Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 13 de 13
1.
Gastroenterology ; 163(1): 270-284, 2022 07.
Article En | MEDLINE | ID: mdl-35301011

BACKGROUND & AIMS: Insulin signaling is known to regulate essential proteostasis mechanisms. METHODS: The analyses here examined effects of insulin signaling in the PiZ mouse model of α1-antitrypsin deficiency in which hepatocellular accumulation and proteotoxicity of the misfolded α1-antitrypsin Z variant (ATZ) causes liver fibrosis and cancer. RESULTS: We first studied the effects of breeding PiZ mice to liver-insulin-receptor knockout (LIRKO) mice (with hepatocyte-specific insulin-receptor gene disruption). The results showed decreased hepatic ATZ accumulation and liver fibrosis in PiZ x LIRKO vs PiZ mice, with reversal of those effects when we bred PiZ x LIRKO mice onto a FOXO1-deficient background. Increased intracellular degradation of ATZ mediated by autophagy was identified as the likely mechanism for diminished hepatic proteotoxicity in PiZ x LIRKO mice and the converse was responsible for enhanced toxicity in PiZ x LIRKO x FOXO1-KO animals. Transcriptomic studies showed major effects on oxidative phosphorylation and autophagy genes, and significant induction of peroxisome proliferator-activated-receptor-γ-coactivator-1α (PGC1α) expression in PiZ-LIRKO mice. Because PGC1α plays a key role in oxidative phosphorylation, we further investigated its effects on ATZ proteostasis in our ATZ-expressing mammalian cell model. The results showed PGC1α overexpression or activation enhances autophagic ATZ degradation. CONCLUSIONS: These data implicate suppression of autophagic ATZ degradation by down-regulation of PGC1α as one mechanism by which insulin signaling exacerbates hepatic proteotoxicity in PiZ mice, and identify PGC1α as a novel target for development of new human α1-antitrypsin deficiency liver disease therapies.


Insulin , Liver , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , alpha 1-Antitrypsin Deficiency , Animals , Insulin/metabolism , Liver/metabolism , Liver/pathology , Liver Cirrhosis/genetics , Liver Cirrhosis/metabolism , Liver Cirrhosis/pathology , Mammals/metabolism , Mice , Mice, Transgenic , Mutation , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Signal Transduction , alpha 1-Antitrypsin Deficiency/genetics , alpha 1-Antitrypsin Deficiency/pathology
2.
PLoS One ; 14(1): e0209748, 2019.
Article En | MEDLINE | ID: mdl-30673724

The classical form of α1-antitrypsin deficiency (ATD) is characterized by intracellular accumulation of the misfolded variant α1-antitrypsin Z (ATZ) and severe liver disease in some of the affected individuals. In this study, we investigated the possibility of discovering novel therapeutic agents that would reduce ATZ accumulation by interrogating a C. elegans model of ATD with high-content genome-wide RNAi screening and computational systems pharmacology strategies. The RNAi screening was utilized to identify genes that modify the intracellular accumulation of ATZ and a novel computational pipeline was developed to make high confidence predictions on repurposable drugs. This approach identified glibenclamide (GLB), a sulfonylurea drug that has been used broadly in clinical medicine as an oral hypoglycemic agent. Here we show that GLB promotes autophagic degradation of misfolded ATZ in mammalian cell line models of ATD. Furthermore, an analog of GLB reduces hepatic ATZ accumulation and hepatic fibrosis in a mouse model in vivo without affecting blood glucose or insulin levels. These results provide support for a drug discovery strategy using simple organisms as human disease models combined with genetic and computational screening methods. They also show that GLB and/or at least one of its analogs can be immediately tested to arrest the progression of human ATD liver disease.


Glyburide/pharmacology , alpha 1-Antitrypsin/drug effects , Animals , Autophagy/drug effects , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/metabolism , Cell Line , Disease Models, Animal , Drug Discovery , Glyburide/analogs & derivatives , Glyburide/therapeutic use , Humans , Liver/metabolism , Liver Cirrhosis/metabolism , Mice , Mice, Transgenic , RNA Interference , alpha 1-Antitrypsin/genetics , alpha 1-Antitrypsin/metabolism , alpha 1-Antitrypsin Deficiency/drug therapy , alpha 1-Antitrypsin Deficiency/genetics
3.
J Biol Chem ; 290(50): 29742-57, 2015 Dec 11.
Article En | MEDLINE | ID: mdl-26494620

Recent studies have shown that autophagy mitigates the pathological effects of proteinopathies in the liver, heart, and skeletal muscle but this has not been investigated for proteinopathies that affect the lung. This may be due at least in part to the lack of an animal model robust enough for spontaneous pathological effects from proteinopathies even though several rare proteinopathies, surfactant protein A and C deficiencies, cause severe pulmonary fibrosis. In this report we show that the PiZ mouse, transgenic for the common misfolded variant α1-antitrypsin Z, is a model of respiratory epithelial cell proteinopathy with spontaneous pulmonary fibrosis. Intracellular accumulation of misfolded α1-antitrypsin Z in respiratory epithelial cells of the PiZ model resulted in activation of autophagy, leukocyte infiltration, and spontaneous pulmonary fibrosis severe enough to elicit functional restrictive deficits. Treatment with autophagy enhancer drugs or lung-directed gene transfer of TFEB, a master transcriptional activator of the autophagolysosomal system, reversed these proteotoxic consequences. We conclude that this mouse is an excellent model of respiratory epithelial proteinopathy with spontaneous pulmonary fibrosis and that autophagy is an important endogenous proteostasis mechanism and an attractive target for therapy.


Autophagy/drug effects , Genetic Therapy , alpha 1-Antitrypsin Deficiency/therapy , Animals , Autophagy/genetics , Disease Models, Animal , Epithelial Cells/pathology , Lung/pathology , Mice , alpha 1-Antitrypsin Deficiency/drug therapy , alpha 1-Antitrypsin Deficiency/pathology
4.
Hepatology ; 62(1): 147-57, 2015 Jul.
Article En | MEDLINE | ID: mdl-25690322

UNLABELLED: In the classical form of α1-antitrypsin deficiency (ATD), aberrant intracellular accumulation of misfolded mutant α1-antitrypsin Z (ATZ) in hepatocytes causes hepatic damage by a gain-of-function, "proteotoxic" mechanism. Whereas some ATD patients develop severe liver disease (SLD) that necessitates liver transplantation, others with the same genetic defect completely escape this clinical phenotype. We investigated whether induced pluripotent stem cells (iPSCs) from ATD individuals with or without SLD could model these personalized variations in hepatic disease phenotypes. Patient-specific iPSCs were generated from ATD patients and a control and differentiated into hepatocyte-like cells (iHeps) having many characteristics of hepatocytes. Pulse-chase and endoglycosidase H analysis demonstrate that the iHeps recapitulate the abnormal accumulation and processing of the ATZ molecule, compared to the wild-type AT molecule. Measurements of the fate of intracellular ATZ show a marked delay in the rate of ATZ degradation in iHeps from SLD patients, compared to those from no liver disease patients. Transmission electron microscopy showed dilated rough endoplasmic reticulum in iHeps from all individuals with ATD, not in controls, but globular inclusions that are partially covered with ribosomes were observed only in iHeps from individuals with SLD. CONCLUSION: iHeps model the individual disease phenotypes of ATD patients with more rapid degradation of misfolded ATZ and lack of globular inclusions in cells from patients who have escaped liver disease. The results support the concept that "proteostasis" mechanisms, such as intracellular degradation pathways, play a role in observed variations in clinical phenotype and show that iPSCs can potentially be used to facilitate predictions of disease susceptibility for more precise and timely application of therapeutic strategies.


Induced Pluripotent Stem Cells/metabolism , Liver Diseases/etiology , alpha 1-Antitrypsin Deficiency/complications , Cells, Cultured , Endoplasmic Reticulum, Rough/metabolism , Humans , Liver Diseases/metabolism , alpha 1-Antitrypsin/metabolism
5.
Hum Mol Genet ; 23(19): 5123-32, 2014 Oct 01.
Article En | MEDLINE | ID: mdl-24838285

α1-Antitrypsin deficiency (ATD) is a common genetic disorder that can lead to end-stage liver and lung disease. Although liver transplantation remains the only therapy currently available, manipulation of the proteostasis network (PN) by small molecule therapeutics offers great promise. To accelerate the drug-discovery process for this disease, we first developed a semi-automated high-throughput/content-genome-wide RNAi screen to identify PN modifiers affecting the accumulation of the α1-antitrypsin Z mutant (ATZ) in a Caenorhabditis elegans model of ATD. We identified 104 PN modifiers, and these genes were used in a computational strategy to identify human ortholog-ligand pairs. Based on rigorous selection criteria, we identified four FDA-approved drugs directed against four different PN targets that decreased the accumulation of ATZ in C. elegans. We also tested one of the compounds in a mammalian cell line with similar results. This methodology also proved useful in confirming drug targets in vivo, and predicting the success of combination therapy. We propose that small animal models of genetic disorders combined with genome-wide RNAi screening and computational methods can be used to rapidly, economically and strategically prime the preclinical discovery pipeline for rare and neglected diseases with limited therapeutic options.


Drug Discovery , Genome-Wide Association Study , RNA Interference , alpha 1-Antitrypsin Deficiency/genetics , alpha 1-Antitrypsin/genetics , Animals , Caenorhabditis elegans , Computational Biology , Disease Models, Animal , Genomics , High-Throughput Screening Assays , Humans , Mutation , Protein Binding , Proteostasis Deficiencies/genetics , Reproducibility of Results , alpha 1-Antitrypsin Deficiency/drug therapy
6.
PLoS One ; 9(1): e87260, 2014.
Article En | MEDLINE | ID: mdl-24498058

The classical form of α1-antitrypsin deficiency (ATD) is associated with hepatic fibrosis and hepatocellular carcinoma. It is caused by the proteotoxic effect of a mutant secretory protein that aberrantly accumulates in the endoplasmic reticulum of liver cells. Recently we developed a model of this deficiency in C. elegans and adapted it for high-content drug screening using an automated, image-based array scanning. Screening of the Library of Pharmacologically Active Compounds identified fluphenazine (Flu) among several other compounds as a drug which reduced intracellular accumulation of mutant α1-antitrypsin Z (ATZ). Because it is representative of the phenothiazine drug class that appears to have autophagy enhancer properties in addition to mood stabilizing activity, and can be relatively easily re-purposed, we further investigated its effects on mutant ATZ. The results indicate that Flu reverses the phenotypic effects of ATZ accumulation in the C. elegans model of ATD at doses which increase the number of autophagosomes in vivo. Furthermore, in nanomolar concentrations, Flu enhances the rate of intracellular degradation of ATZ and reduces the cellular ATZ load in mammalian cell line models. In the PiZ mouse model Flu reduces the accumulation of ATZ in the liver and mediates a decrease in hepatic fibrosis. These results show that Flu can reduce the proteotoxicity of ATZ accumulation in vivo and, because it has been used safely in humans, this drug can be moved rapidly into trials for liver disease due to ATD. The results also provide further validation for drug discovery using C. elegans models that can be adapted to high-content drug screening platforms and used together with mammalian cell line and animal models.


Caenorhabditis elegans/metabolism , Disease Models, Animal , Fluphenazine/pharmacology , alpha 1-Antitrypsin Deficiency/prevention & control , Animals , Animals, Genetically Modified , Antipsychotic Agents/pharmacology , Autophagy/drug effects , Autophagy/genetics , CHO Cells , Caenorhabditis elegans/genetics , Cricetinae , Cricetulus , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HeLa Cells , Hep G2 Cells , Humans , Immunoblotting , Liver/drug effects , Liver/metabolism , Liver/pathology , Mice , Mice, Transgenic , Microscopy, Fluorescence , Mutation , Phagosomes/drug effects , Phagosomes/metabolism , Survival Analysis , alpha 1-Antitrypsin/genetics , alpha 1-Antitrypsin/metabolism , alpha 1-Antitrypsin Deficiency/genetics , alpha 1-Antitrypsin Deficiency/metabolism
7.
J Lipid Res ; 54(2): 514-21, 2013 Feb.
Article En | MEDLINE | ID: mdl-23204298

Colipase is essential for efficient fat digestion. An arginine-to-cysteine polymorphism at position 92 of colipase (Arg92Cys) associates with an increased risk for developing type-2 diabetes through an undefined mechanism. To test our hypothesis that the extra cysteine increases colipase misfolding, thereby altering its intracellular trafficking and function, we expressed Cys92 colipase in HEK293T cells. Less Cys92 colipase is secreted and more is retained intracellularly in an insoluble form compared with Arg92 colipase. Nonreducing gel electrophoresis suggests the folding of secreted Cys92 colipase differs from Arg92 colipase. Cys92 colipase misfolding does not trigger the unfolded protein response (UPR) or endoplasmic reticulum (ER) stress. The ability of secreted Cys92 colipase to stimulate pancreatic triglyceride lipase (PTL) is reduced with all substrates tested, particularly long-chain triglycerides. The reaction of Cys92 colipase with triolein and Intralipid has a much longer lag time, reflecting decreased ability to anchor PTL on those substrates. Our data predicts that humans with the Arg92Cys substitution will secrete less functional colipase into the duodenum and have less efficient fat digestion. Whether inefficient fat digestion or another property of colipase contributes to the risk for developing diabetes remains to be clarified.


Arginine , Colipases/metabolism , Cysteine , Polymorphism, Single Nucleotide , Protein Folding , Colipases/chemistry , Colipases/genetics , Diabetes Mellitus, Type 2/enzymology , Diabetes Mellitus, Type 2/genetics , Dietary Fats/metabolism , Enzyme Stability , HEK293 Cells , Humans , Models, Molecular , Protein Structure, Secondary , Temperature
8.
J Biol Chem ; 287(42): 35341-35350, 2012 Oct 12.
Article En | MEDLINE | ID: mdl-22915586

Neutrophil elastase (NE) is a neutrophil-derived serine proteinase with broad substrate specificity. We have recently demonstrated that NE is capable of entering tumor cell endosomes and processing novel intracellular substrates. In the current study, we sought to determine the mechanism by which NE enters tumor cells. Our results show that NE enters into early endosomal antigen-1(+) endosomes in a dynamin- and clathrin-dependent but flotillin-1- and caveolin-1-independent fashion. Cathepsin G (but not proteinase-3) also enters tumor endosomes via the same mechanism. We utilized (125)I-labeled NE to demonstrate that NE binds to the surface of cancer cells. Incubation of radiolabeled NE with lung cancer cells displays a dissociation constant (K(d)) of 284 nm. Because NE is known to bind to heparan sulfate- and chondroitin sulfate-containing proteoglycans, we treated cells with glycanases to remove these confounding factors, which did not significantly diminish cell surface binding or endosomal entry. Thus, NE and CG bind to the surface of cancer cells, presumably to a cell surface receptor, and subsequently undergo clathrin pit-mediated endocytosis.


Cathepsin G/metabolism , Clathrin/metabolism , Coated Pits, Cell-Membrane/metabolism , Endocytosis , Leukocyte Elastase/metabolism , Lung Neoplasms/metabolism , Neoplasm Proteins/metabolism , Animals , CHO Cells , Cathepsin G/genetics , Caveolin 1/genetics , Caveolin 1/metabolism , Chondroitin Sulfates/genetics , Chondroitin Sulfates/metabolism , Clathrin/genetics , Coated Pits, Cell-Membrane/genetics , Cricetinae , Cricetulus , Humans , Leukocyte Elastase/genetics , Lung Neoplasms/genetics , Neoplasm Proteins/genetics , Protein Binding/genetics , Protein Transport/genetics
9.
Science ; 329(5988): 229-32, 2010 Jul 09.
Article En | MEDLINE | ID: mdl-20522742

In the classical form of alpha1-antitrypsin (AT) deficiency, a point mutation in AT alters the folding of a liver-derived secretory glycoprotein and renders it aggregation-prone. In addition to decreased serum concentrations of AT, the disorder is characterized by accumulation of the mutant alpha1-antitrypsin Z (ATZ) variant inside cells, causing hepatic fibrosis and/or carcinogenesis by a gain-of-toxic function mechanism. The proteasomal and autophagic pathways are known to mediate degradation of ATZ. Here we show that the autophagy-enhancing drug carbamazepine (CBZ) decreased the hepatic load of ATZ and hepatic fibrosis in a mouse model of AT deficiency-associated liver disease. These results provide a basis for testing CBZ, which has an extensive clinical safety profile, in patients with AT deficiency and also provide a proof of principle for therapeutic use of autophagy enhancers.


Autophagy/drug effects , Carbamazepine/pharmacology , Liver Cirrhosis/drug therapy , Liver/metabolism , alpha 1-Antitrypsin Deficiency/metabolism , alpha 1-Antitrypsin/metabolism , Animals , Carbamazepine/administration & dosage , Carbamazepine/therapeutic use , Cell Line , Disease Models, Animal , Endoplasmic Reticulum/metabolism , HeLa Cells , Humans , Liver/drug effects , Liver/pathology , Liver Cirrhosis/etiology , Liver Cirrhosis/metabolism , Liver Cirrhosis/pathology , Mice , Mice, Transgenic , Mutant Proteins/chemistry , Mutant Proteins/metabolism , Phagosomes/drug effects , Phagosomes/ultrastructure , Phenotype , Proteasome Endopeptidase Complex/metabolism , Protein Folding , Solubility , alpha 1-Antitrypsin/chemistry , alpha 1-Antitrypsin/genetics , alpha 1-Antitrypsin Deficiency/complications , alpha 1-Antitrypsin Deficiency/pathology
10.
Ann Clin Biochem ; 46(Pt 2): 172-5, 2009 Mar.
Article En | MEDLINE | ID: mdl-19225029

Interference in immunoassays is a widely recognized problem, which could potentially lead to unnecessary investigations and treatment. We describe a case where interference in a cortisol immunoassay led to a falsely low serum cortisol concentration and interference in the free thyroxine assay led to falsely elevated serum thyroxine concentrations, in the same patient. A 42-year-old woman with documented hypothyroidism underwent a synacthen test for suspected adrenal insufficiency. Previous thyroid function tests had been discordant and difficult to interpret, with elevated thyroxine and non-suppressed thyroid-stimulating hormone. The synacthen test showed a subnormal cortisol response and she was commenced on cortisol replacement. Clinically, it was doubted whether she had true adrenal insufficiency and it was thought that the cortisol results might be artefactually low due to assay interference. Cortisol was measured by an alternative immunoassay, before and after incubation in an antibody blocking tube ('Scantibodies'), after heat treatment and also after treatment with Protein A. The results supported assay interference and cortisol 'replacement' was stopped. Thyroxine had been discontinued although thyroid function tests (TFTs) were significantly different between analytical platforms, also consistent with interference. Thyroxine replacement was restarted and once on a stable dose, the discrepancy in TFTs was also investigated by similar procedures as for cortisol. Good clinician-laboratory interface and laboratory work-up of patients with results that are discordant from the clinical findings can reduce unnecessary investigation and inappropriate treatment.


Addison Disease/diagnosis , Hydrocortisone/blood , Hypothyroidism/diagnosis , Thyroid Function Tests/standards , Thyroxine/blood , Adrenocorticotropic Hormone , Adult , Antibodies, Heterophile/immunology , Female , Humans , Immunoassay/standards , Staphylococcal Protein A , Thyroxine/therapeutic use
11.
J Biol Chem ; 282(38): 27769-80, 2007 Sep 21.
Article En | MEDLINE | ID: mdl-17635928

In the classical form of alpha(1)-antitrypsin deficiency, a mutant protein accumulates in a polymerized form in the endoplasmic reticulum (ER) of liver cells causing liver damage and carcinogenesis by a gain-of-toxic function mechanism. Recent studies have indicated that the accumulation of mutant alpha(1)-antitrypsin Z in the ER specifically activates the autophagic response but not the unfolded protein response and that autophagy plays a critical role in disposal of insoluble alpha(1)-antitrypsin Z. In this study, we used genomic analysis of the liver in a novel transgenic mouse model with inducible expression to screen for changes in gene expression that would potentially define how the liver responds to accumulation of this mutant protein. There was no unfolded protein response. Of several distinct gene expression profiles, marked up-regulation of regulator of G signaling (RGS16) was particularly notable. RGS16 did not increase when model systems were exposed to classical inducers of ER stress, including tunicamycin and calcium ionophore, or when a nonpolymerogenic alpha(1)-antitrypsin mutant accumulated in the ER. RGS16 was up-regulated in livers from patients with alpha(1)-antitrypsin deficiency, and the degree of up-regulation correlated with the hepatic levels of insoluble alpha(1)-antitrypsin Z protein. Taken together, these results indicate that expression of RGS16 is an excellent marker for the distinct form of "ER stress" that occurs in alpha(1)-antitrypsin deficiency, presumably determined by the aggregation-prone properties of the mutant protein that characterizes the deficiency.


Endoplasmic Reticulum/metabolism , RGS Proteins/metabolism , alpha 1-Antitrypsin/chemistry , Animals , HeLa Cells , Hepatocytes/metabolism , Humans , Liver/metabolism , Mice , Mice, Transgenic , Models, Biological , Mutation , Protein Conformation , Protein Denaturation , Signal Transduction
12.
J Biol Chem ; 280(47): 39002-15, 2005 Nov 25.
Article En | MEDLINE | ID: mdl-16183649

In alpha(1)-antitrypsin (alpha1AT) deficiency, a polymerogenic mutant form of the secretory glycoprotein alpha1AT, alpha1ATZ, is retained in the endoplasmic reticulum (ER) of liver cells. It is not yet known how this results in liver injury in a subgroup of deficient individuals and how the remainder of deficient individuals escapes liver disease. One possible explanation is that the "susceptible" subgroup is unable to mount the appropriate protective cellular responses. Here we examined the effect of mutant alpha1ATZ on several potential protective signaling pathways by using cell lines with inducible expression of mutant alpha1AT as well as liver from transgenic mice with liver-specific inducible expression of mutant alpha1AT. The results show that ER retention of polymerogenic mutant alpha1ATZ does not result in an unfolded protein response (UPR). The UPR can be induced in the presence of alpha1ATZ by tunicamycin excluding the possibility that the pathway has been disabled. In striking contrast, ER retention of nonpolymerogenic alpha1AT mutants does induce the UPR. These results indicate that the machinery responsible for activation of the UPR can distinguish the physical characteristics of proteins that accumulate in the ER in such a way that it can respond to misfolded but not relatively ordered polymeric structures. Accumulation of mutant alpha1ATZ does activate specific signaling pathways, including caspase-12 in mouse, caspase-4 in human, NFkappaB, and BAP31, a profile that was distinct from that activated by nonpolymerogenic alpha1AT mutants.


Endoplasmic Reticulum/metabolism , alpha 1-Antitrypsin/genetics , alpha 1-Antitrypsin/metabolism , Animals , Caspase 12 , Caspases/metabolism , Caspases, Initiator , Cell Line , Enzyme Activation , HeLa Cells , Humans , Liver/metabolism , Membrane Proteins/metabolism , Mice , Mice, Transgenic , Mutation , NF-kappa B/metabolism , Protein Folding , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Signal Transduction , Tunicamycin/pharmacology , alpha 1-Antitrypsin Deficiency/genetics , alpha 1-Antitrypsin Deficiency/metabolism
13.
J Gastrointest Surg ; 6(1): 108-15, 2002.
Article En | MEDLINE | ID: mdl-11986026

Our aim was to examine the effects of hepatitis C virus (HCV) infection, a leading cause of end-stage liver disease, and its recurrence after liver transplantation on functional performance and health-related quality of life. Functional performance, liver function, and HCV recurrence were assessed longitudinally in 75 adult transplant recipients (28 with HCV). Quality of life was reported once after transplantation. Functional performance improved through year 2 (P < 0.001) and then declined in those with HCV, whereas the others remained stable (P = 0.05). Time had a positive effect (beta = 0.22, P = 0.05) and HCV infection had a negative effect (beta = -0.28, P = 0.01) on post-transplant functional performance. Educational level (beta = 0.24, P < 0.05) and recent functional performance (beta = 0.31, P = 0.01) had positive effects on quality of life. HCV recurrence was associated with relatively poorer pretransplant functional performance, a greater rate of improvement through month 3 (P < 0.05), and abnormal transaminase values between years 1 and 2 (P < 0.001). Rehospitalization for recurrent HCV was associated with reduced functional performance (P < 0.05). Functional performance improves with time following liver transplantation, but HCV infection exerts an opposing and comparably strong effect. Post-transplant functional performance, in turn, directly affects post-transplant quality of life. Severe, recurrent HCV illness is associated with reduced functional performance.


Hepatitis C, Chronic/complications , Liver Cirrhosis/surgery , Liver Transplantation , Liver Transplantation/adverse effects , Quality of Life , Cohort Studies , Female , Graft Rejection , Graft Survival , Humans , Liver Cirrhosis/etiology , Liver Function Tests , Liver Transplantation/methods , Male , Middle Aged , Probability , Prognosis , Recurrence , Risk Assessment , Sampling Studies , Severity of Illness Index , Treatment Outcome
...