1.Porphyromonas gingivalis bacteremia increases the permeability of the blood-brain barrier via the Mfsd2a/Caveolin-1 mediated transcytosis pathway.
Shuang LEI ; Jian LI ; Jingjun YU ; Fulong LI ; Yaping PAN ; Xu CHEN ; Chunliang MA ; Weidong ZHAO ; Xiaolin TANG
International Journal of Oral Science 2023;15(1):3-3
		                        		
		                        			
		                        			Bacteremia induced by periodontal infection is an important factor for periodontitis to threaten general health. P. gingivalis DNA/virulence factors have been found in the brain tissues from patients with Alzheimer's disease (AD). The blood-brain barrier (BBB) is essential for keeping toxic substances from entering brain tissues. However, the effect of P. gingivalis bacteremia on BBB permeability and its underlying mechanism remains unclear. In the present study, rats were injected by tail vein with P. gingivalis three times a week for eight weeks to induce bacteremia. An in vitro BBB model infected with P. gingivalis was also established. We found that the infiltration of Evans blue dye and Albumin protein deposition in the rat brain tissues were increased in the rat brain tissues with P. gingivalis bacteremia and P. gingivalis could pass through the in vitro BBB model. Caveolae were detected after P. gingivalis infection in BMECs both in vivo and in vitro. Caveolin-1 (Cav-1) expression was enhanced after P. gingivalis infection. Downregulation of Cav-1 rescued P. gingivalis-enhanced BMECs permeability. We further found P. gingivalis-gingipain could be colocalized with Cav-1 and the strong hydrogen bonding between Cav-1 and arg-specific-gingipain (RgpA) were detected. Moreover, P. gingivalis significantly inhibited the major facilitator superfamily domain containing 2a (Mfsd2a) expression. Mfsd2a overexpression reversed P. gingivalis-increased BMECs permeability and Cav-1 expression. These results revealed that Mfsd2a/Cav-1 mediated transcytosis is a key pathway governing BBB BMECs permeability induced by P. gingivalis, which may contribute to P. gingivalis/virulence factors entrance and the subsequent neurological impairments.
		                        		
		                        		
		                        		
		                        			Animals
		                        			;
		                        		
		                        			Rats
		                        			;
		                        		
		                        			Bacteremia/metabolism*
		                        			;
		                        		
		                        			Blood-Brain Barrier/microbiology*
		                        			;
		                        		
		                        			Caveolin 1/metabolism*
		                        			;
		                        		
		                        			Gingipain Cysteine Endopeptidases/metabolism*
		                        			;
		                        		
		                        			Permeability
		                        			;
		                        		
		                        			Porphyromonas gingivalis/pathogenicity*
		                        			;
		                        		
		                        			Transcytosis
		                        			;
		                        		
		                        			Virulence Factors/metabolism*
		                        			
		                        		
		                        	
2.Porphyromonas gingivalis, a periodontal pathogen, impairs post-infarcted myocardium by inhibiting autophagosome-lysosome fusion.
Yuka SHIHEIDO-WATANABE ; Yasuhiro MAEJIMA ; Shun NAKAGAMA ; Qintao FAN ; Natsuko TAMURA ; Tetsuo SASANO
International Journal of Oral Science 2023;15(1):42-42
		                        		
		                        			
		                        			While several previous studies have indicated the link between periodontal disease (PD) and myocardial infarction (MI), the underlying mechanisms remain unclear. Autophagy, a cellular quality control process that is activated in several diseases, including heart failure, can be suppressed by Porphyromonas gingivalis (P.g.). However, it is uncertain whether autophagy impairment by periodontal pathogens stimulates the development of cardiac dysfunction after MI. Thus, this study aimed to investigate the relationship between PD and the development of MI while focusing on the role of autophagy. Neonatal rat cardiomyocytes (NRCMs) and MI model mice were inoculated with wild-type P.g. or gingipain-deficient P.g. to assess the effect of autophagy inhibition by P.g. Wild-type P.g.-inoculated NRCMs had lower cell viability than those inoculated with gingipain-deficient P.g. This study also revealed that gingipains can cleave vesicle-associated membrane protein 8 (VAMP8), a protein involved in lysosomal sensitive factor attachment protein receptors (SNAREs), at the 47th lysine residue, thereby inhibiting autophagy. Wild-type P.g.-inoculated MI model mice were more susceptible to cardiac rupture, with lower survival rates and autophagy activity than gingipain-deficient P.g.-inoculated MI model mice. After inoculating genetically modified MI model mice (VAMP8-K47A) with wild-type P.g., they exhibited significantly increased autophagy activation compared with the MI model mice inoculated with wild-type P.g., which suppressed cardiac rupture and enhanced overall survival rates. These findings suggest that gingipains, which are virulence factors of P.g., impair the infarcted myocardium by cleaving VAMP8 and disrupting autophagy. This study confirms the strong association between PD and MI and provides new insights into the potential role of autophagy in this relationship.
		                        		
		                        		
		                        		
		                        			Mice
		                        			;
		                        		
		                        			Rats
		                        			;
		                        		
		                        			Animals
		                        			;
		                        		
		                        			Porphyromonas gingivalis
		                        			;
		                        		
		                        			Gingipain Cysteine Endopeptidases
		                        			;
		                        		
		                        			Autophagosomes
		                        			;
		                        		
		                        			Myocardium
		                        			;
		                        		
		                        			Periodontal Diseases
		                        			;
		                        		
		                        			Heart Rupture
		                        			
		                        		
		                        	
3.SENP2-mediated SERCA2a deSUMOylation increases calcium overload in cardiomyocytes to aggravate myocardial ischemia/reperfusion injury.
Yuanyuan LUO ; Shuaishuai ZHOU ; Tao XU ; Wanling WU ; Pingping SHANG ; Shuai WANG ; Defeng PAN ; Dongye LI
Chinese Medical Journal 2023;136(20):2496-2507
		                        		
		                        			BACKGROUND:
		                        			Sarcoplasmic reticulum calcium ATPase 2a (SERCA2a) is a key protein that maintains myocardial Ca 2+ homeostasis. The present study aimed to investigate the mechanism underlying the SERCA2a-SUMOylation (small ubiquitin-like modifier) process after ischemia/reperfusion injury (I/RI) in vitro and in vivo .
		                        		
		                        			METHODS:
		                        			Calcium transient and systolic/diastolic function of cardiomyocytes isolated from Serca2a knockout (KO) and wild-type mice with I/RI were compared. SUMO-relevant protein expression and localization were detected by quantitative real-time PCR (RT-qPCR), Western blotting, and immunofluorescence in vitro and in vivo . Serca2a-SUMOylation, infarct size, and cardiac function of Senp1 or Senp2 overexpressed/suppressed adenovirus infected cardiomyocytes, were detected by immunoprecipitation, triphenyltetrazolium chloride (TTC)-Evans blue staining, and echocardiography respectively.
		                        		
		                        			RESULTS:
		                        			The results showed that the changes of Fura-2 fluorescence intensity and contraction amplitude of cardiomyocytes decreased in the I/RI groups and were further reduced in the Serca2a KO + I/RI groups. Senp1 and Senp2 messenger ribose nucleic acid (mRNA) and protein expression levels in vivo and in cardiomyocytes were highest at 6 h and declined at 12 h after I/RI. However, the highest levels in HL-1 cells were recorded at 12 h. Senp2 expression increased in the cytoplasm, unlike that of Senp1. Inhibition of Senp2 protein reversed the I/RI-induced Serca2a-SUMOylation decline, reduced the infarction area, and improved cardiac function, while inhibition of Senp1 protein could not restore the above indicators.
		                        		
		                        			CONCLUSION
		                        			I/RI activated Senp1 and Senp2 protein expression, which promoted Serca2a-deSUMOylation, while inhibition of Senp2 expression reversed Serca2a-SUMOylation and improved cardiac function.
		                        		
		                        		
		                        		
		                        			Animals
		                        			;
		                        		
		                        			Mice
		                        			;
		                        		
		                        			Calcium/metabolism*
		                        			;
		                        		
		                        			Cysteine Endopeptidases/metabolism*
		                        			;
		                        		
		                        			Myocardial Reperfusion Injury/metabolism*
		                        			;
		                        		
		                        			Myocardium/metabolism*
		                        			;
		                        		
		                        			Myocytes, Cardiac/metabolism*
		                        			;
		                        		
		                        			Proteins/metabolism*
		                        			;
		                        		
		                        			Sarcoplasmic Reticulum Calcium-Transporting ATPases/genetics*
		                        			
		                        		
		                        	
4.Effects of deleting peptidoglycan hydrolase genes on the viable cell counts of Bacillus amyloliquefaciens and the yield of alkaline protease.
Xiaojian XU ; Baoyue ZHU ; Xinyue LI ; Jinfang ZHANG ; Wenlong LIU ; Fuping LU ; Yu LI
Chinese Journal of Biotechnology 2022;38(4):1506-1517
		                        		
		                        			
		                        			In order to explore the effect of peptidoglycan hydrolase on the viable cell counts of Bacillus amyloliquefaciens and the yield of alkaline protease, five peptidoglycan hydrolase genes (lytC, lytD, lytE, lytF and lytG) of B. amyloliquefaciens TCCC111018 were knocked out individually. The viable cell counts of the bacteria and their alkaline protease activities before and after gene deletion were determined. The viable cell counts of the knockout mutants BA ΔlytC and BA ΔlytE achieved 1.67×106 CFU/mL and 1.44×106 CFU/mL respectively after cultivation for 60 h, which were 32.5% and 14.3% higher than that of the control strain BA Δupp. Their alkaline protease activities reached 20 264 U/mL and 17 265 U/mL, respectively, which were 43.1% and 27.3% higher than that of the control strain. The results showed that deleting some of the peptidoglycan hydrolase genes effectively maintained the viable cell counts of bacteria and increased the activity of extracellular enzymes, which may provide a new idea for optimization of the microbial host for production of industrial enzymes.
		                        		
		                        		
		                        		
		                        			Bacillus amyloliquefaciens/genetics*
		                        			;
		                        		
		                        			Bacterial Proteins
		                        			;
		                        		
		                        			Cell Count
		                        			;
		                        		
		                        			Endopeptidases/genetics*
		                        			;
		                        		
		                        			N-Acetylmuramoyl-L-alanine Amidase/genetics*
		                        			
		                        		
		                        	
5.Bioactive compounds of Jingfang Granules against SARS-CoV-2 virus proteases 3CLpro and PLpro.
Zhan Peng SHANG ; Yang YI ; Rong YU ; Jing Jing FAN ; Yi Xi HUANG ; Xue QIAO ; Min YE
Journal of Peking University(Health Sciences) 2022;54(5):907-919
		                        		
		                        			OBJECTIVE:
		                        			Jingfang Granules have been recommended for the prevention and treatment of corona virus disease 2019 (COVID-19). Through chemical analysis and bioactivity evaluation, this study aims to elucidate the potential effective components of Jingfang Granules.
		                        		
		                        			METHODS:
		                        			The inhibitory acti-vities of Jingfang Granules extract against 3-chymotrypsin-like protease (3CLpro), papain like protease (PLpro), spike protein receptor-binding domain (S-RBD) and human cyclooxygenase-2 (COX-2) were evaluated using enzyme assay. The antitussive effects were evaluated using the classical ammonia-induced cough model. The chemical constituents of Jingfang Granules were qualitatively and quantitatively analyzed by liquid chromatography-mass spectrometry (LC/MS). The 3CLpro and PLpro inhibitory activities of the major compounds were determined by enzyme assay, molecular docking, and site-directed mutagenesis.
		                        		
		                        			RESULTS:
		                        			Jingfang Granules exhibited 3CLpro and PLpro inhibitory activities, as well as COX-2 inhibitory and antitussive activities. By investigating the MS/MS behaviors of reference standards, a total of fifty-six compounds were characterized in Jingfang Granules. Sixteen of them were unambiguously identified by comparing with reference standards. The contents of the 16 major compounds were also determined, and their total contents were 2 498.8 μg/g. Naringin, nodakenin and neohesperidin were three dominating compounds in Jingfang Granules, and their contents were 688.8, 596.4 and 578.7 μg/g, respectively. In addition, neohesperidin and naringin exhibited PLpro inhibitory activities, and the inhibition rates at 8 μmol/L were 53.5% and 46.1%, respectively. Prim-O-glucosylcimifugin showed significant inhibitory activities against 3CLpro and PLpro, and the inhibitory rates at 8 μmol/L were 76.8% and 78.2%, respectively. Molecular docking indicated that hydrogen bonds could be formed between prim-O-glucosylcimifugin and amino acid residues H163, E166, Q192, T190 of 3CLpro (binding energy, -7.7 kcal/mol) and K157, D164, R166, E167, T301 of PLpro(-7.3 kcal/mol), respectively. Site-directed mutagenesis indicated amino acid residue K157 was a key active site for the interaction between prim-O-glucosylcimifugin and PLpro.
		                        		
		                        			CONCLUSION
		                        			Prim-O-glucosylcimifugin, neohesperidin, and naringin as the major compounds from Jingfang Granules could inhibit severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) virus proteases 3CLpro and PLpro. The results are valuable for rational clinical use of Jingfang Granules.
		                        		
		                        		
		                        		
		                        			Amino Acids
		                        			;
		                        		
		                        			Ammonia
		                        			;
		                        		
		                        			Antitussive Agents
		                        			;
		                        		
		                        			COVID-19
		                        			;
		                        		
		                        			Chymases
		                        			;
		                        		
		                        			Coronavirus 3C Proteases
		                        			;
		                        		
		                        			Cyclooxygenase 2
		                        			;
		                        		
		                        			Cyclooxygenase 2 Inhibitors
		                        			;
		                        		
		                        			Cysteine Endopeptidases/metabolism*
		                        			;
		                        		
		                        			Humans
		                        			;
		                        		
		                        			Molecular Docking Simulation
		                        			;
		                        		
		                        			Papain
		                        			;
		                        		
		                        			Peptide Hydrolases
		                        			;
		                        		
		                        			SARS-CoV-2
		                        			;
		                        		
		                        			Spike Glycoprotein, Coronavirus
		                        			;
		                        		
		                        			Tandem Mass Spectrometry
		                        			
		                        		
		                        	
6.SENP1 induced protein deSUMO modification increased the chemotherapy sensitivity of endometrial cancer side population cells.
Meng Lan YUAN ; Jie BAI ; Cheng Yao LI ; Na XUE ; Xu Hong CHEN ; Feng SHENG ; Xiao Zhi LIU ; Pu LI
Chinese Journal of Oncology 2022;44(12):1362-1368
		                        		
		                        			
		                        			Objective: To inhibit the stemness maintenance potential of endometrial cancer and increase the sensitivity of endometrial cancer side population cells to chemotherapy drugs by inducing extensive deSUMOylation modification of proteins. Methods: Flow cytometry was used to sort and culture CD133(+) CD44(+) KLE endometrial cancer cell clone spheres. Protein expression level of small ubiquitin-related modifier 1 (SUMO1) and two stemness maintenance genes of tumor side population cells, octamer binding transcription factor-4 (Oct4) and sex determining region Y-box2 (Sox2), were detected by western blotting method. Lentivirus-mediated Sentrin/SUMO-specific proteases 1 (SENP1) gene was stably transfected into KLE side population cells. Western blotting was used to detect the protein expressions of SENP1, SUMO1, Oct4 and Sox2. The clone formation rate was compared between KLE side population cells with or without SENP1 overexpression. Flow cytometry was applied to detect cell cycle changes. 3-(4, 5-Dimethylthiazole-2)-2, 5-diphenyl-tetrazolium bromide (MTT) experiment and flow cytometry apoptosis method were used to detect the chemosensitivity of the side population of endometrial cancer cells to cisplatin. Tumor-bearing mouse models of endometrial cancer were established to detect the effect of SENP1 overexpression on the chemotherapy sensitivity of cisplatin. Results: Compared with CD133(-)CD44(-) KLE cells, CD133(+) CD44(+) KLE side population cells could form clonal spheres and express higher levels of SUMO1, Oct4 and Sox2 proteins (P<0.05). Compared with KLE side population cells that were not transfected with SENP1 gene, the expression level of SENP1 protein in KLE side population cells overexpressing SUMO1、Oct4 and Sox2 were lower. The clonal sphere formation rate was reduced from (25.67±5.44)% to (7.46±1.42)%, and cell cycle shifted from G(0)/G(1) phase to G(2) phase. IC(50) of cisplatin decreased from (55.46±6.14) μg/ml to (11.55±3.12) μg/ml, and cell apoptosis rate increased from (9.76±2.09)% to (16.79±3.44)%. Overexpression of SENP1 could reduce the tumorigenesis rate of KLE side population cells in vivo and increase their chemotherapy sensitivity to cisplatin (P<0.05). Conclusion: Overexpression of SENP1 can induce protein deSUMOylation modification, inhibit the stemness maintenance potential of endometrial cancer side population cells, and enhance their chemotherapy sensitivity, which provides a new reference for gene therapy of endometrial cancer.
		                        		
		                        		
		                        		
		                        			Animals
		                        			;
		                        		
		                        			Female
		                        			;
		                        		
		                        			Humans
		                        			;
		                        		
		                        			Mice
		                        			;
		                        		
		                        			Apoptosis
		                        			;
		                        		
		                        			Cell Line, Tumor
		                        			;
		                        		
		                        			Cisplatin/pharmacology*
		                        			;
		                        		
		                        			Cysteine Endopeptidases/metabolism*
		                        			;
		                        		
		                        			Endometrial Neoplasms/genetics*
		                        			;
		                        		
		                        			Side-Population Cells/pathology*
		                        			;
		                        		
		                        			Sumoylation
		                        			
		                        		
		                        	
7.Expression, purification, and characterization of cell-permeable fusion antioxidant enzyme sensitive to matrix metalloproteinases-2/9.
Huocong HE ; Lixiang LIN ; Lingling LI ; Lunqiao WU ; Haiying LIN ; Jianru PAN
Chinese Journal of Biotechnology 2022;38(9):3515-3527
		                        		
		                        			
		                        			Antioxidant enzymes fused with cell-penetrating peptides could enter cells and protect cells from irradiation damage. However, the unselective transmembrane ability of cell-penetrating peptide may also bring antioxidant enzymes into tumor cells, thus protecting tumor cells and consequently reducing the efficacy of radiotherapy. There are active matrix metalloproteinase (MMP)-2 or MMP-9 in most tumor cellular microenvironments. Therefore, a fusion protein containing an MMP-2/9 cleavable substrate peptide X, a cell-penetrating peptide R9, a glutathione S-transferase (GST), and a human Cu, Zn superoxide dismutase (SOD1), was designed and named GST-SOD1-X-R9. In the tumor microenvironment, GST-SOD1-X-R9 would lose its cell-penetrating peptide and could not enter tumor cells due to the cleavage of substrate X by active MMP-2/9, thereby achieving selected entering normal cells. The complete nucleotide sequence of SOD1-X-R9 was synthesized and inserted into the prokaryotic expression vector pGEX-4T-1. The pGEX4T-1-SOD1-X-R9 recombinant plasmid was obtained, and soluble expression of the fusion protein was achieved. GST-SOD1-X-R9 was purified by ammonium sulfate precipitation and GST affinity chromatography. The molecular weight of the fusion protein was approximately 47 kDa, consistent with the theoretical value. The SOD and GST activities were 2 954 U/mg and 328 U/mg, respectively. Stability test suggested that almost no change in either SOD activity or GST activity of GST-SOD1-X-R9 was observed under physiological conditions. The fusion protein could be partially digested by collagenase Ⅳ in solution. Subsequently, the effect of MMP-2/9 activity on transmembrane ability of the fusion protein was tested using 2D and 3D cultured HepG2 cells. Little extracellular MMP-2 activity of HepG2 cells was observed under 2D culture condition. While under the 3D culture model, the size and the MMP-2 activity of the HepG2 tumor spheroid increased daily. GST-SOD1-R9 proteins showed the same transmembrane efficiency in 2D cultured HepG2 cells, but the transmembrane efficiency of GST-SOD1-X-R9 in 3D cultured HepG2 spheres was reduced remarkably. This study provided a basis for further investigating the selectively protective effect of GST-SOD1-X-R9 against oxidative damage in normal cells.
		                        		
		                        		
		                        		
		                        			Ammonium Sulfate
		                        			;
		                        		
		                        			Antioxidants
		                        			;
		                        		
		                        			Cell-Penetrating Peptides/pharmacology*
		                        			;
		                        		
		                        			Endopeptidases
		                        			;
		                        		
		                        			Glutathione Transferase/metabolism*
		                        			;
		                        		
		                        			Humans
		                        			;
		                        		
		                        			Matrix Metalloproteinase 2/genetics*
		                        			;
		                        		
		                        			Matrix Metalloproteinase 9/genetics*
		                        			;
		                        		
		                        			Recombinant Fusion Proteins
		                        			;
		                        		
		                        			Recombinant Proteins
		                        			;
		                        		
		                        			Superoxide Dismutase/metabolism*
		                        			;
		                        		
		                        			Superoxide Dismutase-1
		                        			
		                        		
		                        	
8.Quantitative proteomics reveal the potential biological functions of the deubiquitinating enzyme Ubp14 in Saccharomyces cerevisiae.
Zhaodi LI ; Qiuyan LAN ; Yanchang LI ; Cong XU ; Lei CHANG ; Ping XU ; Changzhu DUAN
Chinese Journal of Biotechnology 2022;38(10):3901-3913
		                        		
		                        			
		                        			Ubiquitination is one of the reversible protein post-translational modifications, in which ubiquitin molecules bind to the target protein in a cascade reaction of ubiquitin activating enzymes, ubiquitin conjugating enzymes, and ubiquitin ligases. The deubiquitinating enzymes (DUBs) remove ubiquitin residues from the substrates, which play key roles in the formation of mature ubiquitin, the removal and trimming of ubiquitin chains, as well as the recycling of free ubiquitin chains. Ubp14, a member of the ubiquitin specific proteases family in Saccharomyces cerevisiae, is mainly responsible for the recycling of intracellular free ubiquitin chains. To investigate its global biological function, a ubp14∆ mutant was constructed by homologous recombination technique. The growth rate of ubp14∆ mutant was lower than that of the wild-type (WT) strain. Using stable isotope labeling by amino acids in cell culture (SILAC) combined with deep coverage proteomics analysis, the differentially expressed proteins of ubp14∆ mutant relative to the wild-type strain were systematically analyzed. A total of 3 685 proteins were identified in this study, and 109 differentially expressed proteins were filtered out by statistical analysis. Gene ontology analysis found that differentially expressed proteins caused by Ubp14 loss were mainly involved in amino acid metabolism, REDOX, heat shock stress and etc, which shed light on the broad biological function of this DUB. This study provides highly reliable proteomic data for further exploring the biological functions of the deubiquitination enzyme Ubp14, and further understanding the relationship between the free ubiquitin homeostasis and biological process regulation.
		                        		
		                        		
		                        		
		                        			Saccharomyces cerevisiae/metabolism*
		                        			;
		                        		
		                        			Proteomics
		                        			;
		                        		
		                        			Endopeptidases/metabolism*
		                        			;
		                        		
		                        			Ubiquitin/metabolism*
		                        			;
		                        		
		                        			Ubiquitination
		                        			;
		                        		
		                        			Proteins/metabolism*
		                        			;
		                        		
		                        			Deubiquitinating Enzymes/metabolism*
		                        			;
		                        		
		                        			Biological Phenomena
		                        			
		                        		
		                        	
9.Key active sites of proteases and protease inhibitors: a review.
Jie ZHANG ; Xi YANG ; Youshan LI
Chinese Journal of Biotechnology 2021;37(2):561-579
		                        		
		                        			
		                        			Proteases are widely found in organisms participating in the decomposition of proteins to maintain the organisms' normal life activities. Protease inhibitors regulate the activities of target proteases by binding to their active sites, thereby affecting protein metabolism. The key amino acid mutations in proteases and protease inhibitors can affect their physiological functions, stability, catalytic activity, and inhibition specificity. More active, stable, specific, environmentally friendly and cheap proteases and protease inhibitors might be obtained by excavating various natural mutants of proteases and protease inhibitors, analyzing their key active sites by using protein engineering methods. Here, we review the studies on proteases' key active sites and protease inhibitors to deepen the understanding of the active mechanism of proteases and their inhibitors.
		                        		
		                        		
		                        		
		                        			Binding Sites
		                        			;
		                        		
		                        			Catalytic Domain
		                        			;
		                        		
		                        			Endopeptidases
		                        			;
		                        		
		                        			Peptide Hydrolases/genetics*
		                        			;
		                        		
		                        			Protease Inhibitors
		                        			;
		                        		
		                        			Proteins
		                        			
		                        		
		                        	
10.Optimization of expression conditions and determination the proteolytic activity of codon-optimized SARS-CoV-2 main protease in Escherichia coli.
Yunyu CHEN ; Zhenghao FU ; Gangan YAN ; Yuan LIN ; Xiaoping LIU
Chinese Journal of Biotechnology 2021;37(4):1334-1345
		                        		
		                        			
		                        			The main protease (Mpro) of SARS-CoV-2 is a highly conserved and mutation-resistant coronaviral enzyme, which plays a pivotal role in viral replication, making it an ideal target for the development of novel broad-spectrum anti-coronaviral drugs. In this study, a codon-optimized Mpro gene was cloned into pET-21a and pET-28a expression vectors. The recombinant plasmids were transformed into E. coli Rosetta(DE3) competent cells and the expression conditions were optimized. The highly expressed recombinant proteins, Mpro and Mpro-28, were purified by HisTrapTM chelating column and its proteolytic activity was determined by a fluorescence resonance energy transfer (FRET) assay. The FRET assay showed that Mpro exhibits a desirable proteolytic activity (25 000 U/mg), with Km and kcat values of 11.68 μmol/L and 0.037/s, respectively. The specific activity of Mpro is 25 times that of Mpro-28, a fusion protein carrying a polyhistidine tag at the N and C termini, indicating additional residues at the N terminus of Mpro, but not at the C terminus, are detrimental to its proteolytic activity. The preparation of active SARS-CoV-2 Mpro through codon-optimization strategy might facilitate the development of the rapid screening assays for the discovery of broad-spectrum anti-coronaviral drugs targeting Mpro.
		                        		
		                        		
		                        		
		                        			COVID-19
		                        			;
		                        		
		                        			Codon/genetics*
		                        			;
		                        		
		                        			Cysteine Endopeptidases/genetics*
		                        			;
		                        		
		                        			Escherichia coli/genetics*
		                        			;
		                        		
		                        			Humans
		                        			;
		                        		
		                        			Peptide Hydrolases
		                        			;
		                        		
		                        			SARS-CoV-2
		                        			;
		                        		
		                        			Viral Nonstructural Proteins/genetics*
		                        			
		                        		
		                        	
            
Result Analysis
Print
Save
E-mail