Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Sci Rep ; 6: 22908, 2016 Mar 11.
Article in English | MEDLINE | ID: mdl-26965911

ABSTRACT

Analysis of environmental DNA (eDNA) enables the detection of species of interest from water and soil samples, typically using species-specific PCR. Here, we describe a method to characterize the biodiversity of a given environment by amplifying eDNA using primer pairs targeting a wide range of taxa and high-throughput sequencing for species identification. We tested this approach on 91 water samples of 40 mL collected along the Cuyahoga River (Ohio, USA). We amplified eDNA using 12 primer pairs targeting mammals, fish, amphibians, birds, bryophytes, arthropods, copepods, plants and several microorganism taxa and sequenced all PCR products simultaneously by high-throughput sequencing. Overall, we identified DNA sequences from 15 species of fish, 17 species of mammals, 8 species of birds, 15 species of arthropods, one turtle and one salamander. Interestingly, in addition to aquatic and semi-aquatic animals, we identified DNA from terrestrial species that live near the Cuyahoga River. We also identified DNA from one Asian carp species invasive to the Great Lakes but that had not been previously reported in the Cuyahoga River. Our study shows that analysis of eDNA extracted from small water samples using wide-range PCR amplification combined with high-throughput sequencing can provide a broad perspective on biological diversity.


Subject(s)
Biodiversity , Classification , DNA/genetics , Metagenomics , Amphibians/classification , Amphibians/genetics , Animals , Birds/classification , Birds/genetics , Carps/classification , Carps/genetics , Computer Simulation , DNA/isolation & purification , Ecosystem , Environmental Monitoring , High-Throughput Nucleotide Sequencing , Mammals/classification , Mammals/genetics , Plants/classification , Rivers , Species Specificity
2.
Clin Infect Dis ; 56(11): 1637-45, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23362296

ABSTRACT

BACKGROUND: In the United States, emtricitabine/tenofovir disoproxil fumarate (FTC/TDF) is a preferred nucleoside reverse transcriptase inhibitor (NRTI) backbone with lamivudine/abacavir (3TC/ABC) as a commonly used alternative. For patients infected with human immunodeficiency virus (HIV-1) virologically suppressed on a boosted protease inhibitor (PI) + 3TC/ABC regimen, the merits of switching to FTC/TDF as the NRTI backbone are unknown. METHODS: SWIFT was a prospective, randomized, open-label 48-week study to evaluate efficacy and safety of switching to FTC/TDF. Subjects receiving 3TC/ABC + PI + ritonavir (RTV) with HIV-1 RNA < 200 c/mL ≥3 months were randomized to continue 3TC/ABC or switch to FTC/TDF. The primary endpoint was time to loss of virologic response (TLOVR) with noninferiority measured by delta of 12%. Virologic failure (VF) was defined as confirmed rebound or the last HIV-1 RNA measurement on study drug ≥200 c/mL. RESULTS: In total, 311 subjects were treated in this study (155 to PI + RTV + FTC/TDF, 156 to PI + RTV + 3TC/ABC). Baseline characteristics were similar between the arms: 85% male, 28% black, median age, 46 years; and median CD4 532 cells/mm(3). By TLOVR through week 48, switching to FTC/TDF was noninferior compared to continued 3TC/ABC (86.4% vs 83.3%, treatment difference 3.0% (95% confidence interval, -5.1% to 11.2%). Fewer subjects on FTC/TDF experienced VF (3 vs 11; P = .034). FTC/TDF showed greater declines in fasting low-density lipoproteins (LDL), total cholesterol (TC), and triglycerides (TG) with significant declines in LDL and TC beginning at week 12 with no TC/HDL ratio change. Switching to FTC/TDF showed improved NCEP thresholds for TC and TG and improved 10-year Framingham TC calculated scores. Decreased estimated glomerular filtration rate [corrected] (eGFR) was observed in both arms with a larger decrease in the FTC/TDF arm. CONCLUSIONS: Switching to FTC/TDF from 3TC/ABC maintained virologic suppression, had fewer VFs, improved lipid parameters and Framingham scores but decreased eGFR. CLINICALTRIALS.GOV IDENTIFIER: NCT00724711.


Subject(s)
Adenine/analogs & derivatives , Anti-Retroviral Agents/administration & dosage , Deoxycytidine/analogs & derivatives , Dideoxynucleosides/administration & dosage , HIV Infections/drug therapy , Lamivudine/administration & dosage , Organophosphonates/administration & dosage , Protease Inhibitors/administration & dosage , Adenine/administration & dosage , Adenine/adverse effects , Adult , Aged , Anti-Retroviral Agents/adverse effects , Antiretroviral Therapy, Highly Active/methods , Biomarkers/blood , Deoxycytidine/administration & dosage , Deoxycytidine/adverse effects , Dideoxynucleosides/adverse effects , Drug Combinations , Emtricitabine , Female , HIV Infections/blood , HIV Infections/urine , Humans , Kaplan-Meier Estimate , Lamivudine/adverse effects , Male , Middle Aged , Organophosphonates/adverse effects , Prospective Studies , Protease Inhibitors/adverse effects , Proteinuria/urine , Risk , Tenofovir
3.
J Evol Biol ; 22(9): 1882-94, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19619164

ABSTRACT

Although lateral gene transfer (LGT) is now recognized as a major force in the evolution of prokaryotes, the contribution of LGT to the evolution and diversification of eukaryotes is less understood. Notably, transfers of complete pathways are believed to be less likely between eukaryotes, because the successful transfer of a pathway requires the physical clustering of functionally related genes. Here, we report that in one of the closest unicellular relatives of animals, the choanoflagellate, Monosiga, three genes whose products work together in the glutamate synthase cycle are of algal origin. The concerted retention of these three independently acquired genes is best explained as the consequence of a series of adaptive replacement events. More generally, this study argues that (i) eukaryote-to-eukaryote transfers of entire metabolic pathways are possible, (ii) adaptive functional replacements of primary pathways can occur, and (iii) functional replacements involving eukaryotic genes are likely to have also contributed to the evolution of eukaryotes. Lastly, these data underscore the potential contribution of algal genes to the evolution of nonphotosynthetic lineages.


Subject(s)
Biological Evolution , Choanoflagellata/genetics , Gene Transfer, Horizontal , Metabolic Networks and Pathways/genetics , Animals , Cation Transport Proteins/genetics , Eukaryota/genetics , Glutamate Synthase/genetics , Glutamate-Ammonia Ligase/genetics
4.
HIV Clin Trials ; 10(2): 76-87, 2009.
Article in English | MEDLINE | ID: mdl-19487177

ABSTRACT

PURPOSE: The KLEAN study extension assessed the long-term efficacy and safety of fosamprenavir-ritonavir (FPV/r) and lopinavir-ritonavir (LPV/r), both administered with abacavir/lamivudine (ABC/3TC) fixed dose combination, over 144 weeks. METHODS: KLEAN was an open-label, noninferiority study that randomised antiretroviral-naïve patients to FPV/r twice daily (bid) or LPV/r bid with ABC/3TC once daily (qd). Patients with a viral load of <400 copies/mL at Week 48 were eligible to participate in the KLEAN study extension (up to 144 weeks) and continued with their previously randomised therapy. RESULTS: The KLEAN study extension (48 to 144 weeks) randomized 199 patients. The proportion of TLOVR responders (HIV-1 RNA <50 copies/mL) at Week 144 was 73% and 60% in the FPV/r and LPV/r arms, respectively. The proportion of TLOVR responders (<50 copies/mL) was the same irrespective of baseline HIV-1 RNA (>100,000 or 100,000 copies/mL). The Week 144 median (interquartile range) change from baseline CD4+ cell count was 300 (236-433) cells/mm3 and 335 (225-444) cells/mm3 in the FPV/r and LPV/r arms, respectively. Diarrhea was the most frequently reported adverse event. A small proportion of patients (FPV/r, 13%; LPV/r, 9%) discontinued study medication due to adverse events. Three patients (FPV/r, 1; LPV/r, 2) experienced virological failure between Week 48 and Week 144. CONCLUSION: The findings of the KLEAN study extension (48 to 144 weeks) support durable viral suppression with both FPV/r and LPV/r treatment regimens when used in combination with ABC/3TC irrespective of viral load at baseline. Both regimens were well tolerated and had similar safety profiles.


Subject(s)
Anti-HIV Agents/standards , HIV Infections/drug therapy , HIV Protease Inhibitors/standards , HIV-1/drug effects , Adult , Aged , Anti-HIV Agents/pharmacology , Anti-HIV Agents/therapeutic use , Carbamates/pharmacology , Carbamates/standards , Carbamates/therapeutic use , Dideoxynucleosides , Drug Combinations , Female , Furans , HIV Infections/virology , HIV Protease Inhibitors/pharmacology , HIV Protease Inhibitors/therapeutic use , HIV-1/genetics , Humans , Lamivudine/pharmacology , Lamivudine/standards , Lamivudine/therapeutic use , Lopinavir , Male , Middle Aged , Organophosphates/pharmacology , Organophosphates/standards , Organophosphates/therapeutic use , Pyrimidinones/pharmacology , Pyrimidinones/standards , Pyrimidinones/therapeutic use , RNA, Viral/blood , Ritonavir/pharmacology , Ritonavir/standards , Ritonavir/therapeutic use , Sulfonamides/pharmacology , Sulfonamides/standards , Sulfonamides/therapeutic use , Viral Load , Young Adult
5.
Antimicrob Agents Chemother ; 45(11): 3021-8, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11600351

ABSTRACT

Human immunodeficiency virus (HIV) protease inhibitors (PIs) are important components of many highly active antiretroviral therapy regimens. However, development of phenotypic and/or genotypic resistance can occur, including cross-resistance to other PIs. Development of resistance takes place because trough levels of free drug are inadequate to suppress preexisting resistant mutant variants and/or to inhibit de novo-generated resistant mutant variants. There is thus a need for new PIs, which are more potent against mutant variants of HIV and show higher levels of free drug at the trough. We have optimized a series of substituted sulfonamides and evaluated the inhibitors against laboratory strains and clinical isolates of HIV type 1 (HIV-1), including viruses with mutations in the protease gene. In addition, serum protein binding was determined to estimate total drug requirements for 90% suppression of virus replication (plasma IC(90)). Two compounds resulting from our studies, designated DPC 681 and DPC 684, are potent and selective inhibitors of HIV protease with IC(90)s for wild-type HIV-1 of 4 to 40 nM. DPC 681 and DPC 684 showed no loss in potency toward recombinant mutant HIVs with the D30N mutation and a fivefold or smaller loss in potency toward mutant variants with three to five amino acid substitutions. A panel of chimeric viruses constructed from clinical samples from patients who failed PI-containing regimens and containing 5 to 11 mutations, including positions 10, 32, 46, 47, 50, 54, 63, 71, 82, 84, and 90 had mean IC(50) values of <20 nM for DPC 681 and DPC 681, respectively. In contrast, marketed PIs had mean IC(50) values ranging from 200 nM (amprenavir) to >900 nM (nelfinavir).


Subject(s)
HIV Infections/virology , HIV Protease Inhibitors/pharmacology , HIV-1/drug effects , Sulfonamides/pharmacology , Administration, Oral , Animals , Blood Proteins/metabolism , Dogs , Drug Resistance, Microbial , Female , Genotype , HIV Protease Inhibitors/pharmacokinetics , Humans , Injections, Intravenous , Male , Protein Binding , Sulfonamides/pharmacokinetics
6.
J Virol ; 75(11): 4999-5008, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11333879

ABSTRACT

Efavirenz (also known as DMP 266 or SUSTIVA) is a potent nonnucleoside inhibitor of human immunodeficiency virus type 1 (HIV-1) reverse transcriptase (RT) activity and of HIV-1 replication in vitro and in vivo. Most patients on efavirenz-containing regimens have sustained antiviral responses; however, rebounds in plasma viral load have been observed in some patients in association with the emergence of mutant strains of HIV-1. Virus isolates from the peripheral blood mononuclear cells (PBMCs) of patients with such treatment failures, as well as recombinant viruses incorporating viral sequences derived from patient plasma, show reduced in vitro susceptibility to efavirenz in association with mutations in the RT gene encoding K103N, Y188L, or G190S/E substitutions. Patterns of RT gene mutations and in vitro susceptibility were similar in plasma virus and in viruses isolated from PBMCs. Variant strains of HIV-1 constructed by site-directed mutagenesis confirmed the role of K103N, G190S, and Y188L substitutions in reduced susceptibility to efavirenz. Further, certain secondary mutations (V106I, V108I, Y181C, Y188H, P225H, and F227L) conferred little resistance to efavirenz as single mutations but enhanced the level of resistance of viruses carrying these mutations in combination with K103N or Y188L. Viruses with K103N or Y188L mutations, regardless of the initial selecting nonnucleoside RT inhibitor (NNRTI), exhibited cross-resistance to all of the presently available NNRTIs (efavirenz, nevirapine, and delavirdine). Some virus isolates from nevirapine or delavirdine treatment failures that lacked K103N or Y188L mutations remained susceptible to efavirenz in vitro, although the clinical significance of this finding is presently unclear.


Subject(s)
Anti-HIV Agents/pharmacology , HIV Infections/virology , HIV-1/drug effects , Oxazines/pharmacology , Reverse Transcriptase Inhibitors/pharmacology , Alkynes , Amino Acid Substitution , Anti-HIV Agents/therapeutic use , Benzoxazines , Cells, Cultured , Clinical Trials, Phase II as Topic , Cohort Studies , Cyclopropanes , Delavirdine/pharmacology , Drug Resistance, Microbial , Drug Resistance, Multiple , Genotype , HIV Infections/drug therapy , HIV Reverse Transcriptase/genetics , HIV-1/enzymology , HIV-1/genetics , Humans , Leukocytes, Mononuclear/virology , Microbial Sensitivity Tests , Molecular Sequence Data , Mutagenesis, Site-Directed , Mutation , Nevirapine/pharmacology , Oxazines/therapeutic use , Reverse Transcriptase Inhibitors/therapeutic use , Treatment Failure
7.
J Med Chem ; 43(10): 2019-30, 2000 May 18.
Article in English | MEDLINE | ID: mdl-10821714

ABSTRACT

A series of 4-alkenyl and 4-alkynyl-3, 4-dihydro-4-(trifluoromethyl)-2-(1H)-quinazolinones were found to be potent non-nucleoside reverse transcriptase inhibitors (NNRTIs) of human immunodeficiency virus type-1 (HIV-1). The 4-alkenyl-3, 4-dihydro-4-(trifluoromethyl)-2-(1H)-quinazolinones DPC 082 and DPC 083 and the 4-alkynyl-3, 4-dihydro-4-(trifluoromethyl)-2-(1H)-quinazolinones DPC 961 and DPC 963 were found to exhibit low nanomolar potency toward wild-type RF virus (IC(90) = 2.0, 2.1, 2.0, and 1.3 nM, respectively) and various single and many multiple amino acid substituted HIV-1 mutant viruses. The increased potency is combined with favorable plasma serum protein binding as demonstrated by improvements in the percent free drug in human plasma when compared to efavirenz: 3.0%, 2.0%, 1.5%, 2. 8%, and 0.2-0.5% for DPC 082, DPC 083, DPC 961, DPC 963, and efavirenz, respectively.


Subject(s)
Anti-HIV Agents/chemical synthesis , HIV Reverse Transcriptase/antagonists & inhibitors , HIV-1/drug effects , Mutation , Quinazolines/chemical synthesis , Reverse Transcriptase Inhibitors/chemical synthesis , Alkynes , Anti-HIV Agents/blood , Anti-HIV Agents/pharmacology , Benzoxazines , Blood Proteins/metabolism , Cyclopropanes , HIV-1/genetics , Humans , Molecular Structure , Oxazines/blood , Oxazines/pharmacology , Protein Binding , Quinazolines/blood , Quinazolines/pharmacology , Reverse Transcriptase Inhibitors/blood , Reverse Transcriptase Inhibitors/pharmacology , Structure-Activity Relationship , Virus Replication/drug effects
8.
J Med Chem ; 41(13): 2411-23, 1998 Jun 18.
Article in English | MEDLINE | ID: mdl-9632373

ABSTRACT

Using the structural information gathered from the X-ray structures of various cyclic urea/HIVPR complexes, we designed and synthesized many nonsymmetrical P2/P2'-substituted cyclic urea analogues. Our efforts concentrated on using an indazole as one of the P2 substituents since this group imparted enzyme (Ki) potency as well as translation into excellent antiviral (IC90) potency. The second P2 substituent was used to adjust the physical and chemical properties in order to maximize oral bioavailability. Using this approach several very potent (IC90 11 nM) and orally bioavailable (F% 93-100%) compounds were discovered (21, 22). However, the resistance profiles of these compounds were inadequate, especially against the double (I84V/V82F) and ritonavir-selected mutant viruses. Further modification of the second P2 substituent in order to increase H-bonding interactions with the backbone atoms of residues Asp 29, Asp 30, and Gly 48 led to analogues with much better resistance profiles. However, these larger analogues were incompatible with the apparent molecular weight requirements for good oral bioavailability of the cyclic urea class of HIVPR inhibitors (MW < 610).


Subject(s)
Anti-HIV Agents , HIV Protease Inhibitors , Indazoles , Urea , Administration, Oral , Animals , Anti-HIV Agents/chemical synthesis , Anti-HIV Agents/chemistry , Anti-HIV Agents/pharmacology , Azepines/pharmacology , Biological Availability , Cell Line , Chromatography, High Pressure Liquid , Dogs , Drug Design , Drug Resistance, Microbial , HIV Protease Inhibitors/chemical synthesis , HIV Protease Inhibitors/chemistry , HIV Protease Inhibitors/pharmacology , HIV-1/drug effects , HIV-1/genetics , Indazoles/chemical synthesis , Indazoles/chemistry , Indazoles/pharmacology , Mutation , RNA, Viral/biosynthesis , Ritonavir/pharmacology , Structure-Activity Relationship , Transcription, Genetic , Urea/analogs & derivatives , Urea/chemical synthesis , Urea/chemistry , Urea/pharmacology
9.
J Med Chem ; 40(10): 1465-74, 1997 May 09.
Article in English | MEDLINE | ID: mdl-9154969

ABSTRACT

We present several novel P1/P1' substituents that can replace the characteristic benzyl P1/P1' moiety of the cyclic urea based HIV protease inhibitor series. These substituents typically provide 5-10-fold improvements in binding affinity compared to the unsubstituted benzyl analogs. The best substituent was the 3,4-(ethylenedioxy)benzyl group. Proper balancing of the molecule's lipophilicity facilitated the transfer of this improved binding affinity into a superior cellular antiviral activity profile. Several analogs were evaluated further for protein binding and resistance liabilities. Compound 18 (IC90 = 8.7 nM) was chosen for oral bioavailability studies based on its log P and solubility profile. A 10 mg/kg dose in dogs provided modest bioavailability with Cmax = 0.22 microg/mL. X-ray crystallographic analysis of two analogs revealed several interesting features responsible for the 3,4-(ethylenedioxy)benzyl-substituted analog's potency: (1) Comparing the two complexes revealed two distinct binding modes for each P1/P1' substituent; (2) The ethylenedioxy moieties are within 3.6 A of Pro 81 providing additional van der Waals contacts missing from the parent structure; (3) The enzyme's Arg 8 side chain moves away from the P1 substituent to accommodate the increased steric volume while maintaining a favorable hydrogen bond distance between the para oxygen substituent and the guanidine NH.


Subject(s)
HIV Protease Inhibitors/chemistry , Urea/analogs & derivatives , Animals , Anti-HIV Agents/chemical synthesis , Anti-HIV Agents/chemistry , Anti-HIV Agents/pharmacokinetics , Anti-HIV Agents/pharmacology , Biological Availability , Cell Line , Crystallography, X-Ray , Dogs , HIV Protease Inhibitors/chemical synthesis , HIV Protease Inhibitors/pharmacokinetics , HIV Protease Inhibitors/pharmacology , Humans , Magnetic Resonance Spectroscopy , Mass Spectrometry , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...