RESUMO
BACKGROUND: Schisandra extract has therapeutic and preventive effects on Alzheimer's disease (AD). Therefore, this study evaluated the anti-AD potential of Schisandrin A (SCH A) using an in vitro cell model. METHODS: SH-SY5Y and SK-N-SH cells were treated with 20 µM amyloid ß-protein (Aß)25-35. The Aß25-35-induced cells were then exposed to different concentrations of SCH A (1, 5, 10, 15 µg/mL). Moreover, to further explore the role of the extracellular signal-regulated kinase (ERK)/mitogen-activated protein kinase (MAPK) pathway in the anti-AD effects of SHC A, SH-SY5Y cells were treated with SCH A following incubation with ERK activator LM22B-10. The impact of SCH A on cell viability and apoptosis was evaluated using 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2-H-tetrazolium bromide (MTT) and flow cytometry. Furthermore, the oxidative stress markers and inflammatory cytokine levels were also assessed. The reactive oxygen species (ROS) levels were examined using 2',7'-Dichlorodihydrofluorescein Diacetate (DCFH-DA) method. Finally, Western blot analysis was employed to evaluate the phospho-ERK1/2 (p-ERK1/2) and ERK1/2. RESULTS: We observed that SCH A treatment (5, 10, 15 µg/mL) substantially increased the cell viability (p < 0.05), and reduced the apoptosis rate (10 and 15 µg/mL) in SH-SY5Y and SK-N-SH cells (p < 0.05). SCH A significantly ameliorated oxidative stress and reduced inflammatory cytokine levels in Aß25-35-induced cells (p < 0.05). Furthermore, SCH A up-regulated the p-ERK1/2 to ERK1/2 ratio in Aß25-35-induced cells. However, LM22B-10 treatment was found to exacerbate this effect of SCH A (p < 0.05). CONCLUSION: SCH A reduces the Aß25-35-induced inflammatory response and oxidative stress in SH-SY5Y and SK-N-SH cells, and the activation of the ERK/MAPK signaling pathway was related to its potential mechanism.
Assuntos
Doença de Alzheimer , Peptídeos beta-Amiloides , Ciclo-Octanos , Inflamação , Lignanas , Estresse Oxidativo , Fragmentos de Peptídeos , Compostos Policíclicos , Estresse Oxidativo/efeitos dos fármacos , Lignanas/farmacologia , Humanos , Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/induzido quimicamente , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Fragmentos de Peptídeos/metabolismo , Compostos Policíclicos/farmacologia , Compostos Policíclicos/uso terapêutico , Ciclo-Octanos/farmacologia , Ciclo-Octanos/uso terapêutico , Inflamação/tratamento farmacológico , Inflamação/induzido quimicamente , Inflamação/metabolismo , Células Tumorais CultivadasRESUMO
Schisandra chinensis (Turcz.) Baill. (S. chinensis) and Schisandra sphenanthera Rehd. et Wils (S. sphenanthera) are called "Wuweizi" in traditional Chinese medicine, and they have distinct clinical applications. To systematically compare the differential characteristics of S. chinensis and S. sphenanthera, this study employed ultra-performance liquid chromatography-quadrupole time-of-flight mass spectrometry (UPLC-QTOF-MS) and gas chromatography-mass spectrometry (GC-MS) to construct chemical profiles of these two species from different regions. In total, 31 non-volatiles and 37 volatiles were identified in S. chinensis, whereas 40 non-volatiles and 34 volatiles were detected in S. sphenanthera. A multivariate statistical analysis showed that the non-volatiles tigloygomisin P, schisandrol A, schisantherin C, and 6-O-benzoylgomisin O and the volatiles ylangene, γ-muurolene, and ß-pinene distinguish these species. Additionally, the metabolism of oxygen free radicals can contribute to the development of various diseases, including cardiovascular and neurodegenerative diseases. Therefore, antioxidant activities were evaluated using 1,1-diphenyl-2-picrylhydrazyl (DPPH) and 2,2'-azino-bis-3-ethylbenzthiazoline-6-sulphonic acid (ABTS) scavenging assays. The results showed that S. sphenanthera exhibited significantly higher antioxidant potential. A gray relational analysis indicated that the key contributors to the antioxidant activity of S. chinensis were schisandrol A, gomisin G, schisantherin C, pregomisin, gomisin J, and schisantherin B. For S. sphenanthera, the key contributors included gomisin K2, schisantherin B, gomisin J, pregomisin, schisantherin C, schisandrin, gomisin G, schisantherin A, schisanhenol, and α-pinene. The identification of the differential chemical markers and the evaluation of the antioxidant activities provide a foundation for further research into the therapeutic applications of these species. This innovative study provides a robust framework for the quality control and therapeutic application of S. chinensis and S. sphenanthera, offering new insights into their medicinal potential.
Assuntos
Antioxidantes , Cromatografia Gasosa-Espectrometria de Massas , Schisandra , Schisandra/química , Antioxidantes/química , Antioxidantes/farmacologia , Antioxidantes/análise , Cromatografia Gasosa-Espectrometria de Massas/métodos , Lignanas/química , Lignanas/análise , Ciclo-Octanos/química , Ciclo-Octanos/análise , Cromatografia Líquida de Alta Pressão/métodos , Compostos Policíclicos/química , Compostos Policíclicos/análise , Monoterpenos Bicíclicos/química , Extratos Vegetais/química , Extratos Vegetais/farmacologia , Monoterpenos/química , Monoterpenos/análise , Compostos Bicíclicos com Pontes/análise , Compostos Bicíclicos com Pontes/química , Medicamentos de Ervas Chinesas/química , Medicamentos de Ervas Chinesas/farmacologiaRESUMO
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.
Assuntos
Ciclo-Octanos , Lignanas , Compostos Policíclicos , Traumatismo por Reperfusão , Lignanas/farmacologia , Lignanas/química , Animais , Ciclo-Octanos/farmacologia , Ciclo-Octanos/química , Compostos Policíclicos/farmacologia , Compostos Policíclicos/química , Camundongos , Traumatismo por Reperfusão/tratamento farmacológico , Traumatismo por Reperfusão/metabolismo , Traumatismo por Reperfusão/patologia , Masculino , Transcriptoma/efeitos dos fármacos , Camundongos Endogâmicos C57BL , Simulação de Acoplamento Molecular , Substâncias Protetoras/farmacologia , Substâncias Protetoras/química , Modelos Animais de Doenças , Apoptose/efeitos dos fármacos , Farmacologia em RedeRESUMO
BACKGROUND: Lung cancer is the leading cause of cancer-related mortality. Cancer poses a significant challenge to human health and remains a persistent and pressing issue. Schisandrin C is one of the active ingredients of Schisandra chinensis and has various biological and pharmacological activities. This study aimed to investigate the effects of Schisandrin C on lung cancer and the underlying mechanism involved. METHODS: A network pharmacology strategy was used to screen the target genes and pathways involved in the relationship between Schisandrin and lung cancer. Next, a single-cell RNA sequencing (scRNA-seq) assay revealed the expression of genes specifically expressed in lung cancer epithelial cells. A549 cells were subsequently treated with Schisandrin C for 24 h or 48 h, cell viability was assessed via MTT and EdU staining experiments, and target gene expression was measured via RT-qPCR and immunofluorescence assays. Moreover, lung cancer patient tissues were observed via multiplex immunofluroscence staining. RESULTS: AKT1, CA9, BRAF, EGFR, ERBB2 and PIK3CA were overlapping target genes for network pharmacology and the scRNA-seq strategy. In vitro, the RT-qPCR results indicated that Schisandrin C inhibited the mRNA expression of the AKT1, CA9, FASN, MMP1, EGFR and BRAF genes. In clinical samples from patients with lung cancer, the expression levels of CA9 and AKT1 were found to be significantly higher in lung tumor tissues than in the adjacent normal (TAN) tissues. Moreover, the administration of an AKT kinase inhibitor reversed the inhibitory effect of Schisandrin C on A549 cells proliferation, whereas the administration of a CA9 inhibitor failed to have a similar effect. CONCLUSIONS: Schisandrin C effectively suppressed the proliferation and viability of A549 cells. Its mechanism was related to the inhibition of the AKT1 signaling pathway.
Assuntos
Proliferação de Células , Ciclo-Octanos , Lignanas , Neoplasias Pulmonares , Compostos Policíclicos , Humanos , Células A549 , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Ciclo-Octanos/farmacologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Lignanas/farmacologia , Lignanas/uso terapêutico , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/patologia , Neoplasias Pulmonares/metabolismo , Compostos Policíclicos/farmacologia , Compostos Policíclicos/uso terapêutico , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais/efeitos dos fármacosRESUMO
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).
Assuntos
Ciclo-Octanos , Frutas , Kadsura , Lignanas , Compostos Fitoquímicos , Lignanas/farmacologia , Lignanas/isolamento & purificação , Frutas/química , Humanos , Kadsura/química , Células Hep G2 , Ciclo-Octanos/farmacologia , Ciclo-Octanos/isolamento & purificação , Estrutura Molecular , Compostos Fitoquímicos/farmacologia , Compostos Fitoquímicos/isolamento & purificação , Substâncias Protetoras/farmacologia , Substâncias Protetoras/isolamento & purificação , Espécies Reativas de Oxigênio/metabolismo , Triglicerídeos , China , Colesterol , Hepatopatia Gordurosa não Alcoólica/tratamento farmacológicoRESUMO
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.
Assuntos
Lesão Pulmonar Aguda , Ciclo-Octanos , Nanopartículas , Dióxido de Silício , Titânio , Animais , Dióxido de Silício/toxicidade , Titânio/toxicidade , Humanos , Ciclo-Octanos/farmacologia , Ciclo-Octanos/uso terapêutico , Camundongos , Lesão Pulmonar Aguda/induzido quimicamente , Lesão Pulmonar Aguda/tratamento farmacológico , Lesão Pulmonar Aguda/patologia , Lesão Pulmonar Aguda/metabolismo , Células A549 , Masculino , Nanopartículas/química , Lignanas/farmacologia , Lignanas/uso terapêutico , Mucina-5AC/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , NF-kappa B/metabolismo , Óxido Nítrico Sintase Tipo II/metabolismo , Silicose/patologia , Silicose/tratamento farmacológico , Silicose/metabolismo , Ciclo-Oxigenase 2/metabolismoRESUMO
Monosubstituted tetrazines are important bioorthogonal reactive tools due to their rapid ligation with trans-cyclooctene. However, their application is limited by the reactivity-stability paradox in biological environments. In this study, we demonstrated that steric effects are crucial in resolving this paradox through theoretical methods and developed a simple synthetic route to validate our computational findings, leading to the discovery of 1,3-azole-4-yl and 1,2-azole-3-yl monosubstituted tetrazines as superior bioorthogonal tools. These new tetrazines surpass previous tetrazines in terms of high reactivities and elevated stabilities. The most stable tetrazine exhibits a reasonable stability (71% remaining after 24 h incubation in cell culture medium) and an exceptionally high reactivity (k2 > 104 M-1 s-1 toward trans-cyclooctene). Due to its good stability in biological systems, a noncanonical amino acid containing such a tetrazine side chain was genetically encoded into proteins site-specifically via an expanded genetic code. The encoded protein can be efficiently labeled using cyclopropane-fused trans-cyclooctene dyes in living mammalian cells with an ultrafast reaction rate exceeding 107 M-1 s-1, making it one of the fastest protein labeling reactions reported to date. Additionally, we showed its superiority through in vivo reactions in living mice, achieving an efficient local anchoring of proteins. These tetrazines are expected to be optimal bioorthogonal reactive tools within living systems.
Assuntos
Ciclo-Octanos , Estrutura Molecular , Humanos , Ciclo-Octanos/química , Ciclo-Octanos/síntese química , Animais , Azóis/química , Azóis/síntese químicaRESUMO
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.
Assuntos
Apoptose , Ciclo-Octanos , Lignanas , Lipopolissacarídeos , Compostos Policíclicos , Células de Sertoli , Animais , Masculino , Ciclo-Octanos/farmacologia , Compostos Policíclicos/farmacologia , Compostos Policíclicos/uso terapêutico , Lignanas/farmacologia , Lignanas/uso terapêutico , Apoptose/efeitos dos fármacos , Camundongos , Células de Sertoli/efeitos dos fármacos , Células de Sertoli/metabolismo , Receptores Androgênicos/metabolismo , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Inflamação/tratamento farmacológico , Inflamação/metabolismo , Inflamação/patologia , Simulação de Acoplamento Molecular , Testículo/efeitos dos fármacos , Testículo/metabolismo , Testículo/patologia , NF-kappa B/metabolismoRESUMO
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.
Assuntos
Estenose da Valva Aórtica , Valva Aórtica , Calcinose , Ciclo-Octanos , Lignanas , Proteína Supressora de Tumor p53 , Animais , Humanos , Masculino , Camundongos , Valva Aórtica/patologia , Valva Aórtica/efeitos dos fármacos , Valva Aórtica/metabolismo , Estenose da Valva Aórtica/tratamento farmacológico , Estenose da Valva Aórtica/patologia , Calcinose/tratamento farmacológico , Calcinose/patologia , Calcinose/metabolismo , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Senescência Celular/efeitos dos fármacos , Ciclo-Octanos/farmacologia , Modelos Animais de Doenças , Inflamação/tratamento farmacológico , Inflamação/patologia , Inflamação/metabolismo , Lignanas/farmacologia , Camundongos Endogâmicos C57BL , Osteogênese/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Transdução de Sinais/efeitos dos fármacos , Proteína Supressora de Tumor p53/metabolismoRESUMO
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.
Assuntos
Doxorrubicina , Sistemas de Liberação de Medicamentos , Verde de Indocianina , Terapia Fototérmica , Pró-Fármacos , Doxorrubicina/farmacologia , Doxorrubicina/química , Terapia Fototérmica/métodos , Humanos , Pró-Fármacos/farmacologia , Pró-Fármacos/química , Animais , Verde de Indocianina/química , Verde de Indocianina/farmacologia , Camundongos , Sistemas de Liberação de Medicamentos/métodos , Linhagem Celular Tumoral , Neoplasias/terapia , Neoplasias/tratamento farmacológico , Ciclo-Octanos/química , Ciclo-Octanos/farmacologia , Camundongos Endogâmicos BALB C , Antineoplásicos/farmacologia , Antineoplásicos/química , FemininoRESUMO
OBJECTIVES: We aimed to compare the stability of the newly developed ß-lactams (cefiderocol) and ß-lactam/ß-lactamase inhibitor combinations (ceftazidime/avibactam, ceftolozane/tazobactam, aztreonam/avibactam, cefepime/taniborbactam, cefepime/zidebactam, imipenem/relebactam, meropenem/vaborbactam, meropenem/nacubactam and meropenem/xeruborbactam) against the most clinically relevant mechanisms of mutational and transferable ß-lactam resistance in Pseudomonas aeruginosa. METHODS: We screened a collection of 61 P. aeruginosa PAO1 derivatives. Eighteen isolates displayed the most relevant mechanisms of mutational resistance to ß-lactams. The other 43 constructs expressed transferable ß-lactamases from genes cloned in pUCP-24. MICs were determined by reference broth microdilution. RESULTS: Cefiderocol and imipenem/relebactam exhibited excellent in vitro activity against all of the mutational resistance mechanisms studied. Aztreonam/avibactam, cefepime/taniborbactam, cefepime/zidebactam, meropenem/vaborbactam, meropenem/nacubactam and meropenem/xeruborbactam proved to be more vulnerable to mutational events, especially to overexpression of efflux operons. The agents exhibiting the widest spectrum of activity against transferable ß-lactamases were aztreonam/avibactam and cefepime/zidebactam, followed by cefepime/taniborbactam, cefiderocol, meropenem/xeruborbactam and meropenem/nacubactam. However, some MBLs, particularly NDM enzymes, may affect their activity. Combined production of certain enzymes (e.g. NDM-1) with increased MexAB-OprM-mediated efflux and OprD deficiency results in resistance to almost all agents tested, including last options such as aztreonam/avibactam and cefiderocol. CONCLUSIONS: Cefiderocol and new ß-lactam/ß-lactamase inhibitor combinations show promising and complementary in vitro activity against mutational and transferable P. aeruginosa ß-lactam resistance. However, the combined effects of efflux pumps, OprD deficiency and efficient ß-lactamases could still result in the loss of all therapeutic options. Resistance surveillance, judicious use of new agents and continued drug development efforts are encouraged.
Assuntos
Antibacterianos , Compostos Azabicíclicos , Cefiderocol , Cefalosporinas , Combinação de Medicamentos , Testes de Sensibilidade Microbiana , Pseudomonas aeruginosa , Inibidores de beta-Lactamases , beta-Lactamases , Pseudomonas aeruginosa/efeitos dos fármacos , Pseudomonas aeruginosa/genética , Pseudomonas aeruginosa/enzimologia , Cefalosporinas/farmacologia , Inibidores de beta-Lactamases/farmacologia , beta-Lactamases/genética , beta-Lactamases/metabolismo , Antibacterianos/farmacologia , Compostos Azabicíclicos/farmacologia , Ciclo-Octanos/farmacologia , Tazobactam/farmacologia , beta-Lactamas/farmacologia , Humanos , Resistência beta-Lactâmica/genética , Ceftazidima/farmacologia , Infecções por Pseudomonas/microbiologia , Infecções por Pseudomonas/tratamento farmacológico , Transferência Genética Horizontal , Cromossomos Bacterianos/genéticaRESUMO
BACKGROUND: In this study, Schisandrin B (SCHB), the main active component of Schisandra chinensis extract (SCE), was taken as the research object. From gene, microRNA (miR-124), and the level of protein expression system to study the influences of microglia phenotype to play the role of nerve inflammation. METHODS: In this study, we investigated the role of miR-124 in regulating microglial polarization alteration and NF-κB/TLR4 signaling and MAPK signaling in the LPS-induced BV2 by PCR, western blot, ELISA, immunofluorescence, and cytometry. RESULTS: SCE and SCHB significantly reduced the NO-releasing, decreased the levels of TNF-α, iNOS, IBA-1, and ratio of CD86+/CD206+, and increased the levels of IL-10, Arg-1. In addition, SCE and SCHB inhibited the nucleus translocation of NF-κB, decreased the expressions of IKK-α, and increased the expressions of IκB-α. Besides, the expressions of TLR4 and MyD88, and the ratios of p-p38/p38, p-ERK/ERK, and p-JNK/JNK were reduced by SCE and SCHB treatments. Furthermore, SCHB upregulated the mRNA levels of miR-124. However, the effects of SCHB were reversed by the miR-124 inhibitor. CONCLUSIONS: These findings suggested SCHB downregulated NF-κB/TLR4/MyD88 signaling pathway and MAPK signaling pathway via miR-124 to restore M1/M2 balance and alleviate depressive symptoms.
Assuntos
Ciclo-Octanos , Lignanas , Lipopolissacarídeos , MicroRNAs , Microglia , NF-kappa B , Compostos Policíclicos , Receptor 4 Toll-Like , MicroRNAs/metabolismo , Ciclo-Octanos/farmacologia , Compostos Policíclicos/farmacologia , Lignanas/farmacologia , Lipopolissacarídeos/farmacologia , Microglia/efeitos dos fármacos , Microglia/metabolismo , Animais , Camundongos , Receptor 4 Toll-Like/metabolismo , NF-kappa B/metabolismo , Linhagem Celular , Transdução de Sinais/efeitos dos fármacos , Schisandra/química , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Extratos Vegetais/farmacologiaRESUMO
Carbapenem-resistant Pseudomonas aeruginosa (CRPa) infection is extremely challenging to manage. Cefepime-zidebactam is a novel combination that can be considered for salvage therapy when no other antimicrobials are susceptible. A 15-y-old boy presented with 56% thermal burns, followed by skin and soft tissue infection, secondary bacteraemia, complicated parapneumonic effusion and endophthalmitis due to CRPa, which was not susceptible to any of the routinely available antibiotics. He was treated with cefepime-zidebactam for 45 d, with which he recovered.
Assuntos
Antibacterianos , Compostos Azabicíclicos , Cefalosporinas , Infecções por Pseudomonas , Pseudomonas aeruginosa , Terapia de Salvação , Humanos , Masculino , Infecções por Pseudomonas/tratamento farmacológico , Antibacterianos/uso terapêutico , Compostos Azabicíclicos/uso terapêutico , Pseudomonas aeruginosa/efeitos dos fármacos , Cefalosporinas/uso terapêutico , Adolescente , Resultado do Tratamento , Queimaduras/tratamento farmacológico , Queimaduras/complicações , Cefepima/uso terapêutico , Combinação de Medicamentos , Piperidinas , Ciclo-OctanosRESUMO
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.
Assuntos
Angiotensina II , Ciclo-Octanos , Lignanas , Camundongos Endogâmicos C57BL , Fator 88 de Diferenciação Mieloide , Miócitos Cardíacos , Compostos Policíclicos , Receptor 4 Toll-Like , Animais , Ciclo-Octanos/farmacologia , Ciclo-Octanos/uso terapêutico , Lignanas/farmacologia , Lignanas/uso terapêutico , Fator 88 de Diferenciação Mieloide/metabolismo , Compostos Policíclicos/farmacologia , Compostos Policíclicos/uso terapêutico , Angiotensina II/metabolismo , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Receptor 4 Toll-Like/metabolismo , Receptor 4 Toll-Like/genética , Camundongos , Masculino , Anti-Inflamatórios/farmacologia , Anti-Inflamatórios/uso terapêutico , Receptor 2 Toll-Like/metabolismo , Receptor 2 Toll-Like/genética , Remodelação Ventricular/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Células Cultivadas , NF-kappa B/metabolismoRESUMO
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.
Assuntos
Sulfato de Dextrana , Doenças Inflamatórias Intestinais , Limosilactobacillus reuteri , Camundongos Endogâmicos C57BL , Probióticos , Animais , Probióticos/administração & dosagem , Probióticos/farmacologia , Probióticos/uso terapêutico , Doenças Inflamatórias Intestinais/terapia , Doenças Inflamatórias Intestinais/tratamento farmacológico , Masculino , Microbioma Gastrointestinal/efeitos dos fármacos , Colite/induzido quimicamente , Colite/tratamento farmacológico , Colite/microbiologia , Colite/terapia , Espécies Reativas de Oxigênio/metabolismo , Camundongos , Humanos , Ciclo-Octanos/administração & dosagem , Ciclo-Octanos/química , Ciclo-Octanos/farmacologia , Ciclo-Octanos/uso terapêuticoRESUMO
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.
Assuntos
Movimento Celular , Proliferação de Células , Ciclo-Octanos , Janus Quinase 2 , Lignanas , Músculo Liso Vascular , Compostos Policíclicos , Fator de Transcrição STAT3 , Transdução de Sinais , Janus Quinase 2/metabolismo , Fator de Transcrição STAT3/metabolismo , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/efeitos dos fármacos , Músculo Liso Vascular/citologia , Lignanas/farmacologia , Transdução de Sinais/efeitos dos fármacos , Ciclo-Octanos/farmacologia , Compostos Policíclicos/farmacologia , Humanos , Miócitos de Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/metabolismo , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Animais , Aterosclerose/patologia , Aterosclerose/metabolismo , Aterosclerose/tratamento farmacológicoRESUMO
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.
Assuntos
Doença de Alzheimer , Disfunção Cognitiva , Glicogênio Sintase Quinase 3 beta , Lignanas , Fator 2 Relacionado a NF-E2 , Fármacos Neuroprotetores , Estresse Oxidativo , Transdução de Sinais , Fator 2 Relacionado a NF-E2/metabolismo , Doença de Alzheimer/tratamento farmacológico , Animais , Glicogênio Sintase Quinase 3 beta/metabolismo , Transdução de Sinais/efeitos dos fármacos , Lignanas/farmacologia , Masculino , Estresse Oxidativo/efeitos dos fármacos , Disfunção Cognitiva/tratamento farmacológico , Fármacos Neuroprotetores/farmacologia , Humanos , Camundongos , Ratos Sprague-Dawley , Ratos , Modelos Animais de Doenças , Schisandra/química , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Ciclo-Octanos/farmacologia , Linhagem Celular Tumoral , Cromonas/farmacologia , Neurônios/efeitos dos fármacos , Neurônios/metabolismoRESUMO
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.
Assuntos
Ciclo-Octanos , Ferroptose , Lignanas , Mastite , Compostos Policíclicos , Transdução de Sinais , Sirtuína 1 , Infecções Estafilocócicas , Staphylococcus aureus , Proteína Supressora de Tumor p53 , Animais , Lignanas/farmacologia , Lignanas/uso terapêutico , Ciclo-Octanos/farmacologia , Ciclo-Octanos/uso terapêutico , Ferroptose/efeitos dos fármacos , Mastite/tratamento farmacológico , Mastite/induzido quimicamente , Mastite/imunologia , Mastite/metabolismo , Compostos Policíclicos/farmacologia , Compostos Policíclicos/uso terapêutico , Feminino , Sirtuína 1/metabolismo , Transdução de Sinais/efeitos dos fármacos , Camundongos , Staphylococcus aureus/efeitos dos fármacos , Proteína Supressora de Tumor p53/metabolismo , Infecções Estafilocócicas/tratamento farmacológico , Infecções Estafilocócicas/imunologia , Anti-Inflamatórios/farmacologia , Anti-Inflamatórios/uso terapêutico , Glândulas Mamárias Animais/efeitos dos fármacos , Glândulas Mamárias Animais/patologia , Glândulas Mamárias Animais/imunologia , Citocinas/metabolismo , Inflamação/tratamento farmacológico , HumanosRESUMO
Bioorthogonal reactions have revolutionized chemical biology by enabling selective chemical transformations within living organisms and cells. This review comprehensively explores bioorthogonal chemistry, emphasizing inverse-electron-demand Diels-Alder (IEDDA) reactions between tetrazines and strained dienophiles and their crucial role in chemical biology and various applications within the human body. This highly reactive and selective reaction finds diverse applications, including cleaving antibody-drug conjugates, prodrugs, proteins, peptide antigens, and enzyme substrates. The versatility extends to hydrogel chemistry, which is crucial for biomedical applications, yet it faces challenges in achieving precise cellularization. In situ activation of cytotoxic compounds from injectable biopolymer belongs to the click-activated protodrugs against cancer (CAPAC) platform, an innovative approach to tumor-targeted prodrug delivery and activation. The CAPAC platform, relying on click chemistry between trans-cyclooctene (TCO) and tetrazine-modified biopolymers, exhibits modularity across diverse tumor characteristics, presenting a promising approach in anticancer therapeutics. The review highlights the importance of bioorthogonal reactions in developing radiopharmaceuticals for positron emission tomography (PET) imaging and theranostics, offering a promising avenue for diverse therapeutic applications.
Assuntos
Reação de Cicloadição , Ciclo-Octanos , Humanos , Ciclo-Octanos/química , Ciclo-Octanos/síntese química , Química Click , Compostos Heterocíclicos com 1 Anel/química , Compostos Heterocíclicos com 1 Anel/síntese química , Antineoplásicos/química , Antineoplásicos/farmacologia , Antineoplásicos/síntese química , Neoplasias/tratamento farmacológico , Neoplasias/diagnóstico por imagem , Estrutura MolecularRESUMO
Inflammation and oxidative stress (OS) are the major pathogenic characteristics of acute kidney injury (AKI). Studies have shown that Schisandrin (Sch) could regulate inflammatory disease. However, the function and mechanism of Sch in AKI progression are still unknown. Here, we investigated Sch's potential effects and mechanism on mice's renal damage and macrophages induced by lipopolysaccharide (LPS). Sch decreased LPS-induced inflammatory factor production while increasing the activity of related antioxidant enzymes in macrophages and mouse kidney tissues. Hematoxylin and eosin staining revealed that Sch may have the ability to profoundly inhibit inflammatory cell invasion and tissue damage caused by LPS in renal tissue. Furthermore, Western blot and immunohistochemical studies showed that Sch exerted its effects mainly through up-regulation of nuclear factor erythroid 2-related factor 2/heme oxygenase-1 and inhibition of Toll-like receptor 4âmitogen-activated protein kinases/nuclear factor-kappa B pathways. Collectively, this study illustrates that Sch suppresses LPS-stimulated AKI by descending inflammation and OS, illuminating prospective AKI treatment options.