dibasic amino acids
Recently Published Documents


TOTAL DOCUMENTS

65
(FIVE YEARS 2)

H-INDEX

19
(FIVE YEARS 0)

Animals ◽  
2021 ◽  
Vol 11 (8) ◽  
pp. 2437
Author(s):  
Simona Kovaříková ◽  
Petr Maršálek ◽  
Kateřina Vrbová

The purpose of this review is to summarize current knowledge on canine and feline cystinuria from available scientific reports. Cystinuria is an inherited metabolic defect characterized by abnormal intestinal and renal amino acid transport in which cystine and the dibasic amino acids ornithine, lysine, and arginine are involved (COLA). At a normal urine pH, ornithine, lysine, and arginine are soluble, but cysteine forms a dimer, cystine, which is relatively insoluble, resulting in crystal precipitation. Mutations in genes coding COLA transporter and the mode of inheritance were identified only in some canine breeds. Cystinuric dogs may form uroliths (mostly in lower urinary tract) which are associated with typical clinical symptoms. The prevalence of cystine urolithiasis is much higher in European countries (up to 14% according to the recent reports) when compared to North America (United States and Canada) where it is approximately 1–3%. Cystinuria may be diagnosed by the detection of cystine urolithiasis, cystine crystalluria, assessment of amino aciduria, or using genetic tests. The management of cystinuria is aimed at urolith removal or dissolution which may be reached by dietary changes or medical treatment. In dogs with androgen-dependent cystinuria, castration will help. In cats, cystinuria occurs less frequently in comparison with dogs.


2021 ◽  
Author(s):  
Yongchan Lee ◽  
Pattama Wiriyasermkul ◽  
Satomi Moriyama ◽  
Deryck J Mills ◽  
Werner Kühlbrandt ◽  
...  

Cystinuria is a genetic disorder characterized by overexcretion of dibasic amino acids and cystine, which causes recurrent kidney stones and occasionally severe kidney failure. Mutations of the two responsible proteins, rBAT and b0,+AT, which comprise system b0,+, are linked to type I and non-type I cystinuria respectively and they exhibit distinct phenotypes due to protein trafficking defects or catalytic inactivation. Although recent structural insights into human b0,+AT-rBAT suggested a model for transport-inactivating mutations, the mechanisms by which type I mutations trigger trafficking deficiencies are not well understood. Here, using electron cryo-microscopy and biochemistry, we discover that Ca2+-mediated higher-order assembly of system b0,+ is the key to its trafficking on the cell surface. We show that Ca2+ stabilizes the interface between two rBAT molecules to mediate super-dimerization, and this in turn facilitates the N-glycan maturation of system b0,+. A common cystinuria mutant T216M and mutations that disrupt the Ca2+ site in rBAT cause the loss of higher-order assemblies, resulting in protein trafficking deficiency. Mutations at the super-dimer interface reproduce the mis-trafficking phenotype, demonstrating that super-dimerization is essential for cellular function. Cell-based transport assays confirmed the importance of the Ca2+ site and super-dimerization, and additionally suggested which residues are involved in cationic amino acid recognition. Taken together, our results provide the molecular basis of type I cystinuria and serve as a guide to develop new therapeutic strategies against it. More broadly, our findings reveal an unprecedented link between transporter oligomeric assembly and trafficking diseases in general.


2018 ◽  
Author(s):  
Jacob Britt ◽  
Ava Saidian ◽  
Dustin Whitaker ◽  
Carter Boyd ◽  
Kyle Wood ◽  
...  

Cystinuria is a relatively rare autosomal recessive disorder that manifests early in life and is associated with the development of kidney stones composed of cystine. It is due to mutations in two genes that are involved in the transport of cystine, neutral, and dibasic amino acids in the proximal tubule of the kidney. Patients are at risk for developing chronic kidney disease. Diagnosis is typically established with stone analysis and quantitative urinary cystine excretion. These patients may form extremely large stones requiring percutaneous nephrolithotomy. Stone-prevention strategies include dietary modifications (increased fluid intake and limitation of sodium and animal protein consumption), urine pH manipulation, and thiol-binding agents. These patients should be followed closely, and preemptive stone removal with ureteroscopy should be considered to limit the necessity for more invasive procedures.   This review contains 2 figures and 38 references. Key Words: a-mercaptopropionyl glycine, amino acid transport, chronic kidney disease, cystinuria, SLC3A1, SLC7A9, thiol-binding agent, urinary pH manipulation


2014 ◽  
Vol 144 (3) ◽  
pp. 282-288 ◽  
Author(s):  
Jessica D. Bihuniak ◽  
Rebecca R. Sullivan ◽  
Christine A. Simpson ◽  
Donna M. Caseria ◽  
Tania B. Huedo-Medina ◽  
...  

2011 ◽  
Vol 58 (2) ◽  
Author(s):  
Małgorzata Ciszewska ◽  
Katarzyna Ruszczyńska ◽  
Marta Oleszczuk ◽  
Nga N Chung ◽  
Ewa Witkowska ◽  
...  

Six hybrid N-ureidoethylamides of octapeptides in which an N-terminal cyclic structure related to enkephalin was elongated by a C-terminal fragment of deltorphin were synthesized on MBHA resin. The synthetic procedure involved deprotection of Boc groups with HCl/dioxane and cleavage of the peptide resin with 45 % TFA in DCM. d-Lys and d-Orn were incorporated in position 2, and Lys, Orn, Dab, or Dap in position 5. The side chains of the dibasic amino function were protected with the Fmoc group. This protection was removed by treatment with 55 % piperidine in DMF, and cyclization was achieved by treatment with bis-(4-nitrophenyl)carbonate. Using various combinations of dibasic amino acids, peptides containing a 17-, 18-, 19- or 20-membered ring structure were obtained. The peptides were tested in the guinea-pig ileum (GPI) and mouse vas deferens (MVD) assays. Diverse opioid activities were observed, depending on the size of the ring. Extension of the enkephalin sequence at the C-terminus by a deltorphin fragment resulted in a change of receptor selectivity in favor of the δ receptor. The conformational propensities of selected peptides were determined using the EDMC method in conjunction with data derived from NMR experiments carried out in water. This approach allowed proper examination of the dynamical behavior of these small peptides. The results were compared with those obtained earlier with corresponding N-(ureidoethyl)pentapeptide amides.


2009 ◽  
Vol 12 (1) ◽  
pp. 15-20 ◽  
Author(s):  
K Popovska-Jankovic ◽  
V Tasic ◽  
R Bogdanovic ◽  
P Miljkovic ◽  
E Baskin ◽  
...  

Five Novel Mutations in Cystinuria Genes SLC3A1 and SLC7A9Cystinuria is an autosomal recessive disorder that is characterized by impaired transport of cystine, lysine, ornithine and arginine in the proximal renal tubule and epithelial cells of the gastrointestinal tract. The transport of these amino acids is mediated by the rBAT/b0,+AT transporter, the subunits of which are encoded by the genes SLC3A1, located on chromosome 2p16.3-21, and SLC7A9, located on chromosome 19q12-13.1. Based on the urinary cystine excretion patterns of obligate heterozygotes, cystinuria is classified into type I (normal amino acid urinary pattern in heterozygotes) and non type I (a variable degree of urinary hyper excretion of cystine and dibasic amino acids in heterozygotes). On the basis of genetic aspects, cystinuria is classified into type A, is caused by mutations in both alleles of SLC3A1; type B, caused by mutations in both alleles of SLC7A9 and type AB, is caused by one mutation in SLC3A1 and one mutation in SLC7A9. Here we present two novel mutations in the SLC3A1 gene (C242R and L573X), which were found in patients from Serbia, and three in the SLC7A9 gene (G73R, V375I, 1048-1051 delACTC), found in patients from Serbia, Macedonia and Turkey, respectively.


2007 ◽  
Vol 293 (3) ◽  
pp. F732-F740 ◽  
Author(s):  
Mariona Font-Llitjós ◽  
Lídia Feliubadaló ◽  
Meritxell Espino ◽  
Ramon Clèries ◽  
Sandra Mañas ◽  
...  

Cystinuria is a hereditary disorder caused by a defect in the apical membrane transport system for cystine and dibasic amino acids in renal proximal tubules and intestine, resulting in recurrent urolithiasis. Mutations in SLC3A1 and SLC7A9 genes, that codify for rBAT/b0,+AT transporter subunits, cause type A and B cystinuria, respectively. In humans, cystinuria treatment is based on the prevention of calculi formation and its dissolution or breakage. Persistent calculi are treated with thiols [i.e., d-penicillamine (DP) and mercaptopropionylglycine (MPG)] for cystine solubilization. We have developed a new protocol with DP to validate our Slc7a9 knockout mouse model for the study of the therapeutic effect of drugs in the treatment of cystine lithiasis. We performed a 5-wk treatment of individually caged lithiasic mutant mice with a previously tested DP dose. To appraise the evolution of lithiasis throughout the treatment a noninvasive indirect method of calculi quantification was developed: calculi mass was quantified by densitometry of X-ray images from cystinuric mice before and after treatment. Urine was collected in metabolic cage experiments to quantify amino acids in DP-treated and nontreated, nonlithiasic mutant mice. We found significant differences between DP-treated and nontreated knockout mice in calculi size and in urinary cystine excretion. Histopathological analysis showed that globally nontreated mutant mice had more severe and diffuse urinary system damage than DP-treated mice. Our results validate the use of this mouse model for testing the efficacy of potential new drugs against cystinuria.


Sign in / Sign up

Export Citation Format

Share Document