GLORIA

GEOMAR Library Ocean Research Information Access

Your email was sent successfully. Check your inbox.

An error occurred while sending the email. Please try again.

Proceed reservation?

Export
Filter
  • Medicine  (3)
Material
Publisher
Language
Years
FID
Subjects(RVK)
  • Medicine  (3)
RVK
  • 1
    In: The Journal of Clinical Pharmacology, Wiley, Vol. 62, No. 11 ( 2022-11), p. 1426-1434
    Abstract: Tigecycline is a broad‐spectrum antibacterial agent. As the incidence of multidrug‐resistant bacterial infections has increased in intensive care units (ICUs) over the past decades, tigecycline is often used in ICUs. Information about tigecycline‐associated hepatotoxicity in ICU patients is limited. To investigate the potential risk factors for tigecycline‐associated hepatotoxicity in ICU patients, 148 patients from 2 centers who had received tigecycline for at least 4 days were retrospectively analyzed. Hepatotoxicity was classified according to the National Cancer Institute Common Terminology Criteria for Adverse Events (5.0) grading system. As a result, 33.8% of patients experienced hepatotoxicity events in the ICU. The multivariate analysis showed that an albumin concentration 〈 25 g/L at baseline (odds ratio, 3.714; 95%CI, 1.082‐12.744; P = .037) and treatment duration (odds ratio, 1.094; 95%CI, 1.032‐1.160; P = .003) were significantly correlated with tigecycline‐associated hepatotoxicity. The median time to onset of hepatotoxicity was 8.0 days. The median duration ICU stay and the in‐hospital mortality rate were not different between the hepatotoxicity group and the nonhepatotoxicity group (33.5 days (interquartile range, 21.0‐72.0) vs 31.0 days (interquartile range, 21‐62.5), P = .850; 38.0% vs 43.8%; P = .504). Therefore, close monitoring of liver function is recommended for patients with baseline albumin concentrations 〈 25 g/L or for patients who receive tigecycline therapy for 〉 8 days.
    Type of Medium: Online Resource
    ISSN: 0091-2700 , 1552-4604
    URL: Issue
    RVK:
    Language: English
    Publisher: Wiley
    Publication Date: 2022
    detail.hit.zdb_id: 2010253-7
    SSG: 15,3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    In: The Journal of Pathology, Wiley, Vol. 258, No. 2 ( 2022-10), p. 164-178
    Abstract: The catalytic subunit of polycomb repressive complex 2 (PRC2), enhancer of zeste homolog 2 (EZH2), has been reported to be involved in angiogenesis in some tumors and autoimmune diseases. However, the mechanisms by which EZH2 regulates peritoneal angiogenesis remain unclear. We detected the expression of EZH2 in clinical samples and the peritoneal tissue of a mouse peritoneal fibrosis model induced by chlorhexidine gluconate (CG). In addition, we further investigated the mechanisms by which inhibition of EZH2 by 3‐deazaneplanocin A (3‐DZNeP) alleviated the CG‐induced peritoneal fibrosis mouse model in vivo and 3‐DZNeP or EZH2 siRNA treatment in cultured human peritoneal mesothelial cells (HPMCs) and human umbilical vein endothelial cells (HUVECs). The expression of EZH2 in the peritoneum of long‐term peritoneal dialysis (PD) patients and the CG‐induced peritoneal fibrosis mouse model was remarkably increased and this was positively associated with higher expression of vascular markers (CD31, CD34, VEGF, p‐VEGFR2). Peritoneal injection of 3‐DZNeP attenuated angiogenesis in the peritoneum of CG‐injured mice; improved peritoneal membrane function; and decreased phosphorylation of STAT3, ERK1/2, and activation of Wnt1/β‐catenin. In in vitro experiments, we demonstrated that inhibition of EZH2 by 3‐DZNeP or EZH2 siRNA decreased tube formation and the migratory ability of HUVECs via two pathways: the Wnt1/β‐catenin pathway and the IL‐6/STAT3 pathway. Suppression of the Wnt1/β‐catenin pathway and the IL‐6/STAT3 pathway subsequently reduced VEGF production in HPMCs. Using specific inhibitors of VEGFR2, ERK1/2, and HIF‐1α, we found that a VEGFR2/ERK1/2/HIF‐1α axis existed and contributed to angiogenesis in vitro . Moreover, phosphorylation of VEGFR2 and activation of the ERK1/2 pathway and HIF‐1α in HUVECs could be suppressed by inhibition of EZH2. Taken together, the results of this study suggest that EZH2 may be a novel target for preventing peritoneal angiogenesis in PD patients. © 2022 The Pathological Society of Great Britain and Ireland.
    Type of Medium: Online Resource
    ISSN: 0022-3417 , 1096-9896
    URL: Issue
    RVK:
    Language: English
    Publisher: Wiley
    Publication Date: 2022
    detail.hit.zdb_id: 1475280-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    In: The Journal of Pathology, Wiley, Vol. 250, No. 1 ( 2020-01), p. 79-94
    Abstract: Dysregulation of histone methyltransferase enhancer of zeste homolog 2 (EZH2) has been implicated in the pathogenesis of many cancers. However, the role of EZH2 in peritoneal fibrosis remains unknown. We investigated EZH2 expression in peritoneal dialysis (PD) patients and assessed its role in peritoneal fibrosis in cultured human peritoneal mesothelial cells (HPMCs) and murine models of peritoneal fibrosis induced by chlorhexidine gluconate (CG) or high glucose peritoneal dialysis fluid (PDF) by using 3‐deazaneplanocin A (3‐DZNeP), and EZH2 conditional knockout mice. An abundance of EZH2 was detected in the peritoneum of patients with PD associated peritonitis and the dialysis effluent of long‐term PD patients, which was positively correlated with expression of TGF‐β1, vascular endothelial growth factor, and IL‐6. EZH2 was found highly expressed in the peritoneum of mice following injury by CG or PDF. In both mouse models, treatment with 3‐DZNeP attenuated peritoneal fibrosis and inhibited activation of several profibrotic signaling pathways, including TGF‐β1/Smad3, Notch1, epidermal growth factor receptor and Src. EZH2 inhibition also inhibited STAT3 and nuclear factor‐κB phosphorylation, and reduced lymphocyte and macrophage infiltration and angiogenesis in the injured peritoneum. 3‐DZNeP effectively improved high glucose PDF‐associated peritoneal dysfunction by decreasing the dialysate‐to‐plasma ratio of blood urea nitrogen and increasing the ratio of dialysate glucose at 2 h after PDF injection to initial dialysate glucose. Moreover, delayed administration of 3‐DZNeP inhibited peritoneal fibrosis progression, reversed established peritoneal fibrosis and reduced expression of tissue inhibitor of metalloproteinase 2, and matrix metalloproteinase‐2 and ‐9. Finally, EZH2‐KO mice exhibited less peritoneal fibrosis than EZH2‐WT mice. In HPMCs, treatment with EZH2 siRNA or 3‐DZNeP suppressed TGF‐β1‐induced upregulation of α‐SMA and Collagen I and preserved E‐cadherin. These results indicate that EZH2 is a key epigenetic regulator that promotes peritoneal fibrosis. Targeting EZH2 may have the potential to prevent and treat peritoneal fibrosis. © 2019 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
    Type of Medium: Online Resource
    ISSN: 0022-3417 , 1096-9896
    URL: Issue
    RVK:
    Language: English
    Publisher: Wiley
    Publication Date: 2020
    detail.hit.zdb_id: 1475280-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
Close ⊗
This website uses cookies and the analysis tool Matomo. More information can be found here...