Six novel mutations in the fumarylacetoacetate hydrolase gene of patients with hereditary tyrosinemia type I

1996 ◽  
Vol 7 (4) ◽  
pp. 367-369 ◽  
Author(s):  
Cynthia Timmers ◽  
Markus Grompe
1994 ◽  
Vol 331 (6) ◽  
pp. 353-357 ◽  
Author(s):  
Markus Grompe ◽  
Maryse St.-Louis ◽  
Sylvie I. Demers ◽  
Muhsen Al-Dhalimy ◽  
Barbara Leclerc ◽  
...  

2019 ◽  
Vol 294 (35) ◽  
pp. 13051-13060 ◽  
Author(s):  
Iratxe Macias ◽  
Ana Laín ◽  
Ganeko Bernardo-Seisdedos ◽  
David Gil ◽  
Esperanza Gonzalez ◽  
...  

1995 ◽  
Vol 4 (2) ◽  
pp. 319-320 ◽  
Author(s):  
Maryse St-Louis ◽  
Jacques Poudrier ◽  
Daniel Phaneuf ◽  
Barbara Lecierc ◽  
Rachel Laframboise ◽  
...  

2021 ◽  
Vol 22 (4) ◽  
pp. 1789
Author(s):  
Jon Gil-Martínez ◽  
Iratxe Macias ◽  
Luca Unione ◽  
Ganeko Bernardo-Seisdedos ◽  
Fernando Lopitz-Otsoa ◽  
...  

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.


2020 ◽  
Vol 21 (6) ◽  
pp. 2136 ◽  
Author(s):  
Dario Balestra ◽  
Daniela Scalet ◽  
Mattia Ferrarese ◽  
Silvia Lombardi ◽  
Nicole Ziliotto ◽  
...  

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.


2012 ◽  
Vol 2012 ◽  
pp. 1-4 ◽  
Author(s):  
Jayesh J. Sheth ◽  
Chitra M. Ankleshwaria ◽  
Rajeshwari Pawar ◽  
Frenny J. Sheth

Carrier of tyrosinemia type I was diagnosed by sequencingFAH(fumarylacetoacetate hydrolase) gene. It leads to the identification of heterozygous status for both c.648C>G (p.Ile216Met) and c.1159G>A (p.Gly387Arg) mutations in exons 8 and 13, respectively, in the parents. The experimental program PolyPhen, SIFT, and MT predicts former missense point mutation as “benign” that creates a potential donor splice site and later one as “probably damaging” which disrupts secondary structure of protein.


2019 ◽  
Vol 116 (44) ◽  
pp. 22229-22236 ◽  
Author(s):  
Shuzhang Yang ◽  
Sandra M. Siepka ◽  
Kimberly H. Cox ◽  
Vivek Kumar ◽  
Marleen de Groot ◽  
...  

Fumarylacetoacetate hydrolase (FAH) is the last enzyme in tyrosine catabolism, and mutations in the FAH gene are associated with hereditary tyrosinemia type I (HT1 or TYRSN1) in humans. In a behavioral screen of N-ethyl-N-nitrosourea mutagenized mice we identified a mutant line which we named “swingshift” (swst, MGI:3611216) with a nonsynonymous point mutation (N68S) in Fah that caused age-dependent disruption of sleep–wake patterns. Mice homozygous for the mutation had an earlier onset of activity (several hours before lights off) and a reduction in total activity and body weight when compared with wild-type or heterozygous mice. Despite abnormal behavioral entrainment to light–dark cycles, there were no differences in the period or phase of the central clock in mutant mice, indicating a defect downstream of the suprachiasmatic nucleus. Interestingly, these behavioral phenotypes became milder as the mice grew older and were completely rescued by the administration of NTBC [2-(2-nitro-4-trifluoromethylbenzoyl)-1,3-cyclohexanedione], an inhibitor of 4-hydroxyphenylpyruvate dioxygenase, which is upstream of FAH. Mechanistically, the swst mutation had no effect on the enzymatic activity of FAH, but rather promoted the degradation of the mutant protein. This led to reduced FAH protein levels and enzymatic activity in the liver and kidney (but not the brain or fibroblasts) of homozygous mice. In addition, plasma tyrosine—but not methionine, phenylalanine, or succinylacetone—increased in homozygous mice, suggesting that swst mutants provide a model of mild, chronic HT1.


Sign in / Sign up

Export Citation Format

Share Document