adult polyglucosan body disease
Recently Published Documents


TOTAL DOCUMENTS

66
(FIVE YEARS 12)

H-INDEX

17
(FIVE YEARS 0)

Author(s):  
Paul Theo Zebhauser ◽  
Isabell Cordts ◽  
Holger Hengel ◽  
Bernhard Haslinger ◽  
Paul Lingor ◽  
...  

AbstractAdult polyglucosan body disease (APBD) is a rare but probably underdiagnosed autosomal recessive neurodegenerative disorder due to pathogenic variants in GBE1. The phenotype is characterized by neurogenic bladder dysfunction, spastic paraplegia, and axonal neuropathy. Additionally, cognitive symptoms and dementia have been reported in APBD but have not been studied systematically. Using exome sequencing, we identified two previously unreported bi-allelic missense GBE1 variants in a patient with severe memory impairment along with the typical non-cognitive symptoms. We were able to confirm a reduction of GBE1 activity in blood lymphocytes. To characterize the neuropsychological profile of patients suffering from APBD, we conducted a systematic review of cognitive impairment in this rare disease. Analysis of 24 cases and case series (in total 58 patients) showed that executive deficits and memory impairment are the most common cognitive symptoms in APBD.


2021 ◽  
Author(s):  
Hilla Vaknin ◽  
Kumudesh Mishra ◽  
Jeevitha D’Souza ◽  
Monzer Marisat ◽  
Uri Sprecher ◽  
...  

AbstractThis work employs Adult Polyglucosan Body Disease (APBD) models to explore the efficacy and mechanism of action of 144DG11, a new polyglucosan-reducing lead compound discovered by a high-throughput screen (HTS). APBD is an adult onset glycogen storage disorder (GSD) manifesting as a debilitating progressive axonopathic leukodystrophy. APBD is caused by glycogen branching enzyme (GBE) deficiency leading to poorly branched and insoluble glycogen inclusions, which precipitate as neuropathogenic polyglucosans (PG). 144DG11 led to prolonged survival and improved motor parameters in a GBE knockin (Gbeys/ys) APBD mouse model. Histopathologically, 144DG11 reduced PG and glycogen levels in brain, liver, heart, and peripheral nerve. Indirect calorimetry experiments revealed that 144DG11 increases carbohydrate burn at the expense of fat burn, suggesting metabolic mobilization of pathogenic PG. These results were also reflected at the cellular level by increased glycolytic, mitochondrial and total ATP production. Mechanistically, we show that the molecular target of 144DG11 is the lysosomal membrane protein LAMP1, whose interaction with the compound, similar to LAMP1 knockdown, enhanced autolysosomal degradation of glycogen and lysosomal acidification. Enhanced mitochondrial activity and lysosomal modifications were also the most pronounced effects of 144DG11 in APBD patient fibroblasts as discovered by image-based multiparametric phenotyping analysis and corroborated by proteomics. In summary, this work presents a broad mechanistic and target-based characterization of 144DG11 in in vivo and cell models of the prototypical GSD APBD. This investigation warrants development of 144DG11 into a safe and efficacious GSD therapy.One Sentence SummaryA new compound, demonstrated to ameliorate APBD in vivo and ex vivo by autophagic catabolism of glycogen, may potentially become a universal drug for glycogen storage disorders.


2021 ◽  
Author(s):  
Emrah Gumusgoz ◽  
Dikran R Guisso ◽  
Sahba Kasiri ◽  
Jun Wu ◽  
Matthew Dear ◽  
...  

SummaryMany adult and most childhood neurological diseases have a genetic basis. CRISPR/Cas9 biotechnology holds great promise in neurological therapy, pending the clearance of major delivery, efficiency and specificity hurdles. We apply CRISPR/Cas9 genome editing in its simplest modality, namely inducing gene sequence disruption, to one adult and one pediatric disease. Adult polyglucosan body disease is a neurodegenerative disease resembling amyotrophic lateral sclerosis. Lafora disease is a severe late childhood onset progressive myoclonus epilepsy. The pathogenic insult in both is formation in the brain of glycogen with overlong branches, which precipitates and accumulates into polyglucosan bodies that drive neuroinflammation and neurodegeneration. We packaged Staphylococcus aureus Cas9 and a guide RNA targeting the glycogen synthase gene Gys1 responsible for brain glycogen branch elongation in AAV9 virus, which we delivered by neonatal intracerebroventricular injection to one mouse model of adult polyglucosan body disease and two mouse models of Lafora disease. This resulted, in all three models, in editing of approximately 17% of Gys1 alleles and a similar extent of reduction of Gys1 mRNA across the brain. The latter led to approximately 50% reductions of GYS1 protein, of abnormal glycogen accumulation and of polyglucosan bodies, as well as corrections of neuroinflammatory markers in all three models. Our work represents proof of principle for virally-delivered CRISPR/Cas9 neurotherapeutics in an adult-onset (adult polyglucosan body) and a childhood-onset (Lafora) neurological diseases.


Author(s):  
Andreia Carvalho ◽  
Joana Nunes ◽  
Ricardo Taipa ◽  
Manuel Melo Pires ◽  
Jorge Pinto Basto ◽  
...  

Author(s):  
Jaspreet Johal ◽  
Ramiro Castro Apolo ◽  
Michael W. Johnson ◽  
Michael R. Persch ◽  
Adam Edwards ◽  
...  

Author(s):  
Paulo Victor Sgobbi Souza ◽  
Bruno Mattos Lombardi Badia ◽  
Igor Braga Farias ◽  
Wladimir Bocca Vieira de Rezende Pinto ◽  
Acary Souza Bulle Oliveira ◽  
...  

2020 ◽  
Vol 7 (11) ◽  
pp. 2186-2198
Author(s):  
Erin E. Chown ◽  
Peixiang Wang ◽  
Xiaochu Zhao ◽  
Justin J. Crowder ◽  
Jordan W. Strober ◽  
...  

2020 ◽  
Vol 3 (1) ◽  
pp. 004-008
Author(s):  
Lawrence Schwartz ◽  
Qing Lu ◽  
Rongzi Liu ◽  
Ruth Kornreich ◽  
Lisa Edelman ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document