Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 64
Filter
1.
Front Neurol ; 11: 596632, 2020.
Article in English | MEDLINE | ID: mdl-33329349

ABSTRACT

Magnetic nanoparticles (MNPs) have potential for enhancing drug delivery in selected cancer patients, including those which have cells that have disseminated within cerebrospinal fluid (CSF) pathways. Here, we present data related to the creation and in vitro use of new two-part MNPs consisting of magnetic gold-iron alloy cores which have streptavidin binding sites, and are coated with biotinylated etoposide. Etoposide was chosen due to its previous use in the CSF and ease of biotinylation. Etoposide magnetic nanoparticles ("Etop-MNPs") were characterized by several different methods, and moved at a distance by surface-walking of MNP clusters, which occurs in response to a rotating permanent magnet. Human cell lines including D283 (medulloblastoma), U138 (glioblastoma), and H2122 (lung adenocarcinoma) were treated with direct application of Etop-MNPs (and control particles), and after remote particle movement. Cell viability was determined by MTT assay and trypan blue exclusion. Results indicated that the biotinylated etoposide was successfully bound to the base MNPs, with the hybrid particle attaining a maximum velocity of 0.13 ± 0.018 cm/sec. Etop-MNPs killed cancer cells in a dose-dependent fashion, with 50 ± 6.8% cell killing of D283 cells (for example) with 24 h of treatment after remote targeting. U138 and H2122 cells were found to be even more susceptible to the killing effect of Etop-MNPs than D283 cells. These findings indicate that the novel Etop-MNPs have a cytotoxic effect, and can be moved relatively rapidly at physiologic distances, using a rotating magnet. While further testing is needed, intrathecal administration of Etop-MNPs holds promise for magnetically-enhanced eradication of cancer cells distributed within CSF pathways, particularly if given early in the course of the disease.

2.
Int J Nanomedicine ; 15: 3071-3085, 2020.
Article in English | MEDLINE | ID: mdl-32431502

ABSTRACT

PURPOSE: Recently, two-dimensional (2D) nanomaterials are gaining tremendous attention as novel antibacterial platforms to combat against continuously evolving antimicrobial resistance levels. Among the family of 2D nanomaterials, black phosphorus (BP) nanosheets have demonstrated promising potential for biomedical applications. However, there is a need to gain nanoscale insights of the antibacterial activity of BP nanosheets which lies at the center of technical challenges. METHODS: Ultra-large BP nanosheets were synthesized by liquid-exfoliation method in the eco-friendly deoxygenated water. Synthesized BP nanosheets were characterized by TEM, AFM, and Raman spectroscopy techniques and their chemical stability was evaluated by EDS and EELS elemental analysis. The antibacterial activity of BP nanosheets was evaluated at nanoscale by the ultramicrotome TEM technique. Further, HAADF-STEM image and EDS elemental line map of the damaged bacterium were utilized to analyze the presence of diagnostic ions. Supportive SEM and ATR-FTIR studies were carried out to confirm the bacterial cell wall damage. In vitro colony counting method was utilized to evaluate the antibacterial performance of ultra-large BP nanosheets. RESULTS: Elemental EELS and EDS analysis of BP nanosheets stored in deoxygenated water confirmed the absence of oxygen peak. TEM studies indicate the various events of bacterial cell damage with the lost cellular metabolism and structural integrity. Colony counting test results show that as-synthesized BP nanosheets (100 µg/mL) can kill ~95% bacteria within 12 hours. CONCLUSION: TEM studies demonstrate the various events of E. coli membrane damage and the loss of structural integrity. These events include the BP nanosheets interaction with the bacterial cell wall, cytoplasmic leakage, detachment of cytoplasm from the cell membrane, reduced density of lipid bilayer and agglomerated DNA structure. The EDS elemental line mapping of the damaged bacterium confirms the disrupted cell membrane permeability and the lost cellular metabolism. SEM micrographs and ATR-FTIR supportive results confirm the bacterial cell wall damage.


Subject(s)
Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Escherichia coli/drug effects , Nanostructures/chemistry , Phosphorus/chemistry , Cell Wall/drug effects , Cell Wall/ultrastructure , Escherichia coli/ultrastructure , Microbial Sensitivity Tests , Microscopy, Atomic Force , Microscopy, Electron, Transmission , Spectrometry, X-Ray Emission , Spectroscopy, Fourier Transform Infrared , Spectrum Analysis, Raman , Water/chemistry
3.
Int J Nanomedicine ; 15: 1929-1938, 2020.
Article in English | MEDLINE | ID: mdl-32256069

ABSTRACT

BACKGROUND: Nanoscale surface roughness has been suggested to have antibacterial and antifouling properties. Several existing models have attempted to explain the antibacterial mechanism of nanoscale rough surfaces without direct observation. Here, conventional and liquid-cell TEM are implemented to observe nanoscale bacteria/surface roughness interaction. The visualization of such interactions enables the inference of possible antibacterial mechanisms. METHODS AND RESULTS: Nanotextures are synthesized on biocompatible polymer microparticles (MPs) via plasma etching. Both conventional and liquid-phase transmission electron microscopy observations suggest that these MPs may cause cell lysis via bacterial binding to a single protrusion of the nanotexture. The bacterium/protrusion interaction locally compromises the cell wall, thus causing bacterial death. This study suggests that local mechanical damage and leakage of the cytosol kill the bacteria first, with subsequent degradation of the cell envelope. CONCLUSION: Nanoscale surface roughness may act via a penetrative bactericidal mechanism. This insight suggests that future research may focus on optimizing bacterial binding to individual nanoscale projections in addition to stretching bacteria between nanopillars. Further, antibacterial nanotextures may find use in novel applications employing particles in addition to nanotextures on fibers or films.


Subject(s)
Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Bacterial Outer Membrane/drug effects , Drug Carriers/chemistry , Bacterial Outer Membrane/ultrastructure , Drug Carriers/pharmacology , Escherichia coli/drug effects , Microplastics/chemistry , Microplastics/pharmacology , Microscopy, Electron, Transmission , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , Surface Properties
4.
J Med Chem ; 61(8): 3350-3369, 2018 04 26.
Article in English | MEDLINE | ID: mdl-29590750

ABSTRACT

Rhodesain (RD) is a parasitic, human cathepsin L (hCatL) like cysteine protease produced by Trypanosoma brucei ( T. b.) species and a potential drug target for the treatment of human African trypanosomiasis (HAT). A library of hCatL inhibitors was screened, and macrocyclic lactams were identified as potent RD inhibitors ( Ki < 10 nM), preventing the cell-growth of Trypanosoma brucei rhodesiense (IC50 < 400 nM). SARs addressing the S2 and S3 pockets of RD were established. Three cocrystal structures with RD revealed a noncovalent binding mode of this ligand class due to oxidation of the catalytic Cys25 to a sulfenic acid (Cys-SOH) during crystallization. The P-glycoprotein efflux ratio was measured and the in vivo brain penetration in rats determined. When tested in vivo in acute HAT model, the compounds permitted up to 16.25 (vs 13.0 for untreated controls) mean days of survival.


Subject(s)
Cathepsin L/antagonists & inhibitors , Cysteine Endopeptidases/metabolism , Cysteine Proteinase Inhibitors/pharmacology , Lactams, Macrocyclic/pharmacology , Trypanocidal Agents/pharmacology , Trypanosoma brucei rhodesiense/drug effects , Animals , Binding Sites , Blood-Brain Barrier/metabolism , Cell Line , Cysteine Endopeptidases/chemistry , Cysteine Proteinase Inhibitors/chemical synthesis , Cysteine Proteinase Inhibitors/chemistry , Cysteine Proteinase Inhibitors/pharmacokinetics , Drug Repositioning , Humans , Lactams, Macrocyclic/chemical synthesis , Lactams, Macrocyclic/chemistry , Lactams, Macrocyclic/pharmacokinetics , Ligands , Male , Mice, Inbred C57BL , Molecular Structure , Rats , Structure-Activity Relationship , Swine , Trypanocidal Agents/chemical synthesis , Trypanocidal Agents/chemistry , Trypanocidal Agents/pharmacokinetics
5.
J Med Syst ; 42(2): 25, 2017 Dec 23.
Article in English | MEDLINE | ID: mdl-29273867

ABSTRACT

Ex vivo heart perfusion has been shown to be an effective means of facilitating the resuscitation and assessment of donor hearts for cardiac transplantation. Over the last ten years however, only a few ex vivo perfusion systems have been developed for this application. While results have been promising, a system capable of facilitating multiple perfusion strategies on the same platform has not yet been realized. In this paper, the design, development and testing of a novel and modular ex vivo perfusion system is described. The system is capable of operating in three unique primary modes: the traditional Langendorff Mode, Pump-Supported Working-Mode, and Passive Afterload Working-Mode. In each mode, physiological hemodynamic parameters can be produced by managing perfusion settings. To evaluate heart viability, six experiments were conducted using porcine hearts and measuring several parameters including: pH, aortic pressure, lactate metabolism, coronary vascular resistance (CVR), and myocardial oxygen consumption. Pressure-volume relationship measurements were used to assess left ventricular contractility in each Working Mode. Hemodynamic and metabolic conditions remained stable and consistent across 4 h of ex vivo heart perfusion on the ex vivo perfusion system, validating the system as a viable platform for future development of novel preservation and assessment strategies.


Subject(s)
Equipment Design , Heart/physiology , Perfusion/methods , Animals , Heart Transplantation/methods , Hemodynamics , Hydrogen-Ion Concentration , Lactic Acid/metabolism , Oxygen Consumption , Swine
6.
J Infect Dev Ctries ; 10(2): 113-5, 2016 Feb 28.
Article in English | MEDLINE | ID: mdl-26927449

ABSTRACT

We have developed a mobile App called ZIKATracker (zikatracker.net) to voluntarily be used to report ZIKV cases on a public or private level. As the Zika virus (ZIKV) infection zones are rapidly expanding across South, Central, and North America, and reports have emerged linking ZIKV infection with developmental defects and neurological sequelae, reporting the movement and sequelae of ZIKV is essential. ZIKATracker is a multi-lingual App (English, French, Spanish, and Portuguese) freely available to anyone worldwide wishing to report a suspected or confirmed case of Zika virus and related symptoms. Knowledge gained from the use of this App will help direct the implementation of mosquito control measures in needed areas, bring aid to those affected by the Zika virus, and understand the movement and sequelae of ZIKV as it spreads through communities and across continents.


Subject(s)
Disease Notification/methods , Mobile Applications , Zika Virus Infection/diagnosis , Humans , North America
7.
J Med Chem ; 59(9): 4087-102, 2016 05 12.
Article in English | MEDLINE | ID: mdl-26878596

ABSTRACT

We present a series of small molecule drug discovery case studies where computational methods were prospectively employed to impact Roche research projects, with the aim of highlighting those methods that provide real added value. Our brief accounts encompass a broad range of methods and techniques applied to a variety of enzymes and receptors. Most of these are based on judicious application of knowledge about molecular conformations and interactions: filling of lipophilic pockets to gain affinity or selectivity, addition of polar substituents, scaffold hopping, transfer of SAR, conformation analysis, and molecular overlays. A case study of sequence-driven focused screening is presented to illustrate how appropriate preprocessing of information enables effective exploitation of prior knowledge. We conclude that qualitative statements enabling chemists to focus on promising regions of chemical space are often more impactful than quantitative prediction.


Subject(s)
Drug Design , Molecular Conformation , Small Molecule Libraries , Structure-Activity Relationship
8.
PLoS Pathog ; 11(10): e1005173, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26448646

ABSTRACT

Seasonal influenza viruses are typically restricted to the human upper respiratory tract whereas influenza viruses with greater pathogenic potential often also target extra-pulmonary organs. Infants, pregnant women, and breastfeeding mothers are highly susceptible to severe respiratory disease following influenza virus infection but the mechanisms of disease severity in the mother-infant dyad are poorly understood. Here we investigated 2009 H1N1 influenza virus infection and transmission in breastfeeding mothers and infants utilizing our developed infant-mother ferret influenza model. Infants acquired severe disease and mortality following infection. Transmission of the virus from infants to mother ferrets led to infection in the lungs and mother mortality. Live virus was also found in mammary gland tissue and expressed milk of the mothers which eventually led to milk cessation. Histopathology showed destruction of acini glandular architecture with the absence of milk. The virus was localized in mammary epithelial cells of positive glands. To understand the molecular mechanisms of mammary gland infection, we performed global transcript analysis which showed downregulation of milk production genes such as Prolactin and increased breast involution pathways indicated by a STAT5 to STAT3 signaling shift. Genes associated with cancer development were also significantly increased including JUN, FOS and M2 macrophage markers. Immune responses within the mammary gland were characterized by decreased lymphocyte-associated genes CD3e, IL2Ra, CD4 with IL1ß upregulation. Direct inoculation of H1N1 into the mammary gland led to infant respiratory infection and infant mortality suggesting the influenza virus was able to replicate in mammary tissue and transmission is possible through breastfeeding. In vitro infection studies with human breast cells showed susceptibility to H1N1 virus infection. Together, we have shown that the host-pathogen interactions of influenza virus infection in the mother-infant dyad initiate immunological and oncogenic signaling cascades within the mammary gland. These findings suggest the mammary gland may have a greater role in infection and immunity than previously thought.


Subject(s)
Animals, Suckling/virology , Host-Parasite Interactions/physiology , Mammary Glands, Animal/virology , Mammary Glands, Human/virology , Orthomyxoviridae Infections/transmission , Animals , Animals, Newborn , Blotting, Western , Cell Line , Disease Models, Animal , Female , Ferrets , Humans , Immunohistochemistry , Influenza A Virus, H1N1 Subtype , Influenza, Human/virology , Lactation , Mammary Glands, Animal/pathology , Microscopy, Confocal , Milk/virology , Mothers , Oligonucleotide Array Sequence Analysis , Orthomyxoviridae Infections/pathology , Pregnancy , Real-Time Polymerase Chain Reaction , Transcriptome
9.
J Infect Dev Ctries ; 8(10): 1301-6, 2014 Oct 15.
Article in English | MEDLINE | ID: mdl-25313607

ABSTRACT

INTRODUCTION: Chemokines are small proteins that regulate different cellular functions, such as leukocyte activation, chemoattraction and inflammation. The chemokine CXCL14 (BRAK) is a highly conserved gene among species and through evolution. It has been shown that CXCL14 is locally upregulated during viral infections, also, it has been found that this chemokine possesses direct antibacterial activities. Nonetheless, the exact role that CXCL14 plays during infection remains elusive. METHODOLOGY: CXCL14 deficient mice were generated in a C57B6/129 background and followed by phenotypic characterization. Later, the effect of CXCL14 deficiency during influenza infection and E. coli challenge was assessed. RESULTS: Other than a slight weight reduction, CXCL14 deficient mice exhibited no phenotypic alterations. CXCL14 deficiency did not influence the outcome of influenza virus infection or challenge with E. coli, and no statistically significant differences in clinical signs, cellular responses and histopathological findings were observed. CONCLUSIONS: CXCL14 does not seem to play a pivotal role during influenza and E. coli infections of the lung; these results are suggestive of functional overlap between CXCL14 and other chemokines that are present during lung infection.


Subject(s)
Chemokines, CXC/deficiency , Escherichia coli Infections/genetics , Escherichia coli Infections/immunology , Orthomyxoviridae Infections/genetics , Orthomyxoviridae Infections/immunology , Pneumonia/pathology , Animals , Body Weight , Disease Models, Animal , Escherichia coli Infections/pathology , Lung/pathology , Mice, Inbred C57BL , Mice, Knockout , Orthomyxoviridae Infections/pathology , Pneumonia/microbiology , Pneumonia/virology , Survival Analysis
10.
Virology ; 464-465: 177-183, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25086242

ABSTRACT

The major burden of influenza morbidity resides within the elderly population. The challenge managing influenza-associated illness in the elderly is the decline of immune function, where mechanisms leading to immunological senescence have not been elucidated. To better represent the immune environment, we investigated clinical morbidity and immune function during sequential homologous and heterologous H1N1 influenza infection in an aged ferret model. Our findings demonstrated experimentally that aged ferrets had significant morbidity during monosubtypic heterologous 2° challenge with significant weight loss and respiratory symptoms. Furthermore, increased clinical morbidity was associated with slower and shorter hemagglutinin antibody generation and attenuated type 1 T-cell gene responses in peripheral blood. These results revealed dampened immune activation during sequential influenza infection in aged ferrets. With the presence of an aged model, dissecting clinical morbidity, viral dynamics and immune response during influenza infection will aid the development of future prophylactics such as age specific influenza vaccines.


Subject(s)
Aging/immunology , Immunity, Heterologous , Influenza A Virus, H1N1 Subtype/immunology , Influenza, Human/immunology , Age Factors , Aged , Animals , Antibodies, Viral/immunology , Disease Models, Animal , Ferrets , Hemagglutinin Glycoproteins, Influenza Virus/administration & dosage , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Humans , Influenza Vaccines/administration & dosage , Influenza Vaccines/genetics , Influenza Vaccines/immunology , Influenza, Human/virology , Male , T-Lymphocytes/immunology
11.
J Gen Virol ; 95(Pt 10): 2127-2139, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24989173

ABSTRACT

Influenza B viruses have become increasingly more prominent during influenza seasons. Influenza B infection is typically considered a mild disease and receives less attention than influenza A, but has been causing 20 to 50 % of the total influenza incidence in several regions around the world. Although there is increasing evidence of mid to lower respiratory tract diseases such as bronchitis and pneumonia in influenza B patients, little is known about the pathogenesis of recent influenza B viruses. Here we investigated the clinical and pathological profiles of infection with strains representing the two current co-circulating B lineages (B/Yamagata and B/Victoria) in the ferret model. Specifically, we studied two B/Victoria (B/Brisbane/60/2008 and B/Bolivia/1526/2010) and two B/Yamagata (B/Florida/04/2006 and B/Wisconsin/01/2010) strain infections in ferrets and observed strain-specific but not lineage-specific pathogenicity. We found B/Brisbane/60/2008 caused the most severe clinical illness and B/Brisbane/60/2008 and the B/Yamagata strains instigated pathology in the middle to lower respiratory tract. Importantly, B/Brisbane/60/2008 established efficient lower respiratory tract infection with high viral burden. Our phylogenetic analyses demonstrate profound reassortment among recent influenza B viruses, which indicates the genetic make-up of B/Brisbane/60/2008 differs from the other strains. This may explain the pathogenicity difference post-infection in ferrets.


Subject(s)
Influenza B virus/physiology , Influenza, Human/virology , Orthomyxoviridae Infections/veterinary , Respiratory Tract Infections/pathology , Respiratory Tract Infections/virology , Animals , Disease Models, Animal , Ferrets , Humans , Influenza B virus/isolation & purification , Orthomyxoviridae Infections/pathology , Orthomyxoviridae Infections/virology
12.
Virology ; 454-455: 78-92, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24725934

ABSTRACT

The role of Group X secreted phospholipase A2 (GX-sPLA2) during influenza infection has not been previously investigated. We examined the role of GX-sPLA2 during H1N1 pandemic influenza infection in a GX-sPLA2 gene targeted mouse (GX(-/-)) model and found that survival after infection was significantly greater in GX(-/-) mice than in GX(+/+) mice. Downstream products of GX-sPLA2 activity, PGD2, PGE2, LTB4, cysteinyl leukotrienes and Lipoxin A4 were significantly lower in GX(-/-) mice BAL fluid. Lung microarray analysis identified an earlier and more robust induction of T and B cell associated genes in GX(-/-) mice. Based on the central role of sPLA2 enzymes as key initiators of inflammatory processes, we propose that activation of GX-sPLA2 during H1N1pdm infection is an early step of pulmonary inflammation and its inhibition increases adaptive immunity and improves survival. Our findings suggest that GX-sPLA2 may be a potential therapeutic target during influenza.


Subject(s)
Group X Phospholipases A2/deficiency , Influenza A Virus, H1N1 Subtype/immunology , Orthomyxoviridae Infections/pathology , Orthomyxoviridae Infections/virology , Animals , B-Lymphocytes/immunology , Disease Models, Animal , Gene Expression Profiling , Group X Phospholipases A2/genetics , Lung/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Microarray Analysis , Survival Analysis , T-Lymphocytes/immunology
13.
J Infect Dev Ctries ; 8(4): 498-509, 2014 Apr 15.
Article in English | MEDLINE | ID: mdl-24727517

ABSTRACT

INTRODUCTION: Conventional methods used to detect and characterize influenza viruses in biological samples face multiple challenges due to the diversity of subtypes and high dissimilarity of emerging strains. Next-generation sequencing (NGS) is a powerful technique that can facilitate the detection and characterization of influenza, however, the sequencing strategy and the procedures of data analysis possess different aspects that require careful consideration. METHODOLOGY: The RNA from the lungs of ferrets infected with influenza A/California/07/2009 was analyzed by next-generation sequencing (NGS) without using specific PCR amplification of the viral sequences. Several bioinformatic approaches were used to resolve the viral genes and detect viral quasispecies. RESULTS: The genomic sequences of influenza virus were characterized to a high level of detail when analyzing the short-reads with either the fast aligner Bowtie2, the general purpose aligner BLASTn or de novo assembly with Abyss. Moreover, when using distant viral sequences as reference, these methods were still able to resolve the viral sequences of a biological sample. Finally, direct sequencing of RNA samples did not provide sufficient coverage of the viral genome to study viral quasispecies, and, therefore, prior amplification of the viral segments by PCR would be required to perform this type of analysis. CONCLUSIONS: the introduction of NGS for virus research allows routine full characterization of viral isolates; however, careful design of the sequencing strategy and the procedures for data analysis are still of critical importance.


Subject(s)
Influenza A Virus, H1N1 Subtype/isolation & purification , RNA, Viral/analysis , Sequence Analysis, RNA/methods , Animals , Computational Biology , Ferrets , Influenza A Virus, H1N1 Subtype/genetics , Software
14.
Virology ; 448: 91-103, 2014 Jan 05.
Article in English | MEDLINE | ID: mdl-24314640

ABSTRACT

Pandemic H1N1 influenza A (H1N1pdm) elicits stronger pulmonary inflammation than previously circulating seasonal H1N1 influenza A (sH1N1), yet mechanisms of inflammatory activation in respiratory epithelial cells during H1N1pdm infection are unclear. We investigated host responses to H1N1pdm/sH1N1 infection and virus entry mechanisms in primary human bronchial epithelial cells in vitro. H1N1pdm infection rapidly initiated a robust inflammatory gene signature (3 h post-infection) not elicited by sH1N1 infection. Protein secretion inhibition had no effect on gene induction. Infection with membrane fusion deficient H1N1pdm failed to induce robust inflammatory gene expression which was rescued with restoration of fusion ability, suggesting H1N1pdm directly triggered the inflammatory signature downstream of membrane fusion. Investigation of intra-virion components revealed H1N1pdm viral RNA (vRNA) triggered a stronger inflammatory phenotype than sH1N1 vRNA. Thus, our study is first to report H1N1pdm induces greater inflammatory gene expression than sH1N1 in vitro due to direct virus-epithelial cell interaction.


Subject(s)
Bronchi/cytology , Cytokines/genetics , Epithelial Cells/immunology , Influenza A Virus, H1N1 Subtype/physiology , Influenza, Human/genetics , Influenza, Human/immunology , Membrane Fusion , Bronchi/immunology , Cells, Cultured , Cytokines/immunology , Epithelial Cells/virology , Humans , Inflammation Mediators/immunology , Influenza A Virus, H1N1 Subtype/genetics , Influenza A Virus, H1N1 Subtype/immunology , Influenza, Human/epidemiology , Influenza, Human/virology , Pandemics
15.
J Med Chem ; 56(23): 9789-801, 2013 Dec 12.
Article in English | MEDLINE | ID: mdl-24224654

ABSTRACT

Starting from the weakly active dual CatS/K inhibitor 5, structure-based design supported by X-ray analysis led to the discovery of the potent and selective (>50,000-fold vs CatK) cyclopentane derivative 22 by exploiting specific ligand-receptor interactions in the S2 pocket of CatS. Changing the central cyclopentane scaffold to the analogous pyrrolidine derivative 57 decreased the enzyme as well as the cell-based activity significantly by 24- and 69-fold, respectively. The most promising scaffold identified was the readily accessible proline derivative (e.g., 79). This compound, with an appealing ligand efficiency (LE) of 0.47, included additional structural modifications binding in the S1 and S3 pockets of CatS, leading to favorable in vitro and in vivo properties. Compound 79 reduced IL-2 production in a transgenic DO10.11 mouse model of antigen presentation in a dose-dependent manner with an ED50 of 5 mg/kg.


Subject(s)
Cathepsins/antagonists & inhibitors , Cysteine Proteinase Inhibitors/chemical synthesis , Animals , Cyclopentanes/chemistry , Cysteine Proteinase Inhibitors/pharmacokinetics , Humans , Mice , Proline/analogs & derivatives , Structure-Activity Relationship
16.
PLoS One ; 8(8): e71761, 2013.
Article in English | MEDLINE | ID: mdl-23967240

ABSTRACT

RATIONALE: Severe influenza remains a major public health threat and is responsible for thousands of deaths annually. Increasing antiviral resistance and limited effectiveness of current therapies highlight the need for new approaches to influenza treatment. Extensive pre-clinical data have shown that mesenchymal stromal (stem) cell (MSC) therapy can induce anti-inflammatory effects and enhance repair of the injured lung. We hypothesized that MSC therapy would improve survival, dampen lung inflammation and decrease acute lung injury (ALI) in a murine model of severe influenza. METHODS: C57Bl/6 mice were infected with influenza A/PuertoRico/8/34 (mouse-adapted H1N1) or influenza A/Mexico/4108/2009 (swine-origin pandemic H1N1) and administered human or mouse MSCs via the tail vein, either pre- or post- infection. MSC efficacy was evaluated as both an independent and adjunctive treatment strategy in combination with the antiviral agent, oseltamivir. Weight loss and survival were monitored. Inflammatory cells, cytokine/chemokines (IFN-γ, CXCL10, CCL2 and CCL5) and markers of ALI (total protein and IgM), were measured in bronchoalveolar lavage fluid and lung parenchyma. RESULTS: Administration of murine MSCs or human MSCs in a prophylactic or therapeutic regimen failed to improve survival, decrease pulmonary inflammation/inflammatory cell counts or prevent ALI in influenza virus-infected mice. MSCs administered in combination with oseltamivir also failed to improve outcomes. CONCLUSIONS: Despite similarities in the clinical presentation and pathobiology of ALI and severe influenza, our findings suggest that MSC therapy may not be effective for prevention and/or treatment of acute severe influenza.


Subject(s)
Cell- and Tissue-Based Therapy , Influenza A Virus, H1N1 Subtype , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , Orthomyxoviridae Infections/therapy , Animals , Antiviral Agents/administration & dosage , Bronchoalveolar Lavage Fluid , Disease Models, Animal , Humans , Male , Mice , Orthomyxoviridae Infections/mortality , Orthomyxoviridae Infections/virology , Pneumonia/pathology , Pneumonia/virology , Treatment Failure
17.
Bioorg Med Chem Lett ; 23(14): 4239-43, 2013 Jul 15.
Article in English | MEDLINE | ID: mdl-23735744

ABSTRACT

A series of amides bearing a variety of amidine head groups was investigated as BACE1 inhibitors with respect to inhibitory activity in a BACE1 enzyme as well as a cell-based assay. Determination of their basicity as well as their properties as substrates of P-glycoprotein revealed that a 2-amino-1,3-oxazine head group would be a suitable starting point for further development of brain penetrating compounds for potential Alzheimer's disease treatment.


Subject(s)
Amides/chemistry , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Aspartic Acid Endopeptidases/antagonists & inhibitors , Protease Inhibitors/chemistry , Alzheimer Disease/drug therapy , Amides/metabolism , Amides/therapeutic use , Amyloid Precursor Protein Secretases/metabolism , Aspartic Acid Endopeptidases/metabolism , Binding Sites , Humans , Molecular Docking Simulation , Protease Inhibitors/metabolism , Protease Inhibitors/therapeutic use , Protein Binding , Protein Structure, Tertiary , Structure-Activity Relationship
18.
ChemMedChem ; 8(6): 967-75, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23658062

ABSTRACT

The cysteine protease rhodesain of Trypanosoma brucei parasites causing African sleeping sickness has emerged as a target for the development of new drug candidates. Based on a triazine nitrile moiety as electrophilic headgroup, optimization studies on the substituents for the S1, S2, and S3 pockets of the enzyme were performed using structure-based design and resulted in inhibitors with inhibition constants in the single-digit nanomolar range. Comprehensive structure-activity relationships clarified the binding preferences of the individual pockets of the active site. The S1 pocket tolerates various substituents with a preference for flexible and basic side chains. Variation of the S2 substituent led to high-affinity ligands with inhibition constants down to 2 nM for compounds bearing cyclohexyl substituents. Systematic investigations on the S3 pocket revealed its potential to achieve high activities with aromatic vectors that undergo stacking interactions with the planar peptide backbone forming part of the pocket. X-ray crystal structure analysis with the structurally related enzyme human cathepsin L confirmed the binding mode of the triazine ligand series as proposed by molecular modeling. Sub-micromolar inhibition of the proliferation of cultured parasites was achieved for ligands decorated with the best substituents identified through the optimization cycles. In cell-based assays, the introduction of a basic side chain on the inhibitors resulted in a 35-fold increase in antitrypanosomal activity. Finally, bioisosteric imidazopyridine nitriles were studied in order to prevent off-target effects with unselective nucleophiles by decreasing the inherent electrophilicity of the triazine nitrile headgroup. Using this ligand, the stabilization by intramolecular hydrogen bonding of the thioimidate intermediate, formed upon attack of the catalytic cysteine residue, compensates for the lower reactivity of the headgroup. The imidazopyridine nitrile ligand showed excellent stability toward the thiol nucleophile glutathione in a quantitative in vitro assay and fourfold lower cytotoxicity than the parent triazine nitrile.


Subject(s)
Cathepsin L/chemistry , Cysteine Endopeptidases/metabolism , Cysteine Proteinase Inhibitors/pharmacology , Imidazoles/pharmacology , Nitriles/pharmacology , Pyridines/pharmacology , Triazines/pharmacology , Trypanosoma brucei brucei/enzymology , Crystallography, X-Ray , Cysteine Proteinase Inhibitors/chemical synthesis , Cysteine Proteinase Inhibitors/chemistry , Dose-Response Relationship, Drug , Humans , Imidazoles/chemical synthesis , Imidazoles/chemistry , Ligands , Models, Molecular , Molecular Structure , Nitriles/chemical synthesis , Nitriles/chemistry , Parasitic Sensitivity Tests , Pyridines/chemical synthesis , Pyridines/chemistry , Structure-Activity Relationship , Triazines/chemical synthesis , Triazines/chemistry , Trypanosoma brucei brucei/drug effects
19.
Acta Crystallogr D Biol Crystallogr ; 69(Pt 6): 1124-37, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23695257

ABSTRACT

The aspartic protease BACE2 is responsible for the shedding of the transmembrane protein Tmem27 from the surface of pancreatic ß-cells, which leads to inactivation of the ß-cell proliferating activity of Tmem27. This role of BACE2 in the control of ß-cell maintenance suggests BACE2 as a drug target for diabetes. Inhibition of BACE2 has recently been shown to lead to improved control of glucose homeostasis and to increased insulin levels in insulin-resistant mice. BACE2 has 52% sequence identity to the well studied Alzheimer's disease target enzyme ß-secretase (BACE1). High-resolution BACE2 structures would contribute significantly to the investigation of this enzyme as either a drug target or anti-target. Surface mutagenesis, BACE2-binding antibody Fab fragments, single-domain camelid antibody VHH fragments (Xaperones) and Fyn-kinase-derived SH3 domains (Fynomers) were used as crystallization helpers to obtain the first high-resolution structures of BACE2. Eight crystal structures in six different packing environments define an ensemble of low-energy conformations available to the enzyme. Here, the different strategies used for raising and selecting BACE2 binders for cocrystallization are described and the crystallization success, crystal quality and the time and resources needed to obtain suitable crystals are compared.


Subject(s)
Amyloid Precursor Protein Secretases/chemistry , Aspartic Acid Endopeptidases/chemistry , Immunoglobulin Fab Fragments/chemistry , Insulin-Secreting Cells/enzymology , Amyloid Precursor Protein Secretases/genetics , Amyloid Precursor Protein Secretases/metabolism , Animals , Area Under Curve , Aspartic Acid Endopeptidases/genetics , Aspartic Acid Endopeptidases/metabolism , Catalytic Domain , Crystallization , Humans , Immunoglobulin Fab Fragments/metabolism , Insulin-Secreting Cells/metabolism , Mice , Models, Molecular , Mutagenesis , Protein Conformation , Surface Plasmon Resonance , X-Ray Diffraction
20.
Sci Rep ; 3: 1698, 2013.
Article in English | MEDLINE | ID: mdl-23608887

ABSTRACT

Evolution of H1N1 influenza A outbreaks of the past 100 years is interesting and significantly complex and details of H1N1 genetic drift remains unknown. Here we investigated the clinical characteristics and immune cross-reactivity of significant historical H1N1 strains. We infected ferrets with H1N1 strains from 1943, 1947, 1977, 1986, 1999, and 2009 and showed each produced a unique clinical signature. We found significant cross-reactivity between viruses with similar HA sequences. Interestingly, A/FortMonmouth/1/1947 antisera cross-reacted with A/USSR/90/1977 virus, thought to be a 1947 resurfaced virus. Importantly, our immunological data that didn't show cross-reactivity can be extrapolated to failure of past H1N1 influenza vaccines, ie. 1947, 1986 and 2009. Together, our results help to elucidate H1N1 immuno-genetic alterations that occurred in the past 100 years and immune responses caused by H1N1 evolution. This work will facilitate development of future influenza therapeutics and prophylactics such as influenza vaccines.


Subject(s)
Ferrets/virology , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Immunity, Innate/immunology , Influenza A Virus, H1N1 Subtype/classification , Influenza A Virus, H1N1 Subtype/immunology , Viral Vaccines/administration & dosage , Animals , Influenza A Virus, H1N1 Subtype/drug effects , Species Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...