Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
1.
Pulm Pharmacol Ther ; 75: 102134, 2022 08.
Article in English | MEDLINE | ID: mdl-35613658

ABSTRACT

Primary ciliary dyskinesia (PCD) is a respiratory disease caused by dysfunction of the cilia with currently no approved treatments. This predominantly autosomal recessive disease is caused by mutations in any one of over 50 genes involved in cilia function; DNAI1 is one of the more frequently mutated genes, accounting for approximately 5-10% of diagnosed PCD cases. A codon-optimized mRNA encoding DNAI1 and encapsulated in a lipid nanoparticle (LNP) was administered to mice via aerosolized inhalation resulting in the expression human DNAI1 in the multiciliated cells of the pseudostratified columnar epithelia. The spatial localization of DNAI1 expression in the bronchioles indicate that delivery of the DNAI1 mRNA transpires the lower airways. In a PCD disease model, exposure to the LNP-encapsulated DNAI1 mRNA resulted in increased ciliary beat frequency using high speed videomicroscopy showing the potential for an mRNA therapeutic to correct cilia function in patients with PCD due to DNAI1 mutations.


Subject(s)
Kartagener Syndrome , Animals , Axonemal Dyneins/genetics , Cilia , Humans , Kartagener Syndrome/diagnosis , Kartagener Syndrome/drug therapy , Kartagener Syndrome/genetics , Liposomes , Mice , Mutation , Nanoparticles , RNA, Messenger
2.
Hepatology ; 60(6): 1826-37, 2014 Dec.
Article in English | MEDLINE | ID: mdl-24811404

ABSTRACT

UNLABELLED: Understanding hepatitis C virus (HCV) replication has been limited by access to serial samples of liver, the primary site of viral replication. Our understanding of how HCV replicates and develops drug-resistant variants in the liver is limited. We studied 15 patients chronically infected with genotype 1 HCV treated with telaprevir (TVR)/pegylated-interferon alpha/ribavirin. Hepatic fine needle aspiration was performed before treatment and at hour 10, days 4 and 15, and week 8 after initiation of antiviral therapy. We measured viral kinetics, resistance patterns, TVR concentrations, and host transcription profiles. All patients completed all protocol-defined procedures that were generally well tolerated. First-phase HCV decline (baseline/treatment day 4) was significantly slower in liver than in plasma (slope plasma: -0.29; liver, -0.009; P < 0.001), whereas second-phase decline (posttreatment days 4-15) did not differ between the two body compartments (-0.11 and -0.15, respectively; P = 0.1). TVR-resistant variants were detected in plasma, but not in liver (where only wild-type virus was detected). Based upon nonstructural protein 3 sequence analysis, no compartmentalization of viral populations was observed between plasma and liver compartments. Gene expression profiling revealed strong tissue-specific expression signatures. Human intrahepatic TVR concentration, measured for the first time, was lower, compared to plasma, on a gram per milliliter basis. We found moderate heterogeneity between HCV RNA levels from different intrahepatic sites, indicating differences in hepatic microenvironments. CONCLUSION: These data support an integrated model for HCV replication wherein the host hepatic milieu and innate immunity control the level of viral replication, and the early antiviral response observed in the plasma is predominantly driven by inhibition of hepatic high-level HCV replication sites.


Subject(s)
Hepacivirus/drug effects , Hepatitis C, Chronic/drug therapy , Liver/virology , Oligopeptides/pharmacokinetics , RNA, Viral/blood , Adolescent , Adult , Aged , Biopsy, Fine-Needle , Drug Resistance, Viral , Female , Gene Expression , Hepacivirus/genetics , Hepatitis C, Chronic/metabolism , Humans , Liver/drug effects , Liver/metabolism , Male , Middle Aged , Models, Statistical , Oligopeptides/therapeutic use , Phylogeny , Treatment Outcome , Young Adult
3.
PLoS Comput Biol ; 10(8): e1003772, 2014 Aug.
Article in English | MEDLINE | ID: mdl-25101970

ABSTRACT

For patients infected with hepatitis C virus (HCV), the combination of the direct-acting antiviral agent telaprevir, pegylated-interferon alfa (Peg-IFN), and ribavirin (RBV) significantly increases the chances of sustained virologic response (SVR) over treatment with Peg-IFN and RBV alone. If patients do not achieve SVR with telaprevir-based treatment, their viral population is often significantly enriched with telaprevir-resistant variants at the end of treatment. We sought to quantify the evolutionary dynamics of these post-treatment resistant variant populations. Previous estimates of these dynamics were limited by analyzing only population sequence data (20% sensitivity, qualitative resistance information) from 388 patients enrolled in Phase 3 clinical studies. Here we add clonal sequence analysis (5% sensitivity, quantitative) for a subset of these patients. We developed a computational model which integrates both the qualitative and quantitative sequence data, and which forms a framework for future analyses of drug resistance. The model was qualified by showing that deep-sequence data (1% sensitivity) from a subset of these patients are consistent with model predictions. When determining the median time for viral populations to revert to 20% resistance in these patients, the model predicts 8.3 (95% CI: 7.6, 8.4) months versus 10.7 (9.9, 12.8) months estimated using solely population sequence data for genotype 1a, and 1.0 (0.0, 1.4) months versus 0.9 (0.0, 2.7) months for genotype 1b. For each individual patient, the time to revert to 20% resistance predicted by the model was typically comparable to or faster than that estimated using solely population sequence data. Furthermore, the model predicts a median of 11.0 and 2.1 months after treatment failure for viral populations to revert to 99% wild-type in patients with HCV genotypes 1a or 1b, respectively. Our modeling approach provides a framework for projecting accurate, quantitative assessment of HCV resistance dynamics from a data set consisting of largely qualitative information.


Subject(s)
Antiviral Agents , Hepacivirus , Hepatitis C , Models, Biological , Oligopeptides , Viral Load/drug effects , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Computational Biology , Drug Resistance, Viral , Drug Therapy, Combination , Hepacivirus/classification , Hepacivirus/drug effects , Hepacivirus/genetics , Hepatitis C/drug therapy , Hepatitis C/virology , Humans , Oligopeptides/pharmacology , Oligopeptides/therapeutic use
4.
J Clin Gastroenterol ; 49(4): 336-44, 2015 Apr.
Article in English | MEDLINE | ID: mdl-24828357

ABSTRACT

BACKGROUND: The phase 3 studies of telaprevir (T) in combination with peginterferon α-2a and ribavirin (PR) in treatment-naive genotype 1 chronic hepatitis C virus-infected patients (ADVANCE/ILLUMINATE) were not designed a priori to assess the effect of race and ethnicity on treatment response. However, these factors are important given the lower sustained virologic response (SVR) rates observed in black and Hispanic/Latino patients treated with PR. GOALS: This retrospective pooled analysis evaluated the effect of race or ethnicity on treatment-naive patient response to telaprevir-based therapy and assessed resistant variant profiles. MATERIALS AND METHODS: This analysis comprised patients enrolled in ADVANCE (N=363) and ILLUMINATE (N=540) who received 12 weeks of telaprevir in combination with PR followed by 12 or 36 weeks of PR alone and patients in ADVANCE (N=361) who received 48 weeks of PR alone. Race and ethnicity were self-reported and not mutually exclusive. RESULTS: Higher SVR rates were observed with telaprevir-based therapy compared with PR in blacks [n=99 (62%) vs. n=28 (29%), respectively] and in Hispanics/Latinos [n=89 (72%) vs. n=38 (39%)]. The SVR was lower in telaprevir-treated blacks [n=99 (62%)] compared with nonblacks [n=791 (78%)] and in Hispanic/Latinos compared with non-Hispanics/Latinos [n=89 (72%) vs. n=801 (76%)]. Low discontinuation rates due to adverse events, including rash and anemia, were observed across subgroups. Resistance profiles were similar among the subgroups. CONCLUSIONS: Treatment-naive black and Hispanic/Latino patients with genotype 1 chronic hepatitis C virus infection may benefit from telaprevir-based therapy, an important finding given the lower SVR rates observed in these patients when they are treated with PR alone.


Subject(s)
Hepatitis C, Chronic/drug therapy , Hepatitis C, Chronic/ethnology , Oligopeptides/therapeutic use , Viral Load/drug effects , Adult , Aged , Antiviral Agents/therapeutic use , Drug Therapy, Combination , Female , Genotype , Hepacivirus/genetics , Humans , Interferon-alpha/therapeutic use , Male , Middle Aged , Polyethylene Glycols/therapeutic use , Recombinant Proteins/therapeutic use , Retrospective Studies , Ribavirin/therapeutic use , Young Adult
5.
J Infect Dis ; 210(12): 1871-80, 2014 Dec 15.
Article in English | MEDLINE | ID: mdl-24943725

ABSTRACT

BACKGROUND: Population sequencing (PS) has shown that telaprevir-resistant variants are not typically detectable at baseline (prevalence, ≤5% of patients), and most variants present at the time of treatment failure are no longer detectable at the end of the study. METHODS: To gain insight into the evolution of telaprevir-resistant variants, their baseline prevalence and persistence after treatment was investigated using a more sensitive, deep-sequencing (DS) technique in a large number of treatment-experienced patients from the REALIZE study who were infected with hepatitis C virus genotype 1. RESULTS: Before treatment initiation, telaprevir-resistant variants (T54A, T54S, or R155K in 1%-2% of the viral population) were detected by DS in a fraction (2%) of patients for whom PS failed to detect resistance; these variants were not necessarily detected at the time of treatment failure. Of 49 patients in whom telaprevir-resistant variants were detected by PS at the time of treatment failure but not at the end of the study, DS revealed the presence of variants (V36A/L/M, T54S, or R155K in 1%-36% of the viral population) in 16 patients (33%) at the end of the study. CONCLUSIONS: Similar to PS findings, DS analysis revealed that the frequency of telaprevir-resistant variants before treatment was also low, and variants detected at the time of treatment failure were no longer detectable in the majority of patients during follow-up.


Subject(s)
Antiviral Agents/therapeutic use , Carrier Proteins/genetics , Hepacivirus/enzymology , Hepacivirus/genetics , Hepatitis C, Chronic/drug therapy , Oligopeptides/therapeutic use , Viral Nonstructural Proteins/genetics , Drug Resistance, Viral , Hepacivirus/isolation & purification , High-Throughput Nucleotide Sequencing , Humans , Intracellular Signaling Peptides and Proteins , Mutation Rate , Prevalence
6.
Antimicrob Agents Chemother ; 58(9): 5456-65, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24982088

ABSTRACT

VX-222, a thiophene-2-carboxylic acid derivative, is a selective nonnucleoside inhibitor of the hepatitis C virus (HCV) NS5B RNA-dependent RNA polymerase. In phase 1 and 2 clinical studies, VX-222 demonstrated effective antiviral efficacy, with substantial reductions in plasma HCV RNA in patients chronically infected with genotype 1 HCV. To characterize the potential for selection of VX-222-resistant variants in HCV-infected patients, the HCV NS5B gene was sequenced at baseline and during and after 3 days of VX-222 dosing (monotherapy) in a phase 1 study. Variants with the substitutions L419C/I/M/P/S/V, R422K, M423I/T/V, I482L/N/T, A486S/T/V, and V494A were selected during VX-222 dosing, and their levels declined over time after the end of dosing. Phenotypic analysis of these variants was conducted using HCV replicons carrying site-directed mutations. Of the 17 variants, 14 showed reduced susceptibility to VX-222 compared with the wild type, with the L419C/S and R422K variants having higher levels of resistance (>200-fold) than the rest of the variants (6.8- to 76-fold). The M423I and A486S variants remained susceptible to VX-222. The 50% effective concentration (EC50) for the L419P variant could not be obtained due to the poor replication of this replicon. The majority of the variants (15/17) were less fit than the wild type. A subset of the variants, predominately the L419S and R422K variants, were observed when the efficacy and safety of VX-222- and telaprevir-based regimens given for 12 weeks were investigated in genotype 1 HCV-infected patients in a phase 2 study. The NS3 and NS5B variants selected during the dual combination therapy showed reduced susceptibility to both telaprevir and VX-222 and had a lower replication capacity than the wild type. The phase 1b study has the ClinicalTrials.gov identifier NCT00911963, and the phase 2a study has ClinicalTrials.gov identifier NCT01080222.


Subject(s)
Antiviral Agents/pharmacology , Cyclohexanols/pharmacology , Genetic Variation/drug effects , Genetic Variation/genetics , Hepacivirus/drug effects , Hepacivirus/genetics , Thiophenes/pharmacology , Viral Nonstructural Proteins/antagonists & inhibitors , Amino Acid Sequence , Base Sequence , Drug Resistance, Viral/drug effects , Drug Resistance, Viral/genetics , Genotype , Hepatitis C/drug therapy , Humans , Molecular Sequence Data , Mutation/drug effects , Mutation/genetics , Oligopeptides/pharmacology , Phenotype , Replicon/drug effects , Replicon/genetics , Virus Replication/drug effects , Virus Replication/genetics
7.
J Virol ; 87(3): 1544-53, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23152524

ABSTRACT

The prevalence of naturally occurring hepatitis C virus (HCV) variants that are less sensitive to direct-acting antiviral (DAA) inhibitors has not been fully characterized. We used population sequence analysis to assess the frequency of such variants in plasma samples from 3,447 DAA-naive patients with genotype 1 HCV. In general, HCV variants with lower-level resistance (3- to 25-fold increased 50% inhibitor concentration [IC(50)]) to telaprevir were observed as the dominant species in 0 to 3% of patients, depending on the specific variant, whereas higher-level resistant variants (>25-fold-increased IC(50)) were not observed. Specific variants resistant to NS5A inhibitors were predominant in up to 6% of patients. Most variants resistant to nucleo(s/t)ide active-site NS5B polymerase inhibitors were not observed, whereas variants resistant to non-nucleoside allosteric inhibitors were observed in up to 18% of patients. The presence of DAA-resistant variants in NS5A, NS5B, or NS3 (including telaprevir-resistant variants), in baseline samples of treatment-naive patients receiving a telaprevir-based regimen in phase 3 studies did not affect the sustained viral response (SVR). Treatment-naive patients with viral populations containing the telaprevir-resistant variants NS3 V36M, T54S, or R155K at baseline achieved a 74% SVR rate, whereas patients with no resistant variants detected prior to treatment achieved a 76% SVR rate. The effect of specific resistant variant frequency on response to various DAA treatments in different patient populations, including interferon nonresponders, should be further studied.


Subject(s)
Antiviral Agents/administration & dosage , Drug Resistance, Viral , Hepacivirus/drug effects , Hepacivirus/isolation & purification , Hepatitis C, Chronic/drug therapy , Hepatitis C, Chronic/virology , Antiviral Agents/pharmacology , Humans , Inhibitory Concentration 50 , Molecular Sequence Data , Mutation, Missense , Plasma/virology , RNA, Viral/genetics , Selection, Genetic , Sequence Analysis, DNA , Treatment Outcome , Viral Nonstructural Proteins/genetics
8.
Clin Infect Dis ; 57(2): 221-9, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23575197

ABSTRACT

BACKGROUND: Telaprevir (TVR), a hepatitis C virus (HCV) NS3/4A protease inhibitor, has been approved to treat genotype 1 HCV. To understand the clinical impact of TVR-resistant variants, we analyzed samples from patients in phase 3 clinical trials to determine the frequency and retention of TVR-resistant variants in patients who did not achieve sustained virologic response (SVR). METHODS: A total of 1797 patients were treated with TVR. Resistant variants (V36A/G/I/L/M, T54A/S, I132V [subtype 1a only], R155G/K/T/M, A156F/N/S/T/V, and D168N) were identified after treatment failure and at visits thereafter, by direct (population) sequencing of the NS3/4A region. Kaplan-Meier analysis was used to determine median time to loss of these variants. RESULTS: Resistant variants were observed in 77% (299/388) of patients who did not achieve SVR. Resistance occurred more commonly in subtype 1a (86%; 232/269) than subtype 1b infections (56%; 67/119). After treatment failure, 355 patients had at least 1 follow-up visit (median follow-up period: 9.6 months). Of patients with resistance at time of failure and at least 1 follow-up visit, 60% (153/254) lost resistance. Kaplan-Meier analysis, including all patients with any sequence data after treatment failure, indicated that median time to wild type was 10.6 months (95% confidence interval [CI], 9.47-12.20) in subtype 1a and 0.9 months (95% CI, 0.00-2.07) in subtype 1b infections. CONCLUSIONS: After failure to achieve SVR with TVR-based treatment, resistant variants are observed in most patients. However, presumably due to the lower fitness of those variants, they tend to be replaced with wild-type virus over time.


Subject(s)
Antiviral Agents/therapeutic use , Drug Resistance, Viral , Hepacivirus/drug effects , Hepatitis C, Chronic/drug therapy , Hepatitis C, Chronic/virology , Oligopeptides/therapeutic use , Antiviral Agents/pharmacology , Carrier Proteins/genetics , Hepacivirus/isolation & purification , Humans , Intracellular Signaling Peptides and Proteins , Mutation Rate , Mutation, Missense , Oligopeptides/pharmacology , Retrospective Studies , Sequence Analysis, DNA , Treatment Failure , Viral Nonstructural Proteins/genetics
9.
Hepatology ; 56(6): 2106-15, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22806681

ABSTRACT

UNLABELLED: In the Phase 3 REALIZE study, 662 genotype 1 hepatitis C virus (HCV)-infected patients with prior peginterferon/ribavirin treatment failure (including relapsers, partial, and null responders) were randomized to 12 weeks of telaprevir given immediately (T12/PR48) or following 4 weeks of peginterferon/ribavirin (lead-in T12/PR48), or 12 weeks of placebo (PR48), combined with a total of 48 weeks of peginterferon alfa-2a/ribavirin. Sustained virologic response (SVR) rates were 64% (T12/PR48), 66% (lead-in T12/PR48), and 17% (PR48). This analysis aimed to characterize treatment outcomes and viral variants emerging in telaprevir-treated patients not achieving SVR. HCV NS3·4A population sequencing was performed at baseline, during treatment, and follow-up. Telaprevir-resistant variants were classified into lower-level (3- to 25-fold 50% inhibitory concentration [IC(50) ] increase: V36A/M, T54A/S, R155I/K/M/T, and A156S) and higher-level (>25-fold IC(50) increase: V36M+R155K and A156T/V) resistance. Resistant variants were uncommon at baseline. Overall, 18% (52%, 19%, and 1% of prior null and partial responders and relapsers, respectively) of telaprevir-treated patients had on-treatment virologic failure, with no significant difference with or without a lead-in. Virologic failure during the telaprevir-treatment phase was predominantly associated with higher-level resistance; virologic failure during the peginterferon/ribavirin-treatment phase was associated with higher- or lower-level, or wildtype variants, depending on genotype. Relapse occurred in 9% of patients completing assigned treatment and was generally associated with lower-level resistant variants or wildtype. Resistant variants were no longer detectable by study end (median follow-up of 11 months) in 58% of non-SVR patients. CONCLUSION: In REALIZE, variants emerging in non-SVR, telaprevir-treated patients were similar irrespective of the use of a lead-in and were consistent with those previously reported. In most patients, resistant variants became undetectable over time.


Subject(s)
Antiviral Agents/therapeutic use , Drug Resistance, Viral , Hepacivirus/genetics , Hepatitis C, Chronic/drug therapy , Hepatitis C, Chronic/virology , Oligopeptides/therapeutic use , Antiviral Agents/pharmacology , Chi-Square Distribution , Double-Blind Method , Drug Resistance, Viral/genetics , Drug Therapy, Combination , Genotype , Hepacivirus/drug effects , Hepatitis C, Chronic/blood , Humans , Inhibitory Concentration 50 , Interferon-alpha/therapeutic use , Kaplan-Meier Estimate , Oligopeptides/pharmacology , Polyethylene Glycols/therapeutic use , RNA, Viral/blood , Recombinant Proteins/therapeutic use , Recurrence , Ribavirin/therapeutic use , Treatment Failure
10.
Mol Biol Evol ; 28(1): 437-47, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20829344

ABSTRACT

The origin and evolution of multidomain proteins are driven by diverse processes including fusion/fission, domain shuffling, and alternative splicing. The 20 aminoacyl-tRNA synthetases (AARS) constitute an ancient conserved family of multidomain proteins. The glutamyl-prolyl tRNA synthetase (EPRS) of bilaterian animals is unique among AARSs, containing two functional enzymes catalyzing ligation of glutamate and proline to their cognate transfer RNAs (tRNAs). The ERS and PRS catalytic domains in multiple bilaterian taxa are linked by variable number of helix-turn-helix domains referred to as WHEP-TRS domains. In addition to its canonical aminoacylation activities, human EPRS exhibits a noncanonical function as an inflammation-responsive regulator of translation. Recently, we have shown that the WHEP domains direct this auxiliary function of human EPRS by interacting with an mRNA stem-loop element (interferon-gamma-activated inhibitor of translation [GAIT] element). Here, we show that EPRS is present in the cnidarian Nematostella vectensis, which pushes the origin of the fused protein back to the cnidarian-bilaterian ancestor, 50-75 My before the origin of the Bilateria. Remarkably, the Nematostella EPRS mRNA is alternatively spliced to yield three isoforms with variable number and sequence of WHEP domains and with distinct RNA-binding activities. Whereas one isoform containing a single WHEP domain binds tRNA, a second binds both tRNA and GAIT element RNA. However, the third isoform contains two WHEP domains and like the human ortholog binds specifically to GAIT element RNA. These results suggest that alternative splicing of WHEP domains in the EPRS gene of the cnidarian-bilaterian ancestor gave rise to a novel molecular function of EPRS conserved during metazoan evolution.


Subject(s)
Amino Acyl-tRNA Synthetases/genetics , Amino Acyl-tRNA Synthetases/metabolism , Cnidaria/enzymology , Cnidaria/genetics , Evolution, Molecular , RNA, Messenger/genetics , RNA, Messenger/metabolism , Alternative Splicing , Amino Acid Sequence , Amino Acyl-tRNA Synthetases/chemistry , Amino Acyl-tRNA Synthetases/classification , Animals , Base Sequence , Gene Duplication , Humans , Molecular Sequence Data , Nucleic Acid Conformation , Phylogeny , Protein Isoforms/chemistry , Protein Isoforms/classification , Protein Isoforms/genetics , Protein Isoforms/metabolism , Sequence Alignment , Surface Plasmon Resonance
11.
Virol J ; 9: 147, 2012 Aug 06.
Article in English | MEDLINE | ID: mdl-22866919

ABSTRACT

BACKGROUND: Development of compensatory mutations within the HIV p7/p1 and p1/p6 protease cleavage site region has been observed in HIV-infected patients treated with protease inhibitors. Mechanisms of fitness compensation may occur in HCV populations upon treatment of HCV protease inhibitors as well. FINDINGS: In this study, we investigated whether substitutions in protease cleavage site regions of HCV occur in response to a treatment regimen containing the NS3/4A protease inhibitor telaprevir (TVR). Evaluation of viral populations from 569 patients prior to treatment showed that the four NS3/4A cleavage sites were well conserved. Few changes in the cleavage site regions were observed in the 159 patients who failed TVR combination treatment, and no residues displayed evidence of directional selection after the acquisition of TVR-resistance. CONCLUSIONS: Cleavage site mutations did not occur after treatment with the HCV protease inhibitor telaprevir.


Subject(s)
Carrier Proteins/genetics , Drug Resistance, Viral , Hepacivirus/genetics , Hepatitis, Chronic/virology , Mutation, Missense , Oligopeptides/administration & dosage , Viral Nonstructural Proteins/genetics , Amino Acid Substitution , Antiviral Agents/administration & dosage , Hepacivirus/enzymology , Hepacivirus/isolation & purification , Hepatitis, Chronic/drug therapy , Humans , Intracellular Signaling Peptides and Proteins , Treatment Failure
12.
Clin Transl Sci ; 14(2): 656-663, 2021 03.
Article in English | MEDLINE | ID: mdl-33278322

ABSTRACT

In previous work, participants with a G970R mutation in cystic fibrosis transmembrane conductance regulator (CFTR) (c.2908G>C) had numerically lower sweat chloride responses during ivacaftor treatment than participants with other CFTR gating mutations. The objective of this substudy was to characterize the molecular defect of the G970R mutation in vitro and assess the benefit of ivacaftor in participants with this mutation. This substudy assessed sweat chloride, spirometry findings, and nasal potential difference on and off ivacaftor treatment in three participants with a G970R/F508del genotype. Intestinal organoids derived from rectal biopsy specimens were used to assess ivacaftor response ex vivo and conduct messenger RNA splice and protein analyses. No consistent or meaningful trends were observed between on-treatment and off-treatment clinical assessments. Organoids did not respond to ivacaftor in forskolin-induced swelling assays; no mature CFTR protein was detected in Western blots. Organoid RNA analysis demonstrated that 3 novel splice variants were created by G970R-CFTR: exon 17 truncation, exons 13-15 and 17 skipping, and intron 17 retention. Functional and molecular analyses indicated that the c.2908G>C mutation caused a cryptic splicing defect. Organoids lacked an ex vivo response with ivacaftor and supported identification of the mechanism underlying the CFTR defect caused by c.2908G>C. Analysis of CFTR mutations indicated that cryptic splicing was a rare cause of mutation misclassification in engineered cell lines. This substudy used organoids as an alternative in vitro model for mutations, such as cryptic splice mutations that cannot be fully assessed using cDNA expressed in recombinant cell systems.


Subject(s)
Aminophenols/administration & dosage , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Cystic Fibrosis/drug therapy , Quinolones/administration & dosage , Adolescent , Adult , Aminophenols/adverse effects , Biopsy , Cell Line , Cells, Cultured , Child , Cystic Fibrosis/genetics , Cystic Fibrosis/pathology , Exons/genetics , Female , Humans , Intestinal Mucosa/cytology , Intestinal Mucosa/pathology , Male , Mutation , Organoids , Precision Medicine/methods , Primary Cell Culture , Quinolones/adverse effects , RNA Splicing , Rectum/cytology , Rectum/pathology , Treatment Outcome , Young Adult
13.
Ann Am Thorac Soc ; 18(3): 433-441, 2021 03.
Article in English | MEDLINE | ID: mdl-33095038

ABSTRACT

Rationale: Ivacaftor's clinical effects in the residual function mutations 3849 + 10kb C→T and D1152H warrant further characterization.Objectives: To evaluate ivacaftor's effect in people with cystic fibrosis aged ≥6 years with 3849 + 10kb C→T or D1152H residual function mutations and to explore the correlation between ivacaftor-induced organoid-based cystic fibrosis transmembrane conductance regulator function measurements and clinical response to ivacaftor.Methods: Participants were randomized (1:1) in this placebo-controlled crossover study; each treatment sequence included two 8-week treatments with an 8-week washout period. The primary endpoint was absolute change in lung clearance index2.5 from baseline through Week 8. Additional endpoints included lung function, patient-reported outcomes, and in vitro intestinal organoid-based measurements of ivacaftor-induced cystic fibrosis transmembrane conductance regulator function.Results: Of 38 participants, 37 completed the study. The primary endpoint was met; the Bayesian posterior probability of improvement in lung clearance index2.5 with ivacaftor versus placebo was >99%. Additional endpoints improved with ivacaftor. Safety findings were consistent with ivacaftor's known safety profile. Dose-dependent swelling was observed in 23 of 25 viable organoid cultures with ivacaftor treatment. Correlations between ivacaftor-induced organoid swelling and clinical endpoints were negligible to low.Conclusions: In people with cystic fibrosis aged ≥6 years with a 3849 + 10kb C→T or D1152H mutation, ivacaftor treatment improved clinical endpoints compared with placebo; however, there was no correlation between organoid swelling and change in clinical endpoints. The organoid assay may assist in identification of ivacaftor-responsive mutations but in this study did not predict magnitude of clinical benefit for individual people with cystic fibrosis with these two mutations.Clinical trial registered with ClinicalTrials.gov (NCT03068312).


Subject(s)
Cystic Fibrosis , Aminophenols/therapeutic use , Bayes Theorem , Cross-Over Studies , Cystic Fibrosis/drug therapy , Cystic Fibrosis/genetics , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Forced Expiratory Volume , Humans , Mutation , Quinolones
14.
Nucleic Acids Res ; 36(Database issue): D607-11, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17982171

ABSTRACT

The starlet sea anemone, Nematostella vectensis, is a basal metazoan organism that has recently emerged as an important model system in developmental biology and evolutionary genomics. StellaBase, the Nematostella Genomics Database (http://stellabase.org), was developed in 2005 as a resource to support the Nematostella research community. Recently, it has become apparent that Nematostella may be a particularly useful system for studying (i) microevolutionary variation in natural populations, and (ii) the functional evolution of human disease genes. We have developed two new databases that will foster such studies: StellaBase Disease (http://stellabase.org/disease) is a relational database that houses 155 904 invertebrate homologous isoforms of human disease genes from four leading genomic model systems (fly, worm, yeast and Nematostella), including 14 874 predicted genes from the sea anemone itself. StellaBase SNP (http://stellabase.org/SNP) is a relational database that describes the location and underlying type of mutation for 20 063 single nucleotide polymorphisms.


Subject(s)
Databases, Genetic , Disease Models, Animal , Genetic Diseases, Inborn/genetics , Polymorphism, Single Nucleotide , Sea Anemones/genetics , Animals , Caenorhabditis elegans/genetics , Drosophila melanogaster/genetics , Genomics , Humans , Internet , Proteins/chemistry , Proteins/genetics , Saccharomyces cerevisiae/genetics , Sequence Homology, Amino Acid , User-Computer Interface
15.
J Am Acad Audiol ; 31(2): 87-95, 2020 02.
Article in English | MEDLINE | ID: mdl-31429400

ABSTRACT

BACKGROUND: Untreated sensorineural hearing loss (SNHL) has been linked to depression, social isolation, anxiety, and a reduction in health-related quality of life (QoL), and is independently associated with cognitive decline. Only one in five persons with SNHL pursues amplification; 76-97% of those having hearing aids report regular or occasional use. Although hearing aid use during all waking hours is advocated for children, recommendations for adults are not as clear. Treatment outcomes, including benefit, satisfaction, and self-efficacy with hearing aids, may be predictors of self-reported hearing aid use, which is useful in clinical practice. PURPOSE: The aim of this study was to determine average hours of self-reported daily hearing aid use by adults and if treatment outcome measures of benefit, satisfaction, and self-efficacy with hearing aids can predict self-reported daily hearing aid use in adults. RESEARCH DESIGN: The present study was a prospective cross-sectional survey with retrospective chart review. STUDY SAMPLE: The study sample consisted of 152 experienced adult advanced digital technology (ADT) hearing aid users between 18 and 90 years of age who were patients in a two-office private practice in California. DATA COLLECTION AND ANALYSIS: A postal survey was sent to 500 experienced adult ADT hearing aid users. Participants completed the Visual Analog Scale for Daily Use of Hearing Aids (VASuse) and validated measures of (1) self-efficacy, (2) satisfaction, and (3) benefit. Retrospective data were collected for all respondents via chart review. Multivariable linear regression was used to explore relationships between treatment outcomes and hearing aid use. RESULTS: Experienced hearing aid users wore their hearing aids an average of 12.0 h/day. Daily hearing aid use was significantly associated with residual participation restriction (RPR) on the International Outcome Inventory for Hearing Aids (IOI-HA) item 5 (p = 0.02). The VASuse was significantly associated with the IOI-HA factor 1, "Me and My Hearing Aids" (p = 0.03), an aggregate measure of satisfaction, benefit, and QoL. CONCLUSIONS: Participants reported wearing their hearing aids an average of 12.0 h/day. Self-reported daily hearing aid use was associated with a combination of satisfaction, benefit, and increased QoL, and with RPR. The interconnectedness of satisfaction, benefit, and QoL positively affected hearing aid use, and greater levels of RPR seemed to discourage hearing aid use. If hearing aid owners are inconsistent or nonusers, then counseling and outcome measures should be used in the domains of satisfaction, benefit, and QoL. Future research should involve additional ADT hearing aid users with different experience levels across various study sites.


Subject(s)
Hearing Aids/statistics & numerical data , Hearing Loss, Sensorineural/rehabilitation , Patient Compliance/statistics & numerical data , Patient Satisfaction/statistics & numerical data , Quality of Life , Adolescent , Adult , Aged , Aged, 80 and over , Cross-Sectional Studies , Female , Humans , Linear Models , Male , Middle Aged , Prospective Studies , Retrospective Studies , Self Efficacy , Surveys and Questionnaires , United States , Young Adult
16.
Mol Biol Evol ; 25(4): 737-47, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18222944

ABSTRACT

Gene families, which encode toxins, are found in many poisonous animals, yet there is limited understanding of their evolution at the nucleotide level. The release of the genome draft sequence for the sea anemone Nematostella vectensis enabled a comprehensive study of a gene family whose neurotoxin products affect voltage-gated sodium channels. All gene family members are clustered in a highly repetitive approximately 30-kb genomic region and encode a single toxin, Nv1. These genes exhibit extreme conservation at the nucleotide level which cannot be explained by purifying selection. This conservation greatly differs from the toxin gene families of other animals (e.g., snakes, scorpions, and cone snails), whose evolution was driven by diversifying selection, thereby generating a high degree of genetic diversity. The low nucleotide diversity at the Nv1 genes is reminiscent of that reported for DNA encoding ribosomal RNA (rDNA) and 2 hsp70 genes from Drosophila, which have evolved via concerted evolution. This evolutionary pattern was experimentally demonstrated in yeast rDNA and was shown to involve unequal crossing-over. Through sequence analysis of toxin genes from multiple N. vectensis populations and 2 other anemone species, Anemonia viridis and Actinia equina, we observed that the toxin genes for each sea anemone species are more similar to one another than to those of other species, suggesting they evolved by manner of concerted evolution. Furthermore, in 2 of the species (A. viridis and A. equina) we found genes that evolved under diversifying selection, suggesting that concerted evolution and accelerated evolution may occur simultaneously.


Subject(s)
Evolution, Molecular , Genome/genetics , Neurotoxins/genetics , Sea Anemones/genetics , Sequence Analysis, DNA , Animals , Base Sequence , Genetic Variation , Geography , Molecular Sequence Data , Phylogeny , Sequence Alignment
17.
J Parasitol ; 95(1): 100-12, 2009 Feb.
Article in English | MEDLINE | ID: mdl-18656992

ABSTRACT

The evolution of parasitism is often accompanied by profound changes to the developmental program. However, relatively few studies have directly examined the developmental evolution of parasitic species from free-living ancestors. The lined sea anemone Edwardsiella lineata is a relatively recently evolved parasite for which closely related free-living outgroups are known, including the starlet sea anemone Nematostella vectensis. The larva of E. lineata parasitizes the ctenophore Mnemiopsis leidyi, and, once embedded in its host, the anemone assumes a novel vermiform body plan. That we might begin to understand how the developmental program of this species has been transformed during the evolution of parasitism, we characterized the gross anatomy, histology, and cnidom of the parasitic stage, post-parasitic larval stage, and adult stage of the E. lineata life cycle. The distinct parasitic stage of the life cycle differs from the post-parasitic larva with respect to overall shape, external ciliation, cnida frequency, and tissue architecture. The parasitic stage and planula both contain holotrichs, a type of cnida not previously reported in Edwardsiidae. The internal morphology of the post-parasitic planula is extremely similar to the adult morphology, with a complete set of mesenterial tissue and musculature despite this stage having little external differentiation. Finally, we observed 2 previously undocumented aspects of asexual reproduction in E. lineata: (1) the parasitic stage undergoes transverse fission via physal pinching, the first report of asexual reproduction in a pre-adult stage in the Edwardsiidae; and (2) the juvenile polyp undergoes transverse fission via polarity reversal, the first time this form of fission has been reported in E. lineata.


Subject(s)
Anthozoa/physiology , Ctenophora/parasitology , Life Cycle Stages/physiology , Animals , Anthozoa/anatomy & histology , Anthozoa/growth & development , Biological Evolution , Host-Parasite Interactions , Larva/anatomy & histology
18.
BMC Evol Biol ; 8: 228, 2008 Aug 05.
Article in English | MEDLINE | ID: mdl-18681949

ABSTRACT

BACKGROUND: Members of the Runx family of transcriptional regulators, which bind DNA as heterodimers with CBFbeta, are known to play critical roles in embryonic development in many triploblastic animals such as mammals and insects. They are known to regulate basic developmental processes such as cell fate determination and cellular potency in multiple stem-cell types, including the sensory nerve cell progenitors of ganglia in mammals. RESULTS: In this study, we detect and characterize the hitherto unexplored Runx/CBFbeta genes of cnidarians and sponges, two basal animal lineages that are well known for their extensive regenerative capacity. Comparative structural modeling indicates that the Runx-CBFbeta-DNA complex from most cnidarians and sponges is highly similar to that found in humans, with changes in the residues involved in Runx-CBFbeta dimerization in either of the proteins mirrored by compensatory changes in the binding partner. In situ hybridization studies reveal that Nematostella Runx and CBFbeta are expressed predominantly in small isolated foci at the base of the ectoderm of the tentacles in adult animals, possibly representing neurons or their progenitors. CONCLUSION: These results reveal that Runx and CBFbeta likely functioned together to regulate transcription in the common ancestor of all metazoans, and the structure of the Runx-CBFbeta-DNA complex has remained extremely conserved since the human-sponge divergence. The expression data suggest a hypothesis that these genes may have played a role in nerve cell differentiation or maintenance in the common ancestor of cnidarians and bilaterians.


Subject(s)
Cnidaria/genetics , Core Binding Factor alpha Subunits/genetics , Core Binding Factor beta Subunit/genetics , Porifera/genetics , Transcription Factors/genetics , Animals , Cnidaria/classification , Contig Mapping , Core Binding Factor alpha Subunits/chemistry , Core Binding Factor beta Subunit/chemistry , Evolution, Molecular , Expressed Sequence Tags , Models, Molecular , Phylogeny , Porifera/classification , Protein Conformation , Transcription Factors/chemistry
19.
Nucleic Acids Res ; 34(Database issue): D495-9, 2006 Jan 01.
Article in English | MEDLINE | ID: mdl-16381919

ABSTRACT

StellaBase, the Nematostella vectensis Genomics Database, is a web-based resource that will facilitate desktop and bench-top studies of the starlet sea anemone. Nematostella is an emerging model organism that has already proven useful for addressing fundamental questions in developmental evolution and evolutionary genomics. StellaBase allows users to query the assembled Nematostella genome, a confirmed gene library, and a predicted genome using both keyword and homology based search functions. Data provided by these searches will elucidate gene family evolution in early animals. Unique research tools, including a Nematostella genetic stock library, a primer library, a literature repository and a gene expression library will provide support to the burgeoning Nematostella research community. The development of StellaBase accompanies significant upgrades to CnidBase, the Cnidarian Evolutionary Genomics Database. With the completion of the first sequenced cnidarian genome, genome comparison tools have been added to CnidBase. In addition, StellaBase provides a framework for the integration of additional species-specific databases into CnidBase. StellaBase is available at http://www.stellabase.org.


Subject(s)
Databases, Genetic , Genomics , Sea Anemones/genetics , Animals , Evolution, Molecular , Gene Expression , Internet , Oligonucleotide Probes/chemistry , Proteins/genetics , Sea Anemones/metabolism , Systems Integration , User-Computer Interface
20.
Biol Bull ; 214(3): 233-54, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18574101

ABSTRACT

Salt marshes are challenging habitats due to natural variability in key environmental parameters including temperature, salinity, ultraviolet light, oxygen, sulfides, and reactive oxygen species. Compounding this natural variation, salt marshes are often heavily impacted by anthropogenic insults including eutrophication, toxic contamination, and coastal development that alter tidal and freshwater inputs. Commensurate with this environmental variability, estuarine animals generally exhibit broader physiological tolerances than freshwater, marine, or terrestrial species. One factor that determines an organism's physiological tolerance is its ability to upregulate "stress-response genes" in reaction to particular stressors. Comparative studies on diverse organisms have identified a number of evolutionarily conserved genes involved in responding to abiotic and biotic stressors. We used homology-based scans to survey the sequenced genome of Nematostella vectensis, the starlet sea anemone, an estuarine specialist, to identify genes involved in the response to three kinds of insult-physiochemical insults, pathogens, and injury. Many components of the stress-response networks identified in triploblastic animals have clear orthologs in the sea anemone, meaning that they must predate the cnidarian-triploblast split (e.g., xenobiotic receptors, biotransformative genes, ATP-dependent transporters, and genes involved in responding to reactive oxygen species, toxic metals, osmotic shock, thermal stress, pathogen exposure, and wounding). However, in some instances, stress-response genes known from triploblasts appear to be absent from the Nematostella genome (e.g., many metal-complexing genes). This is the first comprehensive examination of the genomic stress-response repertoire of an estuarine animal and a member of the phylum Cnidaria. The molecular markers of stress response identified in Nematostella may prove useful in monitoring estuary health and evaluating coastal conservation efforts. These data may also inform conservation efforts on other cnidarians, such as the reef-building corals.


Subject(s)
Adaptation, Physiological/genetics , Genome , Genomics , Sea Anemones/genetics , Animals , Heat-Shock Response/genetics , Osmotic Pressure , Oxidative Stress/genetics , Seawater
SELECTION OF CITATIONS
SEARCH DETAIL