Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 260
Filter
1.
JCI Insight ; 9(12)2024 May 14.
Article in English | MEDLINE | ID: mdl-38912588

ABSTRACT

BackgroundGlycogen storage disease type IV (GSD IV) is an ultrarare autosomal recessive disorder that causes deficiency of functional glycogen branching enzyme and formation of abnormally structured glycogen termed polyglucosan. GSD IV has traditionally been categorized based on primary hepatic or neuromuscular involvement, with hepatic GSD IV subclassified as discrete subtypes: classic (progressive) and nonprogressive.MethodsTo better understand the progression of liver disease in GSD IV, we present clinical and histopathology data from 23 patients from around the world and characterized the liver involvement in the Gbe1ys/ys knockin mouse model.ResultsWe propose an alternative to the established subtype-based terminology for characterizing liver disease in GSD IV and recognize 3 tiers of disease severity: (i) "severe progressive" liver disease, (ii) "intermediate progressive" liver disease, and (iii) "attenuated" liver disease. Analysis of liver pathology revealed that risk for liver failure cannot be predicted from liver biopsy findings alone in individuals affected by GSD IV. Moreover, analysis of postmortem liver pathology from an individual who died over 40 years after being diagnosed with nonprogressive hepatic GSD IV in childhood verified that liver fibrosis did not regress. Last, characterization of the liver involvement in a mouse model known to recapitulate the adult-onset neurodegenerative form of GSD IV (Gbe1ys/ys mouse model) demonstrated hepatic disease.ConclusionOur findings challenge the established subtype-based view of GSD IV and suggest that liver disease severity among patients with GSD IV represents a disease continuum.Trial registrationClinicalTrials.gov NCT02683512FundingNone.


Subject(s)
Disease Models, Animal , Glycogen Storage Disease Type IV , Liver , Adolescent , Adult , Animals , Child , Child, Preschool , Female , Humans , Infant , Male , Mice , Middle Aged , Young Adult , Disease Progression , Glycogen Debranching Enzyme System/genetics , Glycogen Debranching Enzyme System/metabolism , Glycogen Storage Disease Type IV/genetics , Glycogen Storage Disease Type IV/pathology , Glycogen Storage Disease Type IV/metabolism , Liver/pathology , Liver/metabolism , Liver Diseases/pathology , Liver Diseases/metabolism
2.
JCI Insight ; 9(11)2024 May 16.
Article in English | MEDLINE | ID: mdl-38753465

ABSTRACT

Glycogen storage disease type III (GSDIII) is a rare metabolic disorder due to glycogen debranching enzyme (GDE) deficiency. Reduced GDE activity leads to pathological glycogen accumulation responsible for impaired hepatic metabolism and muscle weakness. To date, there is no curative treatment for GSDIII. We previously reported that 2 distinct dual AAV vectors encoding for GDE were needed to correct liver and muscle in a GSDIII mouse model. Here, we evaluated the efficacy of rapamycin in combination with AAV gene therapy. Simultaneous treatment with rapamycin and a potentially novel dual AAV vector expressing GDE in the liver and muscle resulted in a synergic effect demonstrated at biochemical and functional levels. Transcriptomic analysis confirmed synergy and suggested a putative mechanism based on the correction of lysosomal impairment. In GSDIII mice livers, dual AAV gene therapy combined with rapamycin reduced the effect of the immune response to AAV observed in this disease model. These data provide proof of concept of an approach exploiting the combination of gene therapy and rapamycin to improve efficacy and safety and to support clinical translation.


Subject(s)
Dependovirus , Disease Models, Animal , Genetic Therapy , Genetic Vectors , Liver , Sirolimus , Animals , Sirolimus/pharmacology , Sirolimus/therapeutic use , Dependovirus/genetics , Genetic Therapy/methods , Mice , Liver/metabolism , Genetic Vectors/genetics , Genetic Vectors/administration & dosage , Muscle, Skeletal/metabolism , Phenotype , Glycogen Debranching Enzyme System/genetics , Glycogen Debranching Enzyme System/metabolism , Humans , Male
3.
J Agric Food Chem ; 72(19): 11041-11050, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38700846

ABSTRACT

The function of polysaccharides is intimately associated with their size, which is largely determined by the processivity of transferases responsible for their synthesis. A tunnel active center architecture has been recognized as a key factor that governs processivity of several glycoside hydrolases (GHs), e.g., cellulases and chitinases. Similar tunnel architecture is also observed in the Limosilactobacillus reuteri 121 GtfB (Lr121 GtfB) α-glucanotransferase from the GH70 family. The molecular element underpinning processivity of these transglucosylases remains underexplored. Here, we report the synthesis of the smallest (α1 → 4)-α-glucan interspersed with linear and branched (α1 → 6) linkages by a novel 4,6-α-glucanotransferase from L. reuteri N1 (LrN1 GtfB) with an open-clefted active center instead of the tunnel structure. Notably, the loop swapping engineering of LrN1 GtfB and Lr121 GtfB based on their crystal structures clarified the impact of the loop-mediated tunnel/cleft structure at the donor subsites -2 to -3 on processivity of these α-glucanotransferases, enabling the tailoring of both product sizes and substrate preferences. This study provides unprecedented insights into the processivity determinants and evolutionary diversification of GH70 α-glucanotransferases and offers a simple route for engineering starch-converting α-glucanotransferases to generate diverse α-glucans for different biotechnological applications.


Subject(s)
Bacterial Proteins , Glucans , Limosilactobacillus reuteri , Glucans/chemistry , Glucans/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Limosilactobacillus reuteri/enzymology , Limosilactobacillus reuteri/genetics , Limosilactobacillus reuteri/chemistry , Catalytic Domain , Glucosyltransferases/chemistry , Glucosyltransferases/genetics , Glucosyltransferases/metabolism , Protein Engineering , Glycogen Debranching Enzyme System/genetics , Glycogen Debranching Enzyme System/metabolism , Glycogen Debranching Enzyme System/chemistry
4.
J Agric Food Chem ; 72(22): 12607-12617, 2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38785045

ABSTRACT

To explore the roles of loops around active pocket in the reuteran type 4,6-α-glucanotransferase (StGtfB) from S. thermophilus, they were individually or simultaneously replaced with those of an isomalto/maltopolysaccharides type 4,6-α-glucanotransferase from L. reuteri. StGtfB with the replaced loops A1, A2 (A1A2) and A1, A2, B (A1A2B), respectively, showed 1.41- and 0.83-fold activities of StGtfB. Two mutants reduced crystallinity and increased starch disorder at 2, 4, and 8 U/g more than StGtfB and increased DP ≤ 5 short branches of starch by 38.01% at 2 U/g, much more than StGtfB by 4.24%. A1A2B modified starches had the lowest retrogradation over 14 days. A1A2 modified starches had the highest percentage of slowly digestible fractions, ranging from 40.32% to 43.34%. StGtfB and its mutants bind substrates by hydrogen bonding and van der Waals forces at their nonidentical amino acid residues, suggesting that loop replacement leads to a different conformation and changes activity and product structure.


Subject(s)
Bacterial Proteins , Glycogen Debranching Enzyme System , Streptococcus thermophilus , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Biocatalysis , Catalytic Domain , Glycogen Debranching Enzyme System/chemistry , Glycogen Debranching Enzyme System/metabolism , Glycogen Debranching Enzyme System/genetics , Kinetics , Starch/metabolism , Starch/chemistry , Streptococcus thermophilus/enzymology , Streptococcus thermophilus/genetics , Streptococcus thermophilus/chemistry , Streptococcus thermophilus/metabolism , Substrate Specificity
5.
J Biochem ; 176(2): 109-117, 2024 Jul 31.
Article in English | MEDLINE | ID: mdl-38498909

ABSTRACT

Glycogen debranching enzyme is a single polypeptide with distinct catalytic sites for 4-α-glucanotransferase and amylo-α-1,6-glucosidase. To allow phosphorylase to degrade the inner tiers of highly branched glycogen, 4-α-glucanotransferase converts the phosphorylase-limit biantennary branch G-G-G-G-(G-G-G-G↔)G-G- (G: d-glucose, hyphens: α-1,4-linkages; double-headed arrow: α-1,6-linkage) into the G-G-G-G-(G↔)G-G- residue, which is then subjected to amylo-α-1,6-glucosidase to release the remaining G↔ residue. However, while the essential side-chain structure of the 4-α-glucanotransferase donor substrate has been determined to be the G-G-G-G↔ residue (Watanabe, Y., et al. (2008) J. Biochem.143, 435-440), its essential main-chain structure remains to be investigated. In this study, we probed the 4-α-glucanotransferase donor-binding region using novel fluorogenic dextrins Gm-(G4↔)G-Gn-F (F: 1-deoxy-1-[(2-pyridyl)amino]-d-glucitol) and maltohexaose (G6) as the donor and acceptor substrates, respectively. 4-α-Glucanotransferase exhibited maximum activity towards G4-(G4↔)G-F and G4-(G4↔)G-G-F, indicating that recognition of the G4-(G4↔)G-moiety was essential for full enzyme function. Notably, when the 4-α-glucanotransferase activity towards G4-(G4↔)G-G-F was taken as unity, those towards nonbranching dextrins were < 0.001. This indicated that the disproportionation activities towards maltooligosaccharides (Gm) are abnormal behaviours of 4-α-glucanotransferase. Notably, however, these activities have been traditionally measured to identify the 4-α-glucanotransferase mutations causing glycogen storage disease type III. This study provides a basis for more accurate identification.


Subject(s)
Dextrins , Glycogen Debranching Enzyme System , Glycogen Debranching Enzyme System/metabolism , Glycogen Debranching Enzyme System/chemistry , Glycogen Debranching Enzyme System/genetics , Dextrins/metabolism , Dextrins/chemistry , Substrate Specificity
6.
J Clin Invest ; 134(2)2024 Jan 16.
Article in English | MEDLINE | ID: mdl-38015640

ABSTRACT

Glycogen storage disease type III (GSDIII) is a rare inborn error of metabolism affecting liver, skeletal muscle, and heart due to mutations of the AGL gene encoding for the glycogen debranching enzyme (GDE). No curative treatment exists for GSDIII. The 4.6 kb GDE cDNA represents the major technical challenge toward the development of a single recombinant adeno-associated virus-derived (rAAV-derived) vector gene therapy strategy. Using information on GDE structure and molecular modeling, we generated multiple truncated GDEs. Among them, an N-terminal-truncated mutant, ΔNter2-GDE, had a similar efficacy in vivo compared with the full-size enzyme. A rAAV vector expressing ΔNter2-GDE allowed significant glycogen reduction in heart and muscle of Agl-/- mice 3 months after i.v. injection, as well as normalization of histology features and restoration of muscle strength. Similarly, glycogen accumulation and histological features were corrected in a recently generated Agl-/- rat model. Finally, transduction with rAAV vectors encoding ΔNter2-GDE corrected glycogen accumulation in an in vitro human skeletal muscle cellular model of GSDIII. In conclusion, our results demonstrated the ability of a single rAAV vector expressing a functional mini-GDE transgene to correct the muscle and heart phenotype in multiple models of GSDIII, supporting its clinical translation to patients with GSDIII.


Subject(s)
Glycogen Debranching Enzyme System , Glycogen Storage Disease Type III , Humans , Mice , Rats , Animals , Glycogen Storage Disease Type III/genetics , Glycogen Storage Disease Type III/therapy , Glycogen Debranching Enzyme System/genetics , Muscle, Skeletal/metabolism , Glycogen/metabolism , Transgenes
7.
N Biotechnol ; 79: 39-49, 2024 Mar 25.
Article in English | MEDLINE | ID: mdl-38097138

ABSTRACT

4-α-glucanotransferases (4αGTs, EC 2.4.1.25) from glycoside hydrolase family 77 (GH77) catalyze chain elongation of starch amylopectin chains and can be utilized to structurally modify starch to tailor its gelation properties. The potential relationship between the structural design of 4αGTs and functional starch modification is unknown. Here, family GH77 was mined in silico for enzyme candidates based on sub-grouping guided by Conserved Unique Peptide Patterns (CUPP) bioinformatics categorization. From + 12,000 protein sequences a representative set of 27 4αGTs, representing four different domain architectures, different bacterial origins and diverse CUPP groups, was selected for heterologous expression and further study. Most of the enzymes catalyzed starch modification, but their efficacies varied substantially. Five of the 4αGTs were characterized in detail, and their action was compared to that of the industrial benchmark enzyme, Tt4αGT (CUPP 77_1.2), from Thermus thermophilus. Reaction optima of the five 4αGTs ranged from ∼40-60 °C and pH 7.3-9.0. Several were stable for a minimum 4 h at 70 °C. Domain architecture type A proteins, consisting only of a catalytic domain, had high thermal stability and high starch modification ability. All five novel 4αGTs (and Tt4αGT) induced enhanced gelling of potato starch. One, At4αGT from Azospirillum thermophilum (CUPP 77_2.4), displayed distinct starch modifying abilities, whereas T24αGT from Thermus sp. 2.9 (CUPP 77_1.2) modified the starch similarly to Tt4αGT, but slightly more effectively. T24αGT and At4αGT are thus interesting candidates for industrial starch modification. A model is proposed to explain the link between the 4αGT induced molecular modifications and macroscopic starch gelation.


Subject(s)
Glycogen Debranching Enzyme System , Solanum tuberosum , Solanum tuberosum/metabolism , Glycoside Hydrolases , Starch , Glycogen Debranching Enzyme System/genetics , Glycogen Debranching Enzyme System/chemistry , Glycogen Debranching Enzyme System/metabolism , Peptides
8.
Molecules ; 28(3)2023 Jan 30.
Article in English | MEDLINE | ID: mdl-36770986

ABSTRACT

A broad range of enzymes are used to modify starch for various applications. Here, a thermophilic 4-α-glucanotransferase from Thermoproteus uzoniensis (TuαGT) is engineered by N-terminal fusion of the starch binding domains (SBDs) of carbohydrate binding module family 20 (CBM20) to enhance its affinity for granular starch. The SBDs are N-terminal tandem domains (SBDSt1 and SBDSt2) from Solanum tuberosum disproportionating enzyme 2 (StDPE2) and the C-terminal domain (SBDGA) of glucoamylase from Aspergillus niger (AnGA). In silico analysis of CBM20s revealed that SBDGA and copies one and two of GH77 DPE2s belong to well separated clusters in the evolutionary tree; the second copies being more closely related to non-CAZyme CBM20s. The activity of SBD-TuαGT fusions increased 1.2-2.4-fold on amylose and decreased 3-9 fold on maltotriose compared with TuαGT. The fusions showed similar disproportionation activity on gelatinised normal maize starch (NMS). Notably, hydrolytic activity was 1.3-1.7-fold elevated for the fusions leading to a reduced molecule weight and higher α-1,6/α-1,4-linkage ratio of the modified starch. Notably, SBDGA-TuαGT and-SBDSt2-TuαGT showed Kd of 0.7 and 1.5 mg/mL for waxy maize starch (WMS) granules, whereas TuαGT and SBDSt1-TuαGT had 3-5-fold lower affinity. SBDSt2 contributed more than SBDSt1 to activity, substrate binding, and the stability of TuαGT fusions.


Subject(s)
Glycogen Debranching Enzyme System , Starch , Starch/chemistry , Interleukin-1 Receptor-Like 1 Protein , Glycogen Debranching Enzyme System/genetics , Amylopectin
9.
Enzyme Microb Technol ; 164: 110175, 2023 03.
Article in English | MEDLINE | ID: mdl-36516732

ABSTRACT

4,6 α-Glucanotransferase (4,6-α-GTase) and 4,3 α-glucanotransferases (4,3-α-GTase) produced by Lactic Acid Bacteria (LAB) in the GH70 enzyme family have become important due to their catalytic effect on starch and maltodextrins. Their high level of production is necessary for their application at industrial scale. In this respect, both enzymes were expressed extracellularly using Lactococcus lactis as GRAS host. 4,6-α-GTase and 4,3-α-GTase genes from Limosilactobacillus reuteri E81 and Limosilactobacillus fermentum PFC282 respectively were transformed into the plasmid pLEB124 vector having the signal peptide usp45 under the P45 continuous promoter and successfully expressed in Lactococcus lactis MG1363. Western blot screening showed that the relevant enzymes were able to be successfully secreted extracellularly. The Vmax and Km of 4,6-α-GTase were 2.58 µmol min-1 and 0054 mg min-1 whereas 3369 µmol min-1 and 0032 mg min-1 for 4,3-α-GTase respectively. NMR analysis demonstrated the formation of new bonds within the corresponding enzymes. Also, both enzymes were active on maltose, maltoheptaose, maltohexaose and starch and produced malto-oligosaccarides observed by TLC analysis. In conclusion, this study demonstrated first time the extracellular production of 4,6-α-GTase and 4,3-α-GTase with GRAS status that can be useful for starch retrogradation delay and glycaemic index reduction.


Subject(s)
Glycogen Debranching Enzyme System , Lactococcus lactis , Limosilactobacillus reuteri , Lactococcus lactis/genetics , Starch , Glycogen Debranching Enzyme System/genetics , Glycogen Debranching Enzyme System/chemistry , Glucosyltransferases
10.
Neuropathol Appl Neurobiol ; 49(1): e12865, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36456471

ABSTRACT

AIMS: Adult polyglucosan body disease (APBD) is a progressive neurogenetic disorder caused by 1,4-alpha-glucan branching enzyme 1 (GBE1) mutation with an accumulation of polyglucosan bodies (PBs) in the central and peripheral nervous systems as a pathological hallmark. Here, we report two siblings in a family with a GBE1 mutation with prominent frontotemporal lobar degeneration with TAR DNA-binding protein 43 (FTLD-TDP) and ageing-related tau astrogliopathy (ARTAG) copathologies with PBs in the central nervous system. METHODS: Whole-genome sequencing (WGS) followed by Sanger sequencing (SS) was performed on three affected and two unaffected siblings in a pedigree diagnosed with familial frontotemporal dementia. Out of the affected siblings, autopsies were conducted on two cases, and brain samples were used for biochemical and histological analyses. Brain sections were stained with haematoxylin and eosin and immunostained with antibodies against ubiquitin, tau, amyloid ß, α-synuclein, TDP-43 and fused in sarcoma (FUS). RESULTS: A novel single nucleotide deletion in GBE1, c.1280delG, was identified, which is predicted to result in a reading frameshift, p.Gly427Glufs*9. This variant segregated with disease in the family, is absent from population databases and is predicted to cause loss of function, a known genetic mechanism for APBD. The affected siblings showed a greater than 50% decrease in GBE protein levels. Immunohistochemical analysis revealed widespread FTLD-TDP (type A) and ARTAG pathologies as well as PBs in the brains of two affected siblings for whom an autopsy was performed. CONCLUSIONS: This is the first report of a family with several individuals with a FTD clinical phenotype and underlying copathologies of APBD, FTLD-TDP and ARTAG with a segregating GBE1 loss-of-function mutation in affected siblings. The finding of copathologies of APBD and FTLD-TDP suggests these processes may share a disease mechanism resulting from this GBE1 mutation.


Subject(s)
1,4-alpha-Glucan Branching Enzyme , Frontotemporal Dementia , Frontotemporal Lobar Degeneration , Glycogen Debranching Enzyme System , Humans , Frontotemporal Dementia/pathology , 1,4-alpha-Glucan Branching Enzyme/genetics , 1,4-alpha-Glucan Branching Enzyme/metabolism , Amyloid beta-Peptides/metabolism , Frontotemporal Lobar Degeneration/pathology , Brain/pathology , Mutation , DNA-Binding Proteins/metabolism , tau Proteins/metabolism , Glycogen Debranching Enzyme System/genetics , Glycogen Debranching Enzyme System/metabolism
11.
J Microbiol ; 60(4): 375-386, 2022 Apr.
Article in English | MEDLINE | ID: mdl-35157220

ABSTRACT

Vibrio vulnificus MO6-24/O has three genes annotated as debranching enzymes or pullulanase genes. Among them, the gene encoded by VVMO6_03032 (vvde1) shares a higher similarity at the amino acid sequence level to the glycogen debranching enzymes, AmyX of Bacillus subtilis (40.5%) and GlgX of Escherichia coli (55.5%), than those encoded by the other two genes. The vvde1 gene encoded a protein with a molecular mass of 75.56 kDa and purified Vvde1 efficiently hydrolyzed glycogen and pullulan to shorter chains of maltodextrin and maltotriose (G3), respectively. However, it hydrolyzed amylopectin and soluble starch far less efficiently, and ß-cyclodextrin (ß-CD) only rarely. The optimal pH and temperature of Vvde1 was 6.5 and 25°C, respectively. Vvde1 was a cold-adapted debranching enzyme with more than 60% residual activity at 5°C. It could maintain stability for 2 days at 25°C and 1 day at 35°C, but it destabilized drastically at 40°C. The Vvde1 activity was inhibited considerably by Cu2+, Hg2+, and Zn2+, while it was slightly enhanced by Co2+, Ca2+, Ni2+, and Fe2+. The vvde1 knock-out mutant accumulated more glycogen than the wild-type in media supplemented with 1.0% maltodextrin; however, the side chain length distribution of glycogen was similar to that of the wild-type except G3, which was much more abundant in the mutant. Therefore, Vvde1 seemed to debranch glycogen with the degree of polymerization 3 (DP3) as the specific target branch length. Virulence of the pathogen against Caenorhabditis elegans was attenuated significantly by the vvde1 mutation. These results suggest that Vvde1 might be a unique glycogen debranching enzyme that is involved in both glycogen utilization and shaping of glycogen molecules, and contributes toward virulence of the pathogen.


Subject(s)
Glycogen Debranching Enzyme System , Vibrio vulnificus , Amylopectin/metabolism , Glycogen/metabolism , Glycogen Debranching Enzyme System/chemistry , Glycogen Debranching Enzyme System/genetics , Glycogen Debranching Enzyme System/metabolism , Vibrio vulnificus/metabolism , Virulence/genetics
12.
J Agric Food Chem ; 70(6): 1952-1961, 2022 Feb 16.
Article in English | MEDLINE | ID: mdl-35129339

ABSTRACT

Limosilactobacillus reuteri 121 4,6-α-glucanotransferase (Lr121 4,6-α-GTase), belonging to the glycosyl hydrolase (GH) 70 GtfB subfamily, converts starch and maltodextrins into linear isomalto/malto polysaccharides (IMMPs) with consecutive (α1 → 6) linkages. The recent elucidation of its crystal structure allowed identification and analysis of further structural features that determine its reaction and product specificity. Herein, sequence alignments between GtfB enzymes with different product linkage specificities (4,6-α-GTase and 4,3-α-GTase) identified amino acid residues in GH70 homology motifs, which may be critical for reaction and product specificity. Based on these alignments, four Lr121 GtfB-ΔN mutants (I1020M, S1057P, H1056G, and Q1126I) were constructed. Compared to wild-type Lr121 GtfB-ΔN, mutants S1057P and Q1126I had considerably improved catalytic efficiencies. Mutants H1056G and Q1126I showed a 9% decrease and an 11% increase, respectively, in the ratio of (α1 → 6) over (α1 → 4) linkages in maltodextrin-derived products. A change in linkage type (e.g., (α1 → 6) linkages to (α1 → 3) linkages) was not observed. The possible functional roles of these Lr121 GtfB-ΔN residues located around the acceptor substrate-binding subsites are discussed. The results provide new insights into structural determinants of the reaction and product specificity of Lr121 GtfB 4,6-α-GTase.


Subject(s)
Glycogen Debranching Enzyme System , Limosilactobacillus reuteri , Amino Acids , Glycogen Debranching Enzyme System/genetics , Limosilactobacillus reuteri/genetics , Limosilactobacillus reuteri/metabolism , Mutation , Starch , Substrate Specificity
13.
World J Microbiol Biotechnol ; 38(2): 36, 2022 Jan 07.
Article in English | MEDLINE | ID: mdl-34993677

ABSTRACT

4α-Glucanotransferase (4α-GTase) is unique in its ability to form cyclic oligosaccharides, some of which are of industrial importance. Generally, low amount of enzymes is produced by or isolated from their natural sources: animals, plants, and microorganisms. Heterologous expressions of these enzymes, in an attempt to increase their production for applicable uses, have been widely studied since 1980s; however, the expressions are mostly performed in the prokaryotic bacteria, mostly Escherichia coli. Site-directed mutagenesis has added more value to these expressed enzymes to display the desired properties beneficial for their applications. The search for further suitable properties for food application leads to an extended research in expression by another group of host organism, the generally-recognized as safe host including the Bacillus and the eukaryotic yeast systems. Herein, our review focuses on two types of 4α-GTase: the cyclodextrin glycosyltransferase and amylomaltase. The updated studies on the general structure and properties of the two enzymes with emphasis on heterologous expression, mutagenesis for property improvement, and their industrial applications are provided.


Subject(s)
Glycogen Debranching Enzyme System/genetics , Glycogen Debranching Enzyme System/metabolism , Bacillus/enzymology , Bacillus/genetics , Bacteria/enzymology , Escherichia coli/enzymology , Escherichia coli/genetics , Escherichia coli/metabolism , Glucosyltransferases/genetics , Glucosyltransferases/metabolism , Mutagenesis, Site-Directed , Oligosaccharides , Yeasts/enzymology , Yeasts/genetics , Yeasts/metabolism
14.
Hum Mutat ; 43(1): 16-29, 2022 01.
Article in English | MEDLINE | ID: mdl-34633740

ABSTRACT

Autism spectrum disorders (ASD) are neurodevelopmental disorders with an estimated heritability of >60%. Family-based genetic studies of ASD have generally focused on multiple small kindreds, searching for de novo variants of major effect. We hypothesized that molecular genetic analysis of large multiplex families would enable the identification of variants of milder effects. We studied a large multigenerational family of European ancestry with multiple family members affected with ASD or the broader autism phenotype (BAP). We identified a rare heterozygous variant in the gene encoding 1,4-ɑ-glucan branching enzyme 1 (GBE1) that was present in seven of seven individuals with ASD, nine of ten individuals with the BAP, and none of four tested unaffected individuals. We genotyped a community-acquired cohort of 389 individuals with ASD and identified three additional probands. Cascade analysis demonstrated that the variant was present in 11 of 13 individuals with familial ASD/BAP and neither of the two tested unaffected individuals in these three families, also of European ancestry. The variant was not enriched in the combined UK10K ASD cohorts of European ancestry but heterozygous GBE1 deletion was overrepresented in large ASD cohorts, collectively suggesting an association between GBE1 and ASD.


Subject(s)
1,4-alpha-Glucan Branching Enzyme , Autism Spectrum Disorder , Glycogen Debranching Enzyme System , 1,4-alpha-Glucan Branching Enzyme/genetics , Autism Spectrum Disorder/genetics , Exome , Genetic Predisposition to Disease , Glucans , Glycogen Debranching Enzyme System/genetics , Humans
15.
J Agric Food Chem ; 69(48): 14680-14688, 2021 Dec 08.
Article in English | MEDLINE | ID: mdl-34845909

ABSTRACT

4,6-α-Glucanotransferases (4,6-α-GTs) convert amylose V into two types of differently structured products: a linear product connected by continuous α,1 → 6 bonds, such as isomalto/malto-polysaccharide (IMMP), and a highly branched product connected by alternating α,1 → 4 and α,1 → 6 bonds, such as reuteran-like polysaccharide (RLP). The synthesis process of 4,6-α-GT products is unclear, and exploring this process is significant for producing dietary fibers with potential applications. This study identified and expressed Geobacillus sp. 12AMOR1 GtfD-ΔC and Bacillus sporothermodurans GtfC-ΔC. After characterizing their products through 1H NMR and enzymatic fingerprinting, we found that GtfD-ΔC synthesized RLP with 29% α,1 → 6 bonds, and GtfC-ΔC synthesized IMMP with 71% α,1 → 6 bonds. The maltoheptaose incubation experiment showed different chain-length transfer patterns of two 4,6-α-GTs, GtfC-ΔC and GtfD-ΔC, transferring single and multiple glucose residues in each transglycosylation reaction, respectively. Site-directed mutagenesis confirmed that positions S345 and I347 influence the product structure of GtfC-ΔC, and the S345T/I347V mutation changed the GtfC-ΔC product to a linear product connected by alternating α,1 → 4 and α,1 → 6 bonds (pullulan-like polysaccharide) and altered the chain-length transfer pattern of GtfC-ΔC. We proposed that different chain-length transfer patterns between GtfD-ΔC and GtfC-ΔC may explain their differences in product structures. These findings are significant for obtaining the desired dietary fiber by engineering 4,6-α-GT.


Subject(s)
Amino Acids , Glycogen Debranching Enzyme System , Bacillus , Glucosyltransferases , Glycogen Debranching Enzyme System/genetics , Mutation
16.
J Agric Food Chem ; 69(44): 13235-13245, 2021 Nov 10.
Article in English | MEDLINE | ID: mdl-34708648

ABSTRACT

GtfB-type α-glucanotransferase enzymes from glycoside hydrolase family 70 (GH70) convert starch substrates into α-glucans that are of interest as food ingredients with a low glycemic index. Characterization of several GtfBs showed that they differ in product- and substrate specificity, especially with regard to branching, but structural information is limited to a single GtfB, preferring mostly linear starches and featuring a tunneled binding groove. Here, we present the second crystal structure of a 4,6-α-glucanotransferase (Limosilactobacillus reuteri NCC 2613) and an improved homology model of a 4,3-α-glucanotransferase GtfB (L. fermentum NCC 2970) and show that they are able to convert both linear and branched starch substrates. Compared to the previously described GtfB structure, these two enzymes feature a much more open binding groove, reminiscent of and evolutionary closer to starch-converting GH13 α-amylases. Sequence analysis of 287 putative GtfBs suggests that only 20% of them are similarly "open" and thus suitable as broad-specificity starch-converting enzymes.


Subject(s)
Glycogen Debranching Enzyme System , Starch , Glucans , Glycogen Debranching Enzyme System/genetics , Glycoside Hydrolases
17.
Biomolecules ; 11(9)2021 09 09.
Article in English | MEDLINE | ID: mdl-34572549

ABSTRACT

Amylomaltases (4-α-glucanotransferases, E.C. 2.4.1.25) are enzymes which can perform a double-step catalytic process, resulting in a transglycosylation reaction. They hydrolyse glucosidic bonds of α-1,4'-d-glucans and transfer the glucan portion with the newly available anomeric carbon to the 4'-position of an α-1,4'-d-glucan acceptor. The intramolecular reaction produces a cyclic α-1,4'-glucan. Amylomaltases can be found only in prokaryotes, where they are involved in glycogen degradation and maltose metabolism. These enzymes are being studied for possible biotechnological applications, such as the production of (i) sugar substitutes; (ii) cycloamyloses (molecules larger than cyclodextrins), which could potentially be useful as carriers and encapsulating agents for hydrophobic molecules and also as effective protein chaperons; and (iii) thermoreversible starch gels, which could be used as non-animal gelatin substitutes. Extremophilic prokaryotes have been investigated for the identification of amylomaltases to be used in the starch modifying processes, which require high temperatures or extreme conditions. The aim of this article is to present an updated overview of studies on amylomaltases from extremophilic Bacteria and Archaea, including data about their distribution, activity, potential industrial application and structure.


Subject(s)
Archaea/enzymology , Bacteria/enzymology , Extremophiles/enzymology , Glycogen Debranching Enzyme System/metabolism , Amino Acid Sequence , Glycogen Debranching Enzyme System/chemistry , Glycogen Debranching Enzyme System/genetics , Models, Molecular , Mutation/genetics
18.
J Agric Food Chem ; 69(34): 9859-9868, 2021 Sep 01.
Article in English | MEDLINE | ID: mdl-34427087

ABSTRACT

Starch-acting α-glucanotransferase enzymes are of great interest for applications in the food industry. In previous work, we have characterized various 4,6- and 4,3-α-glucanotransferases of the glycosyl hydrolase (GH) family 70 (subfamily GtfB), synthesizing linear or branched α-glucans. Thus far, GtfB enzymes have only been identified in mesophilic Lactobacilli. Database searches showed that related GtfC enzymes occur in Gram-positive bacteria of the genera Exiguobacterium, Bacillus, and Geobacillus, adapted to growth at more extreme temperatures. Here, we report characteristics of the Geobacillus sp. 12AMOR1 GtfC enzyme, with an optimal reaction temperature of 60 °C and a melting temperature of 68 °C, allowing starch conversions at relatively high temperatures. This thermostable 4,6-α-glucanotransferase has a novel product specificity, cleaving off predominantly maltose units from amylose, attaching them with an (α1 → 6)-linkage to acceptor substrates. In fact, this GtfC represents a novel maltogenic α-amylase. Detailed structural characterization of its starch-derived α-glucan products revealed that it yielded a unique polymer with alternating (α1 → 6)/(α1 → 4)-linked glucose units but without branches. Notably, this Geobacillus sp. 12AMOR1 GtfC enzyme showed clear antistaling effects in bread bakery products.


Subject(s)
Geobacillus , Glycogen Debranching Enzyme System , Bread , Geobacillus/genetics , Glucans , Glycogen Debranching Enzyme System/genetics
19.
Mol Genet Genomic Med ; 9(10): e1779, 2021 10.
Article in English | MEDLINE | ID: mdl-34405590

ABSTRACT

BACKGROUND: The condition of uniparental disomy (UPD) occurs when an individual inherits two copies of a chromosome, or part of a chromosome, from one parent. Most cases of uniparental heterodisomy (UPhD) do not cause diseases, whereas cases of uniparental isodisomy (UPiD), while rare, may be pathogenic. Theoretically, UPiD may cause rare genetic diseases in a homozygous recessive manner. METHODS: A 4-year-old girl presented with congenital hearing loss, developmental delay, hepatomegaly, and other clinical features. She and her parents were genetically tested using trio whole exome sequencing (Trio-WES) and copy number variation sequencing (CNV-seq). In addition, we built a structural model to further examine the pathogenicity of the UPiD variants. RESULTS: Trio-WES identified a paternal UPiD in chromosome 1, and two homozygous pathogenic variants AGL c.4284T>G/p.Tyr1428* and USH2A c.6528T>A/p.Tyr2176* in the UPiD region. We further analyzed the pathogenicity of these two variations. The patient was diagnosed with Usher syndrome type 2A (USH2A) and glycogen storage disease type III (GSD3). CONCLUSIONS: Our study reports a rare case of a patient carrying two pathogenic variants of different genes caused by paternal UPiD, supporting the potential application of Trio-WES in detecting and facilitating the diagnosis of UPD.


Subject(s)
Chromosomes, Human, Pair 1 , Glycogen Storage Disease Type III/complications , Glycogen Storage Disease Type III/diagnosis , Paternal Inheritance , Uniparental Disomy , Usher Syndromes/complications , Usher Syndromes/diagnosis , Adult , Biomarkers , Child, Preschool , DNA Copy Number Variations , Female , Genetic Association Studies , Genetic Predisposition to Disease , Glycogen Debranching Enzyme System/chemistry , Glycogen Debranching Enzyme System/genetics , Glycogen Storage Disease Type III/etiology , Glycogen Storage Disease Type III/metabolism , Humans , Male , Middle Aged , Models, Molecular , Pedigree , Sequence Analysis, DNA , Structure-Activity Relationship , Usher Syndromes/etiology , Usher Syndromes/metabolism , Exome Sequencing
20.
J Clin Invest ; 131(12)2021 06 15.
Article in English | MEDLINE | ID: mdl-33945503

ABSTRACT

BACKGROUNDDeciphering the function of the many genes previously classified as uncharacterized open reading frame (ORF) would complete our understanding of a cell's function and its pathophysiology.METHODSWhole-exome sequencing, yeast 2-hybrid and transcriptome analyses, and molecular characterization were performed in this study to uncover the function of the C2orf69 gene.RESULTSWe identified loss-of-function mutations in the uncharacterized C2orf69 gene in 8 individuals with brain abnormalities involving hypomyelination and microcephaly, liver dysfunction, and recurrent autoinflammation. C2orf69 contains an N-terminal signal peptide that is required and sufficient for mitochondrial localization. Consistent with mitochondrial dysfunction, the patients showed signs of respiratory chain defects, and a CRISPR/Cas9-KO cell model of C2orf69 had similar respiratory chain defects. Patient-derived cells revealed alterations in immunological signaling pathways. Deposits of periodic acid-Schiff-positive (PAS-positive) material in tissues from affected individuals, together with decreased glycogen branching enzyme 1 (GBE1) activity, indicated an additional impact of C2orf69 on glycogen metabolism.CONCLUSIONSOur study identifies C2orf69 as an important regulator of human mitochondrial function and suggests that this gene has additional influence on other metabolic pathways.


Subject(s)
Glycogen/metabolism , Loss of Function Mutation , Microcephaly/metabolism , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Open Reading Frames , Animals , Cell Line , Glycogen/genetics , Glycogen Debranching Enzyme System/genetics , Glycogen Debranching Enzyme System/metabolism , Humans , Mice , Mice, Knockout , Microcephaly/genetics , Mitochondria/genetics , Mitochondrial Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL