Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 47
1.
Int J Mol Sci ; 22(4)2021 Feb 11.
Article En | MEDLINE | ID: mdl-33670179

Fumarylacetoacetate hydrolase (FAH) is the fifth enzyme in the tyrosine catabolism pathway. A deficiency in human FAH leads to hereditary tyrosinemia type I (HT1), an autosomal recessive disorder that results in the accumulation of toxic metabolites such as succinylacetone, maleylacetoacetate, and fumarylacetoacetate in the liver and kidney, among other tissues. The disease is severe and, when untreated, it can lead to death. A low tyrosine diet combined with the herbicidal nitisinone constitutes the only available therapy, but this treatment is not devoid of secondary effects and long-term complications. In this study, we targeted FAH for the first-time to discover new chemical modulators that act as pharmacological chaperones, directly associating with this enzyme. After screening several thousand compounds and subsequent chemical redesign, we found a set of reversible inhibitors that associate with FAH close to the active site and stabilize the (active) dimeric species, as demonstrated by NMR spectroscopy. Importantly, the inhibitors are also able to partially restore the normal phenotype in a newly developed cellular model of HT1.


Drug Delivery Systems , Enzyme Inhibitors/pharmacology , Hydrolases/antagonists & inhibitors , Hydrolases/metabolism , Tyrosinemias/drug therapy , Tyrosinemias/enzymology , Animals , Catalytic Domain , Enzyme Inhibitors/chemistry , HEK293 Cells , Humans , Hydrolases/genetics , Mice , Tyrosinemias/genetics
2.
Int J Mol Sci ; 21(6)2020 Mar 20.
Article En | MEDLINE | ID: mdl-32244944

The elucidation of aberrant splicing mechanisms, frequently associated with disease has led to the development of RNA therapeutics based on the U1snRNA, which is involved in 5' splice site (5'ss) recognition. Studies in cellular models have demonstrated that engineered U1snRNAs can rescue different splicing mutation types. However, the assessment of their correction potential in vivo is limited by the scarcity of animal models with the targetable splicing defects. Here, we challenged the U1snRNA in the FAH5961SB mouse model of hepatic fumarylacetoacetate hydrolase (FAH) deficiency (Hereditary Tyrosinemia type I, HT1) due to the FAH c.706G>A splicing mutation. Through minigene expression studies we selected a compensatory U1snRNA (U1F) that was able to rescue this mutation. Intriguingly, adeno-associated virus-mediated delivery of U1F (AAV8-U1F), but not of U1wt, partially rescued FAH splicing in mouse hepatocytes. Consistently, FAH protein was detectable only in the liver of AAV8-U1F treated mice, which displayed a slightly prolonged survival. Moreover, RNA sequencing revealed the negligible impact of the U1F on the splicing profile and overall gene expression, thus pointing toward gene specificity. These data provide early in vivo proof-of-principle of the correction potential of compensatory U1snRNAs in HTI and encourage further optimization on a therapeutic perspective, and translation to other splicing-defective forms of metabolic diseases.


Hydrolases/genetics , RNA Splicing/genetics , RNA, Small Nuclear/genetics , Tyrosinemias/enzymology , Tyrosinemias/genetics , Animals , Cell Line, Tumor , Disease Models, Animal , Mice, Inbred C57BL
3.
Mol Genet Genomic Med ; 7(12): e937, 2019 12.
Article En | MEDLINE | ID: mdl-31568711

BACKGROUND: Tyrosinemia type 1 (HT1, MIM#276700) is caused by a deficiency in fumarylacetoacetate hydrolase (FAH) and it is associated with severe liver and renal disfunction. At present, the mutational FAH (15q25.1, MIM*613871) spectrum underlying HT1 in the Mexican population is unknown. The objective of this study was to determine the FAH genotypes in eight nonrelated Mexican patients with HT1, who were diagnosed clinically. METHODS: Sequencing of FAH and their exon-intron boundaries and in silico protein modeling based on the crystallographic structure of mouse FAH. RESULTS: We identified pathogenic variants in 15/16 studied alleles (93.8%). Nine different variants were found. The most commonly detected HT1-causing allele was NM_000137.2(FAH):c.3G > A or p.(?) [rs766882348] (25%, n = 4/16). We also identified a novel missense variant NM_000137.2(FAH):c.36C > A or p.(Phe12Leu) in a homozygous patient with an early and fatal acute form. The latter was classified as a likely pathogenic variant and in silico protein modeling showed that Phe-12 residue substitution for Leu, produces a repulsion in all possible Leu rotamers, which in turn would lead to a destabilization of the protein structure and possible loss-of-function. CONCLUSION: HT1 patients had a heterogeneous mutational and clinical spectrum and no genotype-phenotype correlation could be established.


Hydrolases/genetics , Mutation, Missense , Tyrosinemias/enzymology , Tyrosinemias/genetics , Alleles , Child, Preschool , Exons , Female , Genotype , Humans , Hydrolases/metabolism , Infant , Introns , Liver/pathology , Male , Mexico/epidemiology , Tyrosinemias/pathology
5.
Cell Transplant ; 28(1): 79-88, 2019 01.
Article En | MEDLINE | ID: mdl-30477316

Orthotopic liver transplantation remains the only curative therapy for inborn errors of metabolism. Given the tremendous success for primary immunodeficiencies using ex-vivo gene therapy with lentiviral vectors, there is great interest in developing similar curative therapies for metabolic liver diseases. We have previously generated a pig model of hereditary tyrosinemia type 1 (HT1), an autosomal recessive disorder caused by deficiency of fumarylacetoacetate hydrolase (FAH). Using this model, we have demonstrated curative ex-vivo gene and cell therapy using a lentiviral vector to express FAH in autologous hepatocytes. To further evaluate the long-term clinical outcomes of this therapeutic approach, we continued to monitor one of these pigs over the course of three years. The animal continued to thrive off the protective drug NTBC, gaining weight appropriately, and maintaining sexual fecundity for the course of his life. The animal was euthanized 31 months after transplantation to perform a thorough biochemical and histological analysis. Biochemically, liver enzymes and alpha-fetoprotein levels remained normal and abhorrent metabolites specific to HT1 remained corrected. Liver histology showed no evidence of tumorigenicity and Masson's trichrome staining revealed minimal fibrosis and no evidence of cirrhosis. FAH-immunohistochemistry revealed complete repopulation of the liver by transplanted FAH-positive cells. A complete histopathological report on other organs, including kidney, revealed no abnormalities. This study is the first to demonstrate long-term safety and efficacy of hepatocyte-directed gene therapy in a large animal model. We conclude that hepatocyte-directed ex-vivo gene therapy is a rational choice for further exploration as an alternative therapeutic approach to whole organ transplantation for metabolic liver disease, including HT1.


Cell- and Tissue-Based Therapy/methods , Genetic Therapy/methods , Hydrolases/metabolism , Tyrosinemias/enzymology , Tyrosinemias/therapy , Animals , Computational Biology , Disease Models, Animal , Hydrolases/genetics , Male , Swine , Tyrosinemias/metabolism
6.
J Med Chem ; 60(10): 4101-4125, 2017 05 25.
Article En | MEDLINE | ID: mdl-28128559

This review mainly focuses on the physiological function of 4-hydroxyphenylpyruvate dioxygenase (HPPD), as well as on the development and application of HPPD inhibitors of several structural classes. Among them, one illustrative example is represented by compounds belonging to the class of triketone compounds. They were discovered by serendipitous observations on weed growth and were developed as bleaching herbicides. Informed reasoning on nitisinone (NTBC, 14), a triketone that failed to reach the final steps of the herbicidal design and development process, allowed it to become a curative agent for type I tyrosinemia (T1T) and to enter clinical trials for alkaptonuria. These results boosted the research of new compounds able to interfere with HPPD activity to be used for the treatment of the tyrosine metabolism-related diseases.


4-Hydroxyphenylpyruvate Dioxygenase/antagonists & inhibitors , Drug Discovery , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Herbicides/chemistry , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Weed Control , 4-Hydroxyphenylpyruvate Dioxygenase/metabolism , Alkaptonuria/drug therapy , Alkaptonuria/enzymology , Animals , Drug Discovery/methods , Enzyme Inhibitors/pharmacokinetics , Enzyme Inhibitors/therapeutic use , Herbicides/metabolism , Humans , Models, Molecular , Plants/drug effects , Plants/enzymology , Small Molecule Libraries/pharmacokinetics , Small Molecule Libraries/therapeutic use , Tyrosinemias/drug therapy , Tyrosinemias/enzymology , Weed Control/methods
8.
Sci Transl Med ; 8(349): 349ra99, 2016 07 27.
Article En | MEDLINE | ID: mdl-27464750

We tested the hypothesis that ex vivo hepatocyte gene therapy can correct the metabolic disorder in fumarylacetoacetate hydrolase-deficient (Fah(-/-)) pigs, a large animal model of hereditary tyrosinemia type 1 (HT1). Recipient Fah(-/-) pigs underwent partial liver resection and hepatocyte isolation by collagenase digestion. Hepatocytes were transduced with one or both of the lentiviral vectors expressing the therapeutic Fah and the reporter sodium-iodide symporter (Nis) genes under control of the thyroxine-binding globulin promoter. Pigs received autologous transplants of hepatocytes by portal vein infusion. After transplantation, the protective drug 2-(2-nitro-4-trifluoromethylbenzyol)-1,3 cyclohexanedione (NTBC) was withheld from recipient pigs to provide a selective advantage for expansion of corrected FAH(+) cells. Proliferation of transplanted cells, assessed by both immunohistochemistry and noninvasive positron emission tomography imaging of NIS-labeled cells, demonstrated near-complete liver repopulation by gene-corrected cells. Tyrosine and succinylacetone levels improved to within normal range, demonstrating complete correction of tyrosine metabolism. In addition, repopulation of the Fah(-/-) liver with transplanted cells inhibited the onset of severe fibrosis, a characteristic of nontransplanted Fah(-/-) pigs. This study demonstrates correction of disease in a pig model of metabolic liver disease by ex vivo gene therapy. To date, ex vivo gene therapy has only been successful in small animal models. We conclude that further exploration of ex vivo hepatocyte genetic correction is warranted for clinical use.


Genetic Therapy/methods , Liver/metabolism , Tyrosinemias/metabolism , Tyrosinemias/therapy , Animals , Cyclohexanones/pharmacology , Disease Models, Animal , Hepatocytes/drug effects , Hepatocytes/metabolism , Hydrolases/genetics , Hydrolases/metabolism , Immunohistochemistry , Nitrobenzoates/pharmacology , Swine , Transplantation, Homologous , Tyrosinemias/enzymology , Tyrosinemias/genetics
9.
Pediatr Int ; 58(10): 1069-1072, 2016 Oct.
Article En | MEDLINE | ID: mdl-27285949

Tyrosinemia type II is a rare autosomal recessive disorder caused by deficiency of tyrosine aminotransferase (TAT). It may occur with ocular and cutaneous symptoms with or without mental retardation, but epileptic seizure is a rare presentation of this disease. Herein we report the clinical, biochemical and genetic features of a 4-year-old boy who presented with afebrile seizure and photophobia. Genomic DNA was obtained from peripheral blood leukocytes from the whole family. Sequencing analysis was performed using the MiSeq next-generation sequencing platform. Sequencing of TAT indicated two new homozygous mutations p.L312P (c.935T>C) and p.T408M (c.1223C>T) for the proband and his asymptomatic sister. During a 2 year follow-up period, the patient had overall poor compliance with protein-restricted diet, but his asymptomatic sister had good compliance with the diet. Cognitive function of the patient worsened steadily, but his asymptomatic sister maintained normal mental status. Tyrosinemia type II should be considered in the differential diagnosis of children presenting with epileptic seizure and photophobia; furthermore, early diagnosis and protein-restricted regimen are important to reduce the risk of long-term complications of tyrosinemia type II such as mental disability.


DNA/genetics , Mutation , Tyrosine Transaminase/genetics , Tyrosinemias/genetics , Child, Preschool , DNA Mutational Analysis , Homozygote , Humans , Male , Pedigree , Tyrosine Transaminase/metabolism , Tyrosinemias/enzymology
11.
Stem Cell Res ; 13(1): 144-53, 2014 Jul.
Article En | MEDLINE | ID: mdl-24879068

Hereditary tyrosinemia type I (HT1) is caused by deficiency in fumarylacetoacetate hydrolase (FAH), an enzyme that catalyzes the last step of tyrosine metabolism. The most severe form of the disease presents acutely during infancy, and is characterized by severe liver involvement, most commonly resulting in death if untreated. Generation of FAH(+/-) pigs was previously accomplished by adeno-associated virus-mediated gene knockout in fibroblasts and somatic cell nuclear transfer. Subsequently, these animals were outbred and crossed to produce the first FAH(-/-) pigs. FAH-deficiency produced a lethal defect in utero that was corrected by administration of 2-(2-nitro-4-trifluoromethylbenzoyl)-1,3 cyclohexanedione (NTBC) throughout pregnancy. Animals on NTBC were phenotypically normal at birth; however, the animals were euthanized approximately four weeks after withdrawal of NTBC due to clinical decline and physical examination findings of severe liver injury and encephalopathy consistent with acute liver failure. Biochemical and histological analyses, characterized by diffuse and severe hepatocellular damage, confirmed the diagnosis of severe liver injury. FAH(-/-) pigs provide the first genetically engineered large animal model of a metabolic liver disorder. Future applications of FAH(-/-) pigs include discovery research as a large animal model of HT1 and spontaneous acute liver failure, and preclinical testing of the efficacy of liver cell therapies, including transplantation of hepatocytes, liver stem cells, and pluripotent stem cell-derived hepatocytes.


Hydrolases/deficiency , Liver Diseases/enzymology , Tyrosinemias/enzymology , Animals , Disease Models, Animal , Female , Gene Knockout Techniques , Genotype , Liver Diseases/metabolism , Male , Pregnancy , Swine
12.
Clin Genet ; 86(2): 167-71, 2014 Aug.
Article En | MEDLINE | ID: mdl-23895425

Hereditary tyrosinemia type I (HT1) is a rare disease caused by a deficiency of fumarylacetoacetate hydrolase (FAH) in the tyrosine catabolic pathway, resulting mainly in hepatic alterations due to accumulation of the toxic metabolites fumarylacetoacetate, maleylacetoacetate and succinylacetone. We have characterized using minigenes four splicing mutations affecting exonic or intronic nucleotides of the FAH gene identified in two HT1 patients. Two of the mutations are novel, c.82-1G>A and c.913G>C and the other two have been previously associated with a splicing defect (c.836A>G and c.1062+5G>A). All mutations were confirmed to affect splicing in minigenes, resulting in exon skipping or activation of a cryptic splice site. We have analyzed the effect of different compounds known to modulate splicing (valproic acid, phenyl butyrate, M344, EIPA, and resveratrol) and the overexpression of splice factors of the SR protein family on the transcriptional profile of the mutant minigenes. For the c.836A>G mutation, a partial recovery of the correctly spliced transcript was observed. These results confirm the relevance of performing functional studies for mutations potentially affecting the splicing process and open the possibility of supplementary therapeutic approaches to diseases caused by splicing defects.


Hydrolases/genetics , Mutation/genetics , RNA Splicing/genetics , Tyrosinemias/enzymology , Tyrosinemias/genetics , Base Sequence , Computer Simulation , Humans , Infant , Molecular Sequence Data
13.
Ideggyogy Sz ; 66(11-12): 415-9, 2013 Nov 30.
Article En | MEDLINE | ID: mdl-24555242

Before the introduction of the NTBC treatment (Orfadine) from two tyrosinemic Hungarian families 1-3 tyrosinemic homozygous male patients died of hepatocellular carcinoma and one patient of hepatocellular carcinoma combined with clear cell renal adenocarcinoma. From the third tyrosinemic family one homozygous girl patient has been treated with NTBC (Orfadine), IMTV-AM, she is symptom-free. Her molecular genetic mutations analysis in the FAH gene detected a common intronel mutation, affecting splicing and of predicted severe effect, IVS6-1 g > t/IVS6-1 g > t with systemic name c.456-1 g > t/c.456-1 g > t (Prof. Magdalena Ugarte).


Cyclohexanones/therapeutic use , Enzyme Inhibitors/therapeutic use , Hydrolases/genetics , Nitrobenzoates/therapeutic use , Tyrosine/blood , Tyrosinemias/genetics , 4-Hydroxyphenylpyruvate Dioxygenase/antagonists & inhibitors , Carcinoma, Hepatocellular/genetics , Carcinoma, Renal Cell/genetics , Child, Preschool , Fatal Outcome , Homozygote , Humans , Hungary , Kidney Neoplasms/genetics , Liver Neoplasms/genetics , Male , Treatment Outcome , Tyrosinemias/diagnosis , Tyrosinemias/drug therapy , Tyrosinemias/enzymology
14.
Cornea ; 32(1): 91-4, 2013 Jan.
Article En | MEDLINE | ID: mdl-22495034

PURPOSE: To describe the confocal microscopic findings in a patient with hereditary tyrosinemia type I (HT-I) treated with 2-(2-nitro-4-trifluoromethylbenzoyl)-1,3-cyclohexanedione (NTBC) who developed corneal crystals. METHODS: In this case study, we describe the confocal microscopic findings in a boy, who was diagnosed with HT-I at the age of 4 months. At 16 years of age, he developed painful corneal lesions in both eyes. On slit-lamp examination, whorl-like branching epithelial corneal lesions were found, staining faintly with fluorescein. His NTBC treatment was stopped and reintroduced at a lower dose after 1 month. The lesions clearly regressed, leaving only mild residual epithelial scarring, without fluorescein staining and without pain. Confocal microscopy was performed in the acute painful stage and in the asymptomatic convalescent stage 5 months later. RESULTS: Confocal microscopy confirmed the presence of slender birefringent spiky crystals in the very superficial corneal epithelium. In the asymptomatic convalescent phase, the crystals clearly persisted on confocal microscopy, although they were barely visible on slit-lamp examination. CONCLUSIONS: This is the first in vivo demonstration by confocal microscopy of corneal crystals present in a patient with proven type I tyrosinemia, under NTBC treatment.


Corneal Opacity/diagnosis , Cyclohexanones/therapeutic use , Enzyme Inhibitors/therapeutic use , Epithelium, Corneal/pathology , Nitrobenzoates/therapeutic use , Tyrosinemias/diagnosis , 4-Hydroxyphenylpyruvate Dioxygenase/antagonists & inhibitors , Adolescent , Corneal Opacity/drug therapy , Corneal Opacity/enzymology , Crystallization , Epithelium, Corneal/drug effects , Epithelium, Corneal/enzymology , Fluorescein/metabolism , Humans , Male , Microscopy, Confocal , Staining and Labeling , Tyrosinemias/drug therapy , Tyrosinemias/enzymology
15.
Neurochem Int ; 61(8): 1370-4, 2012 Dec.
Article En | MEDLINE | ID: mdl-23046746

Tyrosinemia is a rare genetic disease caused by mutations on genes that codify enzymes responsible for tyrosine metabolism. Considering that tyrosinemics patients usually present symptoms associated with central nervous system alterations that ranges from slight decreases in intelligence to severe mental retardation, we decided to investigate whether acute and chronic administration of L-tyrosine in rats would affect acetylcholinesterase mRNA expression and enzymatic activity during their development. In our acute protocol, Wistar rats (10 and 30 days old) were killed one hour after a single intraperitoneal L-tyrosine injection (500 mg/kg) or saline. Chronic administration consisted of L-tyrosine (500 mg/kg) or saline injections 12 h apart for 24 days in Wistar rats (7 days old) and rats were killed 12 h after last injection. Acetylcholinesterase activity was measured by Ellman's method and acetylcholinesterase expression was carried out by a semi-quantitative reverse transcriptase polymerase chain reaction (RT-PCR) assay. We observed that acute (10 and 30 days old rats) and chronic L-tyrosine administration increased acetylcholinesterase activity in serum and all tested brain areas (hippocampus, striatum and cerebral cortex) when compared to control group. Moreover, there was a significant decrease in mRNA levels of acetylcholinesterase in hippocampus was observed after acute protocol (10 and 30 days old rats) and in striatum after chronic protocol. In case these alterations also occur in the brain of the patients, our results may explain, at least in part, the neurological sequelae associated with high plasma concentrations of tyrosine seen in patients affected by tyrosinemia type II.


Acetylcholinesterase/biosynthesis , Tyrosine/pharmacology , Acetylcholinesterase/blood , Acetylcholinesterase/genetics , Animals , Animals, Newborn , Animals, Suckling , Brain Chemistry/drug effects , Disease Models, Animal , Drug Administration Schedule , Enzyme Induction/drug effects , GPI-Linked Proteins/biosynthesis , GPI-Linked Proteins/blood , GPI-Linked Proteins/genetics , Injections, Intraperitoneal , Male , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , RNA, Messenger/biosynthesis , Rats , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction , Tyrosine/administration & dosage , Tyrosinemias/enzymology
16.
Scand J Clin Lab Invest ; 72(5): 369-73, 2012 Sep.
Article En | MEDLINE | ID: mdl-22554029

A total of 28 Norwegians have been diagnosed with hereditary tyrosinaemia type I (HT1) over the last 30 years. In this study, 19 of these patients were investigated. Three novel small deletions were found (NM_000137.1(FAH): c.615delT, p.Phe205LeufsX2, NM_000137.1(FAH): c.744delG, p.Pro249HisfsX55 and NM_000137.1(FAH):c835delC) pGln279ArgfsX25, all of them leading to a change in the reading frame and a premature stop codon. We hereby genetically characterized 51 of the 56 disease-causing alleles, identifying nine different disease-causing mutations in the Norwegian population. We found that 65% of the Norwegian HT1 patients are compound heterozygous for different mutations. Thus, the relatively high incidence of HT1 in Norway of 1 in 74,800 live births is not due to single founder effects or high incidence of parental consanguinity.


Hydrolases/genetics , Sequence Deletion , Tyrosinemias/epidemiology , Tyrosinemias/genetics , Base Sequence , DNA Mutational Analysis , Exons , Female , Gene Frequency , Genetic Association Studies , Humans , Incidence , Male , Models, Molecular , Norway/epidemiology , Protein Structure, Tertiary , Tyrosinemias/enzymology
17.
Mol Genet Metab ; 102(2): 122-5, 2011 Feb.
Article En | MEDLINE | ID: mdl-21112803

OBJECTIVE: Psychomotor impairment has been described in hypertyrosinemia types II and III (HT III). Only recently cognitive deficits have also been reported in hypertyrosinemia type I (HT I). The pathogenic mechanisms responsible are unknown. Since implementation of 2-(2-nitro-4-trifluoromethylbenzoyl)-1,3-cyclohexanedione (NTBC, Nitisinone (Swedish Orphan International)) in the treatment of HT I, plasma tyrosine elevation is a common finding as known from the other hypertyrosinemias. PATIENTS AND METHODS: With elevated tyrosine as suspected pathogenic factor in the development of cognitive deficits, we here investigated tyrosine in the cerebrospinal fluid (CSF) and serotonergic and dopaminergic neurotransmitter levels in three patients with HT I during long-term treatment with Nitisinone. In addition, Nitisinone concentrations in plasma and CSF were measured. We also assessed psychomotor and cognitive development by standardized test systems and brain morphology by magnetic resonance imaging. RESULTS: All patients presented with high tyrosine concentrations in CSF correlating with increased plasma tyrosine levels and a reduced CSF serotonin turnover. MRI revealed no structural abnormalities in the brain. All patients presented with either impaired cognitive development or behavioural abnormalities. CONCLUSIONS: We here outline the need to further study the exact pathogenic mechanisms responsible for the neurotransmitter changes observed in HT type I in order to possibly prevent cognitive dysfunction. Nitisinone has significantly improved outcome and quality of life in HT type I; however, it is also accompanied by elevated plasma and CSF tyrosine. Further studies are essential to identify the necessary dietary tyrosine restriction and the optimal Nitisinone dose.


Cyclohexanones/therapeutic use , Enzyme Inhibitors/therapeutic use , Nitrobenzoates/therapeutic use , Serotonin/metabolism , Tyrosine/cerebrospinal fluid , Tyrosinemias/drug therapy , Tyrosinemias/enzymology , Brain/anatomy & histology , Child , Cyclohexanones/blood , Cyclohexanones/cerebrospinal fluid , Enzyme Inhibitors/blood , Enzyme Inhibitors/cerebrospinal fluid , Humans , Infant , Male , Nitrobenzoates/blood , Nitrobenzoates/cerebrospinal fluid , Psychomotor Performance , Serotonin/analysis , Tyrosine/blood
18.
Eur J Med Genet ; 54(3): 205-8, 2011.
Article En | MEDLINE | ID: mdl-21145993

In the present study we report the clinical features and the molecular genetic investigation of the tyrosine aminotransferase (TAT) gene in a young girl from Croatia with Richner-Hanhart syndrome, mainly suffering from photophobia, hyperkeratosis of the palmes and soles and slight neurological abnormalities. Sequencing analysis of the TAT gene revealed a novel homozygous missense mutation c.1250G>A (p.R417Q) in exon 12, and herewith confirmed the clinical diagnosis. Showing the first symptoms in babyhood, at the age of 8 years it was for the first time clinically diagnosed that the patient suffers from tyrosinemia type II and a therapy with tyrosine and phenylalanine reduced diet has been started successfully. All symptoms disappeared within 2-4 weeks. Since that time, we have been following the girl until today for more than ten years. She is in a good condition, and attends the normal high school program.


Corneal Diseases/genetics , Keratoderma, Palmoplantar/genetics , Mutation, Missense , Tyrosine Transaminase/genetics , Tyrosinemias/genetics , Base Sequence , Corneal Diseases/enzymology , Corneal Diseases/pathology , DNA Mutational Analysis , Female , Humans , Keratoderma, Palmoplantar/enzymology , Keratoderma, Palmoplantar/pathology , Syndrome , Tyrosine Transaminase/deficiency , Tyrosinemias/enzymology , Tyrosinemias/pathology , Young Adult
20.
Gastroenterology ; 139(3): 1019-29, 2010 Sep.
Article En | MEDLINE | ID: mdl-20621682

BACKGROUND & AIMS: Due to the shortage of donor organs, many patients needing liver transplantation cannot receive one. For some liver diseases, hepatocyte transplantation could be a viable alternative, but donor cells currently are procured from the same sources as whole organs, and thus the supply is severely limited. METHODS: Here, we investigated the possibility of isolating viable hepatocytes for liver cell therapy from the plentiful source of morgue cadavers. To determine the utility of this approach, cells were isolated from the livers of non-heart-beating cadaveric mice long after death and transplanted into fumarylacetoacetate hydrolase-deficient mice, a model for the human metabolic liver disease hereditary tyrosinemia type I and a stringent in vivo model for hepatic cell transplantation. RESULTS: Surprisingly, complete and therapeutic liver repopulation could be achieved with hepatocytes derived up to 27 hours post mortem. CONCLUSIONS: Competitive repopulation experiments showed that cadaveric liver cells had a repopulation capacity similar to freshly isolated hepatocytes. Importantly, viable hepatocytes also could be isolated from cadaveric primate liver (monkey and human) efficiently. These data provide evidence that non-heart-beating donors could be a suitable source of hepatocytes for much longer time periods than previously thought possible.


Hepatocytes/transplantation , Hydrolases/deficiency , Liver Regeneration , Liver/enzymology , Tyrosinemias/surgery , Animals , Biomarkers/blood , Cadaver , Cell Proliferation , Cell Separation , Cell Survival , Cells, Cultured , Disease Models, Animal , Hepatocytes/enzymology , Humans , Hydrolases/genetics , Liver/pathology , Macaca mulatta , Mice , Mice, Knockout , Proteins/genetics , RNA, Untranslated , Temperature , Time Factors , Tyrosine/blood , Tyrosinemias/enzymology , Tyrosinemias/genetics , Tyrosinemias/pathology
...