Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 48.198
Filtrer
1.
Arch Microbiol ; 206(7): 336, 2024 Jul 02.
Article de Anglais | MEDLINE | ID: mdl-38954047

RÉSUMÉ

Wild-type Lactococcus lactis strain LAC460 secretes prophage-encoded bacteriocin-like lysin LysL, which kills some Lactococcus strains, but has no lytic effect on the producer. LysL carries two N-terminal enzymatic active domains (EAD), and an unknown C-terminus without homology to known domains. This study aimed to determine whether the C-terminus of LysL carries a cell wall binding domain (CBD) for target specificity of LysL. The C-terminal putative CBD region of LysL was fused with His-tagged green fluorescent protein (HGFPuv). The HGFPuv_CBDlysL gene fusion was ligated into the pASG-IBA4 vector, and introduced into Escherichia coli. The fusion protein was produced and purified with affinity chromatography. To analyse the binding of HGFPuv_CBDLysL to Lactococcus cells, the protein was mixed with LysL-sensitive and LysL-resistant strains, including the LysL-producer LAC460, and the fluorescence of the cells was analysed. As seen in fluorescence microscope, HGFPuv_CBDLysL decorated the cell surface of LysL-sensitive L. cremoris MG1614 with green fluorescence, whereas the resistant L. lactis strains LM0230 and LAC460 remained unfluorescent. The fluorescence plate reader confirmed the microscopy results detecting fluorescence only from four tested LysL-sensitive strains but not from 11 tested LysL-resistant strains. Specific binding of HGFPuv_CBDLysL onto the LysL-sensitive cells but not onto the LysL-resistant strains indicates that the C-terminus of LysL contains specific CBD. In conclusion, this report presents experimental evidence of the presence of a CBD in a lactococcal phage lysin. Moreover, the inability of HGFPuv_CBDLysL to bind to the LysL producer LAC460 may partly explain the host's resistance to its own prophage lysin.


Sujet(s)
Bactériocines , Paroi cellulaire , Lactococcus lactis , Lactococcus lactis/génétique , Lactococcus lactis/métabolisme , Paroi cellulaire/métabolisme , Bactériocines/métabolisme , Bactériocines/génétique , Bactériocines/composition chimique , Escherichia coli/génétique , Escherichia coli/métabolisme , Protéines à fluorescence verte/génétique , Protéines à fluorescence verte/métabolisme , Domaines protéiques , Protéines de fusion recombinantes/génétique , Protéines de fusion recombinantes/métabolisme , Protéines de fusion recombinantes/composition chimique , Liaison aux protéines
2.
J Biotechnol ; 391: 57-63, 2024 Aug 10.
Article de Anglais | MEDLINE | ID: mdl-38851397

RÉSUMÉ

Antigen-presenting cells (APCs) play an important role in virus infection control by bridging innate and adaptive immune responses. Macrophages and dendritic cells (DCs) possess various surface receptors to recognize/internalize antigens, and antibody binding can enhance pathogen-opsonizing uptake by these APCs via interaction of antibody fragment crystallizable (Fc) domains with Fc receptors, evoking profound pathogen control in certain settings. Here, we examined phagocytosis-enhancing potential of Fc domains directly oriented on a retroviral virion/virus-like particle (VLP) surface. We generated an expression vector coding a murine Fc fragment fused to the transmembrane region (TM) of a retroviral envelope protein, deriving expression of the Fc-TM fusion protein on the transfected cell surface and production of virions incorporating the chimeric Fc upon co-transfection. Incubation of Fc-displaying simian immunodeficiency virus (SIV) with murine J774 macrophages and bone marrow-derived DCs derived Fc receptor-dependent enhanced uptake, being visualized by imaging cytometry. Alternative preparation of a murine leukemia virus (MLV) backbone-based Fc-displaying VLP loading an influenza virus hemagglutinin (HA) antigen resulted in enhanced HA internalization by macrophages, stating antigen compatibility of the design. Results show that the Fc-TM fusion molecule can be displayed on certain viruses/VLPs and may be utilized as a molecular adjuvant to facilitate APC antigen uptake.


Sujet(s)
Cellules présentatrices d'antigène , Cellules dendritiques , Fragments Fc des immunoglobulines , Virion , Animaux , Souris , Fragments Fc des immunoglobulines/génétique , Fragments Fc des immunoglobulines/métabolisme , Fragments Fc des immunoglobulines/immunologie , Cellules présentatrices d'antigène/immunologie , Cellules présentatrices d'antigène/métabolisme , Virion/métabolisme , Virion/génétique , Cellules dendritiques/immunologie , Cellules dendritiques/métabolisme , Macrophages/métabolisme , Macrophages/immunologie , Protéines de fusion recombinantes/génétique , Protéines de fusion recombinantes/métabolisme , Protéines de fusion recombinantes/immunologie , Virus de l'immunodéficience simienne/immunologie , Virus de l'immunodéficience simienne/génétique , Glycoprotéine hémagglutinine du virus influenza/immunologie , Glycoprotéine hémagglutinine du virus influenza/génétique , Glycoprotéine hémagglutinine du virus influenza/métabolisme , Lignée cellulaire , Virus de la leucémie murine/génétique , Phagocytose , Humains
3.
Protein Eng Des Sel ; 372024 Jan 29.
Article de Anglais | MEDLINE | ID: mdl-38836499

RÉSUMÉ

Protein developability is requisite for use in therapeutic, diagnostic, or industrial applications. Many developability assays are low throughput, which limits their utility to the later stages of protein discovery and evolution. Recent approaches enable experimental or computational assessment of many more variants, yet the breadth of applicability across protein families and developability metrics is uncertain. Here, three library-scale assays-on-yeast protease, split green fluorescent protein (GFP), and non-specific binding-were evaluated for their ability to predict two key developability outcomes (thermal stability and recombinant expression) for the small protein scaffolds affibody and fibronectin. The assays' predictive capabilities were assessed via both linear correlation and machine learning models trained on the library-scale assay data. The on-yeast protease assay is highly predictive of thermal stability for both scaffolds, and the split-GFP assay is informative of affibody thermal stability and expression. The library-scale data was used to map sequence-developability landscapes for affibody and fibronectin binding paratopes, which guides future design of variants and libraries.


Sujet(s)
Fibronectines , Protéines de fusion recombinantes , Fibronectines/composition chimique , Fibronectines/génétique , Fibronectines/métabolisme , Protéines de fusion recombinantes/génétique , Protéines de fusion recombinantes/composition chimique , Protéines de fusion recombinantes/métabolisme , Protéines à fluorescence verte/génétique , Protéines à fluorescence verte/composition chimique , Protéines à fluorescence verte/métabolisme , Ingénierie des protéines/méthodes , Banque de peptides , Stabilité protéique , Liaison aux protéines , Humains
4.
Proc Natl Acad Sci U S A ; 121(25): e2312499121, 2024 Jun 18.
Article de Anglais | MEDLINE | ID: mdl-38857395

RÉSUMÉ

Ex vivo expansion of human CD34+ hematopoietic stem and progenitor cells remains a challenge due to rapid differentiation after detachment from the bone marrow niche. In this study, we assessed the capacity of an inducible fusion protein to enable sustained ex vivo proliferation of hematopoietic precursors and their capacity to differentiate into functional phagocytes. We fused the coding sequences of an FK506-Binding Protein 12 (FKBP12)-derived destabilization domain (DD) to the myeloid/lymphoid lineage leukemia/eleven nineteen leukemia (MLL-ENL) fusion gene to generate the fusion protein DD-MLL-ENL and retrovirally expressed the protein switch in human CD34+ progenitors. Using Shield1, a chemical inhibitor of DD fusion protein degradation, we established large-scale and long-term expansion of late monocytic precursors. Upon Shield1 removal, the cells lost self-renewal capacity and spontaneously differentiated, even after 2.5 y of continuous ex vivo expansion. In the absence of Shield1, stimulation with IFN-γ, LPS, and GM-CSF triggered terminal differentiation. Gene expression analysis of the obtained phagocytes revealed marked similarity with naïve monocytes. In functional assays, the novel phagocytes migrated toward CCL2, attached to VCAM-1 under shear stress, produced reactive oxygen species, and engulfed bacterial particles, cellular particles, and apoptotic cells. Finally, we demonstrated Fcγ receptor recognition and phagocytosis of opsonized lymphoma cells in an antibody-dependent manner. Overall, we have established an engineered protein that, as a single factor, is useful for large-scale ex vivo production of human phagocytes. Such adjustable proteins have the potential to be applied as molecular tools to produce functional immune cells for experimental cell-based approaches.


Sujet(s)
Différenciation cellulaire , Phagocytes , Humains , Phagocytes/métabolisme , Cellules souches hématopoïétiques/métabolisme , Protéines de fusion oncogènes/génétique , Protéines de fusion oncogènes/métabolisme , Protéines de fusion recombinantes/métabolisme , Protéines de fusion recombinantes/génétique , Protéine de la leucémie myéloïde-lymphoïde/métabolisme , Protéine de la leucémie myéloïde-lymphoïde/génétique , Leucémies/génétique , Leucémies/anatomopathologie , Leucémies/métabolisme , Ingénierie des protéines/méthodes , Phagocytose
5.
Int J Mol Sci ; 25(12)2024 Jun 08.
Article de Anglais | MEDLINE | ID: mdl-38928060

RÉSUMÉ

At present, the magnetic selection of genetically modified cells is mainly performed with surface markers naturally expressed by cells such as CD4, LNGFR (low affinity nerve growth factor receptor), and MHC class I molecule H-2Kk. The disadvantage of such markers is the possibility of their undesired and poorly predictable expression by unmodified cells before or after cell manipulation, which makes it essential to develop new surface markers that would not have such a drawback. Earlier, modified CD52 surface protein variants with embedded HA and FLAG epitope tags (CD52/FLAG and CD52/HA) were developed by the group of Dr. Mazurov for the fluorescent cell sorting of CRISPR-modified cells. In the current study, we tested whether these markers can be used for the magnetic selection of transduced cells. For this purpose, appropriate constructs were created in MigR1-based bicistronic retroviral vectors containing EGFP and DsRedExpress2 as fluorescent reporters. Cytometric analysis of the transduced NIH 3T3 cell populations after magnetic selection evaluated the efficiency of isolation and purity of the obtained populations, as well as the change in the median fluorescence intensity (MFI). The results of this study demonstrate that the surface markers CD52/FLAG and CD52/HA can be effectively used for magnetic cell selection, and their efficiencies are comparable to that of the commonly used LNGFR marker. At the same time, the significant advantage of these markers is the absence of HA and FLAG epitope sequences in cellular proteins, which rules out the spurious co-isolation of negative cells.


Sujet(s)
Antigène CD52 , Protéines de fusion recombinantes , Animaux , Souris , Cellules NIH 3T3 , Antigène CD52/métabolisme , Antigène CD52/génétique , Protéines de fusion recombinantes/génétique , Protéines de fusion recombinantes/métabolisme , Humains , Cytométrie en flux/méthodes , Séparation cellulaire/méthodes , Marqueurs biologiques
6.
J Agric Food Chem ; 72(23): 13360-13370, 2024 Jun 12.
Article de Anglais | MEDLINE | ID: mdl-38830379

RÉSUMÉ

This study reports a peptide design model for engineering fusion-expressed antimicrobial peptides (AMPs) with the AflR dinuclear zinc finger motif to improve the defense against aflatoxins and Aspergillus flavus. The study identified AflR, a Zn2Cys6-type sequence-specific DNA-binding protein, as a key player in the regulation of aflatoxin biosynthesis. By integrating the AflR motif into AMPs, we demonstrate that these novel fusion peptides significantly lower the minimum inhibitory concentrations (MICs) and reduce aflatoxin B1 and B2 levels, outperforming traditional AMPs. Comprehensive analysis, including bioinformatics and structural determination, elucidates the enhanced structure-function relationship underlying their efficacy. Furthermore, the study reveals the possibility that the fusion peptides have the potential to bind to the DNA binding sites of transcriptional regulators, binding DNA sites of key transcriptional regulators, thereby inhibiting genes critical for aflatoxin production. This research not only deepens our understanding of aflatoxin inhibition mechanisms but also presents a promising avenue for developing advanced antifungal agents, which are essential for global food safety and crop protection.


Sujet(s)
Aspergillus flavus , Doigts de zinc , Aspergillus flavus/effets des médicaments et des substances chimiques , Aspergillus flavus/génétique , Aspergillus flavus/métabolisme , Aspergillus flavus/composition chimique , Peptides antimicrobiens/composition chimique , Peptides antimicrobiens/pharmacologie , Peptides antimicrobiens/génétique , Peptides antimicrobiens/métabolisme , Aflatoxines/biosynthèse , Aflatoxines/composition chimique , Aflatoxines/génétique , Ingénierie des protéines , Tests de sensibilité microbienne , Antifongiques/pharmacologie , Antifongiques/composition chimique , Protéines de fusion recombinantes/composition chimique , Protéines de fusion recombinantes/génétique , Protéines de fusion recombinantes/métabolisme , Protéines de fusion recombinantes/pharmacologie
7.
Methods Mol Biol ; 2792: 97-111, 2024.
Article de Anglais | MEDLINE | ID: mdl-38861081

RÉSUMÉ

To measure the kinetic properties of photorespiratory enzymes, it is necessary to work with purified proteins. Protocols to purify photorespiratory enzymes from leaves of various plant species require several time-consuming steps. It is now possible to produce large quantities of recombinant proteins in bacterial cells. They can be rapidly purified as histidine-tagged recombinant proteins by immobilized metal affinity chromatography using Ni2+-NTA-agarose. This chapter describes protocols to purify several Arabidopsis thaliana His-tagged recombinant photorespiratory enzymes (phosphoglycolate phosphatase, glycolate oxidase, and hydroxypyruvate reductase) from Escherichia coli cell cultures using two bacterial strain-plasmid systems: BL21(DE3)-pET and LMG194-pBAD.


Sujet(s)
Protéines d'Arabidopsis , Arabidopsis , Escherichia coli , Hydroxypyruvate reductase , Phosphoric monoester hydrolases , Arabidopsis/génétique , Escherichia coli/génétique , Escherichia coli/métabolisme , Hydroxypyruvate reductase/génétique , Hydroxypyruvate reductase/métabolisme , Hydroxypyruvate reductase/composition chimique , Phosphoric monoester hydrolases/métabolisme , Phosphoric monoester hydrolases/génétique , Phosphoric monoester hydrolases/isolement et purification , Phosphoric monoester hydrolases/composition chimique , Protéines d'Arabidopsis/génétique , Protéines d'Arabidopsis/métabolisme , Protéines d'Arabidopsis/isolement et purification , Protéines d'Arabidopsis/composition chimique , Histidine/métabolisme , Histidine/génétique , Alcohol oxidoreductases/génétique , Alcohol oxidoreductases/métabolisme , Alcohol oxidoreductases/isolement et purification , Alcohol oxidoreductases/composition chimique , Chromatographie d'affinité/méthodes , Protéines recombinantes/métabolisme , Protéines recombinantes/isolement et purification , Protéines recombinantes/génétique , Protéines de fusion recombinantes/génétique , Protéines de fusion recombinantes/isolement et purification , Protéines de fusion recombinantes/métabolisme
8.
Elife ; 132024 Jun 07.
Article de Anglais | MEDLINE | ID: mdl-38847394

RÉSUMÉ

Molecules that facilitate targeted protein degradation (TPD) offer great promise as novel therapeutics. The human hepatic lectin asialoglycoprotein receptor (ASGR) is selectively expressed on hepatocytes. We have previously engineered an anti-ASGR1 antibody-mutant RSPO2 (RSPO2RA) fusion protein (called SWEETS) to drive tissue-specific degradation of ZNRF3/RNF43 E3 ubiquitin ligases, which achieved hepatocyte-specific enhanced Wnt signaling, proliferation, and restored liver function in mouse models, and an antibody-RSPO2RA fusion molecule is currently in human clinical trials. In the current study, we identified two new ASGR1- and ASGR1/2-specific antibodies, 8M24 and 8G8. High-resolution crystal structures of ASGR1:8M24 and ASGR2:8G8 complexes revealed that these antibodies bind to distinct epitopes on opposing sides of ASGR, away from the substrate-binding site. Both antibodies enhanced Wnt activity when assembled as SWEETS molecules with RSPO2RA through specific effects sequestering E3 ligases. In addition, 8M24-RSPO2RA and 8G8-RSPO2RA efficiently downregulate ASGR1 through TPD mechanisms. These results demonstrate the possibility of combining different therapeutic effects and degradation mechanisms in a single molecule.


Sujet(s)
Récepteurs des asialoglycoprotéines , Protéolyse , Ubiquitin-protein ligases , Voie de signalisation Wnt , Humains , Ubiquitin-protein ligases/métabolisme , Ubiquitin-protein ligases/génétique , Récepteurs des asialoglycoprotéines/métabolisme , Animaux , Souris , Cristallographie aux rayons X , Hépatocytes/métabolisme , Protéines de fusion recombinantes/métabolisme , Protéines de fusion recombinantes/génétique , Protéines et peptides de signalisation intercellulaire
9.
Methods Enzymol ; 699: 121-161, 2024.
Article de Anglais | MEDLINE | ID: mdl-38942501

RÉSUMÉ

The step catalyzed by terpene synthases is a well-recognized and significant bottleneck in engineered terpenoid bioproduction. Consequently, substantial efforts have been devoted towards increasing metabolic flux catalyzed by terpene synthases, employing strategies such as gene overexpression and protein engineering. Notably, numerous studies have demonstrated remarkable titer improvements by applying translational fusion, typically by fusing the terpene synthase with a prenyl diphosphate synthase that catalyzes the preceding step in the pathway. The main appeal of the translational fusion approach lies in its simplicity and orthogonality to other metabolic engineering tools. However, there is currently limited understanding of the underlying mechanism of flux enhancement, owing to the unpredictable and often protein-specific effects of translational fusion. In this chapter, we discuss practical considerations when engineering translationally fused terpene synthases, drawing insights from our experience and existing literature. We also provide detailed experimental workflows and protocols based on our previous work in budding yeast (Saccharomyces cerevisiae). Our intention is to encourage further research into the translational fusion of terpene synthases, anticipating that this will contribute mechanistic insights not only into the activity, behavior, and regulation of terpene synthases, but also of other enzymes.


Sujet(s)
Alkyl et aryl transferases , Génie métabolique , Saccharomyces cerevisiae , Alkyl et aryl transferases/génétique , Alkyl et aryl transferases/métabolisme , Génie métabolique/méthodes , Saccharomyces cerevisiae/génétique , Saccharomyces cerevisiae/métabolisme , Protéines de fusion recombinantes/génétique , Protéines de fusion recombinantes/métabolisme , Terpènes/métabolisme , Biosynthèse des protéines , Ingénierie des protéines/méthodes
10.
Sci Adv ; 10(26): eadl2492, 2024 Jun 28.
Article de Anglais | MEDLINE | ID: mdl-38924395

RÉSUMÉ

Bioproduction of 1-alkenes from naturally abundant free fatty acids offers a promising avenue toward the next generation of hydrocarbon-based biofuels and green commodity chemicals. UndB is the only known membrane-bound 1-alkene-producing enzyme, with great potential for 1-alkene bioproduction, but the enzyme exhibits limited turnovers, thus restricting its widespread usage. Here, we explore the molecular basis of the limitation of UndB activity and substantially improve its catalytic power. We establish that the enzyme undergoes peroxide-mediated rapid inactivation during catalysis. To counteract this inactivation, we engineered a chimeric membrane enzyme by conjugating UndB with catalase that protected UndB against peroxide and enhanced its number of turnovers tremendously. Notably, our chimeric enzyme is the only example of a membrane enzyme successfully engineered with catalase. We subsequently constructed a whole-cell biocatalytic system and achieved remarkable efficiencies (up to 95%) in the biotransformation of a wide range of fatty acids (both aliphatic and aromatic) into corresponding 1-alkenes with numerous biotechnological applications.


Sujet(s)
Alcènes , Biocatalyse , Catalase , Alcènes/métabolisme , Catalase/génétique , Catalase/métabolisme , Acides gras/métabolisme , Ingénierie des protéines/méthodes , Escherichia coli/génétique , Escherichia coli/métabolisme , Protéines de fusion recombinantes/métabolisme , Protéines de fusion recombinantes/génétique
11.
J Vet Med Sci ; 86(7): 744-747, 2024 Jul 02.
Article de Anglais | MEDLINE | ID: mdl-38749739

RÉSUMÉ

The red fluorescent protein (rfp)-blasticidin deaminase (bsd) fusion gene was transfected into Babesia ovata by electroporation with the plasmid DNA and selected with 15 µg/mL of blasticidin S under the in vitro culture condition. The transfected parasite with episomal DNA was selected and cultured for further analysis based on the presence of the rfp-bsd fusion gene by PCR and expression of the fusion protein by immunofluorescence antibody test under fluorescence microscopy for 2 months after the transfection. The results are the first, to our knowledge, to demonstrate the expression and stability of the episomal rfp-bsd fusion gene under the control of actin promoter as a selectable marker for the transfection system in B. ovata.


Sujet(s)
Babesia , Protéines luminescentes , , Transfection , Protéines luminescentes/génétique , Protéines luminescentes/métabolisme , Babesia/génétique , Protéines de fusion recombinantes/métabolisme , Protéines de fusion recombinantes/génétique , Animaux , Plasmides/génétique
12.
Int J Biol Macromol ; 269(Pt 1): 131989, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38697425

RÉSUMÉ

Uric acid is the end product of purine metabolism in humans due to inactivation of the uricase determined by the mutated uricase gene. Uricase catalyzes the conversion of uric acid into water-soluble allantoin that is easily excreted by the kidneys. Hyperuricemia occurs when the serum concentration of uric acid exceeds its solubility (7 mg/dL). However, modifications to improve the uricase activity is under development for treating the hyperuricemia. Here we designed 7 types of human-porcine chimeric uricase by multiple sequence comparisons and targeted mutagenesis. An optimal human-porcine chimeric uricase mutant (uricase-10) with both high activity (6.33 U/mg) and high homology (91.45 %) was determined by enzyme activity measurement. The engineering uricase was further modified with PEGylation to improve the stability of recombinant protein drugs and reduce immunogenicity, uricase-10 could be more suitable for the treatment of gout and hyperuricemia theoretically.


Sujet(s)
Polyéthylène glycols , Protéines de fusion recombinantes , Urate oxidase , Animaux , Humains , Hyperuricémie/traitement médicamenteux , Hyperuricémie/génétique , Protéines mutantes/composition chimique , Protéines mutantes/génétique , Protéines mutantes/métabolisme , Mutation , Polyéthylène glycols/composition chimique , Ingénierie des protéines/méthodes , Protéines de fusion recombinantes/génétique , Protéines de fusion recombinantes/composition chimique , Protéines de fusion recombinantes/métabolisme , Protéines recombinantes/composition chimique , Protéines recombinantes/génétique , Protéines recombinantes/métabolisme , Solubilité , Urate oxidase/composition chimique , Urate oxidase/génétique , Urate oxidase/métabolisme , Acide urique/métabolisme
13.
Anticancer Res ; 44(6): 2567-2575, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38821612

RÉSUMÉ

BACKGROUND/AIM: Protein phosphatase and tensin homolog (PTEN) is a tumor suppressor protein with potential to be a new biotechnological drug for PTEN-deficient cancer treatment. This study aimed to develop PTEN-based chimeric proteins (CPP-PTEN-THP) for human epidermal growth factor receptor 2 (HER2)-positive breast cancer treatment, addressing current limitations like inadequate delivery, poor tumor penetration, and low selectivity, while assessing their potential HER2-specific anticancer effects. MATERIALS AND METHODS: pCEFL-EGFP vector was used for both TAT-PTEN-LTV and KLA-PTEN-LTV construction. Non-contact co-cultures were employed using HEK-293T cells for protein expression, and HCC-1954 and MCF-7 cell lines for cytotoxicity testing. Protein detection was analyzed by western blotting and a docking prediction analysis was performed to infer the interactions. RESULTS: Endogenous and recombinant PTEN protein expression was confirmed in cell lysates. A 54-kDa signal matching the theoretical size of PTEN was detected, showing a greater level in TAT-PTEN-LTV (215.1±26.45%) and KLA-PTEN-LTV (129.2±1.44%) compared to endogenous PTEN. After the noncontact co-culture method, cytotoxic studies showed HCC-1954 preferential cell inhibition growth, with 25.95±0.9% and 12.25±1.29% inhibition by KLA-PTEN-LTV and TAT-PTEN-LTV respectively, compared to MCF-7 cells. An LTV-HER2 interaction model was proposed, inferring that LTV interactions are mainly due to the Pro, Trp, and Tyr residues that target HER2. CONCLUSION: The developed PTEN-based chimeric proteins have HER2-specific anticancer activity against HCC-1954 cells.


Sujet(s)
Phosphohydrolase PTEN , Récepteur ErbB-2 , Protéines de fusion recombinantes , Humains , Phosphohydrolase PTEN/métabolisme , Phosphohydrolase PTEN/génétique , Récepteur ErbB-2/métabolisme , Récepteur ErbB-2/génétique , Protéines de fusion recombinantes/pharmacologie , Protéines de fusion recombinantes/métabolisme , Protéines de fusion recombinantes/génétique , Antinéoplasiques/pharmacologie , Lignée cellulaire tumorale , Cellules HEK293 , Cellules MCF-7 , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/métabolisme , Tumeurs du sein/anatomopathologie , Tumeurs du sein/génétique , Femelle , Simulation de docking moléculaire , Techniques de coculture
14.
Nat Biomed Eng ; 8(5): 499-512, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38693431

RÉSUMÉ

Bispecific T-cell engagers (BiTEs) bring together tumour cells and cytotoxic T cells by binding to specific cell-surface tumour antigens and T-cell receptors, and have been clinically successful for the treatment of B-cell malignancies. Here we show that a BiTE-sialidase fusion protein enhances the susceptibility of solid tumours to BiTE-mediated cytolysis of tumour cells via targeted desialylation-that is, the removal of terminal sialic acid residues on glycans-at the BiTE-induced T-cell-tumour-cell interface. In xenograft and syngeneic mouse models of leukaemia and of melanoma and breast cancer, and compared with the parental BiTE molecules, targeted desialylation via the BiTE-sialidase fusion proteins enhanced the formation of immunological synapses, T-cell activation and T-cell-mediated tumour-cell cytolysis in the presence of the target antigen. The targeted desialylation of tumour cells may enhance the potency of therapies relying on T-cell engagers.


Sujet(s)
Sialidase , Animaux , Sialidase/métabolisme , Humains , Souris , Lignée cellulaire tumorale , Protéines de fusion recombinantes/métabolisme , Protéines de fusion recombinantes/génétique , Lymphocytes T/immunologie , Femelle , Anticorps bispécifiques/immunologie , Anticorps bispécifiques/pharmacologie , Activation des lymphocytes , Acide N-acétyl-neuraminique/métabolisme , Acide N-acétyl-neuraminique/composition chimique , Tests d'activité antitumorale sur modèle de xénogreffe , Lymphocytes T cytotoxiques/immunologie , Récepteurs aux antigènes des cellules T/métabolisme , Récepteurs aux antigènes des cellules T/immunologie
15.
Methods Enzymol ; 697: 293-319, 2024.
Article de Anglais | MEDLINE | ID: mdl-38816127

RÉSUMÉ

Assembly of de novo peptides designed from scratch is in a semi-rational manner and creates artificial supramolecular structures with unique properties. Considering that the functions of various proteins in living cells are highly regulated by their assemblies, building artificial assemblies within cells holds the potential to simulate the functions of natural protein assemblies and engineer cellular activities for controlled manipulation. How can we evaluate the self-assembly of designed peptides in cells? The most effective approach involves the genetic fusion of fluorescent proteins (FPs). Expressing a self-assembling peptide fused with an FP within cells allows for evaluating assemblies through fluorescence signal. When µm-scale assemblies such as condensates are formed, the peptide assemblies can be directly observed by imaging. For sub-µm-scale assemblies, fluorescence correlation spectroscopy analysis is more practical. Additionally, the fluorescence resonance energy transfer (FRET) signal between FPs is valuable evidence of proximity. The decrease in fluorescence anisotropy associated with homo-FRET reveals the properties of self-assembly. Furthermore, by combining two FPs, one acting as a donor and the other as an acceptor, the heteromeric interaction between two different components can be studied through the FRET signal. In this chapter, we provide detailed protocols, from designing and constructing plasmid DNA expressing the peptide-fused protein to analysis of self-assembly in living cells.


Sujet(s)
Transfert d'énergie par résonance de fluorescence , Protéines luminescentes , Peptides , Protéines de fusion recombinantes , Transfert d'énergie par résonance de fluorescence/méthodes , Peptides/composition chimique , Peptides/métabolisme , Protéines de fusion recombinantes/génétique , Protéines de fusion recombinantes/métabolisme , Protéines de fusion recombinantes/composition chimique , Humains , Protéines luminescentes/génétique , Protéines luminescentes/composition chimique , Protéines luminescentes/métabolisme , Protéines à fluorescence verte/génétique , Protéines à fluorescence verte/métabolisme , Protéines à fluorescence verte/composition chimique , Plasmides/génétique
16.
Microb Cell Fact ; 23(1): 155, 2024 May 27.
Article de Anglais | MEDLINE | ID: mdl-38802857

RÉSUMÉ

BACKGROUND: Rhizomucor miehei (RM) lipase is a regioselective lipase widely used in food, pharmaceutical and biofuel industries. However, the high cost and low purity of the commercial RM lipase limit its industrial applications. Therefore, it is necessary to develop cost-effective strategies for large-scale preparation of this lipase. The present study explored the high-level expression of RM lipase using superfolder green fluorescent protein (sfGFP)-mediated Escherichia coli secretion system. RESULTS: The sfGFP(-15) mutant was fused to the C-terminus of RM lipase to mediate its secretion expression. The yield of the fusion protein reached approximately 5.1 g/L with high-density fermentation in 5-L fermentors. Unlike conventional secretion expression methods, only a small portion of the target protein was secreted into the cell culture while majority of the fusion protein was still remained in the cytoplasm. However, in contrast to intracellular expression, the target protein in the cytoplasm could be transported efficiently to the supernatant through a simple washing step with equal volume of phosphate saline (PBS), without causing cell disruption. Hence, the approach facilitated the downstream purification step of the recombinant RM lipase. Moreover, contamination or decline of the engineered strain and degradation or deactivation of the target enzyme can be detected efficiently because they exhibited bright green fluorescence. Next, the target protein was immobilized with anion-exchange and macropore resins. Diethylaminoethyl sepharose (DEAE), a weak-basic anion-exchange resin, exhibited the highest bind capacity but inhibited the activity of RM lipase dramatically. On the contrary, RM lipase fixed with macropore resin D101 demonstrated the highest specific activity. Although immobilization with D101 didn't improve the activity of the enzyme, the thermostability of the immobilized enzyme elevated significantly. The immobilized RM lipase retained approximately 90% of its activity after 3-h incubation at 80 °C. Therefore, D101 was chosen as the supporting material of the target protein. CONCLUSION: The present study established a highly efficient strategy for large-scale preparation of RM lipase. This innovative technique not only provides high-purity RM lipase at a low cost but also has great potential as a platform for the preparation of lipases in the future.


Sujet(s)
Escherichia coli , Triacylglycerol lipase , Rhizomucor , Triacylglycerol lipase/génétique , Triacylglycerol lipase/métabolisme , Triacylglycerol lipase/composition chimique , Rhizomucor/enzymologie , Rhizomucor/génétique , Escherichia coli/génétique , Escherichia coli/métabolisme , Enzymes immobilisées/métabolisme , Enzymes immobilisées/génétique , Enzymes immobilisées/composition chimique , Protéines à fluorescence verte/métabolisme , Protéines à fluorescence verte/génétique , Protéines de fusion recombinantes/génétique , Protéines de fusion recombinantes/métabolisme , Protéines de fusion recombinantes/composition chimique , Protéines de fusion recombinantes/biosynthèse , Fermentation
17.
Food Chem ; 453: 139695, 2024 Sep 30.
Article de Anglais | MEDLINE | ID: mdl-38788651

RÉSUMÉ

Alginate lyases with high activity and good thermostability are lacking for the preparation of alginate oligosaccharides (AOS) with various biological activities. We constructed a fusion alginate lyase with both endo-and exo-activities. AlyRm6A-Zu7 was successfully constructed by connecting the highly thermostable AlyRm6A to a new exotype lyase, AlyZu7. The fusion enzyme exhibited high catalytic activity and thermostability. It transformed sodium alginate into oligosaccharides with degrees of polymerization (DP) of 2-4 while producing 4-deoxy-L-erythro-5-hexoseulose uronic acid (DEH). The maximum reducing sugar, AOS, and DP1 + DEH yields were 75 %, 45 %, and 40 %, respectively. Molecular docking confirmed the formation of a stable complex between the substrate and AlyRm6A-Zu7. Protein interactions increased the thermostability of AlyZu7. This work provides new insights into the industrial formation of AOS and monosaccharide DEH using thermally stable fusion enzymes, which has a positive effect in the fields of functional oligosaccharide production and biofuel formation.


Sujet(s)
Alginates , Stabilité enzymatique , Simulation de docking moléculaire , Oligosaccharides , Polysaccharide-lyases , Oligosaccharides/composition chimique , Oligosaccharides/métabolisme , Polysaccharide-lyases/composition chimique , Polysaccharide-lyases/génétique , Polysaccharide-lyases/métabolisme , Alginates/composition chimique , Alginates/métabolisme , Protéines bactériennes/composition chimique , Protéines bactériennes/génétique , Protéines bactériennes/métabolisme , Protéines de fusion recombinantes/composition chimique , Protéines de fusion recombinantes/métabolisme , Protéines de fusion recombinantes/génétique , Biocatalyse
18.
Microbiology (Reading) ; 170(5)2024 May.
Article de Anglais | MEDLINE | ID: mdl-38787390

RÉSUMÉ

Archaeal cell biology is an emerging field expected to identify fundamental cellular processes, help resolve the deep evolutionary history of cellular life, and contribute new components and functions in biotechnology and synthetic biology. To facilitate these, we have developed plasmid vectors that allow convenient cloning and production of proteins and fusion proteins with flexible, rigid, or semi-rigid linkers in the model archaeon Haloferax volcanii. For protein subcellular localization studies using fluorescent protein (FP) tags, we created vectors incorporating a range of codon-optimized fluorescent proteins for N- or C-terminal tagging, including GFP, mNeonGreen, mCherry, YPet, mTurquoise2 and mScarlet-I. Obtaining functional fusion proteins can be challenging with proteins involved in multiple interactions, mainly due to steric interference. We demonstrated the use of the new vector system to screen for improved function in cytoskeletal protein FP fusions, and identified FtsZ1-FPs that are functional in cell division and CetZ1-FPs that are functional in motility and rod cell development. Both the type of linker and the type of FP influenced the functionality of the resulting fusions. The vector design also facilitates convenient cloning and tandem expression of two genes or fusion genes, controlled by a modified tryptophan-inducible promoter, and we demonstrated its use for dual-colour imaging of tagged proteins in H. volcanii cells. These tools should promote further development and applications of archaeal molecular and cellular biology and biotechnology.


Sujet(s)
Protéines d'archée , Clonage moléculaire , Vecteurs génétiques , Haloferax volcanii , Protéines luminescentes , Plasmides , Haloferax volcanii/génétique , Haloferax volcanii/métabolisme , Vecteurs génétiques/génétique , Vecteurs génétiques/métabolisme , Protéines luminescentes/génétique , Protéines luminescentes/métabolisme , Protéines d'archée/génétique , Protéines d'archée/métabolisme , Plasmides/génétique , Plasmides/métabolisme , Protéines de fusion recombinantes/génétique , Protéines de fusion recombinantes/métabolisme , Expression des gènes , Protéines à fluorescence verte/génétique , Protéines à fluorescence verte/métabolisme
19.
Protein Expr Purif ; 221: 106506, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-38772430

RÉSUMÉ

Influenza poses a substantial health risk, with infants and the elderly being particularly susceptible to its grave impacts. The primary challenge lies in its rapid genetic evolution, leading to the emergence of new Influenza A strains annually. These changes involve punctual mutations predominantly affecting the two main glycoproteins: Hemagglutinin (HA) and Neuraminidase (NA). Our existing vaccines target these proteins, providing short-term protection, but fall short when unexpected pandemics strike. Delving deeper into Influenza's genetic makeup, we spotlight the nucleoprotein (NP) - a key player in the transcription, replication, and packaging of RNA. An intriguing characteristic of the NP is that it is highly conserved across all Influenza A variants, potentially paving the way for a more versatile and broadly protective vaccine. We designed and synthesized a novel NP-Hoc fusion protein combining Influenza A nucleoprotein and T4 phage Hoc, cloned using Gibson assembly in E. coli, and purified via ion affinity chromatography. Simultaneously, we explore the T4 coat protein Hoc, typically regarded as inconsequential in controlled viral replication. Yet, it possesses a unique ability: it can link with another protein, showcasing it on the T4 phage coat. Fusing these concepts, our study designs, expresses, and purifies a novel fusion protein named NP-Hoc. We propose this protein as the basis for a new generation of vaccines, engineered to guard broadly against Influenza A. The excitement lies not just in the immediate application, but the promise this holds for future pandemic resilience, with NP-Hoc marking a significant leap in adaptive, broad-spectrum influenza prevention.


Sujet(s)
Bactériophage T4 , Escherichia coli , Protéines de fusion recombinantes , Bactériophage T4/génétique , Bactériophage T4/composition chimique , Bactériophage T4/métabolisme , Protéines de fusion recombinantes/génétique , Protéines de fusion recombinantes/composition chimique , Protéines de fusion recombinantes/isolement et purification , Protéines de fusion recombinantes/métabolisme , Protéines de fusion recombinantes/biosynthèse , Escherichia coli/génétique , Escherichia coli/métabolisme , Expression des gènes , Protéines nucléocapside/génétique , Protéines nucléocapside/composition chimique , Protéines nucléocapside/métabolisme , Virus de la grippe A/génétique , Virus de la grippe A/métabolisme , Vaccins antigrippaux/génétique , Vaccins antigrippaux/biosynthèse , Vaccins antigrippaux/immunologie , Vaccins antigrippaux/composition chimique , Humains , Protéines de liaison à l'ARN/génétique , Protéines de liaison à l'ARN/métabolisme , Protéines de liaison à l'ARN/composition chimique , Protéines de liaison à l'ARN/isolement et purification
20.
Protein Expr Purif ; 221: 106501, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-38782081

RÉSUMÉ

Nanobodies are single-variable domain antibodies with excellent properties, which are evolving as versatile tools to guide cognate antigens in vitro and in vivo for biological research, diagnosis, and treatment. Given their simple structure, nanobodies are readily produced in multiple systems. However, selecting an appropriate expression system is crucial because different conditions might cause proteins to produce different folds or post-translational modifications (PTMs), and these differences often result in different functions. At present, the strategies of PTMs are rarely reported. The GFP nanobody can specifically target the GFP protein. Here, we engineered a GFP nanobody fused with 6 × His tag and Fc tag, respectively, and expressed in bacteria and mammalian cells. The 6 × His-GFP-nanobody was produced from Escherichia coli at high yields and the pull-down assay indicated that it can precipitate the GFP protein. Meanwhile, the Fc-GFP-nanobody can be expressed in HEK293T cells, and the co-immunoprecipitation experiment can trace and target the GFP-tagged protein in vivo. Furthermore, some different PTMs in antigen-binding regions have been identified after using mass spectrometry (MS) to analyze the GFP nanobodies, which are expressed in prokaryotes and eukaryotes. In this study, a GFP nanobody was designed, and its binding ability was verified by using the eukaryotic and prokaryotic protein expression systems. In addition, this GFP nanobody was transformed into a useful instrument for more in-depth functional investigations of GFP fusion proteins. MS was further used to explore the reason for the difference in binding ability, providing a novel perspective for the study of GFP nanobodies and protein expression purification.


Sujet(s)
Escherichia coli , Protéines à fluorescence verte , Maturation post-traductionnelle des protéines , Protéines de fusion recombinantes , Anticorps à domaine unique , Humains , Protéines à fluorescence verte/génétique , Protéines à fluorescence verte/métabolisme , Protéines à fluorescence verte/composition chimique , Anticorps à domaine unique/génétique , Anticorps à domaine unique/composition chimique , Anticorps à domaine unique/biosynthèse , Anticorps à domaine unique/isolement et purification , Anticorps à domaine unique/immunologie , Cellules HEK293 , Escherichia coli/génétique , Escherichia coli/métabolisme , Protéines de fusion recombinantes/génétique , Protéines de fusion recombinantes/composition chimique , Protéines de fusion recombinantes/isolement et purification , Protéines de fusion recombinantes/métabolisme , Protéines de fusion recombinantes/biosynthèse , Ingénierie des protéines/méthodes , Expression des gènes
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...