-
Clayton Werner posted an update a month ago
To assess the efficacy of a new glycyrrhizin acid (GA)-eluting stent, in vivo experiments were conducted to measure strut coverage and neointimal growth. Rabbits (n = 20), New Zealand White, with atherosclerotic iliac artery plaques, were randomly divided into three groups, distinguished by the type of stent implanted: bare-metal stents (BMSs), rapamycin-eluting stents, and GA-eluting stents. After 28 days of in vivo intravascular ultrasound (IVUS) assessment, the vessels were collected for scanning electron microscopy (SEM) analysis and histological preparation. After four weeks of observation, the groups’ stent and external elastic lamina (EEL) areas were analyzed for comparative purposes. In contrast to bare metal stents (BMSs), rapamycin- or GA-eluting stents produced a reduction in the neointimal area, with GA-eluting stents yielding the smallest reduction. A disproportionately larger number of uncovered struts were associated with rapamycin-eluting stents, when contrasted with GA-eluting and bare-metal stents. Endothelial nitric oxide synthase (eNOS) expression levels were notably higher in GA-eluting stents when compared to BMSs and rapamycin-eluting stents, even as endothelial coverage between struts remained equivalent across the groups. In addition, stents that released GA exhibited a marked improvement in re-endothelialization and arterial healing compared to those releasing rapamycin, as observed in a rabbit atherosclerotic model. The study’s results highlight that the GA-coated stent used successfully reduced intimal hyperplasia and enhanced re-endothelialization.
Over thousands of years, traditional Chinese medicine has utilized Aconite to treat diarrhea. Whether the anti-diarrheal action of aconite aqueous extract (AA) is associated with regulation of gut microbiota or bile acid metabolism is currently undetermined. This study investigated whether AA’s anti-diarrheal properties are linked to its influence on gut microbiota composition and bile acid dynamics. Researchers examined the therapeutic impact of AA in a mouse model of diarrhea by measuring various factors, including body weight, stool water content, diarrhea severity, intestinal motility, colon tissue structure, and immunohistochemical markers in the colon. Subsequently, high-throughput sequencing of 16S rRNA was performed to determine the impact of AA on the gut microbial community, and targeted metabolomics was used to investigate the effect of AA on the metabolism of bile acids. Results from AA treatment revealed a decrease in fecal water content and diarrhea, a reduced intestinal transit rate, a decrease in colon pathology, and an increase in the amounts of AQP3 and AQP4 in the colon tissue. On top of that, AA showcased the proficiency to regulate the intricate balance of the gut microbiota. atpase pathway The effects were characterized by an increase in its richness (as determined by the ACE and Chao1 indices), a modification of the gut microbiota community structure (analyzed using PCA, PCoA, and NMDS), a rise in the relative abundances of norank f Muribaculaceae, Ruminococcus, Lachnospiraceae NK4A136 group, Prevotellaceae UCG-001, and norank f norank o Clostridia UCG-014, and a decrease in the relative abundances of Escherichia-Shigella, unclassified f Ruminococcaceae, Ruminococcus torques group, and Parasutterella. Essentially, AA notably enhanced fecal TCA (a primary bile acid) and DCA, LCA, GDCA, dehydro-LCA, and 12-keto-LCA (secondary bile acids), effectively re-establishing BA homeostasis. Besides, AA amplified the proportions of DCA/CA, DCA/TCA, and LCA/CDCA; concomitantly, it reduced the proportions of TLCA/LCA, GLCA/LCA, and TUDCA/UDCA. The observed anti-diarrheal effect of AA was contingent upon the recovery of gut microbiota and the maintenance of BA metabolic equilibrium. This study’s data reveals connections between AA methodologies and effective diarrheal treatment.
Baicalein, a 56,7-trihydroxyflavone compound used in traditional Chinese medicine, displays a number of pharmacological and biological activities, including, but not limited to, anti-inflammatory and anti-fibrotic properties. Although baicalein may hold therapeutic promise, its impact on peritoneal fibrosis has not yet been clinically investigated. Network pharmacology and molecular docking analyses were used in this study to assess baicalein’s impact on and underlying mechanisms of peritoneal fibrosis in peritoneal dialysis patients. Validation of the results encompassed both animal models and cultured human mesothelial cell lines. According to network analysis, nine shared genes are present in both baicalein’s targets and the human peritoneum RNA-seq dataset, comprising four encapsulating peritoneal sclerosis samples and four control samples. The elevated expression of MMP2, BAX, ADORA3, HIF1A, PIM1, CA12, and ALOX5 in the peritoneum with encapsulating peritoneal sclerosis, when compared to controls, indicates their potential as crucial targets for baicalein’s therapeutic effect against peritoneal fibrosis. Through KEGG and GO enrichment analyses, baicalein’s capacity to combat peritoneal fibrosis was determined to be through its influence on cell proliferation, inflammatory reactions, and the AGE-RAGE pathway. Analysis via molecular docking highlighted a significant potential for baicalein to bind to MMP2, consistent with the predictive model. Significantly, in a mouse model of peritoneal fibrosis generated by intraperitoneal injection of 425% glucose dialysis solution, baicalein treatment demonstrably reduced peritoneal fibrosis. This reduction manifested as less collagen deposition, lower protein expression of -smooth muscle actin and fibronectin, and attenuated peritoneal thickness, likely through a decrease in MMP2 expression, thus highlighting baicalein’s potential as a therapeutic agent for peritoneal dialysis-related fibrosis.
Nonsteroidal anti-inflammatory drugs (NSAIDs) and acetaminophen, as non-opioid analgesics, are frequently used to manage post-surgical pain experienced after a dental implant procedure. Yet, the relative pain-relieving power of common non-steroidal anti-inflammatory drugs and acetaminophen, when taken without a prescription, for individuals undergoing implant procedures, is presently unknown. We investigated the differential analgesic and anti-inflammatory responses to naproxen sodium and acetaminophen after the surgical procedure of placing one or two dental implants. In a double-blind, randomized trial, adult patients (n = 15 per group) underwent treatment with either naproxen sodium (440 mg loading dose + 220 mg every eight hours) or acetaminophen (1000 mg every six hours, maximum daily dose 3000 mg) for the three days subsequent to implant placement. The study medication’s impact on pain was monitored via a 0-10 pain scale assessment every 20 minutes for six hours post-treatment. The rescue medication Tramadol was dispensed in 50 mg tablets. Interleukin (IL)-6, IL-8, and IL-1 levels were ascertained in plasma and gingival crevicular fluid (GCF) samples taken pre-operatively and at 0, 1, 2, 4, 6, 24, and 72 hours post-operatively. A significant decrease in pain scores was observed in patients treated with naproxen sodium, in comparison to the acetaminophen group. Following surgical intervention, plasma and gingival crevicular fluid levels of inflammatory mediators escalated, subsequently declining to levels close to pre-operative values within 72 hours. Six hours post-operative, patients receiving naproxen sodium displayed substantially lower plasma IL-6 levels compared to those given acetaminophen. The levels of inflammatory mediators within gingival crevicular fluid remained unchanged regardless of the treatment group. Correlating the number of implants placed and body mass index (BMI) revealed their respective effects on inflammatory mediator levels in plasma and gingival crevicular fluid. In the reduction of post-operative pain and systemic inflammation after the placement of one or two dental implants, naproxen sodium outperformed acetaminophen. Further investigation is required to ascertain the applicability of these findings to more intricate implant procedures and the impact they have on clinical outcomes after implant insertion. Researchers utilize ClinicalTrials.gov for clinical trial registration, thereby enhancing the visibility and accountability of their studies. Concerning the identifier, it is NCT04694300.
A malignant tumor, gastric cancer (GC), is unfortunately characterized by its poor prognosis and high prevalence. Patients with GC may experience extended survival thanks to combined treatment approaches. Flavanoid acacetin demonstrably inhibits multiple forms of cancer cells, yet the underlying mechanisms remain largely obscure. Network pharmacology, coupled with RNA sequencing, was instrumental in predicting acacetin’s targets, which were further validated via drug affinity responsive target stability (DARTS), cellular thermal shift assay (CETSA), and molecular docking simulations. The biological roles of acacetin in MKN45 and MGC803 cell lines were explored through the use of TUNEL assays, crystal staining procedures, and colony formation assays. By employing reverse experiments, the pathways influenced by acacetin were validated. In a study using a xenotransplanted subcutaneous tumor model, the in vivo antitumor activity of acacetin was measured. From the study’s results, we found that acacetin directly binds to the EGFR protein, identified from over a dozen predicted targets. Beyond that, acacetin caused a change in the level of phosphorylated epidermal growth factor receptor. In the laboratory setting, acacetin stimulated the programmed cell death of gastric cancer cells. Indeed, EGFR agonists effectively reversed the inhibitory effects of acacetin on STAT3 and ERK signaling. Acacetin’s in vivo influence on tumor pEGFR protein levels led to a reduction in the size of GC xenograft tumors, without any clear evidence of toxicity. Our research findings indicate that acacetin directly impacts EGFR in GC cells. The antitumor effects of acacetin were linked to its inhibition of EGFR phosphatase activity, observed both in vitro and in vivo.