Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 86
Filter
Add more filters










Publication year range
1.
Arch Pharm (Weinheim) ; 356(6): e2200592, 2023 Jun.
Article in English | MEDLINE | ID: mdl-37021586

ABSTRACT

The "magic methyl effect" strategy was used to design a series of 5-alkyl-2-pyrazol-oxazolidin-4-one derivatives as novel hepatitis B virus (HBV) capsid assembly modulators. Most of these compounds exhibited potent HBV inhibitory activities with low cytotoxicities in HepG2.2.15 cells. The most promising compounds 9d and 10b had single-digit nanomolar IC50 values with a high selectivity index. Compared with the lead compound (3.0%), they caused 15% and 18% decreases in HBe antigen secretion at 1.0 µM, respectively. In addition, compounds 9d and 10b possessed good pharmacokinetic profiles with oral bioavailability values of 56.1% and 48.9%, respectively. These results indicated that the two compounds were potential therapeutic agents for HBV infection.


Subject(s)
Capsid , Hepatitis B virus , Virus Assembly , Antiviral Agents/pharmacology , Structure-Activity Relationship , Capsid Proteins/pharmacology , Virus Replication
2.
J Comput Aided Mol Des ; 36(3): 193-203, 2022 03.
Article in English | MEDLINE | ID: mdl-35262811

ABSTRACT

We have identified novel HIV-1 capsid inhibitors targeting the PF74 binding site. Acting as the building block of the HIV-1 capsid core, the HIV-1 capsid protein plays an important role in the viral life cycle and is an attractive target for antiviral development. A structure-based virtual screening workflow for hit identification was employed, which includes docking 1.6 million commercially-available drug-like compounds from the ZINC database to the capsid dimer, followed by applying two absolute binding free energy (ABFE) filters on the 500 top-ranked molecules from docking. The first employs the Binding Energy Distribution Analysis Method (BEDAM) in implicit solvent. The top-ranked compounds are then refined using the Double Decoupling method in explicit solvent. Both docking and BEDAM refinement were carried out on the IBM World Community Grid as part of the FightAIDS@Home project. Using this virtual screening workflow, we identified 24 molecules with calculated binding free energies between - 6 and - 12 kcal/mol. We performed thermal shift assays on these molecules to examine their potential effects on the stability of HIV-1 capsid hexamer and found that two compounds, ZINC520357473 and ZINC4119064 increased the melting point of the latter by 14.8 °C and 33 °C, respectively. These results support the conclusion that the two ZINC compounds are primary hits targeting the capsid dimer interface. Our simulations also suggest that the two hit molecules may bind at the capsid dimer interface by occupying a new sub-pocket that has not been exploited by existing CA inhibitors. The possible causes for why other top-scored compounds suggested by ABFE filters failed to show measurable activity are discussed.


Subject(s)
Anti-HIV Agents , HIV-1 , Anti-HIV Agents/chemistry , Anti-HIV Agents/pharmacology , Capsid/metabolism , Capsid Proteins/metabolism , Capsid Proteins/pharmacology , Molecular Docking Simulation , Protein Binding , Solvents , Workflow
3.
Sci Rep ; 11(1): 16579, 2021 08 16.
Article in English | MEDLINE | ID: mdl-34400669

ABSTRACT

Recombinant MrNV capsid protein has been shown to effectively deliver plasmid DNA and dsRNA into Sf9 insect cells and shrimp tissues. To extend its application to cancer cell-targeting drug delivery, we created three different types of chimeric MrNV virus-like particles (VLPs) (R-MrNV, I-MrNV, and E-MrNV) that have specificity toward the epidermal growth factor receptor (EGFR), a cancer cell biomarker, by incorporating the EGFR-specific GE11 peptide at 3 different locations within the host cell recognition site of the capsid. All three chimeric MrNV-VLPs preserved the ability to form a mulberry-like VLP structure and to encapsulate EGFP DNA plasmid with an efficiency comparable to that previously reported for normal MrNV (N-MrNV). Compared to N-MrNV, the chimeric R-MrNV and E-MrNV carrying the exposed GE-11 peptide showed a significantly enhanced binding and internalization abilities that were specific towards EGFR expression in colorectal cancer cells (SW480). Specific targeting of chimeric MrNV to EGFR was proven by both EGFR silencing with siRNA vector and a competition with excess GE-11 peptide as well as the use of EGFR-negative colorectal cells (SW620) and breast cancer cells (MCF7). We demonstrated here that both chimeric R-MrNV and E-MrNV could be used to encapsulate cargo such as exogenous DNA and deliver it specifically to EGFR-positive cells. Our study presents the potential use of surface-modified VLPs of shrimp virus origin as nanocontainers for targeted cancer drug delivery.


Subject(s)
Adenocarcinoma/drug therapy , Capsid Proteins/pharmacology , Colorectal Neoplasms/drug therapy , Molecular Targeted Therapy , Neoplasm Proteins/antagonists & inhibitors , Nodaviridae/chemistry , Peptides/pharmacology , Recombinant Fusion Proteins/pharmacology , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Cell Line, Tumor , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , DNA, Recombinant/administration & dosage , DNA, Recombinant/genetics , Drug Compounding , Drug Delivery Systems , Drug Design , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/chemistry , ErbB Receptors/genetics , Humans , Neoplasm Proteins/chemistry , Neoplasm Proteins/genetics , Recombinant Fusion Proteins/genetics
4.
J Biol Chem ; 296: 100653, 2021.
Article in English | MEDLINE | ID: mdl-33845047

ABSTRACT

The transcription terminator Rho regulates many physiological processes in bacteria, such as antibiotic sensitivity, DNA repair, RNA remodeling, and so forth, and hence, is a potential antimicrobial target, which is unexplored. The bacteriophage P4 capsid protein, Psu, moonlights as a natural Rho antagonist. Here, we report the design of novel peptides based on the C-terminal region of Psu using phenotypic screening methods. The resultant 38-mer peptides, in addition to containing mutagenized Psu sequences, also contained plasmid sequences, fused to their C termini. Expression of these peptides inhibited the growth of Escherichia coli and specifically inhibited Rho-dependent termination in vivo. Peptides 16 and 33 exhibited the best Rho-inhibitory properties in vivo. Direct high-affinity binding of these two peptides to Rho also inhibited the latter's RNA-dependent ATPase and transcription termination functions in vitro. These two peptides remained functional even if eight to ten amino acids were deleted from their C termini. In silico modeling and genetic and biochemical evidence revealed that these two peptides bind to the primary RNA-binding site of the Rho hexamer near its subunit interfaces. In addition, the gene expression profiles of these peptides and Psu overlapped significantly. These peptides also inhibited the growth of Mycobacteria and inhibited the activities of Rho proteins from Mycobacterium tuberculosis, Xanthomonas, Vibrio cholerae, and Salmonella enterica. Our results showed that these novel anti-Rho peptides mimic the Rho-inhibition function of the ∼42-kDa dimeric bacteriophage P4 capsid protein, Psu. We conclude that these peptides and their C-terminal deletion derivatives could provide a basis on which to design novel antimicrobial peptides.


Subject(s)
Capsid Proteins/pharmacology , Drug Design , Escherichia coli Proteins/antagonists & inhibitors , Escherichia coli/metabolism , Mycobacterium tuberculosis/drug effects , Peptide Fragments/pharmacology , Terminator Regions, Genetic , Xanthomonas/drug effects , Amino Acid Sequence , Escherichia coli/genetics , Escherichia coli/growth & development , Mycobacterium tuberculosis/growth & development , Peptide Library , Plasmids , Protein Binding , Sequence Homology , Xanthomonas/growth & development
5.
Front Immunol ; 11: 606569, 2020.
Article in English | MEDLINE | ID: mdl-33343580

ABSTRACT

Cervical cancer remains a global health burden despite the introduction of highly effective vaccines for the prophylaxis of causative human papillomavirus infection (HPV). Current efforts to eradicate cervical cancer focus on the development of broadly protective, cost-effective approaches. HPV minor capsid protein L2 is being recognized as a promising alternative to the major capsid protein L1 because of its ability to induce responses against a wider range of different HPV types. However, a major limitation of L2 as a source of cross-neutralizing epitopes is its lower immunogenicity compared to L1 when assembled into VLPs. Various approaches have been proposed to overcome this limitation, we developed and tested ferritin-based bio-nanoparticles displaying tandemly repeated L2 epitopes from eight different HPV types grafted onto the surface of Pyrococcus furiosus thioredoxin (Pf Trx). Genetic fusion of the Pf Trx-L2(8x) module to P. furiosus ferritin (Pf Fe) did not interfere with ferritin self-assembly into an octahedral structure composed by 24 protomers. In guinea pigs and mice, the ferritin super-scaffolded, L2 antigen induced a broadly neutralizing antibody response covering 14 oncogenic and two non-oncogenic HPV types. Immune-responsiveness lasted for at least one year and the resulting antibodies also conferred protection in a cervico-vaginal mouse model of HPV infection. Given the broad organism distribution of thioredoxin and ferritin, we also verified the lack of cross-reactivity of the antibodies elicited against the scaffolds with human thioredoxin or ferritin. Altogether, the results of this study point to P. furiosus ferritin nanoparticles as a robust platform for the construction of peptide-epitope-based HPV vaccines.


Subject(s)
Alphapapillomavirus/drug effects , Antibodies, Viral/blood , Bacterial Proteins/pharmacology , Broadly Neutralizing Antibodies/blood , Capsid Proteins/pharmacology , Ferritins/pharmacology , Oncogene Proteins, Viral/pharmacology , Papillomavirus Infections/prevention & control , Papillomavirus Vaccines/pharmacology , Alphapapillomavirus/genetics , Alphapapillomavirus/immunology , Animals , Antibody Specificity , Bacterial Proteins/genetics , Bacterial Proteins/immunology , Capsid Proteins/genetics , Capsid Proteins/immunology , Epitopes , Female , Ferritins/genetics , Ferritins/immunology , Guinea Pigs , Immunization , Immunogenicity, Vaccine , Mice, Inbred BALB C , Nanoparticles , Oncogene Proteins, Viral/genetics , Oncogene Proteins, Viral/immunology , Papillomavirus Infections/blood , Papillomavirus Infections/immunology , Papillomavirus Vaccines/genetics , Papillomavirus Vaccines/immunology , Sf9 Cells , Spodoptera , Thioredoxins/genetics , Thioredoxins/immunology , Thioredoxins/pharmacology , Time Factors , Vaccines, DNA/pharmacology
6.
J Virol ; 94(22)2020 10 27.
Article in English | MEDLINE | ID: mdl-32847863

ABSTRACT

Induction of necroptosis by mammalian reovirus requires both type I interferon (IFN)-signaling and viral replication events that lead to production of progeny genomic double-stranded RNA (dsRNA). The reovirus outer capsid protein µ1 negatively regulates reovirus-induced necroptosis by limiting RNA synthesis. To determine if the outer capsid protein σ3, which interacts with µ1, also functions in regulating necroptosis, we used small interfering RNA (siRNA)-mediated knockdown. Similarly to what was observed in diminishment of µ1 expression, knockdown of newly synthesized σ3 enhances necroptosis. Knockdown of σ3 does not impact reovirus RNA synthesis. Instead, this increase in necroptosis following σ3 knockdown is accompanied by an increase in IFN production. Furthermore, ectopic expression of σ3 is sufficient to block IFN expression following infection. Surprisingly, the capacity of σ3 protein to bind dsRNA does not impact its capacity to diminish production of IFN. Consistent with this, infection with a virus harboring a mutation in the dsRNA binding domain of σ3 does not result in enhanced production of IFN or necroptosis. Together, these data suggest that σ3 limits the production of IFN to control innate immune signaling and necroptosis following infection through a mechanism that is independent of its dsRNA binding capacity.IMPORTANCE We use mammalian reovirus as a model to study how virus infection modulates innate immune signaling and cell death induction. Here, we sought to determine how viral factors regulate these processes. Our work highlights a previously unknown role for the reovirus outer capsid protein σ3 in limiting the induction of a necrotic form of cell death called necroptosis. Induction of cell death by necroptosis requires production of interferon. The σ3 protein limits the induction of necroptosis by preventing excessive production of interferon following infection.


Subject(s)
Capsid Proteins/metabolism , Cell Death/drug effects , Interferons/metabolism , Reoviridae/physiology , Animals , Capsid Proteins/genetics , Capsid Proteins/pharmacology , Cell Line , HEK293 Cells , HeLa Cells , Humans , Mice , RNA, Double-Stranded/genetics , RNA, Small Interfering/metabolism , Reoviridae/genetics , Signal Transduction , Virus Replication
7.
Protein Expr Purif ; 175: 105687, 2020 11.
Article in English | MEDLINE | ID: mdl-32681952

ABSTRACT

A promising candidate for tumor targeted toxins is the chicken anemia-derived protein apoptin that induces tumor-specific apoptosis. It was aimed to design a novel apoptin-based targeted toxin by genetic fusion of apoptin with the tumor-directed ligand epidermal growth factor (EGF) using Escherichia coli as expression host. However, apoptin is highly hydrophobic and tends to form insoluble aggregates. Therefore, three different apoptin-EGF variants were generated. The fusion protein hexa-histidine (His)-apoptin-EGF (HAE) was expressed in E. coli and purified under denaturing conditions due to inclusion bodies. The protein solubility was improved by maltose-binding protein (MBP) or glutathione S-transferase. The protein MBP-apoptin-EGFHis (MAEH) was found favorable as a targeted toxin regarding final yield (4-6 mg/L) and stability. MBP was enzymatically removed using clotting factor Xa, which resulted in low yield and poor separation. MAEH was tested on target and non-target cell lines. The targeted tumor cell line A431 showed significant toxicity with an IC50 of 69.55 nM upon incubation with MAEH while fibroblasts and target receptor-free cells remained unaffected. Here we designed a novel EGF receptor targeting drug with high yield, purity and stability.


Subject(s)
Antineoplastic Agents , Capsid Proteins , Epidermal Growth Factor , Neoplasms/drug therapy , Recombinant Fusion Proteins , Animals , Antineoplastic Agents/isolation & purification , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , Capsid Proteins/biosynthesis , Capsid Proteins/genetics , Capsid Proteins/isolation & purification , Capsid Proteins/pharmacology , Cell Line, Tumor , Epidermal Growth Factor/biosynthesis , Epidermal Growth Factor/genetics , Epidermal Growth Factor/isolation & purification , Epidermal Growth Factor/pharmacology , Humans , Mice , NIH 3T3 Cells , Neoplasms/metabolism , Neoplasms/pathology , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/isolation & purification , Recombinant Fusion Proteins/pharmacology
8.
Proc Natl Acad Sci U S A ; 116(47): 23437-23443, 2019 11 19.
Article in English | MEDLINE | ID: mdl-31685638

ABSTRACT

Antibiotic resistance has become one of the major threats to global health. Photodynamic inactivation (PDI) develops little antibiotic resistance; thus, it becomes a promising strategy in the control of bacterial infection. During a PDI process, light-induced reactive oxygen species (ROS) damage the membrane components, leading to the membrane rupture and bacteria death. Due to the short half-life and reaction radius of ROS, achieving the cell-membrane intercalation of photosensitizers is a key challenge for PDI of bacteria. In this work, a tetraphenylethylene-based discrete organoplatinum(II) metallacycle (1) acts as a photosensitizer with aggregation-induced emission. It self-assembles with a transacting activator of transduction (TAT) peptide-decorated virus coat protein (2) through electrostatic interactions. This assembly (3) exhibits both ROS generation and strong membrane-intercalating ability, resulting in significantly enhanced PDI efficiency against bacteria. By intercalating in the bacterial cell membrane or entering the bacteria, assembly 3 decreases the survival rate of gram-negative Escherichia coli to nearly zero and that of gram-positive Staphylococcus aureus to ∼30% upon light irradiation. This study has wide implications from the generation of multifunctional nanomaterials to the control of bacterial infection, especially for gram-negative bacteria.


Subject(s)
Acids, Acyclic/pharmacology , Anti-Bacterial Agents/pharmacology , Capsid Proteins/pharmacology , Cell Membrane/drug effects , Escherichia coli/drug effects , Gene Products, tat/pharmacology , Organoplatinum Compounds/pharmacology , Photosensitizing Agents/pharmacology , Staphylococcus aureus/drug effects , Stilbenes/pharmacology , Acids, Acyclic/chemistry , Electron Spin Resonance Spectroscopy , Escherichia coli/radiation effects , Escherichia coli/ultrastructure , Microscopy, Electron , Photochemotherapy/methods , Reactive Oxygen Species , Staphylococcus aureus/radiation effects , Staphylococcus aureus/ultrastructure , Static Electricity , Tobacco Mosaic Virus
9.
Vet Microbiol ; 235: 295-300, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31383316

ABSTRACT

Tembusu virus (TMUV), a member of the genus flavivirus, primarily causes egg-drop syndrome in ducks and is associated with low disease mortality but high morbidity. The commercially available live vaccines for treating TMUV currently include the main WF100, HB, and FX2010-180P strains, and efficient treatment and/or preventative measures are still urgently needed. Capsid-targeted viral inactivation (CTVI) is a conceptually powerful new antiviral strategy that is based on two proteins from the capsid protein of a virus and a crucial effector molecule. The effector molecule can destroy the viral DNA/RNA or interfere with the proper folding of key viral proteins, while the capsid protein mainly plays a role in viral integration and assembly; the fusion proteins are incorporated into virions during packaging. This study aimed to explore the potential use of this strategy in duck TMUV. Our results revealed that these fusion proteins can be expressed in susceptible BHK21 cells without cytotoxicity and possess excellent Ca2+-dependent nuclease activity, and their expression is also detectable in DF-1 cells. Compared to those in the negative controls (BHK21 and BHK21/pcDNA3.1(+) cells), the numbers of viral RNA copies in TMUV-infected BHK21/Cap-SNase and BHK21/Cap-Linker-SNase cells were reduced by 48 h, and the effect of Cap-Linker-SNase was superior to that of Cap-SNase. As anticipated, these results suggest that these fusion proteins contribute to viral resistance to treatment. Thus, CTVI might be applicable for TMUV inhibition as a novel antiviral therapeutic candidate during viral infection.


Subject(s)
Capsid Proteins/pharmacology , Micrococcal Nuclease/pharmacology , Viral Fusion Proteins/pharmacology , Virus Inactivation/drug effects , Virus Replication/drug effects , Animals , Capsid Proteins/genetics , Cell Line , Ducks , Flavivirus , Flavivirus Infections/drug therapy , Flavivirus Infections/virology , Micrococcal Nuclease/genetics
10.
ACS Appl Mater Interfaces ; 10(42): 35745-35759, 2018 Oct 24.
Article in English | MEDLINE | ID: mdl-30360122

ABSTRACT

Cervical cancer remains the second-most prevalent female malignancy around the world, leading to a great majority of cancer-related mortality that occurs mainly in developing countries. Developing an effective and low-cost vaccine against human papillomavirus (HPV) infection, especially in medically underfunded areas, is urgent. Compared with vaccines based on HPV L1 viruslike particles (VLPs) in the market, recombinant HPV L1 pentamer expressed in Escherichia coli represents a promising and potentially cost-effective vaccine for preventing HPV infection. Hybrid particles comprising a polymer core and lipid shell have shown great potential compared to conventional aluminum salts adjuvant and is urgently needed for HPV L1 pentamer vaccines. It is well-reported that particle sizes are crucial in regulating immune responses. Nevertheless, reports on the relationship between the particulate size and the resultant immune response have been in conflict, and there is no answer to how the size of particles regulates specific immune response for HPV L1 pentamer-based candidate vaccines. Here, we fabricated HPV 16 L1 pentamer-loaded poly(d,l-lactide- co-glycolide) (PLGA)/lecithin hybrid particles with uniform sizes (0.3, 1, and 3 µm) and investigated the particle size effects on antigen release, activation of lymphocytes, dendritic cells (DCs) activation and maturation, follicular helper CD4+ T (TFH) cells differentiation, and release of pro-inflammatory cytokines and chemokines. Compared with the other particle sizes, 1 µm particles induced more powerful antibody protection and yielded more persistent antibody responses, as well as more heightened anamnestic responses upon repeat vaccination. The superior immune responses might be attributed to sustainable antigen release and robust antigen uptake and transport and then further promoted a series of cascade reactions, including enhanced DCs maturation, increased lymphocytes activation, and augmented TFH cells differentiation in draining lymph nodes (DLNs). Here, a powerful and economical platform for HPV vaccine and a comprehensive understanding of particle size effect on immune responses for HPV L1 pentamer-based candidate vaccines are provided.


Subject(s)
Capsid Proteins , Human papillomavirus 16/immunology , Immunity, Cellular , Nanoparticles/chemistry , Oncogene Proteins, Viral , Papillomavirus Vaccines , Vaccination , Animals , B-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/immunology , Capsid Proteins/chemistry , Capsid Proteins/immunology , Capsid Proteins/pharmacology , Dendritic Cells/immunology , Female , Humans , Mice , Mice, Inbred BALB C , Oncogene Proteins, Viral/chemistry , Oncogene Proteins, Viral/immunology , Oncogene Proteins, Viral/pharmacology , Papillomavirus Vaccines/chemistry , Papillomavirus Vaccines/immunology , Papillomavirus Vaccines/pharmacology , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer/pharmacology
11.
J Biol Chem ; 293(39): 15221-15232, 2018 09 28.
Article in English | MEDLINE | ID: mdl-30108178

ABSTRACT

Porcine circovirus type 2 (PCV2) is one of the smallest, nonenveloped, single-stranded DNA viruses. The PCV2 capsid protein (Cap) is the sole viral structural protein and main antigenic determinant. Previous sequence analysis has revealed that the N terminus of the PCV2 Cap contains a nuclear localization signal (NLS) enriched in positively charged residues. Here, we report that PCV2's NLS can function as a cell-penetrating peptide (CPP). We observed that this NLS can carry macromolecules, e.g. enhanced GFP (EGFP), into cells when they are fused to the NLS, indicating that it can function as a CPP, similar to the classical CPP derived from HIV type 1 transactivator of transcription protein (HIV TAT). We also found that the first 17 residues of the NLS (NLS-A) have a key role in cellular uptake. In addition to entering cells via multiple endocytic processes, NLS-A was also rapidly internalized via direct translocation enabled by increased membrane permeability and was evenly distributed throughout cells when its concentration in cell cultures was ≥10 µm Of note, cellular NLS-A uptake was ∼10 times more efficient than that of HIV TAT. We inferred that the externalized NLS of the PCV2 Cap may accumulate to a high concentration (≥10 µm) at a local membrane area, increasing membrane permeability to facilitate viral entry into the cell to release its genome into a viral DNA reproduction center. We conclude that NLS-A has potential as a versatile vehicle for shuttling foreign molecules into cells, including pharmaceuticals for therapeutic interventions.


Subject(s)
Capsid Proteins/genetics , Cell-Penetrating Peptides/genetics , Nuclear Localization Signals/genetics , rev Gene Products, Human Immunodeficiency Virus/genetics , Animals , Antibodies, Viral/chemistry , Antibodies, Viral/immunology , Capsid Proteins/chemistry , Capsid Proteins/pharmacology , Cell-Penetrating Peptides/chemistry , Cell-Penetrating Peptides/pharmacology , Circovirus/chemistry , Circovirus/genetics , DNA, Single-Stranded/chemistry , DNA, Single-Stranded/genetics , DNA, Viral/chemistry , DNA, Viral/genetics , DNA, Viral/pharmacology , Epitopes/chemistry , Epitopes/genetics , Epitopes/immunology , Humans , Nuclear Localization Signals/chemistry , Swine , rev Gene Products, Human Immunodeficiency Virus/chemistry
12.
J Allergy Clin Immunol ; 142(6): 1808-1817.e3, 2018 12.
Article in English | MEDLINE | ID: mdl-29522849

ABSTRACT

BACKGROUND: Altered epithelial physical and functional barrier properties along with TH1/TH2 immune dysregulation are features of allergic asthma. Regulation of junction proteins to improve barrier function of airway epithelial cells has the potential for alleviation of allergic airway inflammation. OBJECTIVE: We sought to determine the immunomodulatory effect of knob protein of the adenoviral capsid on allergic asthma and to investigate its mechanism of action on airway epithelial junction proteins and barrier function. METHODS: Airway inflammation, including junction protein expression, was evaluated in allergen-challenged mice with and without treatment with knob. Human bronchial epithelial cells were exposed to knob, and its effects on expression of junction proteins and barrier integrity were determined. RESULTS: Administration of knob to allergen-challenged mice suppressed airway inflammation (eosinophilia, airway hyperresponsiveness, and IL-5 levels) and prevented allergen-induced loss of airway epithelial occludin and E-cadherin expression. Additionally, knob decreased expression of TH2-promoting inflammatory mediators, specifically IL-33, by murine lung epithelial cells. At a cellular level, treatment of human bronchial epithelial cells with knob activated c-Jun N-terminal kinase, increased expression of occludin and E-cadherin, and enhanced epithelial barrier integrity. CONCLUSION: Increased expression of junction proteins mediated by knob leading to enhanced epithelial barrier function might mitigate the allergen-induced airway inflammatory response, including asthma.


Subject(s)
Capsid Proteins/pharmacology , Capsid Proteins/therapeutic use , Epithelial Cells/drug effects , Adenoviridae , Aged , Animals , Bronchi/cytology , Bronchoalveolar Lavage Fluid/immunology , Cadherins/metabolism , Cell Line , Cytokines/immunology , Eosinophilia/immunology , Epithelial Cells/metabolism , Female , Humans , Male , Mice, Inbred BALB C , Middle Aged , Occludin/metabolism , Recombinant Proteins/pharmacology , Recombinant Proteins/therapeutic use , Respiratory Hypersensitivity/drug therapy , Respiratory Hypersensitivity/immunology
13.
Curr Med Chem ; 25(30): 3540-3559, 2018.
Article in English | MEDLINE | ID: mdl-29521208

ABSTRACT

BACKGROUND: Research in the field of antitumor chemotherapeutics pursues a key issue, drug selectivity for cancer cells. In the last 20 years, a group of proteins has attracted scientific interest as cancer chemotherapeutics due to their ability to specifically kill cancer cells while leaving normal cells undamaged. One of these proteins is apoptin. METHODS: In this study, the recent available literature regarding cell death mechanisms induced by apoptin has been reviewed. Delivering this drug to tumor cells is a challenge because it spontaneously forms soluble non-covalent aggregates. This led us to include in this review the different approaches for obtaining the maximum efficiency of apoptin entry to cancer cells. RESULTS: This review provides an up-to-date summary of the mechanisms by which apoptin induces selective apoptosis in tumor cells while leaving normal cells undamaged. It highlights the relationship between the apoptosis mechanism induced by this protein and its functional motifs. Apoptin has been described as an intrinsically disordered protein, which explains its ability to interact with multiple partners and affect multiple pathways inside the cell. Characterization of the different partners and pathways induced by apoptin has begun to shed light on the molecular basis of apoptin's tumor-selective cytotoxicity. CONCLUSION: The findings confirm the interest in apoptin as a potentially safe antitumor drug. Research still needed to be conducted to find an effective way to deliver apoptin for use in clinics.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Capsid Proteins/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/metabolism , Capsid Proteins/chemistry , Capsid Proteins/metabolism , Cell Cycle Checkpoints/drug effects , Cyclin-Dependent Kinases/chemistry , Cyclin-Dependent Kinases/metabolism , DNA/chemistry , DNA/metabolism , DNA Damage/drug effects , Drug Carriers/chemistry , Humans , Neoplasms/metabolism , Neoplasms/pathology
14.
Biochem Biophys Res Commun ; 498(1): 221-227, 2018 03 25.
Article in English | MEDLINE | ID: mdl-29501489

ABSTRACT

Breast cancer is the leading cause of cancer incidence and cancer-related mortality among women and is becoming a major public health problem around the world. The current study aims to investigate the possible role and mechanism of recombinant Apoptin (rApoptin), a potential anticancer candidate that minimally impacts normal cells, in the breast cancer cell proliferation and apoptosis in vitro and in vivo. We found that rApoptin could effectively inhibit the proliferation and apoptosis in MCF-7 and MDA-MB-231 cells in vitro, which was further confirmed by flow cytometry analysis. Apoptin partially inhibited MCF-7 cell xenograft tumor development in vivo. Furthermore, we found via western blot that rApoptin-induced apoptosis in MCF-7 and MDA-MB-231 cells was associated with the phosphorylation of Nur77 (p-Nur77) and Akt (p-Akt). In addition, compared with the control groups, rApoptin-treated tissues showed significantly higher expression of Bax and Cyt c while Bcl-2 expression was decreased by rApoptin treatment. Together, our results are the first to demonstrate that rApoptin was able to effectively induce breast cancer cell apoptosis both in vitro and in vivo and that this activity could be regulated by the phosphorylation of Nur77 and Akt and the mitochondrial pathway. Our findings highlight the potential application of rApoptin as a breast cancer treatment.


Subject(s)
Apoptosis/drug effects , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Capsid Proteins/pharmacology , Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Recombinant Proteins/pharmacology , Animals , Carcinogenesis/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Humans , Mice, Nude , Phosphorylation/drug effects
15.
J Bacteriol ; 200(1)2018 01 01.
Article in English | MEDLINE | ID: mdl-29038252

ABSTRACT

Rho is a hexameric molecular motor that functions as a conserved transcription terminator in the majority of bacterial species and is a potential drug target. Psu is a bacteriophage P4 capsid protein that inhibits Escherichia coli Rho by obstructing its ATPase and translocase activities. In this study, we explored the anti-Rho activity of Psu for Rho proteins from different pathogens. Sequence alignment and homology modeling of Rho proteins from pathogenic bacteria revealed the conserved nature of the Psu-interacting regions in all these proteins. We chose Rho proteins from various pathogens, including Mycobacterium smegmatis, Mycobacterium bovis, Mycobacterium tuberculosis, Xanthomonas campestris, Xanthomonas oryzae, Corynebacterium glutamicum, Vibrio cholerae, Salmonella enterica, and Pseudomonas syringae The purified recombinant Rho proteins of these organisms showed variable rates of ATP hydrolysis on poly(rC) as the substrate and were capable of releasing RNA from the E. coli transcription elongation complexes. Psu was capable of inhibiting these two functions of all these Rho proteins. In vivo pulldown assays revealed direct binding of Psu with many of these Rho proteins. In vivo expression of psu induced killing of M. smegmatis, M. bovis, X. campestris, and E. coli expressing S. enterica Rho indicating Psu-induced inhibition of Rho proteins of these strains under physiological conditions. We propose that the "universal" inhibitory function of the Psu protein against the Rho proteins from both Gram-negative and Gram-positive bacteria could be useful for designing peptides with antimicrobial functions and that these peptides could contribute to synergistic antibiotic treatment of the pathogens by compromising the Rho functions.IMPORTANCE Bacteriophage-derived protein factors modulating different bacterial processes could be converted into unique antimicrobial agents. Bacteriophage P4 capsid protein Psu is an inhibitor of the E. coli transcription terminator Rho. Here we show that apart from antagonizing E. coli Rho, Psu is able to inhibit Rho proteins from various phylogenetically unrelated Gram-negative and Gram-positive pathogens. Upon binding to these Rho proteins, Psu inhibited them by affecting their ATPase and RNA release functions. The expression of Psu in vivo kills various pathogens, such as Mycobacterium and Xanthomonas species. Hence, Psu could be useful for identifying peptide sequences with anti-Rho activities and might constitute part of synergistic antibiotic treatment against pathogens.


Subject(s)
Capsid Proteins/pharmacology , Gram-Negative Bacteria/drug effects , Gram-Positive Bacteria/drug effects , Rho Factor/antagonists & inhibitors , Transcription Termination, Genetic/drug effects , Adenosine Triphosphatases/antagonists & inhibitors , Bacteriophages/chemistry , Capsid Proteins/genetics , Capsid Proteins/metabolism , DNA-Directed RNA Polymerases/metabolism , Escherichia coli/genetics , Escherichia coli/pathogenicity , Gram-Negative Bacteria/genetics , Gram-Negative Bacteria/pathogenicity , Gram-Positive Bacteria/genetics , Gram-Positive Bacteria/pathogenicity , Mycobacterium/drug effects , Mycobacterium tuberculosis/drug effects , Mycobacterium tuberculosis/genetics , Mycobacterium tuberculosis/pathogenicity , Protein Binding , Recombinant Proteins/metabolism , Rho Factor/genetics , Rho Factor/metabolism , Sequence Homology, Nucleic Acid , Vibrio cholerae/drug effects , Vibrio cholerae/genetics , Vibrio cholerae/pathogenicity , Xanthomonas/drug effects
16.
Biomed Res Int ; 2017: 7658970, 2017.
Article in English | MEDLINE | ID: mdl-29119112

ABSTRACT

Improving vaccine immunogenicity by targeting antigens to dendritic cells has recently emerged as a new design strategy in vaccine development. In this study, the VP1 gene of foot-and-mouth disease virus (FMDV) serotype A was fused with the gene encoding human immunodeficiency virus (HIV) membrane glycoprotein gp120 or C2-V3 domain of hepatitis C virus (HCV) envelope glycoprotein E2, both of which are DC-SIGN-binding glycoproteins. After codon optimization, the VP1 protein and the two recombinant VP1-gp120 and VP1-E2 fusion proteins were expressed in Sf9 insect cells using the insect cell-baculovirus expression system. Western blotting showed that the VP1 protein and two recombinant VP1-gp120 and VP1-E2 fusion proteins were correctly expressed in the Sf9 insect cells and had good reactogenicity. Guinea pigs were then immunized with the purified proteins, and the resulting humoral and cellular immune responses were analyzed. The VP1-gp120 and VP1-E2 fusion proteins induced significantly higher specific anti-FMDV antibody levels than the VP1 protein and stronger cell-mediated immune responses. This study provides a new perspective for the development of novel FMDV subunit vaccines.


Subject(s)
Capsid Proteins , Foot-and-Mouth Disease Virus , HIV Envelope Protein gp120 , Immunogenicity, Vaccine , Recombinant Fusion Proteins , Viral Vaccines , Animals , Capsid Proteins/biosynthesis , Capsid Proteins/genetics , Capsid Proteins/immunology , Capsid Proteins/pharmacology , Cell Adhesion Molecules , Foot-and-Mouth Disease Virus/genetics , Foot-and-Mouth Disease Virus/immunology , Guinea Pigs , HIV Envelope Protein gp120/biosynthesis , HIV Envelope Protein gp120/genetics , HIV Envelope Protein gp120/immunology , HIV Envelope Protein gp120/pharmacology , HIV-1/genetics , Hepacivirus , Lectins, C-Type , Receptors, Cell Surface , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/pharmacology , Sf9 Cells , Spodoptera , Viral Vaccines/biosynthesis , Viral Vaccines/genetics , Viral Vaccines/pharmacology
17.
Nanomedicine ; 13(8): 2475-2484, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28842374

ABSTRACT

Opportunistic bacteria that cause life-threatening infections are still a central problem associated with a healthcare setting. Bacteriophage capsid immobilization on nanostructured polymers maximizes its tail exposure and looks promising in applications toward skin-infections as alternative to antibiotics standardly used. The main goal of this work was to investigate the covalent immobilization of vB_Pae_Kakheti25 bacteriophage capsid on polycaprolactone (PCL) nanofibers (non-woven textile), as a potential effective antimicrobial, laundry resistant and non-toxic dressing for biomedical use. Surface analyses showed that the immobilization of vB_Pae_Kakheti25 bacteriophage capsid on PCL nanofibres oriented bacteriophage tails to interact with bacteria. Furthermore, antimicrobial assays showed a very effective 6 log bacterial reduction, which was equivalent to 99.9999%, after immediate and 2 hours of contact, even following 25 washing cycles (due to covalent bond). The activity of PCL-vB_Pae_Kakheti25 against P. aeruginosa was immediate and its reduction was complete.


Subject(s)
Anti-Infective Agents/pharmacology , Bacteriophages , Bandages , Capsid Proteins/pharmacology , Immobilized Proteins/pharmacology , Wound Infection/prevention & control , Animals , Anti-Infective Agents/chemistry , BALB 3T3 Cells , Bacteriophages/chemistry , Bandages/microbiology , Bandages/virology , Capsid Proteins/chemistry , Cell Line , Humans , Immobilized Proteins/chemistry , Mice , Models, Molecular , Nanofibers/chemistry , Nanofibers/ultrastructure , Polyesters/chemistry , Pseudomonas Infections/prevention & control , Pseudomonas aeruginosa/drug effects
18.
Arch Microbiol ; 199(8): 1121-1131, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28444417

ABSTRACT

Chlamydia trachomatis infection is one of the most prevalent sexually transmitted diseases. Our research pertains to the inhibitory effect and molecular mechanism of the chlamydiaphage capsid protein VP1 on the growth of Chlamydia trachomatis. In this research, the capsid protein VP1 of the guinea-pig conjunctivitis chlamydiaphage phiCPG1 was expressed, purified and identified, and then, it was applied to the cultivation of different serovars of Chlamydia trachomatis and Chlamydia psittaci. The inhibitory effect was observed in each serovar of Chlamydia trachomatis (D, E, F, G, H, I, K, and L2) and Chlamydia psittaci inoculated with VP1 protein. The inhibition affection of VP1 on the growth of Chlamydia trachomatis was caused by the changes of expressions of some related proteins including 36 proteins up-regulated and 81 proteins down-regulated in the development cycle of Ct through the label-free test, and the transcription levels of these proteins, including Hc1, pmpD, and MOMP, were confirmed by RT-PCR. It provides information that is essential for understanding the mechanism of chlamydiaphage capsid protein VP1 on chlamydia and a new direction for further clinical treatment of chlamydial infection.


Subject(s)
Bacterial Outer Membrane Proteins/metabolism , Bacteriophages/metabolism , Capsid Proteins/pharmacology , Chlamydia trachomatis/drug effects , Chlamydia trachomatis/growth & development , Animals , Chlamydia Infections/drug therapy , Chlamydia trachomatis/virology , Guinea Pigs , Humans , Up-Regulation
19.
J Huazhong Univ Sci Technolog Med Sci ; 37(1): 115-121, 2017 Feb.
Article in English | MEDLINE | ID: mdl-28224434

ABSTRACT

The researches on chlamydia in recent years show that chlamydia bacteriophage may be a potential and effective means to solve the clinical infection of chlamydia trachomatis (Ct). We investigated the biological effect of chlamydiaphage phiCPG1 capsid protein Vp1 on Ct both in McCoy cells and genital tract of mice. Different concentrations of Vp1 were co-incubated with Ct E serotype strain in McCoy cells. Female BALB/c mice were used to establish Ct E strain-induced urogenital infection model. They were randomly divided into five groups and given different treatments on the fifth day after Ct inoculation. Animals in groups 1 and 2 were given 30 µL different concentrations of Vp1 in the genital tract respectively, those in group 3 were intramuscularly injected with 30 µL Vp1, those in the infected group did not receive any intervention, and those in the control group received 30 µL PBS in the genital tract. The vaginal discharge was collected to identify the live chlamydia by cell culture and gene fragment by real time PCR different days after infection. Inhibition rate of 100 µg/mL and 50 µg/mL Vp1 proteins against Ct E strain in the McCoy cell cultures was 91% and 79% respectively. The number of intracellular Ct inclusion in the McCoy cells co-cultured with vaginal discharge of group 1 and group 2 was less than in the infected group, and that in group 1 was less than in group 2, on the 7th day after Ct inoculation. Real-time PCR showed that chlamydia concentration of the vaginal discharge in group 2 was lower than in the infected group, and that in group 1 was lower than in group 2 on the 10th day. It was suggested that Vp1 capsid proteins had inhibitory effect on the proliferation of Ct serovar E strain in cell culture and mouse genital tract.


Subject(s)
Bacteriophages/metabolism , Capsid Proteins/administration & dosage , Chlamydia Infections/prevention & control , Chlamydia trachomatis/drug effects , Genitalia, Female/virology , Animals , Capsid Proteins/pharmacology , Cell Line , Chlamydia Infections/microbiology , Chlamydia trachomatis/genetics , Chlamydia trachomatis/virology , Disease Models, Animal , Female , In Vitro Techniques , Mice , Mice, Inbred BALB C , Random Allocation
20.
Invest New Drugs ; 35(3): 260-268, 2017 06.
Article in English | MEDLINE | ID: mdl-28120180

ABSTRACT

Apoptin is a nonstructural protein encoded by one of the three open reading frames of the chicken anemia virus genome. It has attracted a great deal of interest due to its ability to induce apoptosis in multiple transformed and malignant mammalian cell lines without affecting primary and non-transformed cells. However, the use of Apoptin as an anticancer drug is restricted by its strong tendency to aggregate. A number of methods to overcome this problem have been proposed, including transduction techniques to deliver the Apoptin gene into tumor cells, but all such methods have certain drawbacks. Here we describe that a truncated variant of Apoptin, lacking residues 1 to 43, is a soluble, non-aggregating protein that maintains most of the biological properties of wild-type Apoptin when transfected into cells. We show that the cytotoxic effect of this variant is also present when it is added exogenously to cancer cells, but not to normal cells. In addition to the interest this protein has attracted as a promising therapeutic strategy, it is also an excellent model to study the structural properties of Apoptin and how they relate to its mechanism of action.


Subject(s)
Antineoplastic Agents/pharmacology , Capsid Proteins/chemistry , Capsid Proteins/pharmacology , Apoptosis/drug effects , Capsid Proteins/genetics , Cell Line , Cell Line, Tumor , DNA/metabolism , Escherichia coli/genetics , Humans , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...