Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
PLoS Pathog ; 14(12): e1007439, 2018 12.
Article in English | MEDLINE | ID: mdl-30576397

ABSTRACT

Arenaviruses are a significant cause of hemorrhagic fever, an often-fatal disease for which there is no approved antiviral therapy. Lassa fever in particular generates high morbidity and mortality in West Africa, where the disease is endemic, and a recent outbreak in Nigeria was larger and more geographically diverse than usual. We are developing LHF-535, a small-molecule viral entry inhibitor that targets the arenavirus envelope glycoprotein, as a therapeutic candidate for Lassa fever and other hemorrhagic fevers of arenavirus origin. Using a lentiviral pseudotype infectivity assay, we determined that LHF-535 had sub-nanomolar potency against the viral envelope glycoproteins from all Lassa virus lineages, with the exception of the glycoprotein from the LP strain from lineage I, which was 100-fold less sensitive than that of other strains. This reduced sensitivity was mediated by a unique amino acid substitution, V434I, in the transmembrane domain of the envelope glycoprotein GP2 subunit. This position corresponds to the attenuation determinant of Candid#1, a live-attenuated Junín virus vaccine strain used to prevent Argentine hemorrhagic fever. Using a virus-yield reduction assay, we determined that LHF-535 potently inhibited Junín virus, but not Candid#1, and the Candid#1 attenuation determinant, F427I, regulated this difference in sensitivity. We also demonstrated that a daily oral dose of LHF-535 at 10 mg/kg protected mice from a lethal dose of Tacaribe virus. Serial passage of Tacaribe virus in LHF-535-treated Vero cells yielded viruses that were resistant to LHF-535, and the majority of drug-resistant viruses exhibited attenuated pathogenesis. These findings provide a framework for the clinical development of LHF-535 as a broad-spectrum inhibitor of arenavirus entry and provide an important context for monitoring the emergence of drug-resistant viruses.


Subject(s)
Antiviral Agents/pharmacology , Lassa Fever , Lassa virus/genetics , Virulence/drug effects , Virulence/genetics , Animals , Chlorocebus aethiops , Drug Resistance, Viral/drug effects , Drug Resistance, Viral/genetics , HEK293 Cells , Humans , Lassa virus/drug effects , Mice , Mutation , Vero Cells , Viral Envelope Proteins/genetics
2.
Methods Mol Biol ; 1604: 393-403, 2018.
Article in English | MEDLINE | ID: mdl-28986850

ABSTRACT

The majority of viruses causing hemorrhagic fever in humans are Risk Group 3 or 4 pathogens and, therefore, can only be handled in biosafety level 3 or 4 (BSL-3/4) containment laboratories. The restricted number of such laboratories, the substantial financial requirements to maintain them, and safety concerns for the laboratory workers pose formidable challenges for rapid medical countermeasure discovery and evaluation. BSL-2 surrogate systems are a less challenging, cheap, and fast alternative to the use of live high-consequence viruses for dissecting and targeting individual steps of viral lifecycles with a diminished threat to the laboratory worker. Typical surrogate systems are virion-like particles (VLPs), transcriptionally active ("infectious") VLPs, minigenome systems, recombinant heterotypic viruses encoding proteins of target viruses, and vesiculoviral or retroviral pseudotype systems. Here, we outline the use of retroviral pseudotypes for identification of antivirals against BSL-4 pathogens.


Subject(s)
Hemorrhagic Fevers, Viral/virology , Antiviral Agents/therapeutic use , Hemorrhagic Fevers, Viral/drug therapy , Humans , Retroviridae/drug effects , Retroviridae/genetics , Virus Internalization/drug effects
3.
Antiviral Res ; 145: 24-32, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28645623

ABSTRACT

Filoviruses, consisting of Ebola virus, Marburg virus and Cuevavirus, cause severe hemorrhagic fevers in humans with high mortality rates up to 90%. Currently, there is no approved vaccine or therapy available for the prevention and treatment of filovirus infection in humans. The recent 2013-2015 West African Ebola epidemic underscores the urgency to develop antiviral therapeutics against these infectious diseases. Our previous study showed that GPCR antagonists, particularly histamine receptor antagonists (antihistamines) inhibit Ebola and Marburg virus entry. In this study, we screened a library of 1220 small molecules with predicted antihistamine activity, identified multiple compounds with potent inhibitory activity against entry of both Ebola and Marburg viruses in human cancer cell lines, and confirmed their anti-Ebola activity in human primary cells. These small molecules target a late-stage of Ebola virus entry. Further structure-activity relationship studies around one compound (cp19) reveal the importance of the coumarin fused ring structure, especially the hydrophobic substituents at positions 3 and/or 4, for its antiviral activity, and this identified scaffold represents a favorable starting point for the rapid development of anti-filovirus therapeutic agents.


Subject(s)
Antiviral Agents/pharmacology , Coumarins/chemistry , Coumarins/pharmacology , Ebolavirus/drug effects , Histamine Antagonists/pharmacology , Marburgvirus/drug effects , Virus Internalization/drug effects , Animals , Antiviral Agents/chemistry , Cell Line , Cell Line, Tumor , Coumarins/analysis , Drug Discovery , Hemorrhagic Fever, Ebola/drug therapy , High-Throughput Screening Assays , Histamine Antagonists/chemistry , Humans , Hydrophobic and Hydrophilic Interactions , Marburg Virus Disease/drug therapy , Small Molecule Libraries , Structure-Activity Relationship
4.
J Virol ; 91(13)2017 07 01.
Article in English | MEDLINE | ID: mdl-28381571

ABSTRACT

A dynamic actin cytoskeleton is necessary for viral entry, intracellular migration, and virion release. For HIV-1 infection, during entry, the virus triggers early actin activity by hijacking chemokine coreceptor signaling, which activates a host dependency factor, cofilin, and its kinase, the LIM domain kinase (LIMK). Although knockdown of human LIM domain kinase 1 (LIMK1) with short hairpin RNA (shRNA) inhibits HIV infection, no specific small-molecule inhibitor of LIMK has been available. Here, we describe the design and discovery of novel classes of small-molecule inhibitors of LIMK for inhibiting HIV infection. We identified R10015 as a lead compound that blocks LIMK activity by binding to the ATP-binding pocket. R10015 specifically blocks viral DNA synthesis, nuclear migration, and virion release. In addition, R10015 inhibits multiple viruses, including Zaire ebolavirus (EBOV), Rift Valley fever virus (RVFV), Venezuelan equine encephalitis virus (VEEV), and herpes simplex virus 1 (HSV-1), suggesting that LIMK inhibitors could be developed as a new class of broad-spectrum antiviral drugs.IMPORTANCE The actin cytoskeleton is a structure that gives the cell shape and the ability to migrate. Viruses frequently rely on actin dynamics for entry and intracellular migration. In cells, actin dynamics are regulated by kinases, such as the LIM domain kinase (LIMK), which regulates actin activity through phosphorylation of cofilin, an actin-depolymerizing factor. Recent studies have found that LIMK/cofilin are targeted by viruses such as HIV-1 for propelling viral intracellular migration. Although inhibiting LIMK1 expression blocks HIV-1 infection, no highly specific LIMK inhibitor is available. This study describes the design, medicinal synthesis, and discovery of small-molecule LIMK inhibitors for blocking HIV-1 and several other viruses and emphasizes the feasibility of developing LIMK inhibitors as broad-spectrum antiviral drugs.


Subject(s)
Antiviral Agents/pharmacology , Enzyme Inhibitors/pharmacology , HIV-1/drug effects , Lim Kinases/antagonists & inhibitors , Virus Release/drug effects , Virus Replication/drug effects , Antiviral Agents/chemical synthesis , Antiviral Agents/isolation & purification , Cells, Cultured , Ebolavirus/drug effects , Encephalitis Virus, Venezuelan Equine/drug effects , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/isolation & purification , HIV-1/physiology , Herpesvirus 1, Human/drug effects , Humans , Microbial Sensitivity Tests , Molecular Docking Simulation , Rift Valley fever virus/drug effects
5.
Sci Rep ; 6: 34475, 2016 Sep 30.
Article in English | MEDLINE | ID: mdl-27686742

ABSTRACT

Diverse pathogenic agents often utilize overlapping host networks, and hub proteins within these networks represent attractive targets for broad-spectrum drugs. Using bacterial toxins, we describe a new approach for discovering broad-spectrum therapies capable of inhibiting host proteins that mediate multiple pathogenic pathways. This approach can be widely used, as it combines genetic-based target identification with cell survival-based and protein function-based multiplex drug screens, and concurrently discovers therapeutic compounds and their protein targets. Using B-lymphoblastoid cells derived from the HapMap Project cohort of persons of African, European, and Asian ancestry we identified host caspases as hub proteins that mediate the lethality of multiple pathogenic agents. We discovered that an approved drug, Bithionol, inhibits host caspases and also reduces the detrimental effects of anthrax lethal toxin, diphtheria toxin, cholera toxin, Pseudomonas aeruginosa exotoxin A, Botulinum neurotoxin, ricin, and Zika virus. Our study reveals the practicality of identifying host proteins that mediate multiple disease pathways and discovering broad-spectrum therapies that target these hub proteins.

6.
Nature ; 531(7594): 381-5, 2016 Mar 17.
Article in English | MEDLINE | ID: mdl-26934220

ABSTRACT

The most recent Ebola virus outbreak in West Africa, which was unprecedented in the number of cases and fatalities, geographic distribution, and number of nations affected, highlights the need for safe, effective, and readily available antiviral agents for treatment and prevention of acute Ebola virus (EBOV) disease (EVD) or sequelae. No antiviral therapeutics have yet received regulatory approval or demonstrated clinical efficacy. Here we report the discovery of a novel small molecule GS-5734, a monophosphoramidate prodrug of an adenosine analogue, with antiviral activity against EBOV. GS-5734 exhibits antiviral activity against multiple variants of EBOV and other filoviruses in cell-based assays. The pharmacologically active nucleoside triphosphate (NTP) is efficiently formed in multiple human cell types incubated with GS-5734 in vitro, and the NTP acts as an alternative substrate and RNA-chain terminator in primer-extension assays using a surrogate respiratory syncytial virus RNA polymerase. Intravenous administration of GS-5734 to nonhuman primates resulted in persistent NTP levels in peripheral blood mononuclear cells (half-life, 14 h) and distribution to sanctuary sites for viral replication including testes, eyes, and brain. In a rhesus monkey model of EVD, once-daily intravenous administration of 10 mg kg(-1) GS-5734 for 12 days resulted in profound suppression of EBOV replication and protected 100% of EBOV-infected animals against lethal disease, ameliorating clinical disease signs and pathophysiological markers, even when treatments were initiated three days after virus exposure when systemic viral RNA was detected in two out of six treated animals. These results show the first substantive post-exposure protection by a small-molecule antiviral compound against EBOV in nonhuman primates. The broad-spectrum antiviral activity of GS-5734 in vitro against other pathogenic RNA viruses, including filoviruses, arenaviruses, and coronaviruses, suggests the potential for wider medical use. GS-5734 is amenable to large-scale manufacturing, and clinical studies investigating the drug safety and pharmacokinetics are ongoing.


Subject(s)
Alanine/analogs & derivatives , Antiviral Agents/therapeutic use , Hemorrhagic Fever, Ebola/drug therapy , Macaca mulatta/virology , Ribonucleotides/therapeutic use , Adenosine Monophosphate/analogs & derivatives , Alanine/pharmacokinetics , Alanine/pharmacology , Alanine/therapeutic use , Amino Acid Sequence , Animals , Antiviral Agents/pharmacokinetics , Antiviral Agents/pharmacology , Cell Line, Tumor , Ebolavirus/drug effects , Female , HeLa Cells , Hemorrhagic Fever, Ebola/prevention & control , Humans , Male , Molecular Sequence Data , Organ Specificity , Prodrugs/pharmacokinetics , Prodrugs/pharmacology , Prodrugs/therapeutic use , Ribonucleotides/pharmacokinetics , Ribonucleotides/pharmacology
7.
Sci Rep ; 5: 13476, 2015 Aug 27.
Article in English | MEDLINE | ID: mdl-26310922

ABSTRACT

A longstanding and still-increasing threat to the effective treatment of infectious diseases is resistance to antimicrobial countermeasures. Potentially, the targeting of host proteins and pathways essential for the detrimental effects of pathogens offers an approach that may discover broad-spectrum anti-pathogen countermeasures and circumvent the effects of pathogen mutations leading to resistance. Here we report implementation of a strategy for discovering broad-spectrum host-oriented therapies against multiple pathogenic agents by multiplex screening of drugs for protection against the detrimental effects of multiple pathogens, identification of host cell pathways inhibited by the drug, and screening for effects of the agent on other pathogens exploiting the same pathway. We show that a clinically used antimalarial drug, Amodiaquine, discovered by this strategy, protects host cells against infection by multiple toxins and viruses by inhibiting host cathepsin B. Our results reveal the practicality of discovering broadly acting anti-pathogen countermeasures that target host proteins exploited by pathogens.


Subject(s)
Antigens, Bacterial/pharmacology , Bacterial Toxins/pharmacology , Host-Pathogen Interactions/drug effects , Viruses/drug effects , Amodiaquine/chemistry , Amodiaquine/pharmacology , Animals , Cathepsin B/metabolism , Cell Death/drug effects , Cytosol/drug effects , Cytosol/metabolism , Drug Approval , Ebolavirus/drug effects , Endosomes/drug effects , Endosomes/metabolism , HeLa Cells , Humans , Metabolome/drug effects , Mice , Models, Biological , RAW 264.7 Cells , United States , United States Food and Drug Administration
8.
ACS Infect Dis ; 1(6): 264-71, 2015 Jun 12.
Article in English | MEDLINE | ID: mdl-27622742

ABSTRACT

Herein we report on a diazachrysene class of small molecules that exhibit potent antiviral activity against the Ebola (EBOV) virus. The antiviral compounds are easily synthesized, and the most active compounds have excellent in vitro activity (0.34-0.70 µM) and are significantly less lipophilic than their predecessors. The three most potent diazachrysene antivirals do not exhibit any toxicity in vivo and protected 70-90% of the mice at 10 mg/kg following EBOV challenge. Together, these studies suggest that diazachrysenes are a promising class of compounds for hit to lead optimization and as potential Ebola therapeutics.

9.
Bioorg Med Chem Lett ; 23(21): 5840-3, 2013 Nov 01.
Article in English | MEDLINE | ID: mdl-24064500

ABSTRACT

Previously we reported the optimization of antiviral scaffolds containing benzimidazole and related heterocycles possessing activity against a variety of arenaviruses. These series of compounds were discovered through an HTS campaign of a 400,000 small molecule library using lentivirus-based pseudotypes incorporated with the Lassa virus envelope glycoprotein (LASV GP). This screening also uncovered an alternate series of very potent arenavirus inhibitors based upon an acylhydrazone scaffold. Subsequent SAR analysis of this chemical series involved various substitutions throughout the chemical framework along with assessment of the preferred stereochemistry. These studies led to an optimized analog (ST-161) possessing subnanomolar activity against LASV and submicromolar activity against a number of other viruses in the Arenaviridae family.


Subject(s)
Antiviral Agents/chemistry , Antiviral Agents/pharmacology , Hydrazones/chemistry , Hydrazones/pharmacology , Lassa virus/drug effects , Acylation , Drug Discovery , Humans , Lassa Fever/drug therapy , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology
10.
J Microbiol Methods ; 94(2): 125-132, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23685397

ABSTRACT

Vibrio tubiashii has been linked to disease outbreaks in molluscan species, including oysters, geoducks, and clams. In particular, oyster hatcheries in the Pacific Northwest have been plagued by intermittent vibriosis since 2006. Accurate detection of vibrios, including V. tubiashii, is critical to the hatcheries in order to allow for rapid remediation efforts. The current methods for detection of Vibrio spp. are not ideal for use at the hatchery. Plating samples require time and is not sensitive to extracelluar pathogenic products, such as the secreted zinc-metalloprotease, VtpA. Other sensitive methods to detect bacteria, such as qPCR, require a high level of laboratory skills and expensive supplies that are prohibitive for use at hatchery sites. Thus, hatcheries would benefit from a sensitive, simple method to detect V. tubiashii and its secreted toxin. Here, we describe the development of two inexpensive and highly specific tests for the shellfish-toxic zinc-metalloprotease secreted by V. tubiashii: enzyme-linked immunoassays (ELISA) and a lateral flow immunoassay (dipstick assay). Both technologies rely on a set of monoclonal antibodies used in a sandwich format, with the capture antibody recognizing a different epitope than the detection antibody on the mature VtpA protein. Both assays are quantitative and give colorimetric readouts. The sandwich ELISA was sensitive when VtpA was diluted into PBS, but was markedly less sensitive in conditions that correlate with the environment of hatchery-derived samples, such as in the presence of seawater, algae, or oyster larvae. In contrast, the dipstick assay remained very sensitive in the presence of these contaminants, is less work-intensive, and much more rapid, making this format the preferred assay method for detecting VtpA on site in a hatchery or environmental setting.


Subject(s)
Bacterial Proteins/analysis , Immunoassay/methods , Metalloproteases/analysis , Ostreidae/microbiology , Seafood/microbiology , Vibrio/isolation & purification , Animals , Antibodies, Monoclonal/analysis , Bacterial Proteins/metabolism , Enzyme-Linked Immunosorbent Assay/instrumentation , Enzyme-Linked Immunosorbent Assay/methods , Immunoassay/instrumentation , Metalloproteases/metabolism , Mice , Mice, Inbred BALB C , Ostreidae/chemistry , Seafood/analysis , Vibrio/enzymology , Vibrio/metabolism , Zinc/metabolism
11.
Bioorg Med Chem Lett ; 23(3): 744-9, 2013 Feb 01.
Article in English | MEDLINE | ID: mdl-23265895

ABSTRACT

A chemically diverse library of about 400,000 small molecules was screened for antiviral activity against lentiviral pseudotypes with the Lassa virus envelope glycoprotein (LASV GP) gene incorporated. High-throughput screening resulted in discovery of a hit compound (ST-37) possessing a benzimidazole core which led to a potent compound series. Herein, we report SAR studies which involved structural modifications to the phenyl rings and methylamino linker portion attached to the benzimidazole core. Many analogs in this study possessed single digit nanomolar potency against LASV pseudotypes. Compounds in this benzimidazole series also exhibited nanomolar antiviral activity against pseudotypes generated from other arenavirus envelopes indicating the potential for development of a broad-spectrum inhibitor. Ultimately, lead compound ST-193 was identified and later found to be efficacious in a lethal LASV guinea pig model showing superior protection compared to ribavirin treatment.


Subject(s)
Antiviral Agents/chemical synthesis , Antiviral Agents/pharmacology , Arenavirus/drug effects , Benzimidazoles/chemistry , Drug Discovery , Animals , Antiviral Agents/chemistry , Benzimidazoles/chemical synthesis , Benzimidazoles/pharmacology , Disease Models, Animal , Guinea Pigs , Small Molecule Libraries
12.
Bioorg Med Chem Lett ; 23(3): 750-6, 2013 Feb 01.
Article in English | MEDLINE | ID: mdl-23265900

ABSTRACT

A series of potent arenavirus inhibitors sharing a benzimidazole core were previously reported by our group. SAR studies were expanded beyond the previous analysis, which involved the attached phenyl rings and methylamino linker portion, to include modifications focused on the benzimidazole core. These changes included the introduction of various substituents to the bicyclic benzimidazole ring system along with alternate core heterocycles. Many of the analogs containing alternate nitrogen-based bicyclic ring systems were found to retain antiviral potency compared to the benzimidazole series from which we derived our lead compound, ST-193. In fact, 21 h, built on an imidazopyridine core, possessed a near tenfold increase in potency against Lassa virus pseudotypes compared to ST-193. As found with the benzimidazole series, broad-spectrum arenavirus activity was also observed for a number of the analogs discovered during this study.


Subject(s)
Antiviral Agents/chemical synthesis , Antiviral Agents/pharmacology , Arenavirus/drug effects , Benzimidazoles/chemistry , Drug Discovery , Heterocyclic Compounds/chemical synthesis , Antiviral Agents/chemistry , Benzimidazoles/chemical synthesis , Benzimidazoles/pharmacology , Heterocyclic Compounds/chemistry , Heterocyclic Compounds/pharmacology , Lassa virus/drug effects , Structure-Activity Relationship
13.
Bioorg Med Chem Lett ; 22(13): 4263-72, 2012 Jul 01.
Article in English | MEDLINE | ID: mdl-22664128

ABSTRACT

A series of acylthiourea derivatives were designed, synthesized, and evaluated for broad-spectrum antiviral activity with selected viruses from Poxviridae (vaccinia virus) and two different genera of the family Bunyaviridae (Rift Valley fever and La Crosse viruses). A compound selected from a library screen, compound 1, displayed submicromolar antiviral activity against both vaccinia virus (EC(50)=0.25 µM) and La Crosse virus (EC(50)=0.27 µM) in cytopathic effect (CPE) assays. SAR analysis was performed to further improve antiviral potency and to optimize drug-like properties of the initial hits. During our analysis, we identified 26, which was found to be nearly fourfold more potent than 1 against both vaccinia and La Crosse viruses. Selected compounds were further tested to more fully characterize the spectrum of antiviral activity. Many of these possessed single digit micromolar and sub-micromolar antiviral activity against a diverse array of targets, including influenza virus (Orthomyxoviridae), Tacaribe virus (Arenaviridae), and dengue virus (Flaviviridae).


Subject(s)
Antiviral Agents/chemistry , Thiourea/chemistry , Antiviral Agents/chemical synthesis , Antiviral Agents/pharmacology , Arenavirus/drug effects , Dengue Virus/drug effects , La Crosse virus/drug effects , Orthomyxoviridae/drug effects , Structure-Activity Relationship , Thiourea/chemical synthesis , Thiourea/pharmacology , Vaccinia virus/drug effects
14.
Dis Aquat Organ ; 85(2): 123-31, 2009 Jun 10.
Article in English | MEDLINE | ID: mdl-19694172

ABSTRACT

Vibrio tubiashii, a pathogen of shellfish larvae and juveniles, produces several extracellular products. Here, we document that culture supernatants of several marine Vibrio species showed toxicity to oyster larvae. Treatment of these supernatants with EDTA not only severely diminished proteolytic activities, but also dramatically reduced toxicity to the larvae. Culture supernatants of metalloprotease-deficient mutants of V. tubiashii, V. cholerae, and V. splendidus were impaired in their ability to cause larval death compared to the wild type strains. Culture supernatants of Pseudomonas aeruginosa, known to contain several secreted proteases, showed virtually no toxicity to oyster larvae. Purified V. tubiashii protease A (VtpA), but not the prototype metalloprotease, thermolysin from Bacillus thermoproteolyticus, was highly toxic to the larvae. In addition, toxicity of purified VtpA was much greater for 6-d-old oyster larvae than for 16-d-old larvae. Together, these results indicated that culture supernatants of a variety of Vibrio species are highly toxic to oyster larvae and that the production of a metalloprotease is required for this effect. We propose that there are, as yet uncharacterized, specific substrates contained in larval tissue that are degraded by VtpA as well as certain homologous metalloproteases produced by other marine Vibrio species which, in turn, may contribute to vibriosis.


Subject(s)
Crassostrea/drug effects , Crassostrea/microbiology , Metalloproteases/toxicity , Vibrio/physiology , Vibrio/pathogenicity , Aging , Animals , Bacillus/physiology , Culture Media/toxicity , Larva/drug effects , Larva/microbiology , Metalloproteases/isolation & purification , Metalloproteases/metabolism , Pseudomonas aeruginosa/physiology , Substrate Specificity , Vibrio/enzymology
15.
J Microbiol Methods ; 76(3): 262-8, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19135097

ABSTRACT

Vibrio tubiashii has recently re-emerged as a pathogen of bivalve larvae, causing a marked increase in the mortality of these species within shellfish rearing facilities. This has resulted in substantial losses of seed production and thus created the need for specific as well as sensitive detection methods for this pathogen. In this project, quantitative PCR (qPCR) primers were developed and optimized based upon analysis of the V. tubiashii vtpA gene sequence, encoding a metalloprotease known to cause larval mortality. Standard curves were developed utilizing dilutions of known quantities of V. tubiashii cells that were compared to colony forming unit (CFU) plate counts. The assay was optimized for detection of vtpA with both lab-grown V. tubiashii samples and filter-captured environmental seawater samples seeded with V. tubiashii. In addition, the primers were confirmed to specifically detect only V. tubiashii when tested against a variety of non-target Vibrio species. Validation of the assay was completed by analyzing samples obtained from a shellfish hatchery. The development of this rapid and sensitive assay for quantitative detection of V. tubiashii will accurately determine levels of this bacterium in a variety of seawater samples, providing a useful tool for oyster hatcheries and a method to assess the presence of this bacterium in the current turbulent ocean environment.


Subject(s)
Metalloproteases/genetics , Polymerase Chain Reaction/methods , Vibrio/isolation & purification , Animals , Base Sequence , Colony Count, Microbial , Crassostrea/microbiology , DNA Primers/genetics , DNA, Bacterial/analysis , Genes, Bacterial , Molecular Sequence Data , Seawater/microbiology , Sequence Analysis, DNA , Vibrio/genetics
16.
J Immunol Methods ; 343(1): 1-12, 2009 Mar 31.
Article in English | MEDLINE | ID: mdl-19152798

ABSTRACT

Drug-induced mitochondrial toxicity can occur as a result of inhibition of mitochondrial DNA (mtDNA) replication as with certain nucleoside reverse transcriptase inhibitors or inhibition of mtDNA-encoded protein synthesis as with certain antibacterials. Both types of dysfunction have the overall effect of reducing the level of proteins encoded by mtDNA. A lateral-flow immunoassay which measures the levels of both a mtDNA-encoded protein and a nuclear DNA-encoded protein allows simple and rapid determination of the ratio of these 2 proteins and, hence, identifies changes in mtDNA-encoded protein levels. Here, we describe an assay that compares the level of Complex IV (cytochrome c oxidase), a mitochondrial protein which has 3 subunits encoded by mtDNA and made by mitochondrial ribosomes, with that of frataxin, a protein encoded by nuclear DNA and made by cytosolic ribosomes. We tested a selection of antibacterials and antiretrovirals in cells and show that the ratio of Complex IV: frataxin decreases when a drug inhibits either mtDNA replication or mtDNA-encoded protein synthesis. The results obtained with the assay were confirmed by Western blotting and immunocytochemical analysis. The assay has high reproducibility, requires small amounts of sample, is quantitative, and is able to identify drugs which ultimately lead to a decrease in mtDNA-encoded proteins.


Subject(s)
DNA Replication/drug effects , DNA, Mitochondrial/drug effects , Protein Biosynthesis/drug effects , Anti-Bacterial Agents/pharmacology , Anti-Retroviral Agents/pharmacology , Cell Line , Electron Transport Complex IV/biosynthesis , Humans , Immunoassay , Iron-Binding Proteins/biosynthesis , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondrial Proteins/biosynthesis , Reproducibility of Results , Frataxin
SELECTION OF CITATIONS
SEARCH DETAIL
...