Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Virus Genes ; 56(2): 150-167, 2020 Apr.
Article in English | MEDLINE | ID: mdl-32076918

ABSTRACT

The viruses historically implicated or currently considered as candidates for misuse in bioterrorist events are poxviruses, filoviruses, bunyaviruses, orthomyxoviruses, paramyxoviruses and a number of arboviruses causing encephalitis, including alpha- and flaviviruses. All these viruses are of concern for public health services when they occur in natural outbreaks or emerge in unvaccinated populations. Recent events and intelligence reports point to a growing risk of dangerous biological agents being used for nefarious purposes. Public health responses effective in natural outbreaks of infectious disease may not be sufficient to deal with the severe consequences of a deliberate release of such agents. One important aspect of countermeasures against viral biothreat agents are the antiviral treatment options available for use in post-exposure prophylaxis. These issues were adressed by the organizers of the 16th Medical Biodefense Conference, held in Munich in 2018, in a special session on the development of drugs to treat infections with viruses currently perceived as a threat to societies or associated with a potential for misuse as biothreat agents. This review will outline the state-of-the-art methods in antivirals research discussed and provide an overview of antiviral compounds in the pipeline that are already approved for use or still under development.


Subject(s)
Antiviral Agents/therapeutic use , Arboviruses/drug effects , Bioterrorism/prevention & control , Virus Diseases/drug therapy , Arboviruses/pathogenicity , Filoviridae/drug effects , Filoviridae/pathogenicity , Humans , Orthobunyavirus/drug effects , Orthobunyavirus/pathogenicity , Orthomyxoviridae/drug effects , Orthomyxoviridae/pathogenicity , Paramyxovirinae/drug effects , Paramyxovirinae/pathogenicity , Poxviridae/drug effects , Poxviridae/pathogenicity , Virus Diseases/virology
2.
Free Radic Biol Med ; 152: 551-560, 2020 05 20.
Article in English | MEDLINE | ID: mdl-31775023

ABSTRACT

Idebenone has recently been investigated as a drug therapy for Leber's hereditary optic neuropathy (LHON), a rare genetic mitochondrial disease that causes rapid and progressive bilateral vision loss. Although several studies have shown that idebenone can promote vision recovery in patients with LHON, the evidence for the efficacy of idebenone is still limited. Idebenone failed to demonstrate superiority over placebo in the primary end-points of the only published randomised, double-blind, placebo-controlled trial. There appears to be a patient-specific response to idebenone with high variability in therapeutic outcomes. A recent study suggested that the cytosolic enzyme NAD(P)H: quinone acceptor oxidoreductase (NQO1) is the major enzyme involved in the activation of idebenone, and the beneficial effects of idebenone are dependent on the expression of NQO1. Here, we confirm the NQO1-dependent activity of idebenone, but we also show, for the first time, that the cytotoxicity of idebenone is linked to cellular expression of NQO1. Upon idebenone administration, cells deficient in NQO1 show a marked decrease in viability in comparison to NQO1 expressing cells, with idebenone causing ROS production and deleterious effects on ATP levels and cell viability. In addition, our data highlights that only cells expressing NQO1 can significantly activate idebenone, indicating that other proposed metabolic activation pathways, such as complex II and glycerol-3-phosphate dehydrogenase, do not play a significant role in idebenone activation. Furthermore, we provide evidence of idebenone-induced toxicity in the retina ex-vivo, which can be explained by the variation of NQO1 expression between different cell types in the mouse retina. Idebenone mediated cell rescue in the rotenone ex vivo model also indicated that this drug has a narrow therapeutic window. These findings will help to guide the development of future therapies and drug delivery strategies including intra-ocular administration. The specific dependence of idebenone activity on NQO1 may also explain the variation in patient outcomes in clinical trials.


Subject(s)
Antioxidants , Ubiquinone , Animals , Antioxidants/pharmacology , Cell Death , Humans , Mice , NAD(P)H Dehydrogenase (Quinone)/genetics , Retina , Ubiquinone/analogs & derivatives , Ubiquinone/pharmacology
3.
Oncogene ; 37(2): 231-240, 2018 01 11.
Article in English | MEDLINE | ID: mdl-28925395

ABSTRACT

Highly expressed in cancer protein 1 (Hec1) is a subunit of the kinetochore (KT)-associated Ndc80 complex, which ensures proper segregation of sister chromatids at mitosis by mediating the interaction between KTs and microtubules (MTs). HEC1 mRNA and protein are highly expressed in many malignancies as part of a signature of chromosome instability. These properties render Hec1 a promising molecular target for developing therapeutic drugs that exert their anticancer activities by producing massive chromosome aneuploidy. A virtual screening study aimed at identifying small molecules able to bind at the Hec1-MT interaction domain identified one positive hit compound and two analogs of the hit with high cytotoxic, pro-apoptotic and anti-mitotic activities. The most cytotoxic analog (SM15) was shown to produce chromosome segregation defects in cancer cells by inhibiting the correction of erroneous KT-MT interactions. Live cell imaging of treated cells demonstrated that mitotic arrest and segregation abnormalities lead to cell death through mitotic catastrophe and that cell death occurred also from interphase. Importantly, SM15 was shown to be more effective in inducing apoptotic cell death in cancer cells as compared to normal ones and effectively reduced tumor growth in a mouse xenograft model. Mechanistically, cold-induced MT depolymerization experiments demonstrated a hyper-stabilization of both mitotic and interphase MTs. Molecular dynamics simulations corroborate this finding by showing that SM15 can bind the MT surface independently from Hec1 and acts as a stabilizer of both MTs and KT-MT interactions. Overall, our studies represent a clear proof of principle that MT-Hec1-interacting compounds may represent novel powerful anticancer agents.


Subject(s)
Antineoplastic Agents/pharmacology , Microtubules/drug effects , Neoplasms/drug therapy , Nuclear Proteins/antagonists & inhibitors , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Chromosomal Instability/drug effects , Chromosomal Instability/genetics , Chromosome Segregation/drug effects , Computer Simulation , Cytoskeletal Proteins , Drug Screening Assays, Antitumor/methods , Humans , Inhibitory Concentration 50 , Interphase/drug effects , Kinetochores/metabolism , Male , Mice , Mice, Nude , Microtubules/metabolism , Mitosis/drug effects , Molecular Docking Simulation , Neoplasms/pathology , Nuclear Proteins/chemistry , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Protein Domains/drug effects , Xenograft Model Antitumor Assays
4.
Luminescence ; 27(3): 234-41, 2012.
Article in English | MEDLINE | ID: mdl-22362656

ABSTRACT

Bioluminescence, the emission of light from live organisms, occurs in 18 phyla and is the major communication system in the deep sea. It has appeared independently many times during evolution but its origins remain unknown. Coelenterazine bioluminescence discovered in luminous jellyfish is the most common chemistry causing bioluminescence in the sea, occurring in seven phyla. Sequence similarities between coelenterazine luciferases and photoproteins from different phyla are poor (often < 5%). The aim of this study was to examine albumin that binds organic substances as a coelenterazine luciferase to test the hypothesis that the evolutionary origin of a bioluminescent protein was the result of the formation of a solvent cage containing just a few key amino acids. The results show for the first time that bovine and human albumin catalysed coelenterazine chemiluminescence consistent with a mono-oxygenase, whereas gelatin and haemoglobin, an oxygen carrier, had very weak activity. Insulin also catalysed coelenterazine chemiluminescence and was increased by Zn(2+). Albumin chemiluminescence was heat denaturable, exhibited saturable substrate characteristics and was inhibited by cations that bound these proteins and by drugs that bind to human albumin drug site I. Molecular modelling confirmed the coelenterazine binding site and identified four basic amino acids: lys195, arg222, his242 and arg257, potentially important in binding and catalysis similar to naturally occurring coelenterazine bioluminescent proteins. These results support the 'solvent cage' hypothesis for the evolutionary origin of enzymatic coelenterazine bioluminescent proteins. They also have important consequences in diseases such as diabetes, gut disorders and food intolerance where a mono-oxygenase could affect cell surface proteins.


Subject(s)
Albumins/chemistry , Albumins/metabolism , Imidazoles/chemistry , Luminescence , Mixed Function Oxygenases/metabolism , Pyrazines/chemistry , Animals , Catalysis , Cattle , Enzyme Activation , Gelatin/chemistry , Hemoglobins/chemistry , Humans , Imidazoles/metabolism , Luminescent Measurements , Mixed Function Oxygenases/chemistry , Models, Molecular , Pyrazines/metabolism , Zinc/chemistry
5.
Article in English | MEDLINE | ID: mdl-18058549

ABSTRACT

The aim of the present work is to investigate through molecular modelling the possible role of the human enzyme Hint1 in the final P-N bond cleavage of phosphoramidate ProTides, which would lead to the intracellular delivery of unmasked nucleoside analogue monophosphates. Herein, we report our preliminary analysis based on docking studies of (E)-5-(2-bromovinyl)-2'-deoxyuridine (BVdU) related aminoacyl phosphates with Hint1 and the effect of the amino acid moiety on the enzyme-substrate binding affinity.


Subject(s)
Amides/metabolism , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/metabolism , Phosphoric Acids/metabolism , Bromodeoxyuridine/analogs & derivatives , Bromodeoxyuridine/chemistry , Bromodeoxyuridine/metabolism , Catalytic Domain , Humans , Models, Molecular , Protein Conformation , Substrate Specificity
6.
J Mol Graph Model ; 25(1): 17-29, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16310386

ABSTRACT

[Arg(91), Ala(96)] MBP(87-99) is an altered peptide ligand (APL) of myelin basic protein (MBP), shown to actively inhibit experimental autoimmune encephalomyelitis (EAE), which is studied as a model of multiple sclerosis (MS). The APL has been rationally designed by substituting two of the critical residues for recognition by the T-cell receptor. A conformational analysis of the APL has been sought using a combination of 2D NOESY nuclear magnetic resonance (NMR) experiments and detailed molecular dynamics (MD) calculations, in order to comprehend the stereoelectronic requirements for antagonistic activity, and to propose a putative bioactive conformation based on spatial proximities of the native peptide in the crystal structure. The proposed structure presents backbone similarity with the native peptide especially at the N-terminus, which is important for major histocompatibility complex (MHC) binding. Primary (Val(87), Phe(90)) and secondary (Asn(92), Ile(93), Thr(95)) MHC anchors occupy the same region in space, whereas T-cell receptor (TCR) contacts (His(88), Phe(89)) have different orientation between the two structures. A possible explanation, thus, of the antagonistic activity of the APL is that it binds to MHC, preventing the binding of myelin epitopes, but it fails to activate the TCR and hence to trigger the immunologic response. NMR experiments coupled with theoretical calculations are found to be in agreement with X-ray crystallography data and open an avenue for the design and synthesis of novel peptide restricted analogues as well as peptide mimetics that rises as an ultimate goal.


Subject(s)
Models, Molecular , Myelin Basic Protein/chemistry , Peptide Fragments/chemistry , Amino Acid Sequence , Amino Acids, Aromatic/chemistry , Animals , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Humans , Ligands , Molecular Sequence Data , Myelin Basic Protein/therapeutic use , Peptide Fragments/therapeutic use , Protein Conformation
9.
Antivir Chem Chemother ; 12(2): 77-89, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11527045

ABSTRACT

Bicyclic furano pyrimidine nucleosides have been found to be highly potent and selective inhibitors of varicella zoster virus (VZV). They are inactive against herpes simplex virus and have been known for several decades as (unwanted) synthetic by-products in the Pd-catalysed coupling of acetylenes to 5-iodo nucleosides. These fluorescent bicyclic nucleosides are now established as a new family of potent antivirals. They are unusual in that they exhibit complete specificity for VZV and require an alkyl (or alkylaryl) side-chain for biological activity. The latter requirement confers extremely high lipophilicities on these compounds, unknown amongst chemotherapeutic nucleosides, which may be of considerable importance in formulation, dosing and tissue distribution. The most potent compounds reported are p-alkylaryl compounds, with EC50 values below 1 nM versus VZV and selectivity index values of around 1,000,000. Here, we review the discovery, synthesis, characterization, antiviral profile, SAR, mechanism of action and development prospects for this new family of antivirals.


Subject(s)
Antiviral Agents/chemistry , Antiviral Agents/pharmacology , Furans/pharmacology , Herpesvirus 3, Human/drug effects , Pyrimidine Nucleosides/chemistry , Pyrimidine Nucleosides/pharmacology , Antiviral Agents/chemical synthesis , Deoxyribose/chemistry , Furans/chemical synthesis , Furans/chemistry , Magnetic Resonance Spectroscopy , Pyrimidine Nucleosides/chemical synthesis , Solubility , Spectrometry, Fluorescence , Substrate Specificity
10.
Bioorg Med Chem Lett ; 10(11): 1215-7, 2000 Jun 05.
Article in English | MEDLINE | ID: mdl-10866384

ABSTRACT

Preliminary SAR studies on the side chain of a new class of antiviral nucleosides have shown that terminal substitution in the side-chain, with a halogen atom, lead to potent and highly specific anti-VZV agents.


Subject(s)
Antiviral Agents/pharmacology , Halogens/chemistry , Herpesvirus 3, Human/drug effects , Nucleosides/pharmacology , Antiviral Agents/chemistry , Nucleosides/chemistry , Structure-Activity Relationship
11.
Antivir Chem Chemother ; 11(6): 383-93, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11227996

ABSTRACT

We have recently reported the discovery of an entirely new category of potent antivaricella-zoster virus agents based on novel deoxynucleoside analogues bearing unusual fluorescent bicyclic furo base moieties. Initial studies revealed an absolute requirement of a long alkyl side-chain, with an optimal length of C8-C10, for antiviral activity. However, the impact of this requirement on the physical properties of these compounds is high: inherent lipophilicity and extremely poor aqueous solubility, which may limit the use of these nucleosides as drugs. In order to address this issue, we have now prepared a new series of analogues, bearing ether and glycol type side-chains, designed to improve the aqueous solubility of the compounds. Synthesis of target nucleosides involved Pd-catalysed coupling of terminal alkynes with 5-iodo-2'-deoxyuridine. The 5-alkynyl nucleosides thus obtained were then treated with copper (I) iodide to produce the desired bicyclic analogues. As anticipated, the new compounds exhibited a dramatic increase in aqueous solubility, although antiviral activity was significantly reduced. A possible correlation between antiviral activity and overall compound lipophilicity is discussed.


Subject(s)
Antiviral Agents/chemical synthesis , Antiviral Agents/pharmacology , Herpesvirus 3, Human/drug effects , Pyrimidine Nucleosides/antagonists & inhibitors , Pyrimidine Nucleosides/chemical synthesis , Humans
12.
J Med Chem ; 42(22): 4479-84, 1999 Nov 04.
Article in English | MEDLINE | ID: mdl-10579812

ABSTRACT

We herein report the discovery of an entirely new category of potent antiviral agents based on novel deoxynucleoside analogues with unusual bicyclic base moieties. Target structures, previously known as byproducts in Pd-catalyzed coupling of terminal alkynes with 5-iodo-nucleosides, are recognized herein for the first time to be potent and selective inhibitors of varicella-zoster virus (VZV) in vitro. As an unusual structure-activity relationship we noted the absolute requirement of a long alkyl side chain, with an optimum length of C(8)-C(10), for antiviral activity. We thus report the synthesis and characterization of a series of chain-modified analogues and their extensive in vitro evaluation. The lead compounds have a ca. 300-fold enhancement in anti-VZV activity over the reference compound acyclovir, with no detectable in vitro cytotoxicity. The novel structure of these compounds, coupled with their ease of synthesis, excellent antiviral profile, and promising physical properties, makes them of great interest for possible antiviral drug development.


Subject(s)
Antiviral Agents/chemical synthesis , Herpesvirus 3, Human/drug effects , Nucleosides/chemical synthesis , Antiviral Agents/chemistry , Antiviral Agents/pharmacology , Cell Line , Cell Survival/drug effects , Humans , Nucleosides/chemistry , Nucleosides/pharmacology , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...