Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Pediatr Qual Saf ; 7(5): e600, 2022.
Article in English | MEDLINE | ID: mdl-36168514

ABSTRACT

Mother's own milk (MOM) reduces complications of preterm birth. Despite high initiation rates of expression, half of preterm infants do not receive MOM at discharge. Frequent outreach and a short message service (SMS) have improved MOM provision in term dyads. We aimed to improve MOM provision rate from 61% to >80% by implementing standardized lactation education and Breastfeeding & Lactation Outreach via SMS Supporting Mothers (BLOSSoM). Methods: The baseline period was June 2019 to April 2020. A multidisciplinary team implemented PDSA cycles: education/documentation (standardized lactation education and education documentation, May 2020-April 2021), and BLOSSoM (SMS program providing educational texts/videos, reminders, 2-way communication with neonatal intensive care unit (NICU) lactation, May 2021-December 2021). The primary outcome was MOM provision at NICU discharge/transfer for infants younger than 34 weeks, as analyzed on the SPC chart. BLOSSoM participants evaluated the program using a 5-point Likert scale. Results: Demographic and clinical characteristics were unchanged among the three periods. However, the monthly MOM provision rate improved from 61% to 81%. Eighty-seven percent of BLOSSoM participants completed the evaluation with 83% rating the program most supportive, 78% rating the videos as the most helpful, followed by team check-ins (54%) and 2-way texting (24%). Conclusions: Using a multidisciplinary approach, we improved the monthly MOM provision rate at discharge/transfer for preterm infants. SMS providing educational texts/media and 2-way communication supporting lactating NICU mothers was critical to our success. Providing another method of communication through SMS was well accepted and valued by the majority.

2.
Article in English | MEDLINE | ID: mdl-31160281

ABSTRACT

Substitutions at residue Y181 in HIV-1 reverse transcriptase (RT), in particular, Y181C, Y181I, and Y181V, are associated with nonnucleoside RT inhibitor (NNRTI) cross-resistance. In this study, we used kinetic and thermodynamic approaches, in addition to molecular modeling, to gain insight into the mechanisms by which these substitutions confer resistance to nevirapine (NVP), efavirenz (EFV), and rilpivirine (RPV). Using pre-steady-state kinetics, we found that the dissociation constant (Kd ) values for inhibitor binding to the Y181C and Y181I RT-template/primer (T/P) complexes were significantly reduced. In the presence of saturating concentrations of inhibitor, the Y181C RT-T/P complex incorporated the next correct deoxynucleoside triphosphate (dNTP) more efficiently than the wild-type (WT) complex, and this phenotype correlated with decreased mobility of the RT on the T/P substrate. Interestingly, we found that the Y181F substitution in RT-which represents a transitional mutation between Y181 and Y181I/V, or a partial revertant-conferred hypersusceptibility to EFV and RPV at both the virus and enzyme levels. EFV and RPV bound more tightly to Y181F RT-T/P. Furthermore, inhibitor-bound Y181F RT-T/P was less efficient than the WT complex in incorporating the next correct dNTP, and this could be attributed to increased mobility of Y181F RT on the T/P substrate. Collectively, our data highlight the key role that Y181 in RT plays in NNRTI binding.


Subject(s)
HIV Reverse Transcriptase/genetics , HIV-1/enzymology , Reverse Transcriptase Inhibitors/pharmacology , Alkynes , Anti-HIV Agents/pharmacology , Benzoxazines/pharmacology , Cyclopropanes , Drug Resistance, Viral/genetics , Fluorescence Polarization , HIV-1/drug effects , HIV-1/genetics , Humans , Mutation/genetics , Nevirapine/pharmacology , Rilpivirine/pharmacology
3.
Proc Natl Acad Sci U S A ; 112(22): 6979-84, 2015 Jun 02.
Article in English | MEDLINE | ID: mdl-26038551

ABSTRACT

Fragment-based screening methods can be used to discover novel active site or allosteric inhibitors for therapeutic intervention. Using saturation transfer difference (STD) NMR and in vitro activity assays, we have identified fragment-sized inhibitors of HIV-1 reverse transcriptase (RT) with distinct chemical scaffolds and mechanisms compared to nonnucleoside RT inhibitors (NNRTIs) and nucleoside/nucleotide RT inhibitors (NRTIs). Three compounds were found to inhibit RNA- and DNA-dependent DNA polymerase activity of HIV-1 RT in the micromolar range while retaining potency against RT variants carrying one of three major NNRTI resistance mutations: K103N, Y181C, or G190A. These compounds also inhibit Moloney murine leukemia virus RT but not the Klenow fragment of Escherichia coli DNA polymerase I. Steady-state kinetic analyses demonstrate that one of these fragments is a competitive inhibitor of HIV-1 RT with respect to deoxyribonucleoside triphosphate (dNTP) substrate, whereas a second compound is a competitive inhibitor of RT polymerase activity with respect to the DNA template/primer (T/P), and consequently also inhibits RNase H activity. The dNTP competing RT inhibitor retains activity against the NRTI-resistant mutants K65R and M184V, demonstrating a drug resistance profile distinct from the nucleotide competing RT inhibitors indolopyridone-1 (INDOPY-1) and 4-dimethylamino-6-vinylpyrimidine-1 (DAVP-1). In antiviral assays, the T/P competing compound inhibits HIV-1 replication at a step consistent with an RT inhibitor. Screening of additional structurally related compounds to the three fragments led to the discovery of molecules with improved potency against HIV-1 RT. These fragment inhibitors represent previously unidentified scaffolds for development of novel drugs for HIV-1 prevention or treatment.


Subject(s)
Drug Discovery/methods , HIV-1/enzymology , Prodrugs/isolation & purification , Reverse Transcriptase Inhibitors/isolation & purification , Reverse Transcriptase Inhibitors/pharmacology , DNA Primers/genetics , Electrophoretic Mobility Shift Assay , Magnetic Resonance Spectroscopy , Prodrugs/analysis , Reverse Transcriptase Inhibitors/analysis , Ribonuclease H/antagonists & inhibitors , Small Molecule Libraries , Virus Replication/drug effects
4.
Nucleic Acids Res ; 42(18): 11687-96, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25232099

ABSTRACT

Non-nucleoside reverse transcriptase (RT) inhibitors (NNRTIs) are routinely used to treat HIV-1 infection, yet their mechanism of action remains unclear despite intensive investigation. In this study, we developed complementary single-molecule fluorescence and ensemble fluorescence anisotropy approaches to discover how NNRTIs modulate the intra-molecular conformational changes and inter-molecular dynamics of RT-template/primer (T/P) and RT-T/P-dNTP complexes. We found that NNRTI binding to RT induces opening of the fingers and thumb subdomains, which increases the dynamic sliding motion of the enzyme on the T/P and reduces dNTP binding affinity. Further, efavirenz promotes formation of the E138-K101 salt bridge between the p51 and p66 subunits of RT, which contributes to opening of the thumb/fingers subdomains. Engineering a more polar salt bridge between p51 and p66 resulted in even greater increases in the thumb/fingers opening, RT sliding, dNTP binding disruption and in vitro and in vivo RT inhibition than were observed with wild-type RT. We also observed that K103N, a clinically relevant NNRTI resistance mutation, does not prevent binding between efavirenz and RT-T/P but instead allows formation of a stable and productive RT-T/P-dNTP complex, possibly through disruption of the E138-K101 salt bridge. Collectively, these data describe unique structure-activity-resistance relationships that could be exploited for drug development.


Subject(s)
HIV Reverse Transcriptase/antagonists & inhibitors , HIV Reverse Transcriptase/chemistry , Reverse Transcriptase Inhibitors/pharmacology , Alkynes , Allosteric Regulation , Benzoxazines/pharmacology , Cyclopropanes , DNA Primers , Deoxyribonucleotides/metabolism , Fluorescence Polarization , HIV Reverse Transcriptase/genetics , HIV Reverse Transcriptase/metabolism , Mutation , Protein Subunits/chemistry , Templates, Genetic
5.
Antiviral Res ; 107: 31-4, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24746459

ABSTRACT

The nonnucleoside reverse transcriptase (RT) inhibitor rilpivirine (RPV) has been co-formulated with emtricitabine and tenofovir disoproxil fumarate for initial therapy of HIV-1-infected individuals. RPV, formulated as a long-acting nanosuspension, will also be assessed for its ability to prevent HIV-1 infection in resource limited settings. In this study, we determined whether any pre-existing genetic differences occurred among different HIV-1 subtypes at residues in RT associated with decreased virologic response to RPV. We found that the E138A substitution occurs more frequently in subtype C (range: 5.9-7.5%) than B (range: 0-2.3%) sequences from both treatment-naïve and -experienced individuals (p<0.01) in 4 independent genotype databases. In one of the databases (Stanford University), E138K and E138Q were also more common in RTI-experienced subtype C sequences (1.0% and 1.1%, respectively) than in subtype B sequences (0.3% and 0.6%, respectively). E138A/K/Q in subtype C decreased RPV susceptibility 2.9-, 5.8-, and 5.4-fold, respectively. Taken together, these data suggest that E138A could impact treatment or prevention strategies that include RPV in geographic areas where subtype C infection is prevalent.


Subject(s)
Anti-HIV Agents/pharmacology , Drug Resistance, Viral , HIV Reverse Transcriptase/genetics , HIV-1/enzymology , Mutation, Missense , Nitriles/pharmacology , Pyrimidines/pharmacology , Amino Acid Substitution , HIV Infections/drug therapy , HIV Infections/virology , HIV Reverse Transcriptase/metabolism , HIV-1/classification , HIV-1/genetics , Humans , Rilpivirine
6.
PLoS One ; 9(1): e84964, 2014.
Article in English | MEDLINE | ID: mdl-24489654

ABSTRACT

Combination antiretroviral therapy (cART) can effectively suppress HIV-1 replication, but the latent viral reservoir in resting memory CD4(+) T cells is impervious to cART and represents a major barrier to curing HIV-1 infection. Reactivation of latent HIV-1 represents a possible strategy for elimination of this reservoir. In this study we describe the discovery of 1,2,9,10-tetramethoxy-7H-dibenzo[de,g]quinolin-7-one (57704) which reactivates latent HIV-1 in several cell-line models of latency (J89GFP, U1 and ACH-2). 57704 also increased HIV-1 expression in 3 of 4 CD8(+)-depleted blood mononuclear cell preparations isolated from HIV-1-infected individuals on suppressive cART. In contrast, vorinostat increased HIV-1 expression in only 1 of the 4 donors tested. Importantly, 57704 does not induce global T cell activation. Mechanistic studies revealed that 57704 reactivates latent HIV-1 via the phosphatidylinositol 3-kinase (PI3K)/protein kinase B (Akt) signaling pathway. 57704 was found to be an agonist of PI3K with specificity to the p110α isoform, but not the p110ß, δ or γ isoforms. Taken together, our work suggests that 57704 could serve as a scaffold for the development of more potent activators of latent HIV-1. Furthermore, it highlights the involvement of the PI3K/Akt pathway in the maintenance of HIV-1 latency.


Subject(s)
Anti-HIV Agents/pharmacology , CD4-Positive T-Lymphocytes/drug effects , Class Ia Phosphatidylinositol 3-Kinase/metabolism , HIV-1/drug effects , Heterocyclic Compounds, 4 or More Rings/pharmacology , Quinolones/pharmacology , Virus Activation/drug effects , CD4-Positive T-Lymphocytes/enzymology , CD4-Positive T-Lymphocytes/virology , CD8-Positive T-Lymphocytes/virology , Cell Line , Class Ia Phosphatidylinositol 3-Kinase/genetics , Drug Discovery , Drug Therapy, Combination , Enzyme Activation/drug effects , Gene Expression , HIV Infections/drug therapy , HIV Infections/enzymology , HIV Infections/virology , HIV-1/physiology , Histone Deacetylase Inhibitors/pharmacology , Humans , Hydroxamic Acids/pharmacology , Lymphocyte Activation , Lymphocyte Depletion , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Virus Latency , Vorinostat
7.
Antimicrob Agents Chemother ; 58(4): 2430-3, 2014.
Article in English | MEDLINE | ID: mdl-24419343

ABSTRACT

We characterized the relative fitness of multiple nonnucleoside reverse transcriptase (RT) inhibitor (NNRTI)-resistant HIV-1 variants in the presence of etravirine (ETV), rilpivirine (RPV), and/or the nucleoside RT inhibitor emtricitabine (FTC) by simultaneous competitive culture and 454 deep sequencing. The E138A substitution, typically associated with decreased virologic responses to ETV- and RPV-containing regimens, confers a clear fitness advantage to the virus in the presence of FTC and decreases FTC susceptibility 4.7-fold.


Subject(s)
Anti-HIV Agents/pharmacology , Deoxycytidine/analogs & derivatives , HIV Reverse Transcriptase/genetics , HIV Reverse Transcriptase/metabolism , Deoxycytidine/pharmacology , Drug Resistance, Viral/genetics , Emtricitabine , Humans , Virus Replication/drug effects , Virus Replication/genetics
8.
Antimicrob Agents Chemother ; 58(3): 1797-9, 2014.
Article in English | MEDLINE | ID: mdl-24366751

ABSTRACT

Although approved by the U.S. Food and Drug Administration, enfuvirtide is rarely used in combination antiretroviral therapies (cART) to treat HIV-1 infection, primarily because of its intense dosing schedule that requires twice-daily subcutaneous injection. Here, we describe the development of enfuvirtide-loaded, degradable poly(lactic-co-glycolic) acid microparticles that provide linear in vitro release of the drug over an 18-day period. This sustained-release formulation could make enfuvirtide more attractive for use in cART.


Subject(s)
HIV Envelope Protein gp41/administration & dosage , HIV Fusion Inhibitors/administration & dosage , Peptide Fragments/administration & dosage , Cell-Derived Microparticles/ultrastructure , Delayed-Action Preparations , Enfuvirtide , HIV Infections/drug therapy , HIV-1/drug effects , Humans , In Vitro Techniques , Microscopy, Confocal , Microscopy, Electron, Scanning
9.
Retrovirology ; 10: 70, 2013 Jul 09.
Article in English | MEDLINE | ID: mdl-23835323

ABSTRACT

BACKGROUND: Uncoating of the HIV-1 core plays a critical role during early post-fusion stages of infection but is poorly understood. Microscopy-based assays are unable to easily distinguish between intact and partially uncoated viral cores. RESULTS: In this study, we used 5-ethynyl uridine (EU) to label viral-associated RNA during HIV production. At early time points after infection with EU-labeled virions, the viral-associated RNA was stained with an EU-specific dye and was detected by confocal microscopy together with viral proteins. We observed that detection of the viral-associated RNA was specific for EU-labeled virions, was detected only after viral fusion with target cells, and occurred after an initial opening of the core. In vitro staining of cores showed that the opening of the core allowed the small molecule dye, but not RNase A or antibodies, inside. Also, staining of the viral-associated RNA, which is co-localized with nucleocapsid, decays over time after viral infection. The decay rate of RNA staining is dependent on capsid (CA) stability, which was altered by CA mutations or a small molecule inducer of HIV-1 uncoating. While the staining of EU-labeled RNA was not affected by inhibition of reverse transcription, the kinetics of core opening of different CA mutants correlated with initiation of reverse transcription. Analysis of the E45A CA mutant suggests that initial core opening is independent of complete capsid disassembly. CONCLUSIONS: Taken together, our results establish a novel RNA accessibility-based assay that detects an early event in HIV-1 uncoating and can be used to further define this process.


Subject(s)
HIV-1/physiology , Microscopy, Confocal/methods , Virology/methods , Virus Uncoating , Cell Line , Humans , RNA, Viral/analysis , Staining and Labeling/methods , Uridine/analogs & derivatives , Uridine/metabolism , Viral Proteins/analysis
10.
J Virol ; 87(15): 8805-7, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23720723

ABSTRACT

We applied an efficient method to characterize the relative fitness levels of multiple nonnucleoside reverse transcriptase (NNRTI)-resistant HIV-1 variants by simultaneous competitive culture and 454 deep sequencing. Using this method, we show that the Y181V mutation in the HIV-1 reverse transcriptase in particular confers a clear selective advantage to the virus over 14 other NNRTI resistance mutations in the presence of etravirine in vitro.


Subject(s)
Anti-HIV Agents/pharmacology , Drug Resistance, Viral , HIV Reverse Transcriptase/genetics , HIV-1/physiology , Pyridazines/pharmacology , Reverse Transcriptase Inhibitors/pharmacology , Virus Replication , Cell Line , HIV-1/genetics , HIV-1/growth & development , High-Throughput Nucleotide Sequencing , Humans , Nitriles , Pyrimidines , RNA, Viral/genetics , Virulence , Virus Cultivation
11.
J Biol Chem ; 286(25): 22211-8, 2011 Jun 24.
Article in English | MEDLINE | ID: mdl-21531716

ABSTRACT

Deacetylation of histone proteins at the HIV type 1 (HIV-1) long terminal repeat (LTR) by histone deactylases (HDACs) can promote transcriptional repression and virus latency. As such, HDAC inhibitors (HDACI) could be used to deplete reservoirs of persistent, quiescent HIV-1 proviral infection. However, the development of HDACI to purge latent HIV-1 requires knowledge of the HDAC isoforms contributing to viral latency and the development of inhibitors specific to these isoforms. In this study, we identify the HDACs responsible for HIV-1 latency in Jurkat J89GFP cells using a chemical approach that correlates HDACI isoform specificity with their ability to reactivate latent HIV-1 expression. We demonstrate that potent inhibition or knockdown of HDAC1, an HDAC isoform reported to drive HIV-1 into latency, was not sufficient to de-repress the viral LTR. Instead, we found that inhibition of HDAC3 was necessary to activate latent HIV-1. Consistent with this finding, we identified HDAC3 at the HIV-1 LTR by chromatin immunoprecipitation. Interestingly, we show that valproic acid is a weak inhibitor of HDAC3 (IC(50) = 5.5 mm) relative to HDAC1 (IC(50) = 170 µm). Because the total therapeutic concentration of valproic acid ranges from 275 to 700 µm in adults, these data may explain why this inhibitor has no effect on the decay of latent HIV reservoirs in patients. Taken together, our study suggests an important role for HDAC3 in HIV-1 latency and, importantly, describes a chemical approach that can readily be used to identify the HDAC isoforms that contribute to HIV-1 latency in other cell types.


Subject(s)
HIV-1/drug effects , HIV-1/physiology , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/metabolism , Virus Activation/drug effects , Virus Latency/drug effects , Adult , HIV Long Terminal Repeat/genetics , HIV-1/enzymology , HIV-1/genetics , Histone Deacetylases/chemistry , Humans , Hydroxamic Acids/pharmacology , Jurkat Cells , Peptides, Cyclic/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/chemistry , Protein Isoforms/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Substrate Specificity , Valproic Acid/pharmacology
12.
Retrovirology ; 7: 40, 2010 May 07.
Article in English | MEDLINE | ID: mdl-20459669

ABSTRACT

BACKGROUND: Lemay et al recently reported that the RNA binding protein HuR directly interacts with the ribonuclease H (RNase H) domain of HIV-1 reverse transcriptase (RT) and influences the efficiency of viral reverse transcription (Lemay et al., 2008, Retrovirology 5:47). HuR is a member of the embryonic lethal abnormal vision protein family and contains 3 RNA recognition motifs (RRMs) that bind AU-rich elements (AREs). To define the structural determinants of the HuR-RT interaction and to elucidate the mechanism(s) by which HuR influences HIV-1 reverse transcription activity in vitro, we cloned and purified full-length HuR as well as three additional protein constructs that contained the N-terminal and internal RRMs, the internal and C-terminal RRMs, or the C-terminal RRM only. RESULTS: All four HuR proteins were purified and characterized by biophysical methods. They are well structured and exist as monomers in solution. No direct protein-protein interaction between HuR and HIV-1 RT was detected using NMR titrations with 15N labeled HuR variants or the 15N labeled RNase H domain of HIV-1 RT. Furthermore, HuR did not significantly affect the kinetics of HIV-1 reverse transcription in vitro, even on RNA templates that contain AREs. CONCLUSIONS: Our results suggest that HuR does not impact HIV-1 replication through a direct protein-protein interaction with the viral RT.


Subject(s)
Antigens, Surface/metabolism , HIV Reverse Transcriptase/metabolism , HIV-1/physiology , Protein Interaction Mapping , RNA, Viral/metabolism , RNA-Binding Proteins/metabolism , Reverse Transcription , Antigens, Surface/genetics , Antigens, Surface/isolation & purification , Cloning, Molecular , ELAV Proteins , ELAV-Like Protein 1 , Humans , Nuclear Magnetic Resonance, Biomolecular , Protein Multimerization , RNA-Binding Proteins/genetics , RNA-Binding Proteins/isolation & purification , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism
13.
Mol Cancer Ther ; 7(1): 143-51, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18202017

ABSTRACT

2-[(1-methylpropyl)dithio]-1H-imidazole (IV-2) is a known inhibitor of the thioredoxin system. It causes the oxidation of cysteine residues from both thioredoxin reductase and thioredoxin, with only the latter leading to irreversible inhibition of protein function. Although IV-2 is considered to be the first specific inhibitor of thioredoxin to undergo evaluation in cancer patients (under the name PX-12), it is unclear whether the oxidative ability of IV-2 is limited to proteins of the thioredoxin family. The current study investigated the specificity of IV-2 by examining its interaction with tubulin, a protein in which cysteine oxidation causes loss of polymerization competence. The cellular effects of IV-2 were examined in MCF-7 breast cancer and endothelial cells (human umbilical vein endothelial cells). Immunocytochemistry revealed a loss of microtubule structure with Western blot analysis confirming that treated cells contained a higher proportion of unpolymerized tubulin. Cell-free tubulin polymerization assays showed a dose-dependent inhibition of tubulin polymerization and depolymerization of preformed microtubules, confirming a direct interaction between IV-2 and tubulin. Further investigation of the tubulin interaction, through analysis of sulfhydryl reactivity and disulfide bond formation, suggested that IV-2 acts through the oxidation of cysteines in tubulin. Biochemical assays indicated that the oxidative properties of IV-2 are not limited to thioredoxin and tubulin, as cysteine-dependent proteases were also inhibited. Breast cancer cells with thioredoxin silenced by short interfering RNA remained sensitive to IV-2, albeit at higher antiproliferative GI50 values than in cells with normal thioredoxin function. These findings show that modulation of targets other than thioredoxin contribute to the effects of IV-2 on proliferating cells.


Subject(s)
Cysteine/metabolism , Disulfides/pharmacology , Imidazoles/pharmacology , Tubulin/metabolism , Breast Neoplasms/pathology , Cell Survival/drug effects , Cells, Cultured , Disulfides/chemistry , Ficain/antagonists & inhibitors , Ficain/metabolism , Humans , Imidazoles/chemistry , Microtubules/drug effects , Molecular Structure , Oxidation-Reduction/drug effects , Papain/antagonists & inhibitors , Papain/metabolism , Protease Inhibitors/pharmacology , RNA, Small Interfering/genetics , Sensitivity and Specificity , Thioredoxins/genetics , Thioredoxins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...