arginase deficiency
Recently Published Documents


TOTAL DOCUMENTS

54
(FIVE YEARS 6)

H-INDEX

18
(FIVE YEARS 1)

JIMD Reports ◽  
2021 ◽  
Author(s):  
Nandaki Keshavan ◽  
Michelle Wood ◽  
Lucy M. Alderson ◽  
Mario Cortina‐Borja ◽  
Rachel Skeath ◽  
...  

2021 ◽  
Vol 12 ◽  
Author(s):  
Kuntal Sen ◽  
Afrouz A. Anderson ◽  
Matthew T. Whitehead ◽  
Andrea L. Gropman

The urea cycle disorders (UCD) are rare genetic disorder due to a deficiency of one of six enzymes or two transport proteins that act to remove waste nitrogen in form of ammonia from the body. In this review, we focus on neuroimaging studies in OTCD and Arginase deficiency, two of the UCD we have extensively studied. Ornithine transcarbamylase deficiency (OTCD) is the most common of these, and X-linked. Hyperammonemia (HA) in OTCD is due to deficient protein handling. Cognitive impairments and neurobehavioral disorders have emerged as the major sequelae in Arginase deficiency and OTCD, especially in relation to executive function and working memory, impacting pre-frontal cortex (PFC). Clinical management focuses on neuroprotection from HA, as well as neurotoxicity from other known and yet unclassified metabolites. Prevention and mitigation of neurological injury is a major challenge and research focus. Given the impact of HA on neurocognitive function of UCD, neuroimaging modalities, especially multi-modality imaging platforms, can bring a wealth of information to understand the neurocognitive function and biomarkers. Such information can further improve clinical decision making, and result in better therapeutic interventions. In vivo investigations of the affected brain using multimodal neuroimaging combined with clinical and behavioral phenotyping hold promise. MR Spectroscopy has already proven as a tool to study biochemical aberrations such as elevated glutamine surrounding HA as well as to diagnose partial UCD. Functional Near Infrared Spectroscopy (fNIRS), which assesses local changes in cerebral hemodynamic levels of cortical regions, is emerging as a non-invasive technique and will serve as a surrogate to fMRI with better portability. Here we review two decades of our research using non-invasive imaging and how it has contributed to an understanding of the cognitive effects of this group of genetic conditions.


Author(s):  
Patricia Jones ◽  
Khushbu Patel ◽  
Dinesh Rakheja
Keyword(s):  

2019 ◽  
Vol 116 (42) ◽  
pp. 21150-21159 ◽  
Author(s):  
Brian Truong ◽  
Gabriella Allegri ◽  
Xiao-Bo Liu ◽  
Kristine E. Burke ◽  
Xuling Zhu ◽  
...  

Arginase deficiency is caused by biallelic mutations in arginase 1 (ARG1), the final step of the urea cycle, and results biochemically in hyperargininemia and the presence of guanidino compounds, while it is clinically notable for developmental delays, spastic diplegia, psychomotor function loss, and (uncommonly) death. There is currently no completely effective medical treatment available. While preclinical strategies have been demonstrated, disadvantages with viral-based episomal-expressing gene therapy vectors include the risk of insertional mutagenesis and limited efficacy due to hepatocellular division. Recent advances in messenger RNA (mRNA) codon optimization, synthesis, and encapsulation within biodegradable liver-targeted lipid nanoparticles (LNPs) have potentially enabled a new generation of safer, albeit temporary, treatments to restore liver metabolic function in patients with urea cycle disorders, including ARG1 deficiency. In this study, we applied such technologies to successfully treat an ARG1-deficient murine model. Mice were administered LNPs encapsulating human codon-optimized ARG1 mRNA every 3 d. Mice demonstrated 100% survival with no signs of hyperammonemia or weight loss to beyond 11 wk, compared with controls that perished by day 22. Plasma ammonia, arginine, and glutamine demonstrated good control without elevation of guanidinoacetic acid, a guanidino compound. Evidence of urea cycle activity restoration was demonstrated by the ability to fully metabolize an ammonium challenge and by achieving near-normal ureagenesis; liver arginase activity achieved 54% of wild type. Biochemical and microscopic data showed no evidence of hepatotoxicity. These results suggest that delivery of ARG1 mRNA by liver-targeted nanoparticles may be a viable gene-based therapeutic for the treatment of arginase deficiency.


JCI Insight ◽  
2019 ◽  
Vol 4 (17) ◽  
Author(s):  
Xiao-Bo Liu ◽  
Jillian R. Haney ◽  
Gloria Cantero ◽  
Jenna R. Lambert ◽  
Marcos Otero-Garcia ◽  
...  

2019 ◽  
Vol 14 (3) ◽  
pp. 133
Author(s):  
LeemaPauline Cornelius ◽  
Vivekasaravanan Raju ◽  
Asir Julin

2018 ◽  
Vol 26 ◽  
pp. 110-114 ◽  
Author(s):  
Amanda Jichlinski ◽  
Lindsay Clarke ◽  
Matthew T. Whitehead ◽  
Andrea Gropman

2018 ◽  
Vol 124 (2) ◽  
pp. 114-123 ◽  
Author(s):  
Stephanie A.K. Angarita ◽  
Brian Truong ◽  
Suhail Khoja ◽  
Matthew Nitzahn ◽  
Abha K. Rajbhandari ◽  
...  

2017 ◽  
Vol 20 (6) ◽  
pp. 517-521 ◽  
Author(s):  
Matthew Koo ◽  
Gerald S Lipshutz ◽  
Stephen D Cederbaum ◽  
Charles Lassman

2016 ◽  
Vol 36 (25) ◽  
pp. 6680-6690 ◽  
Author(s):  
Gloria Cantero ◽  
Xiao-Bo Liu ◽  
Ronald F. Mervis ◽  
Maria T. Lazaro ◽  
Stephen D. Cederbaum ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document