• Title/Summary/Keyword: anti-BNP

Search Result 2, Processing Time 0.02 seconds

Functional Expression of Anti-BNP scFv in E. coli Cytoplasm for the Detection of B-type Natriuretic Peptide (B-type natriuretic peptide 분석을 위한 항 BNP scFv 항체의 대장균 세포질 내에서의 기능적 발현)

  • Maeng, Bo-Hee;Nam, Dong-Hyun;Kim, Yong-Hwan
    • KSBB Journal
    • /
    • v.24 no.6
    • /
    • pp.591-597
    • /
    • 2009
  • B-type natriuretic peptide is a neurohormone secreted in the cardiac ventricles. BNP levels are elevated in patients with ventricular dysfunction. Therefore, the concentration of BNP is important factor to reflect diagnosis and prognosis for cardiovascular disease. In this respect, anti-BNP scFv is an urgent requirement for early diagnosis in the field of biosensor. Herein, the genetic codes of anti-BNP scFv were chemically synthesized and cloned into both pET22b (+) and pColdⅣ vector, respectively. The recombinant scFv was successfully expressed as a functional form in cytoplasm of E. coli and detected through Western blot and ELISA. The highest level of functional expression of anti-BNP scFv was achieved using pET22b (+) vector at $15^{\circ}C$ by addition of 0.1 mM IPTG. Additionally, being exposed to both BNP and ANP, anti-BNP scFv specifically captured only BNP. Therefore, anti-BNP scFv expressed in this study will be applied to measure the concentration of BNP as a diagnostic recognition molecule.

Ginsenoside F1 attenuates pirarubicin-induced cardiotoxicity by modulating Nrf2 and AKT/Bcl-2 signaling pathways

  • Yang Zhang;Jiulong Ma;Shan Liu;Chen Chen;Qi Li;Meng Qin;Liqun Ren
    • Journal of Ginseng Research
    • /
    • v.47 no.1
    • /
    • pp.106-116
    • /
    • 2023
  • Background: Pirarubicin (THP) is an anthracycline antibiotic used to treat various malignancies in humans. The clinical usefulness of THP is unfortunately limited by its dose-related cardiotoxicity. Ginsenoside F1 (GF1) is a metabolite formed when the ginsenosides Re and Rg1 are hydrolyzed. However, the protective effects and underlying mechanisms of GF1 on THP-induced cardiotoxicity remain unclear. Methods: We investigated the anti-apoptotic and anti-oxidative stress effects of GF1 on an in vitro model, using H9c2 cells stimulated by THP, plus trigonelline or AKT inhibitor imidazoquinoxaline (IMQ), as well as an in vivo model using THP-induced cardiotoxicity in rats. Using an enzyme-linked immunosorbent test, the levels of malondialdehyde (MDA), brain natriuretic peptide (BNP), creatine kinase (CK-MB), cardiac troponin (c-TnT), lactate dehydrogenase (LDH), superoxide dismutase (SOD) and glutathione (GSH) were determined. Nuclear factor (erythroid-derived2)-like 2 (Nrf2) and the expression of Nrf2 target genes, including heme oxygenase-1 (HO-1), glutathione-S-transferase (Gst), glutamate-cysteine ligase modifier subunit (GCLM), and expression levels of AKT/Bcl-2 signaling pathway proteins were detected using Western blot analysis. Results: THP-induced myocardial histopathological damage, electrocardiogram (ECG) abnormalities, and cardiac dysfunction were reduced in vivo by GF1. GF1 also decreased MDA, BNP, CK-MB, c-TnT, and LDH levels in the serum, while raising SOD and GSH levels. GF1 boosted Nrf2 nuclear translocation and Nrf2 target gene expression, including HO-1, Gst, and GCLM. Furthermore, GF1 regulated apoptosis by activating AKT/Bcl-2 signaling pathways. Employing Nrf2 inhibitor trigonelline and AKT inhibitor IMQ revealed that GF1 lacked antioxidant and anti-apoptotic effects. Conclusion: In conclusion, GF1 was found to alleviate THP-induced cardiotoxicity via modulating Nrf2 and AKT/Bcl-2 signaling pathways, ultimately alleviating myocardial oxidative stress and apoptosis.