Volume 8, Issue 4 (Vol.8 No.4 Jan 2020)                   rbmb.net 2020, 8(4): 347-357 | Back to browse issues page

PMID: 32582792

XML Print


National Center for Biotechnology,Kurgalzhyn road, 13/5, Astana, 010000, Kazakhstan & L.N.GumilyovEurasian National University, Satpayev st., 2, Astana, 010008, Kazakhstan.
Abstract:   (3399 Views)
Background: The programmed cell deathprotein 1 (PD-1), which is a member of the CD28 receptor family, can negatively regulate antitumor immune responses by interacting with its ligands, PD-L1 or PD-L2. The PD-1–PD-L1 signaling pathway is a checkpoint mechanism that plays essential roles in downregulating immune responses in cancerous tissues. Thus, blocking this signaling pathway leads to enhanced antitumor immunity, potentially preventing tumor progression.
Methods: We synthesized the extracellular domain of the PD-1 receptor (rPD-1) de novoby using a two-step polymerase chain reaction and the Phusion® DNA polymerase. The synthesized gene was cloned into the pET28 expression plasmid and transformed into competent Escherichia coli. Purification of rPD-1 was performed by metal-affinity chromatography, using a HisTrap column.Purified rPD-1 was characterized by western blotting and mass spectrometry using the SwissProt database and the Mascot program.
Results: Designed and synthesized construct of rPD-1 was 500 bpin size. Analysis of the electrophoresis data of purified rPD-1 showed the presence of a protein with a molecular mass of 21 kDa. Mass spectrometry data using the SwissProt database and the Mascot program outputted the highest-scoring sequence to correspond to rPD-1.
Conclusions: Synthesized de novo rPD-1 may have potential therapeutic applications in enhancing antitumor immune responses
Full-Text [PDF 557 kb]   (2689 Downloads)    
Type of Article: Original Article | Subject: Molecular Biology
Received: 2019/08/14 | Accepted: 2019/08/29 | Published: 2020/05/10

Rights and permissions
Creative Commons License This work is licensed under a Creative Commons Attribution-NonCommercial 4.0 International License.