxacin in both oral and IV formulations in the treatment of CABP. The United States Food and Drug Administration (FDA), European Medicines Agency (EMA), and Health Canada have each approved lefamulin for the treatment of CABP. A Phase II clinical trial has been completed for the treatment of ABSSSI, while the pediatric program is in Phase I. The most common adverse effects of lefamulin include mild-to-moderate gastrointestinal-related events such as nausea and diarrhea. Lefamulin represents a safe and effective option for treating CABP in cases of antimicrobial resistance to first-line therapies, clinical failure, or intolerance/adverse effects to currently used agents. Clinical experience and ongoing clinical investigation will allow clinicians and antimicrobial stewardship programs to optimally use lefamulin in the treatment of CABP.


Subject(s)
Anti-Bacterial Agents/therapeutic use , Community-Acquired Infections/drug therapy , Diterpenes/therapeutic use , Pneumonia, Bacterial/drug therapy , Polycyclic Compounds/therapeutic use , Thioglycolates/therapeutic use , Administration, Oral , Anti-Bacterial Agents/administration & dosage , Diterpenes/administration & dosage , Humans , Injections, Intravenous , Polycyclic Compounds/administration & dosage , Thioglycolates/administration & dosage
20.
Cancer Genomics Proteomics ; 17(5): 517-527, 2020.
Article in English | MEDLINE | ID: mdl-32859630

ABSTRACT

BACKGROUND/AIM: Deregulated activation of signaling through the RAS/RAF/mitogen-activated protein kinase/extracellular signal-regulated kinase (RAS/RAF/MEK/ERK) and signal transducer and activator of transcription (STAT) pathways is involved in numerous hematological malignancies, making it an attractive therapeutic target. This study aimed to assess the effect of the combination of ERK2 inhibitor VX-11e and STAT3 inhibitor STA-21 on acute lymphoblastic leukemia cell lines REH and MOLT-4. MATERIALS AND METHODS: REH and MOLT-4 cell lines were cultured with each drug alone and in combination. Cell viability, ERK activity, cell cycle distribution, apoptosis and oxidative stress induction were assessed by flow cytometry. Protein levels of STAT3, phospho-STAT3, protein tyrosine phosphatase 4A3 (PTP4A3), survivin, p53 and p21 were determined by western blotting. RESULTS: VX-11e in combination with STA-21 significantly inhibited cell viability, induced G0/G1 cell-cycle arrest, enhanced production of reactive oxygen species, and induced apoptosis. These effects were associated with an increased level of p21 protein in REH cells and with reduced levels of phopho-STAT3, survivin and PTP4A3 proteins in MOLT-4 cells. CONCLUSION: Our findings provide a rationale for combined inhibition of RAS/RAF/MEK/ERK and STAT3 pathways in order to enhance anticancer effects against acute lymphoblastic leukemia cells.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , STAT3 Transcription Factor/antagonists & inhibitors , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Humans , MAP Kinase Signaling System/drug effects , Polycyclic Compounds/pharmacology , Polycyclic Compounds/therapeutic use , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Pyrimidines/pharmacology , Pyrimidines/therapeutic use , Pyrroles/pharmacology , Pyrroles/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...