Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
Mol Ther ; 31(7): 1994-2004, 2023 07 05.
Article in English | MEDLINE | ID: mdl-36805083

ABSTRACT

Gene therapy with an adeno-associated virus serotype 8 (AAV8) vector (AAV8-LSPhGAA) could eliminate the need for enzyme replacement therapy (ERT) by creating a liver depot for acid α-glucosidase (GAA) production. We report initial safety and bioactivity of the first dose (1.6 × 1012 vector genomes/kg) cohort (n = 3) in a 52-week open-label, single-dose, dose-escalation study (NCT03533673) in patients with late-onset Pompe disease (LOPD). Subjects discontinued biweekly ERT after week 26 based on the detection of elevated serum GAA activity and the absence of clinically significant declines per protocol. Prednisone (60 mg/day) was administered as immunoprophylaxis through week 4, followed by an 11-week taper. All subjects demonstrated sustained serum GAA activities from 101% to 235% of baseline trough activity 2 weeks following the preceding ERT dose. There were no treatment-related serious adverse events. No subject had anti-capsid T cell responses that decreased transgene expression. Muscle biopsy at week 24 revealed unchanged muscle glycogen content in two of three subjects. At week 52, muscle GAA activity for the cohort was significantly increased (p < 0.05). Overall, these initial data support the safety and bioactivity of AAV8-LSPhGAA, the safety of withdrawing ERT, successful immunoprophylaxis, and justify continued clinical development of AAV8-LSPhGAA therapy in Pompe disease.


Subject(s)
Glycogen Storage Disease Type II , Humans , alpha-Glucosidases/genetics , alpha-Glucosidases/metabolism , Antibodies/genetics , Enzyme Replacement Therapy/methods , Genetic Therapy/methods , Glycogen Storage Disease Type II/therapy , Glycogen Storage Disease Type II/drug therapy , Liver/metabolism
2.
Biochim Biophys Acta ; 1850(10): 2103-10, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25445708

ABSTRACT

BACKGROUND: Tremendous progress has been made in the past 20 years in understanding the roles played by immunophilins, and in particular the cyclophilins, in supporting the replication cycles of human viruses. A growing body of genetic and biochemical evidence and data from clinical trials confirm that cyclophilins are essential cofactors that contribute to establishing a permissive environment within the host cell that supports the replication of HIV-1 and HCV. Cyclophilin A regulates HIV-1 replication kinetics and infectivity, modulates sensitivity to host restriction factors, and cooperates in the transit of the pre-integration complex into the nucleus of infected cells. Cyclophilin A is an essential cofactor whose expression supports HCV-specific RNA replication in human hepatocytes. GENERAL SIGNIFICANCE: Peptidyl-prolyl isomerase inhibitors have been used in clinical trials to validate cyclophilins as antiviral targets for the treatment of HIV-1 and Chronic Hepatitis C virus infection and as molecular probes to identify the roles played by immunophilins in supporting the replication cycles of human viruses. SCOPE OF REVIEW: This review summarizes emerging research that defines the functions of immunophilins in supporting the replication cycles of HIV-1, HCV, HBV, coronaviruses, and other viral pathogens and describes new information that suggests a role for immunophilins in regulating innate immune responses against chronic viral infection. MAJOR CONCLUSIONS: The dependence on cyclophilins by evolutionarily distinct viruses for accomplishing various steps in replication such as viral entry, initiation of genomic nucleic acid replication, viral genome uncoating, nuclear import and nuclear entry, emphasizes the potential of cyclophilin inhibitors as therapeutic agents. This article is part of a Special Issue entitled Proline-directed Foldases: Cell Signaling Catalysts and Drug Targets.


Subject(s)
Cyclophilin A/metabolism , RNA Virus Infections/metabolism , RNA Viruses/physiology , RNA, Viral/biosynthesis , Virus Internalization , Virus Replication/physiology , Animals , Cyclophilin A/genetics , Humans , RNA Virus Infections/genetics , RNA Viruses/pathogenicity
3.
J Hepatol ; 58(1): 16-23, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22902549

ABSTRACT

BACKGROUND & AIMS: Cyclophilin A (CypA) is vital for HCV replication. Cyp inhibitors successfully decrease viral loads in HCV-infected patients. However, their mechanisms of action remain unknown. Since interferon (IFN) can also suppress HCV replication, we asked whether a link between CypA and the IFN response exists. METHODS: We used cellular and recombinant pulldown approaches to investigate the possibility of a specific association of CypA with host ligands. RESULTS: We found for the first time that CypA binds to a major component of the IFN response - the IFN regulatory factor 9 (IRF9). IRF9 is the DNA-binding component of the transcriptional IFN-stimulated gene factor 3 (ISGF3). CypA binds directly to IRF9 via its peptidyl-prolyl isomerase (PPIase) pocket. Cyp inhibitors such as cyclosporine A (CsA) or non-immunosuppressive derivates such as alisporivir and SCY-635, prevent IRF9-CypA complex formation. CypA binds to the C-terminal IRF-association-domain (IAD), but not to the DNA-binding or linker domains of IRF9. Remarkably, CypA associates with the multimeric ISGF3 complex. We also obtained evidence that CypA neutralization enhances IFN-induced transcription. Interestingly, the hepatitis C virus (HCV) non-structural 5A (NS5A) protein, which is known to modulate the IFN response, competes with IRF9 for CypA binding and can prevent the formation of IRF9-CypA complexes. CONCLUSIONS: This study demonstrates for the first time that CypA binds specifically to a component of the Janus kinase/signal transducer and activator of transcription (JAK/STAT) pathway, IRF9. This study also reveals a novel opportunity of HCV to modulate the IFN response via NS5A.


Subject(s)
Cyclophilin A/metabolism , Hepacivirus/growth & development , Hepatitis C, Chronic/virology , Interferon-Stimulated Gene Factor 3, gamma Subunit/metabolism , Viral Nonstructural Proteins/metabolism , Binding Sites/physiology , Binding, Competitive/physiology , Hep G2 Cells , Hepacivirus/genetics , Hepacivirus/metabolism , Hepatitis C, Chronic/genetics , Hepatitis C, Chronic/metabolism , Humans , Interferon-Stimulated Gene Factor 3, gamma Subunit/genetics , Interferons/metabolism , Janus Kinases/metabolism , Ligands , STAT1 Transcription Factor/metabolism , STAT2 Transcription Factor/metabolism , Signal Transduction/physiology , Transcriptional Activation/physiology , Viral Load/physiology , Viral Nonstructural Proteins/genetics , Virus Replication/physiology
4.
Open Virol J ; 6: 97-102, 2012.
Article in English | MEDLINE | ID: mdl-23230455

ABSTRACT

In this study, we established a flow cytometry live cell-based assay that permits the screening of hepatitis C virus (HCV) inhibitors. Specifically, we created a stable cell line, which harbors a subgenomic replicon encoding an NS5A-YFP fusion protein. This system allows direct measurement of YFP fluorescence in live hepatoma cells in which the HCV replicon replicates. We demonstrated that this stable fluorescent system permits the rapid and sensitive quantification of HCV replication inhibition by direct-acting antiviral agents (DAA) including protease and NS5A inhibitors and host-targeting antiviral agents (HTA) including cyclophilin inhibitors. This flow cytometry-based live cell assay is well suited for multiple applications such as the evaluation of HCV replication as well as antiviral drug screening.

5.
Viruses ; 4(11): 2558-77, 2012 Oct 29.
Article in English | MEDLINE | ID: mdl-23202494

ABSTRACT

The advent of the replicon system together with advances in cell culture have contributed significantly to our understanding of the function of virally-encoded structural and nonstructural proteins in the replication cycle of the hepatitis C virus. In addition, in vitro systems have been used to identify several host proteins whose expression is critical for supporting such diverse activities as viral entry, RNA replication, particle assembly, and the release of infectious virions. Among all known host proteins that participate in the HCV replication cycle, cyclophilins are unique because they constitute the only host target that has formed the basis of pharmaceutical drug discovery and drug development programs. The introduction of the nonimmunosuppressive cyclophilin inhibitors into clinical testing has confirmed the clinical utility of CsA-based inhibitors for the treatment of individuals with chronic hepatitis C infection and has yielded new insights into their mechanism(s) of action. This review describes the biochemical evidence for the potential roles played by cyclophilins in supporting HCV RNA replication and summarizes clinical trial results obtained with the first generation of nonimmunosuppressive cyclophilin inhibitors.


Subject(s)
Antiviral Agents/therapeutic use , Cyclophilins/antagonists & inhibitors , Hepatitis C, Chronic/drug therapy , Antiviral Agents/administration & dosage , Clinical Trials, Phase I as Topic , Clinical Trials, Phase II as Topic , Cyclophilins/metabolism , Hepacivirus/physiology , Humans , Treatment Outcome
6.
Antimicrob Agents Chemother ; 56(10): 5113-21, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22802259

ABSTRACT

Alisporivir is the most advanced host-targeting antiviral cyclophilin (Cyp) inhibitor in phase III studies and has demonstrated a great deal of promise in decreasing hepatitis C virus (HCV) viremia in infected patients. In an attempt to further elucidate the mechanism of action of alisporivir, HCV replicons resistant to the drug were selected. Interestingly, mutations constantly arose in domain II of NS5A. To demonstrate that these mutations are responsible for drug resistance, they were reintroduced into the parental HCV genome, and the resulting mutant viruses were tested for replication in the presence of alisporivir or in the absence of the alisporivir target, CypA. We also examined the effect of the mutations on NS5A binding to itself (oligomerization), CypA, RNA, and NS5B. Importantly, the mutations did not affect any of these interactions. Moreover, the mutations did not preserve NS5A-CypA interactions from alisporivir rupture. NS5A mutations alone render HCV only slightly resistant to alisporivir. In sharp contrast, when multiple NS5A mutations are combined, significant resistance was observed. The introduction of multiple mutations in NS5A significantly restored viral replication in CypA knockdown cells. Interestingly, the combination of NS5A mutations renders HCV resistant to all classes of Cyp inhibitors. This study suggests that a combination of multiple mutations in domain II of NS5A rather than a single mutation is required to render HCV significantly and universally resistant to Cyp inhibitors. This in accordance with in vivo data that suggest that alisporivir is associated with a low potential for development of viral resistance.


Subject(s)
Antiviral Agents/pharmacology , Cyclosporine/pharmacology , Hepacivirus/drug effects , Hepacivirus/genetics , Viral Nonstructural Proteins/genetics , Amino Acid Sequence , Cell Line, Tumor , Drug Resistance, Viral/genetics , Genotype , Humans , Molecular Sequence Data , Mutation , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Amino Acid
7.
Dermatol Online J ; 18(5): 5, 2012 May 15.
Article in English | MEDLINE | ID: mdl-22630575

ABSTRACT

Anaplastic large cell lymphoma (ALCL) is classified as a CD30 positive non-Hodgkin lymphoma. Systemic ALCL (S-ALCL) is further subdivided into two subgroups based on anaplastic lymphoma kinase (ALK) expression. In systemic ALCL, positive ALK expression correlates with a favorable prognosis, whereas negative ALK expression correlates with poorer overall survival. By definition, primary cutaneous ALCL (cut-ALCL) is limited to the skin and is uniformly ALK-negative. Cut-ALCL closely resembles LyP with regards to its benign clinical course and CD30 positivity. We describe a unique case of ALK-negative (ALK-) S-ALCL presenting with cutaneous disseminated dome-shaped papules.


Subject(s)
Lymphoma, Large-Cell, Anaplastic/enzymology , Lymphoma, Large-Cell, Anaplastic/pathology , Receptor Protein-Tyrosine Kinases/analysis , Skin/pathology , Anaplastic Lymphoma Kinase , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Bone Marrow/pathology , Cyclophosphamide/administration & dosage , Dexamethasone/administration & dosage , Diagnosis, Differential , Fatal Outcome , Humans , Lymphocytes/pathology , Lymphoma, Large-Cell, Anaplastic/diagnosis , Lymphoma, Large-Cell, Anaplastic/drug therapy , Lymphoma, Primary Cutaneous Anaplastic Large Cell/diagnosis , Lymphoma, Primary Cutaneous Anaplastic Large Cell/enzymology , Lymphoma, Primary Cutaneous Anaplastic Large Cell/pathology , Male , Middle Aged , Salvage Therapy , Skin Neoplasms/enzymology , Skin Neoplasms/pathology
8.
Antimicrob Agents Chemother ; 56(7): 3888-97, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22585215

ABSTRACT

The nonimmunosuppressive cyclophilin (Cyp) inhibitor SCY-635 blocks hepatitis C virus (HCV) replication both in vitro and in vivo and represents a novel potent anti-HCV agent. However, its mechanism of action remains to be fully elucidated. A growing body of evidence suggests that cyclophilin A (CypA) is absolutely necessary for HCV replication and that the HCV nonstructural 5A (NS5A) protein serves as a main viral ligand for CypA. In this study, we examined the effect of SCY-635 on HCV replication. Specifically, we asked whether SCY-635 blocks HCV replication by targeting CypA-NS5A interactions. We also investigated the possibility that HCV can escape SCY-635 selection pressure and whether this resistance influences either CypA-NS5A interactions or the dependence of HCV on CypA. We found not only that SCY-635 efficiently inhibits HCV replication, but it is sufficient alone to clear HCV replicon-containing cells. We found that SCY-635 prevents CypA-NS5A interactions in a dose-dependent manner. SCY-635 prevents the contact between CypA and NS5A derived from genotypes 1 to 3. Together, these data suggest that NS5A-CypA interactions control HCV replication and that SCY-635 blocks viral replication by preventing the formation of these complexes. We also found that NS5A mutant proteins found in SCY-635-resistant HCV replicons behave similarly to wild-type NS5A in terms of both CypA binding and SCY-635-mediated dissociation and inhibition of CypA binding. However, the NS5A mutations found in SCY-635-resistant HCV replicons rescued viral replication in CypA-knockdown cells, suggesting that the NS5A mutations, which arose in vitro under SCY-635 selection, do not alter the binding affinity of CypA for NS5A. These specific mutations in NS5A eliminate the dependence of HCV RNA replication on the expression of host CypA.


Subject(s)
Antiviral Agents/pharmacology , Cyclophilin A/pharmacology , Cyclosporins/pharmacology , Hepacivirus/metabolism , Viral Nonstructural Proteins/metabolism , Enzyme-Linked Immunosorbent Assay , Hepacivirus/drug effects , Hepacivirus/genetics , Virus Replication/drug effects
9.
J Hepatol ; 57(1): 47-54, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22425702

ABSTRACT

BACKGROUND & AIMS: SCY-635 is a non-immunosuppressive analog of cyclosporin A that inhibits cyclophilins A and B and hepatitis C virus (HCV) replication in vitro. In a phase 1b multi-dose escalation study, we evaluated the safety, plasma pharmacokinetics, and antiviral activity of 15 days of monotherapy with SCY-635 in adults with chronic genotype 1 HCV infection. METHODS: Twenty adults with chronic HCV genotype 1 were randomized to SCY-635 oral doses of 100, 200, or 300 mg three times daily for 15 days. RESULTS: No dose-limiting clinical or laboratory toxicities were identified. On day 15, the mean decline in plasma viremia was 2.24±1.74 log(10) IU/ml with SCY-635 900 mg/d. Individual antiviral responses correlated with host IL28B genotype. Post hoc analyses indicated treatment with SCY-635 increased plasma protein concentrations of interferon α (IFNα), IFNs λ(1) and λ(3), and 2'5' oligoadenylate synthetase 1 (2'5'OAS-1), with the greatest increases in IL28B CC and CT subjects. Changes in plasma concentrations for all markers were coincident with changes in the plasma concentration of SCY-635. Peaks of IFNs α, λ(1), and λ(3) and 2'5'OAS-1 were observed within 2 h after drug administration. In replicon cells, SCY-635 enhanced secretion of type I and type III IFNs and increased the expression of IFN-stimulated genes (ISG). CONCLUSIONS: These studies establish clinical proof of concept for SCY-635 as a novel antiviral agent and suggest that restoration of the host innate immune response to chronic hepatitis C infection may represent a major mechanism through which cyclophilin inhibitors exert clinical antiviral activity.


Subject(s)
Antiviral Agents/administration & dosage , Cyclophilin A/antagonists & inhibitors , Cyclophilins/antagonists & inhibitors , Cyclosporins/administration & dosage , Hepacivirus/drug effects , Hepatitis C, Chronic/drug therapy , Adult , Aged , Antiviral Agents/adverse effects , Antiviral Agents/pharmacokinetics , Carcinoma, Hepatocellular , Cell Line, Tumor , Cyclosporins/adverse effects , Cyclosporins/pharmacokinetics , Dose-Response Relationship, Drug , Female , Genotype , Hepacivirus/genetics , Hepacivirus/growth & development , Hepatitis C, Chronic/immunology , Humans , Interferon-alpha/blood , Interferon-beta/blood , Interferon-gamma/blood , Interferons , Interleukins/genetics , Liver Neoplasms , Male , Middle Aged , Treatment Outcome
10.
Curr Opin Investig Drugs ; 11(8): 911-8, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20721833

ABSTRACT

Cyclophilins (Cyps) constitute one of the three families of peptidyl prolyl isomerase enzymes. CypA is the prototypical member of the Cyp family and is the predominant Cyp expressed in human cells. Recent studies indicate that CypA has an essential role in supporting HCV-specific RNA replication and protein expression. CypA interacts with several virally expressed proteins, including the non-structural (NS) proteins NS2, NS5A and NS5B, and may regulate diverse activities ranging from polypeptide processing to viral assembly. The introduction of non-immunosuppressive Cyp inhibitors into clinical trials confirms that Cyp inhibition is a valid strategy for developing novel therapeutics for the treatment of chronic HCV infection. This review describes the cyclophilin protein family and the potential roles played by cyclophilins in supporting HCV RNA replication and protein expression, as well as the initial clinical results obtained with a novel series of non-immunosuppressive cyclophilin inhibitors that established the clinical proof of concept for this emerging class of therapeutic agents.


Subject(s)
Cyclophilins/antagonists & inhibitors , Cyclophilins/metabolism , Cyclosporine/therapeutic use , Hepacivirus/physiology , Hepatitis C/drug therapy , Hepatitis C/virology , Cell Line , Clinical Trials as Topic , Cyclophilin A/antagonists & inhibitors , Cyclophilin A/genetics , Cyclophilin A/metabolism , Cyclosporine/chemistry , Cyclosporine/metabolism , Cyclosporine/pharmacology , Cyclosporins/metabolism , Cyclosporins/pharmacology , Cyclosporins/therapeutic use , Hepacivirus/drug effects , Hepacivirus/genetics , Hepacivirus/metabolism , Humans , Peptidylprolyl Isomerase/antagonists & inhibitors , Peptidylprolyl Isomerase/metabolism , RNA, Viral/biosynthesis , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/metabolism , Viral Proteins/antagonists & inhibitors , Viral Proteins/metabolism , Virus Replication/drug effects
11.
Antimicrob Agents Chemother ; 54(2): 660-72, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19933795

ABSTRACT

SCY-635 is a novel nonimmunosuppressive cyclosporine-based analog that exhibits potent suppression of hepatitis C virus (HCV) replication in vitro. SCY-635 inhibited the peptidyl prolyl isomerase activity of cyclophilin A at nanomolar concentrations but showed no detectable inhibition of calcineurin phosphatase activity at concentrations up to 2 microM. Metabolic studies indicated that SCY-635 did not induce the major cytochrome P450 enzymes 1A2, 2B6, and 3A4. SCY-635 was a weak inhibitor and a poor substrate for P-glycoprotein. Functional assays with stimulated Jurkat cells and stimulated human peripheral blood mononuclear cells indicated that SCY-635 is a weaker inhibitor of interleukin-2 secretion than cyclosporine. A series of two-drug combination studies was performed in vitro. SCY-635 exhibited synergistic antiviral activity with alpha interferon 2b and additive antiviral activity with ribavirin. SCY-635 was shown to be orally bioavailable in multiple animal species and produced blood and liver concentrations of parent drug that exceeded the 50% effective dose determined in the bicistronic con1b-derived replicon assay. These results suggest that SCY-635 warrants further investigation as a novel therapeutic agent for the treatment of individuals who are chronically infected with HCV.


Subject(s)
Antiviral Agents/pharmacology , Cyclosporins/chemistry , Hepacivirus/drug effects , Hepacivirus/genetics , Immunosuppressive Agents/chemistry , RNA, Viral/genetics , Animals , Antiviral Agents/chemistry , Antiviral Agents/pharmacokinetics , Antiviral Agents/therapeutic use , Cell Line , Cells, Cultured , Cyclophilin A/metabolism , Cyclosporins/pharmacokinetics , Cyclosporins/pharmacology , Cyclosporins/therapeutic use , Dogs , Dose-Response Relationship, Drug , Enzyme Activation/drug effects , Haplorhini , Hepatitis C/drug therapy , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Interleukin-2/metabolism , Jurkat Cells/drug effects , Jurkat Cells/metabolism , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/metabolism , Mice , Molecular Structure , Rats , Virus Replication/drug effects
12.
Article in English | MEDLINE | ID: mdl-15583538

ABSTRACT

The present article reviews the association between microbial colonization of the oral cavity and the lungs in critically ill patients that develop ventilator-associated pneumonia (VAP) in the intensive care unit (ICU) setting. The risk factors and microorganisms associated with VAP are presented. The role of oral colonization of VAP-associated pathogens (VAP-AP) in the development of VAP is examined. We explore the potential factors involved in oral colonization of VAP-AP, which are atypical bacteria for the oral cavity. Strategies for the prevention or moderation of oral colonization of VAP-AP have had limited success. We need a deeper understanding of the pathophysiology of VAP in order to reduce the morbidity, mortality, and cost from this common complication in ICU medicine and surgery.


Subject(s)
Bacterial Physiological Phenomena , Mouth/microbiology , Pneumonia, Bacterial/microbiology , Respiration, Artificial/adverse effects , Critical Care , Critical Illness , Gram-Negative Bacterial Infections/microbiology , Gram-Negative Bacterial Infections/prevention & control , Humans , Lung/microbiology , Pneumonia, Bacterial/prevention & control , Risk Factors
13.
J Pharmacol Exp Ther ; 311(3): 929-38, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15273255

ABSTRACT

Epithelial sodium channel (ENaC) blockers have been proposed as a therapy to restore mucus clearance (MC) in cystic fibrosis (CF) airways. The therapeutic effects of the first generation ENaC blocker, amiloride, in CF patients, however, were minimal. Because the failure of amiloride reflected both its low potency and short duration of action on airway surfaces, we investigated whether the increased potency of benzamil and phenamil would produce more favorable pharmacodynamic properties. In vitro potency, maximal efficacy, rate of recovery from maximal block of ENaC, and rate of drug absorption were compared for amiloride, benzamil, and phenamil in cultured human and ovine bronchial epithelial cells. In both human and ovine bronchial epithelia, the rank order of potency was benzamil > phenamil >> amiloride, the maximal efficacy was benzamil = phenamil = amiloride, the recovery to baseline sodium transport was phenamil < benzamil << amiloride, and the rate of drug absorption was phenamil > benzamil >> amiloride. Based on greater potency, benzamil was compared with amiloride in in vivo pharmacodynamic studies in sheep, including tracheal mucus velocity (TMV) and MC. Benzamil enhanced MC and TMV, but acute potency or duration of effect did not exceed that of amiloride. In conclusion, our data support the hypothesis that ENaC blocker aerosol therapy increases MC. However, rapid absorption of benzamil from the mucosal surface offset its greater potency, making it equieffective with amiloride in vivo. More potent, less absorbable, third generation ENaC blockers will be required for an effective aerosol CF pharmacotherapy.


Subject(s)
Amiloride/analogs & derivatives , Amiloride/therapeutic use , Cystic Fibrosis/drug therapy , Lung Diseases/drug therapy , Sodium Channel Blockers/therapeutic use , Absorption , Amiloride/pharmacokinetics , Animals , Bronchi/drug effects , Bronchi/metabolism , Cystic Fibrosis/complications , Electrophysiology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelial Sodium Channels , Epithelium/drug effects , Epithelium/metabolism , Female , Humans , In Vitro Techniques , Lung Diseases/etiology , Mannitol/metabolism , Microscopy, Confocal , Mucus/physiology , Muscle Contraction/drug effects , Sheep , Sodium/metabolism , Sodium Channels/metabolism , Viscosity
SELECTION OF CITATIONS
SEARCH DETAIL
...