Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
Expert Opin Ther Targets ; : 1-23, 2024 Jun 07.
Article in English | MEDLINE | ID: mdl-38828744

ABSTRACT

BACKGROUND: Hypertension worsens outcomes in SARS-CoV-2 patients. Sartans, a type of antihypertensive angiotensin receptor blocker-(ARB), reduce COVID-19 morbidity and mortality by targeting angiotensin-converting enzyme-2 (ACE2). This study aimed to evaluate the antiviral and antihypertensive effects of nirmatrelvir, commercial sartans (candesartan, losartan, and losartan carboxylic (Exp3174)), and newly synthesized sartans (benzimidazole-N-biphenyl carboxyl (ACC519C) and benzimidazole-N-biphenyl tetrazole (ACC519T)), compared to nirmatrelvir, the antiviral component of Paxlovid. RESEARCH DESIGN AND METHODS: Surface plasmon resonance (SPR) and enzymatic studies assessed drug effects on ACE2. Antiviral abilities were tested with SARS-CoV-2-infected Vero E6 cells, and antihypertensive effects were evaluated using angiotensin II-contracted rabbit iliac arteries. RESULTS: Benzimidazole-based candesartan and ACC519C showed antiviral activity comparable to nirmatrelvir (95% inhibition). Imidazole-based losartan, Exp3174, and ACC519T were less potent (75%-80% and 50%, respectively), with Exp3174 being the least effective. SPR analysis indicated high sartans-ACE2 binding affinity. Candesartan and nirmatrelvir combined had greater inhibitory and cytopathic effects (3.96%) than individually (6.10% and 5.08%). ACE2 enzymatic assays showed varying effects of novel sartans on ACE2. ACC519T significantly reduced angiotensin II-mediated contraction, unlike nirmatrelvir and ACC519T(2). CONCLUSION: This study reports the discovery of a new class of benzimidazole-based sartans that significantly inhibit SARS-CoV-2, likely due to their interaction with ACE2.

2.
Front Immunol ; 15: 1382576, 2024.
Article in English | MEDLINE | ID: mdl-38779661

ABSTRACT

Monoclonal antibodies targeting immune checkpoints have revolutionized oncology. Yet, the effectiveness of these treatments varies significantly among patients, and they are associated with unexpected adverse events, including hyperprogression. The murine research model used in drug development fails to recapitulate both the functional human immune system and the population heterogeneity. Hence, a novel model is urgently needed to study the consequences of immune checkpoint blockade. Dogs appear to be uniquely suited for this role. Approximately 1 in 4 companion dogs dies from cancer, yet no antibodies are commercially available for use in veterinary oncology. Here we characterize two novel antibodies that bind canine PD-1 with sub-nanomolar affinity as measured by SPR. Both antibodies block the clinically crucial PD-1/PD-L1 interaction in a competitive ELISA assay. Additionally, the antibodies were tested with a broad range of assays including Western Blot, ELISA, flow cytometry, immunofluorescence and immunohistochemistry. The antibodies appear to bind two distinct epitopes as predicted by molecular modeling and peptide phage display. Our study provides new tools for canine oncology research and a potential veterinary therapeutic.


Subject(s)
Antibodies, Monoclonal , Programmed Cell Death 1 Receptor , Dogs , Animals , Programmed Cell Death 1 Receptor/immunology , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/metabolism , Antibodies, Monoclonal/immunology , Humans , Immune Checkpoint Inhibitors/immunology , Immune Checkpoint Inhibitors/pharmacology , B7-H1 Antigen/immunology , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/metabolism , Epitopes/immunology , Dog Diseases/immunology , Dog Diseases/drug therapy , Protein Binding , Neoplasms/immunology , Neoplasms/veterinary , Neoplasms/drug therapy
3.
Transl Oncol ; 42: 101892, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38359715

ABSTRACT

The PD-1/PD-L1 complex belongs to the group of inhibitory immune checkpoints and plays a critical role in immune regulation. The PD-1/PD-L1 axis is also responsible for immune evasion of cancer cells, and this complex is one of the main targets of immunotherapies used in oncology. Treatment using immune checkpoint inhibitors is mainly based on antibodies. This approach has great therapeutic potential; however, it also has major drawbacks and can induce immune-related adverse events. Thus, there is a strong need for alternative, non-antibody-based therapies using small molecules, peptides, or peptidomimetics. In the present study, we designed, synthesized, and evaluated a set of PD-1-targeting peptides based on the sequence and structure of PD-L1. The binding of these peptides to PD-1 was investigated using SPR and ELISA. We also assessed their ability to compete with PD-L1 for binding to PD-1 and their inhibitory properties against the PD-1/PD-L1 complex at the cellular level. The best results were obtained for the peptide PD-L1(111-127)(Y112C-I126C), named (L11), which displaced PD-L1 from binding to PD-1 in the competitive assay and inhibited the formation of the PD-1/PD-L1 complex. The (L11) peptide also exhibited strong affinity for PD-1. NMR studies revealed that (L11) does not form a well-defined secondary structure; however, MD simulation indicated that (L11) binds to PD-1 at the same place as PD-L1. After further optimization of the structure, the peptide inhibitor obtained in this study could also be used as a potential therapeutic compound targeting the PD-1/PD-L1 axis.

4.
Front Microbiol ; 14: 1307349, 2023.
Article in English | MEDLINE | ID: mdl-38098664

ABSTRACT

Tailocins are nanomolecular machines with bactericidal activity. They are produced by bacteria to contribute to fitness in mixed communities, and hence, they play a critical role in their ecology in a variety of habitats. Here, we characterized the new tailocin produced by Dickeya dadantii strain 3937, a well-characterized member of plant pathogenic Soft Rot Pectobacteriaceae (SRP). Tailocins induced in D. dadantii were ca. 166 nm long tubes surrounded by contractive sheaths with baseplates having tail fibers at one end. A 22-kb genomic cluster involved in their synthesis and having high homology to the cluster coding for the tail of the Peduovirus P2 was identified. The D. dadantii tailocins, termed dickeyocins P2D1 (phage P2-like dickeyocin 1), were resistant to inactivation by pH (3.5-12), temperature (4-50°C), and elevated osmolarity (NaCl concentration: 0.01-1 M). P2D1 could kill a variety of different Dickeya spp. but not any strain of Pectobacterium spp. tested and were not toxic to Caenorhabditis elegans.

5.
Nucleic Acids Res ; 51(19): 10551-10567, 2023 10 27.
Article in English | MEDLINE | ID: mdl-37713613

ABSTRACT

For DNA replication initiation in Bacteria, replication initiation proteins bind to double-stranded DNA (dsDNA) and interact with single-stranded DNA (ssDNA) at the replication origin. The structural-functional relationship of the nucleoprotein complex involving initiator proteins is still elusive and different models are proposed. In this work, based on crosslinking combined with mass spectrometry (MS), the analysis of mutant proteins and crystal structures, we defined amino acid residues essential for the interaction between plasmid Rep proteins, TrfA and RepE, and ssDNA. This interaction and Rep binding to dsDNA could not be provided in trans, and both are important for dsDNA melting at DNA unwinding element (DUE). We solved two crystal structures of RepE: one in a complex with ssDNA DUE, and another with both ssDNA DUE and dsDNA containing RepE-specific binding sites (iterons). The amino acid residues involved in interaction with ssDNA are located in the WH1 domain in stand ß1, helices α1 and α2 and in the WH2 domain in loops preceding strands ß1' and ß2' and in these strands. It is on the opposite side compared to RepE dsDNA-recognition interface. Our data provide evidence for a loop-back mechanism through which the plasmid replication initiator molecule accommodates together dsDNA and ssDNA.


Subject(s)
DNA, Single-Stranded , DNA-Binding Proteins , DNA, Single-Stranded/genetics , DNA-Binding Proteins/metabolism , DNA Replication , Plasmids/genetics , DNA/genetics , DNA/metabolism , Amino Acids/genetics
6.
Biomed Pharmacother ; 165: 115161, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37473684

ABSTRACT

Immune checkpoints can be divided into co-stimulatory and co-inhibitory molecules that regulate the activation and effector functions of T cells. The co-inhibitory pathways mediated by ICPs are used by cancer cells to escape from immune surveillance, and therefore the blockade of these receptor/ligand interactions is one of the strategies used in the treatment of cancer. The two main pathways currently under investigation are CTLA-4/CD80/CD86 and PD-1/PD-L1, and the monoclonal Abs targeting them have shown potent immunomodulatory effects and activity in clinical environments. Another interesting target in cancer treatment is the BTLA/HVEM complex. Binding of BTLA protein on T cells to HVEM on cancer cells leads to inhibition of T cell proliferation and cytokine production. In the presented work, we focused on blocking the HVEM protein using BTLA-derived peptides. Based on the crystal structure of the BTLA/HVEM complex and MM/GBSA analysis performed here, we designed and synthesized peptides, specifically fragments of BTLA protein. We subsequently checked the inhibitory capacities of these compounds using ELISA and a cellular reporter platform. Two of these peptides, namely BTLA(35-43) and BTLA(33-64)C58Abu displayed the most promising properties, and we therefore performed further studies to evaluate their affinity to HVEM protein, their stability in plasma and their effect on viability of human PBMCs. In addition, the 3D structure for the peptide BTLA(33-64)C58Abu was determined using NMR. Obtained data confirmed that the BTLA-derived peptides could be the basis for future drugs and their immunomodulatory potential merits further examination.


Subject(s)
Receptors, Immunologic , Receptors, Tumor Necrosis Factor, Member 14 , Humans , Receptors, Immunologic/metabolism , Receptors, Tumor Necrosis Factor, Member 14/chemistry , Receptors, Tumor Necrosis Factor, Member 14/metabolism , T-Lymphocytes , Peptides/chemistry , Protein Binding
7.
Chemistry ; 29(55): e202300970, 2023 Oct 02.
Article in English | MEDLINE | ID: mdl-37332024

ABSTRACT

In this work, we present studies on relatively new and still not well-explored potential anticancer targets which are shelterin proteins, in particular the TRF1 protein can be blocked by in silico designed "peptidomimetic" molecules. TRF1 interacts directly with the TIN2 protein, and this protein-protein interaction is crucial for the proper functioning of telomere, which could be blocked by our novel modified peptide molecules. Our chemotherapeutic approach is based on assumption that modulation of TRF1-TIN2 interaction may be more harmful for cancer cells as cancer telomeres are more fragile than in normal cells. We have shown in vitro within SPR experiments that our modified peptide PEP1 molecule interacts with TRF1, presumably at the site originally occupied by the TIN2 protein. Disturbance of the shelterin complex by studied molecule may not in short term lead to cytotoxic effects, however blocking TRF1-TIN2 resulted in cellular senescence in cellular breast cancer lines used as a cancer model. Thus, our compounds appeared useful as starting model compounds for precise blockage of TRF proteins.


Subject(s)
Shelterin Complex , Telomeric Repeat Binding Protein 2 , Telomeric Repeat Binding Protein 1/chemistry , Telomeric Repeat Binding Protein 1/genetics , Telomeric Repeat Binding Protein 1/metabolism , Telomere/metabolism , Peptides/pharmacology
8.
Int J Mol Sci ; 24(9)2023 May 08.
Article in English | MEDLINE | ID: mdl-37176159

ABSTRACT

This study is an extension of current research into a novel class of synthetic antihypertensive drugs referred to as "bisartans", which are bis-alkylated imidazole derivatives bearing two symmetric anionic biphenyltetrazoles. Research to date indicates that bisartans are superior to commercially available hypertension drugs, since the former undergo stronger docking to angiotensin-converting enzyme 2 (ACE2). ACE2 is the key receptor involved in SARS-CoV-2 entry, thus initiating COVID-19 infection and in regulating levels of vasoactive peptides such as angiotensin II and beneficial heptapeptides A(1-7) and Alamandine in the renin-angiotensin system (RAS). In previous studies using in vivo rabbit-iliac arterial models, we showed that Na+ or K+ salts of selected Bisartans initiate a potent dose-response inhibition of vasoconstriction. Furthermore, computational studies revealed that bisartans undergo stable binding to the vital interfacial region between ACE2 and the SARS-CoV-2 "receptor binding domain" (i.e., the viral RBD). Thus, bisartan homologs are expected to interfere with SARS-CoV-2 infection and/or suppress disease expression in humans. The primary goal of this study was to investigate the role of tetrazole in binding and the network of amino acids of SARS-CoV-2 Spike RBD-ACE2 complex involved in interactions with sartans. This study would, furthermore, allow the expansion of the synthetic space to create a diverse suite of new bisartans in conjunction with detailed computational and in vitro antiviral studies. A critical role for tetrazole was uncovered in this study, shedding light on the vital importance of this group in the binding of sartans and bisartans to the ACE2/Spike complex. The in silico data predicting an interaction of tetrazole-containing sartans with ACE2 were experimentally validated by the results of surface plasmon resonance (SPR) analyses performed with a recombinant human ACE2 protein.


Subject(s)
COVID-19 , Animals , Humans , Rabbits , SARS-CoV-2/metabolism , Antihypertensive Agents/pharmacology , Antihypertensive Agents/therapeutic use , Antihypertensive Agents/metabolism , Angiotensin-Converting Enzyme 2/metabolism , Angiotensin II Type 1 Receptor Blockers , Binding Sites , Protein Binding
9.
Front Microbiol ; 14: 1328842, 2023.
Article in English | MEDLINE | ID: mdl-38249469

ABSTRACT

Although the mechanism of DNA replication initiation has been investigated for over 50 years, many important discoveries have been made related to this process in recent years. In this mini-review, we discuss the current state of knowledge concerning the structure of the origin region in bacterial chromosomes and plasmids, recently discovered motifs recognized by replication initiator proteins, and proposed in the literature models describing initial origin opening. We review structures of nucleoprotein complexes formed by replication initiators at chromosomal and plasmid replication origins and discuss their functional implications. We also discuss future research challenges in this field.

10.
Sci Rep ; 12(1): 18497, 2022 11 02.
Article in English | MEDLINE | ID: mdl-36323731

ABSTRACT

The present research shows the antitumor activity of a protein-polysaccharide complex Venetin-1 obtained from the coelomic fluid of Dendrobaena veneta earthworms against A549 cancer cells. The investigations are a continuation of experiments on the antitumor activity of coelomic fluid obtained from this species. The Venetin-1 nanoparticle was obtained after thermal treatment of the coelomic fluid, separation from coelomocytes, filtration, and lyophilization. The preparation showed a selective effect on cancer cells, whereas normal cells were unaffected. Venetin-1 was effective against the lung cancer cells at doses of 31.3 and 62.5 µg/ml, and the results were imaged using light microscopy and scanning electron microscopy (SEM). The cells died mainly via the apoptosis pathway. Necrotic cells appeared sporadically in the microscopic view. SEM imaging revealed complete destruction of the A549 cells after the incubation with Venetin-1. The atomic force microscopy (AFM) analyses showed changes in the topography, peak force error images, and Young's modulus (elasticity) of the A549 cells after the incubation with Venetin-1. The transmission electron cryomicroscopy (Cryo-TEM) analysis indicated a polymeric nature of the analyzed preparation. The samples of Venetin-1 showed a very homogeneous size profile with the microparticle size of approximately 58.23 nm. A significant decrease in Venetin-1 binding to sphingomyelin was observed. Venetin-1 lost its pore-forming activity or deactivation of the pore-forming activity occurred. This confirms the absence of hemolytic capacity of Venetin-1 towards red blood cells. The conducted analyses show the suitability of the obtained complex for biomedical research. The next step will consist in analyses of the effect of Venetin-1 on the immune system in mice.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Nanoparticles , Oligochaeta , Animals , Mice , Humans , Oligochaeta/physiology , Carcinoma, Non-Small-Cell Lung/drug therapy , Lung Neoplasms/drug therapy , A549 Cells
11.
Bioorg Chem ; 128: 106047, 2022 11.
Article in English | MEDLINE | ID: mdl-35963023

ABSTRACT

Over the past few years, many molecules such as monoclonal antibodies, affibodies, nanobodies, and small compounds have been designed and tested as inhibitors of PD-1/PD-L1 complex formation. Some of them have been successfully implemented into clinical oncology practice. However, the majority of these compounds have disadvantages and limitations, such as high production price, potential for immunogenicity and/or prolonged clearance. Thus, new inhibitors of the PD-1/PD-L1 immune checkpoints are needed. Recently, peptides emerged as potential novel approach for blocking receptor/ligand interaction. In the presented studies we have designed, synthesised and tested peptides, which are potential inhibitors of the PD-1/PD-L1 axis. The amino acid sequences of the designed peptides were based on the binding sites of PD-1 to PD-L1, as determined by the crystal structure of the protein complex and also based on MM/GBSA analysis. Interactions of the peptides with PD-L1 protein were confirmed using SPR, while their inhibitory properties were studied using cell-based PD-1/PD-L1 immune checkpoint blockade assays. The characterization of the peptides has shown that the peptides PD-1(119-142)T120C-E141C, PD-1(119-142)C123-S137C and PD-1(122-138)C123-S137C strongly bind to PD-L1 protein and disrupt the interaction of the proteins. PD-1(122-138)C123-S137C peptide was shown to have the best inhibitory potential from the panel of peptides. Its 3D NMR structure was determined and the binding site to PD-L1 was established using molecular modelling methods. Our results indicate that the PD-1 derived peptides are able to mimic the PD-1 protein and inhibit PD-1/PD-L1 complex formation.


Subject(s)
B7-H1 Antigen , Neoplasms , B7-H1 Antigen/metabolism , Humans , Immunotherapy/methods , Neoplasms/therapy , Peptides/chemistry , Peptides/pharmacology , Programmed Cell Death 1 Receptor/chemistry , Programmed Cell Death 1 Receptor/metabolism
12.
Bioorg Chem ; 122: 105748, 2022 05.
Article in English | MEDLINE | ID: mdl-35325694

ABSTRACT

Cancer immunotherapy using blockade of immune checkpoints is mainly based on monoclonal antibodies. Despite the tremendous success achieved by using those molecules to block immune checkpoint proteins, antibodies possess some weaknesses, which means that there is still a need to search for new compounds as alternatives to antibodies. Many current approaches are focused on use of peptides/peptidomimetics to destroy receptor/ligand interactions. Our studies concern blockade of the BTLA/HVEM complex, which generates an inhibitory effect on the immune response resulting in tolerance to cancer cells. To design inhibitors of such proteins binding we based our work on the amino acid sequence and structure of a ligand of HVEM protein, namely glycoprotein D, which possesses the same binding site on HVEM as BTLA protein. To disrupt the BTLA and HVEM interaction we designed several peptides, all fragments of glycoprotein D, and tested their binding to HVEM using SPR and their ability to inhibit the BTLA/HVEM complex formation using ELISA tests and cellular reporter platforms. That led to identification of two peptides, namely gD(1-36)(K10C-D30C) and gD(1-36)(A12C-L25C), which interact with HVEM and possess blocking capacities. Both peptides are not cytotoxic to human PBMCs, and show stability in human plasma. We also studied the 3D structure of the gD(1-36)(K10C-D30C) peptide using NMR and molecular modeling methods. The obtained data reveal that it possesses an unstructured conformation and binds to HVEM in the same location as gD and BTLA. All these results suggest that peptides based on the binding fragment of gD protein represent promising immunomodulation agents for future cancer immunotherapy.


Subject(s)
Receptors, Immunologic , Receptors, Tumor Necrosis Factor, Member 14 , Amino Acid Sequence , Binding Sites , Glycoproteins , Humans , Receptors, Immunologic/chemistry , Receptors, Immunologic/metabolism , Receptors, Tumor Necrosis Factor, Member 14/chemistry , Receptors, Tumor Necrosis Factor, Member 14/metabolism
13.
Anal Chem ; 93(29): 10084-10089, 2021 07 27.
Article in English | MEDLINE | ID: mdl-34264066

ABSTRACT

We report for the first time on in situ transduction of electrochemical responses of ion-selective electrodes, operating under non-zero-current conditions, to emission change signals. The proposed novel-type PVC-based membrane comprises a dispersed redox and emission active ion-to-electron transducer. The electrochemical trigger applied induces a redox process of the transducer, inducing ion exchange between the membrane and the solution, resulting also in change of its emission spectrum. It is shown that electrochemical signals recorded for ion-selective electrodes operating under voltammetric/coulometric conditions correlate with emission intensity changes recorded in the same experiments. Moreover, the proposed optical readout offers extended linear response range compared to electrical signals recorded in voltammetric or coulometric mode.


Subject(s)
Electrons , Ion-Selective Electrodes , Electrodes , Oxidation-Reduction
14.
BMC Res Notes ; 14(1): 275, 2021 Jul 19.
Article in English | MEDLINE | ID: mdl-34281605

ABSTRACT

OBJECTIVE: The ability to form nucleoprotein complexes is a fundamental activity of DNA replication initiation proteins. They bind within or nearby the region of replication origin what results in melting of a double-stranded DNA (dsDNA) and formation of single-stranded DNA (ssDNA) region where the replication machinery can assemble. For prokaryotic initiators it was shown that they interact with the formed ssDNA and that this interaction is required for the replication activity. The ability to interact with ssDNA was also shown for Saccharomyces cerevisiae replication initiation protein complex ORC. For Archaea, which combine features of both prokaryotic and eukaryotic organisms, there was no evidence whether DNA replication initiators can interact with ssDNA. We address this issue in this study. RESULTS: Using purified Orc1 protein from Aeropyrum pernix (ApOrc1) we analyzed its ability to interact with ssDNA containing sequence of an AT-rich region of the A. pernix origin Ori1 as well as with homopolymers of thymidine (polyT) and adenosine (polyA). The Bio-layer interferometry, surface plasmon resonance and microscale thermophoresis showed that the ApOrc1 can interact with ssDNA and it binds preferentially to T-rich ssDNA. The hydrolysis of ATP is not required for this interaction.


Subject(s)
DNA, Single-Stranded , Origin Recognition Complex , Archaea/metabolism , DNA Replication , Origin Recognition Complex/genetics , Origin Recognition Complex/metabolism , Protein Binding , Replication Origin
15.
Int J Mol Sci ; 22(5)2021 Mar 04.
Article in English | MEDLINE | ID: mdl-33806461

ABSTRACT

The present study aimed to synthesize novel polycationic polymers composed of N-substituted L-2,3-diaminopropionic acid residues (DAPEGs) and investigate their cell permeability, cytotoxicity, and DNA-binding ability. The most efficient cell membrane-penetrating compounds (O2Oc-Dap(GO2)n-O2Oc-NH2, where n = 4, 6, and 8) showed dsDNA binding with a binding constant in the micromolar range (0.3, 3.4, and 0.19 µM, respectively) and were not cytotoxic to HB2 and MDA-MB-231 cells. Selected compounds used in the transfection of a GFP plasmid showed high transfection efficacy and minimal cytotoxicity. Their interaction with plasmid DNA and the increasing length of the main chain of tested compounds strongly influenced the organization and shape of the flower-like nanostructures formed, which were unique for 5/6-FAM-O2Oc-[Dap(GO2)]8-O2Oc-NH2 and typical for large proteins.


Subject(s)
Cell Membrane Permeability/physiology , Nucleic Acids/metabolism , Polymers/pharmacology , beta-Alanine/analogs & derivatives , Cell Line , Cell Line, Tumor , Humans , Nanostructures/chemistry , Plasmids/metabolism , Transfection/methods , beta-Alanine/pharmacology
16.
Nucleic Acids Res ; 49(6): 3394-3408, 2021 04 06.
Article in English | MEDLINE | ID: mdl-33660784

ABSTRACT

An essential feature of replication initiation proteins is their ability to bind to DNA. In this work, we describe a new domain that contributes to a replication initiator sequence-specific interaction with DNA. Applying biochemical assays and structure prediction methods coupled with DNA-protein crosslinking, mass spectrometry, and construction and analysis of mutant proteins, we identified that the replication initiator of the broad host range plasmid RK2, in addition to two winged helix domains, contains a third DNA-binding domain. The phylogenetic analysis revealed that the composition of this unique domain is typical within the described TrfA-like protein family. Both in vitro and in vivo experiments involving the constructed TrfA mutant proteins showed that the newly identified domain is essential for the formation of the protein complex with DNA, contributes to the avidity for interaction with DNA, and the replication activity of the initiator. The analysis of mutant proteins, each containing a single substitution, showed that each of the three domains composing TrfA is essential for the formation of the protein complex with DNA. Furthermore, the new domain, along with the winged helix domains, contributes to the sequence specificity of replication initiator interaction within the plasmid replication origin.


Subject(s)
DNA Helicases/chemistry , DNA Helicases/metabolism , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , DNA/metabolism , Trans-Activators/chemistry , Trans-Activators/metabolism , Models, Molecular , Protein Binding , Protein Domains
17.
J Orthop Trauma ; 34 Suppl 2: S19-S20, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32639343

ABSTRACT

This case demonstrates a recognized association between an acetabular injury pattern and underlying morphology of the hip. In the patient discussed, hyperflexion of the hip results in the engagement of the present CAM lesion, and the resulting subluxation leads to a fracture of the posterior wall and instability of the hip. This combination of pathologies was addressed with a surgical dislocation approach to address both the CAM lesion and fix the posterior wall.


Subject(s)
Fractures, Bone , Hip Dislocation , Acetabulum/diagnostic imaging , Acetabulum/surgery , Hip Dislocation/diagnostic imaging , Hip Dislocation/etiology , Hip Dislocation/surgery , Humans
18.
Int J Mol Sci ; 21(2)2020 Jan 18.
Article in English | MEDLINE | ID: mdl-31963646

ABSTRACT

Immune checkpoints are crucial in the maintenance of antitumor immune responses. The activation or blockade of immune checkpoints is dependent on the interactions between receptors and ligands; such interactions can provide inhibitory or stimulatory signals, including the enhancement or suppression of T-cell proliferation, differentiation, and/or cytokine secretion. B-and T-lymphocyte attenuator (BTLA) is a lymphoid-specific cell surface receptor which is present on T-cells and interacts with herpes virus entry mediator (HVEM), which is present on tumor cells. The binding of HVEM to BTLA triggers an inhibitory signal which attenuates the immune response. This feature is interesting for studying the molecular interactions between HVEM and BTLA, as they may be targeted for novel immunotherapies. This work was based on the crystal structure of the BTLA/HVEM complex showing that BTLA binds the N-terminal cysteine-rich domain of HVEM. We investigated the amino acid sequence of HVEM and used molecular modeling methods to develop inhibitors of the BTLA/HVEM interaction. We synthesized novel compounds and determined their ability to interact with the BTLA protein and inhibit the formation of the BTLA/HVEM complex. Our results suggest that the HVEM (14-39) peptide is a potent inhibitor of the formation of the BTLA/HVEM protein complex.


Subject(s)
Disulfides/chemistry , Peptides/pharmacology , Receptors, Immunologic/metabolism , Receptors, Tumor Necrosis Factor, Member 14/metabolism , Binding Sites/drug effects , Crystallography, X-Ray , Humans , Models, Molecular , Molecular Docking Simulation , Peptides/chemical synthesis , Peptides/chemistry , Protein Binding/drug effects , Protein Conformation , Receptors, Immunologic/chemistry , Receptors, Tumor Necrosis Factor, Member 14/chemistry
19.
Sci Rep ; 8(1): 15287, 2018 10 16.
Article in English | MEDLINE | ID: mdl-30327496

ABSTRACT

The activity of type II toxin-antitoxin systems (TA), which are responsible for many important features of bacterial cells, is based on the differences between toxin and antitoxin stabilities. The antitoxin lability results from bacterial protease activity. Here, we investigated how particular Escherichia coli cytosolic proteases, namely, Lon, ClpAP, ClpXP, and ClpYQ, affect the stability of both the toxin and antitoxin components of the parDE system from the broad host range plasmid RK2. The results of our in vivo and in vitro experiments show that the ParD antitoxin is degraded by the ClpAP protease, and dsDNA stimulates this process. The ParE toxin is not degraded by any of these proteases and can therefore cause growth inhibition of plasmid-free cells after an unequal plasmid distribution during cell division. We also demonstrate that the ParE toxin interaction with ParD prevents antitoxin proteolysis by ClpAP; however, this interaction does not prevent the ClpAP interaction with ParD. We show that ClpAP protease homologs affect plasmid stability in other bacterial species, indicating that ClpAP is a universal activator of the parDE system and that ParD is a universal substrate for ClpAP.


Subject(s)
Bacterial Toxins/metabolism , DNA-Binding Proteins/metabolism , Endopeptidase Clp/metabolism , Escherichia coli Proteins/metabolism , Escherichia coli/physiology , Toxin-Antitoxin Systems , Caulobacter crescentus/genetics , DNA/physiology , Escherichia coli/enzymology , Escherichia coli/genetics , Plasmids , Protein Binding , Proteolysis , Pseudomonas putida/genetics
20.
J Biol Chem ; 292(18): 7507-7518, 2017 05 05.
Article in English | MEDLINE | ID: mdl-28292931

ABSTRACT

Lon protease previously has been shown to interact with DNA, but the role of this interaction for Lon proteolytic activity has not been characterized. In this study, we used truncated Escherichia coli Lon constructs, bioinformatics analysis, and site-directed mutagenesis to identify Lon domains and residues crucial for Lon binding with DNA and effects on Lon proteolytic activity. We found that deletion of Lon's ATPase domain abrogated interactions with DNA. Substitution of positively charged amino acids in this domain in full-length Lon with residues conferring a net negative charge disrupted binding of Lon to DNA. These changes also affected the degradation of nucleic acid-binding protein substrates of Lon, intracellular localization of Lon, and cell morphology. In vivo tests revealed that Lon-DNA interactions are essential for Lon activity in cell division control. In summary, we demonstrate that the ability of Lon to bind DNA is determined by its ATPase domain, that this binding is required for processing protein substrates in nucleoprotein complexes, and that Lon may help regulate DNA replication in response to growth conditions.


Subject(s)
Adenosine Triphosphatases/metabolism , DNA Replication/physiology , DNA, Bacterial/biosynthesis , DNA-Binding Proteins/metabolism , Escherichia coli Proteins/metabolism , Escherichia coli/enzymology , Protease La/metabolism , Adenosine Triphosphatases/genetics , Cell Division/physiology , DNA, Bacterial/genetics , DNA-Binding Proteins/genetics , Escherichia coli/genetics , Escherichia coli Proteins/genetics , Protease La/genetics , Protein Domains
SELECTION OF CITATIONS
SEARCH DETAIL
...