Skip to main content
Erschienen in: Journal of Inherited Metabolic Disease 4/2016

Open Access 30.05.2016 | SSIEM 2015

From structural biology to designing therapy for inborn errors of metabolism

verfasst von: Wyatt W. Yue

Erschienen in: Journal of Inherited Metabolic Disease | Ausgabe 4/2016

Abstract

At the SSIEM Symposium in Istanbul 2010, I presented an overview of protein structural approaches in the study of inborn errors of metabolism (Yue and Oppermann 2011). Five years on, the field is going strong with new protein structures, uncovered catalytic functions and novel chemical matters for metabolic enzymes, setting the stage for the next generation of drug discovery. This article aims to update on recent advances and lessons learnt on inborn errors of metabolism via the protein-centric approach, citing examples of work from my group, collaborators and co-workers that cover diverse pathways of transsulfuration, cobalamin and glycogen metabolism. Taking into consideration that many inborn errors of metabolism result in the loss of enzyme function, this presentation aims to outline three key principles that guide the design of small molecule therapy in this technically challenging field: (1) integrating structural, biochemical and cell-based data to evaluate the wide spectrum of mutation-driven enzyme defects in stability, catalysis and protein-protein interaction; (2) studying multi-domain proteins and multi-protein complexes as examples from nature, to learn how enzymes are activated by small molecules; (3) surveying different regions of the enzyme, away from its active site, that can be targeted for the design of allosteric activators and inhibitors.
Hinweise
Communicated by: Avihu Boneh
Presented at the Annual Symposium of the Society for the Study of Inborn Errors of Metabolism, Lyon, France, September 1-4, 2015.

Introduction: Structure biology of metabolic enzymes is advancing

The field of structural biology has come a long way since the first protein crystal structure, that of sperm whale myoglobin, in 1958. The Protein Data Bank, a public repository of structural data determined from x-ray crystallography, nuclear magnetic resonance as well as electron microscopy (EM), has recently reached its 100,000th entry in 2014. Protein structure determination has nowadays become a streamlined process replete with technological advances that allow automation, parallelization and miniaturization of constituent steps (Su et al 2015). Examples of such pioneering development include heterologous expression systems to generate multi-component protein complexes, automated chromatography platforms for purification towards homogeneity, remedial strategies for crystallization of protein samples, as well as the implementation of high quality x-ray and electron diffraction sources worldwide.
In modern days, the term ‘structural biology’ is more appropriately coined to cover the toolkit of biophysical and biochemical, in addition to structural methods, that can probe the oligomeric assembly (e.g. size exclusion, analytical ultracentrifugation), conformational changes (e.g. spectroscopy), enzyme catalysis (e.g. Michaelis-Menton kinetics), as well as ligand/protein binding (e.g. isothermal titration calorimetry, surface plasmon resonance) features of proteins. Structural biology has therefore been instrumental in delineating the molecular functions and mechanisms of diverse target proteins, including the hundreds of human metabolic enzymes associated with inborn errors of metabolism (IEMs) (Kang and Stevens 2009; Yue and Oppermann 2011). Adopting a family-wide and pathway-wide approach in target protein selection (Osman and Edwards 2014), the Structural Genomics Consortium has to date determined nearly 50 structures of human IEM-linked metabolic enzymes (Table 1), as a first step towards developing a mechanistic understanding and therapeutic advancement of these rare genetic diseases. Our recent addition to this IEM repertoire includes structural determination of six protein players (MUT, MMAA, MCEE, MMACHC, MMADHC, MTR) involved in the different stages of the processing, trafficking and assembly of the vitamin B12 cofactor to its two destination enzymes (Froese et al 2010; Froese et al 2012; Froese et al 2015a, b). Inherited defect in each gene of this intricate B12 processing pathway gives rise to the metabolic disorders of methylmalonic aciduria and homocystinuria (Froese and Gravel 2010).
Table 1
Crystal structures of metabolic enzymes that are associated with inborn errors of metabolism, as determined by the SGC Oxford group of Metabolic and Rare Diseases and deposited in the public domain. Unless specified otherwise, all structures listed are of human proteins
Target name and description
Genbank ID
PDB IDs
Length
Structure region
Associated disorders (OMIM)
AASS
aminoadipate-semialdehyde synthase
13027640
to be deposited
926
455–926
Hyperlysinemia type I (238700)
ACACA
acetyl-CoA carboxylase alpha
38679960
2YL2, 4ASI
2383
118–654 (2YL2), 1608–2375 (4ASI)
ACACA deficiency (613933)
ACADS
acyl-CoA dehydrogenase, short chain
4557233
2VIG
412
30–412
ACADS deficiency (201470)
ACADSB
acyl-CoA dehydrogenase, short/branched chain
4501859
2JIF
432
52–432
ACADSB deficiency 2-methylbutyryl glycinuria (610006)
ACADVL
acyl-CoA dehydrogenase, very long chain
4557235
2UXW
655
72–655
ACADVL deficiency (201475)
ADA
adenosine deaminase
47078295
3IAR
363
5–363
Severe combined immunodeficiency (102700)
ALDH7A1
aldehyde dehydrogenase 7 family, member A1
4557343
2J6L
511
1–499
Pyridoxine-dependent epilepsy (266100)
CBS
cystathionine-beta-synthase
4557415
4UUU, 4COO
551
406–547 (4UUU), 1–551 (4COO)
homocystinuria due to CBS deficiency (236200)
CRYBB3
crystallin, beta B3
4758074
3QK3
211
21–199
Cataract congenital nuclear autosomal recessive type 2 (609741)
ENO3
enolase 3
153267427
2XSX
434
1–434
Glycogen storage disorder type 13 (612932)
FH
fumarate hydratase
19743875
3EO4
510
44–510
Fumarase deficiency (606812), MCUL (150800), HLRCC (605839)
FKBP14
peptidyl-prolyl cis-trans isomerase FKBP14
8923659
4DIP
211
19–140
Ehlers-Danlos syndrome types VIA and VIB (614557)
GALT
galactose-1-phosphate uridylyltransferase
22165416
5IN3
379
1–379
Classical galactosemia (230400)
GBE1
glucan (1,4-alpha-), branching enzyme 1
189458812
5CLT, 4BZY, 5CLW
702
38–700
Glycogen storage disorder type IV (232500), Adult polyglucosan body disease (263570)
GLRX5
glutaredoxin 5
42516576
2WEM, 2WUL
157
35–150
Anemia sideroblastic pyridoxine-refractory (205950)
GMDS
GDP-mannose 4,6-dehydratase
9087147
1T2A
372
23–372
Cerebellar vermis hypoplasia (602884)
GPD1L
glycerol-3-phosphate dehydrogenase 1-like protein
24307999
2PLA
351
1–349
Brugada syndrome type 2 (611777)
GYG1
glycogenin 1
12652581
3T7O,3T7O,3U2V,3U2U,3U2X,3U2T,3RMW,3RMV,3QVB,3U2W,3Q4S,3T7N,3T7M
350
1–262
Glycogen storage disease type 15 (613507)
HIBCH
3-hydroxyisobutyryl-CoA hydrolase
37594471
3BPT
386
42–386
HIBCH deficiency (250620)
HMGCS2
3-hydroxy-3-methylglutaryl-CoA synthase 2
5031751
2V4W, 2WYA
508
51–508
HMGCS deficiency (605911)
HPD
4-hydroxyphenylpyruvate dioxygenase
4504477
3ISQ
393
8–393
Tyrosinemia type 3 (276710), hawkinsinuria (140350)
HPGD
hydroxyprostaglandin dehydrogenase
1203982
2GDZ
266
3–256
Primary hypertrophic osteoathropathy (259100), isolated congenital nail clubbing (119900)
HSD17B10
hydroxyacyl-CoA dehydrogenase, type II
4758504
2O23
261
1–261
2-methyl-3-hydroxybutyryl-CoA dehydrogenase deficiency (300438), mental retardation syndromic X-linked type 10 (300220)
HSD17B4
hydroxysteroid (17-beta) dehydrogenase 4
4504505
1ZBQ
736
1–304
D-bifunctional protein deficiency (261515)
ISPD
isoprenoid synthase domain containing
157412259
4CVH
451
43–451
Muscular dystrophy-dystroglycanopathy congenital with brain and eye anomalies A7 (614643), Muscular dystrophy-dystroglycanopathy limb-girdle C7 (616052)
MAT1A
methionine adenosyltransferase I, alpha
4557737
2OBV
395
16–395
methionine adenosyltransferase deficiency (250850)
1MCCC1-MCCC2
3-methylcrotonoyl-CoA carboxylase complex (MCCC1* and MCCC2†)
13518228* 11545863†
to be deposited
725* and 563†
48–716* and 18–563†
3-methylcrotonoyl-CoA carboxylase 1 deficiency (210200)
MCEE
methylmalonyl CoA epimerase
188035928
3RMU
176
45–176
Methylmalonyl-CoA epimerase deficiency (251120)
MLYCD
malonyl-CoA decarboxylase
6912498
2YGW
454
1–451
MLYCD deficiency (248360)
MMAA
methylmalonic aciduria type A
26892295
2WWW
418
72–418
Methylmalonic aciduria type cblA (251100)
MMACHC
methylmalonic aciduria cblC type, with homocystinuria
153070822
3SOM
282
1–282
Methylmalonic aciduria cblC
2MMADHC
methylmalonic aciduria (cobalamin deficiency) cblD type, with homocystinuria
19527054
5A4R
296
129–296
Methylmalonic aciduria and homocystinuria type cblD (277410)
1MOCS2A-MOCS2B
molybdopterin synthase complex (MOCS2A* and MOCS2B†)
28631173* 4758732†
to be deposited
88* and 188†
9–88* and 27–179†
Molybdenum cofactor deficiency type B (252150)
MOCS2B
molybdopterin synthase catalytic subunit large subunit MOCS2B
4758732
4AP8
188
27–179
Molybdenum cofactor deficiency type B (252150)
MTR
5-methyltetrahydrofolate-homocysteine methyltransferase
4557765
4CCZ
1265
16–657
Homocystinuria-megaloblastic anemia, cblG complementation type (250940)
MUT
methylmalonyl CoA mutase
4557767
3BIC, 2XIJ, 2XIQ
750
12–750
Methylmalonic aciduria type mut (251000)
OXCT1
3-oxoacid CoA transferase
4557817
3DLX
520
40–520
SCOT deficiency (245050)
PAH
phenylalanine hydroxylase
4557819
5FII
452
19–118
Phenylketonuria (261600)
PCCA
propionyl-CoA carboxylase, alpha
65506442
2JKU
728
659–728
Propionic acidemia type I (606054)
PHGDH
phosphoglycerate dehydrogenase
23308577
2G76
533
4–315
PHGDH deficiency (601815)
PHYH
phytanoyl-CoA 2-hydroxylase
5453884
2A1X
338
31–338
Refsum disease (266500)
PTS
6-pyruvoyltetrahydropterin synthase
4506331
3I2B
145
7–145
BH4-deficient hyperphenylalaninemia type A (261640)
PYCR1
pyrroline-5-carboxylate reductase 1
24797097
2IZZ
319
1–300
Cutis laxa autosomal recessive type 2B (612940)
PYCS
pyrroline-5-carboxylate synthetase
21361368
2H5G
795
356–795
Mental retardation-joint hypermobility-skin laxity with or without metabolic abnormalities (612652)
SNX14B
sorting nexin 14 [isoform b]
24797143
4BGJ
893
505–649
Spinocerebellar ataxia, autosomal recessive, 20 (616354)
SPR
sepiapterin reductase
4507185
1Z6Z
261
1–248
Dystonia DOPA-responsive due to SPR deficiency (612716)
TGM1
transglutaminase 1
4507475
2XZZ
817
693–787
Ichthyosis lamellar type 1 (242300)
TH
tyrosine hydroxylase
88900503
2XSN
497
163–497
Segawa syndrome (605407)
TPH2
neuronal tryptophan hydroxylase
31795563
4V06
490
148–490
Attention deficit-hyperactivity disorder 7 (613003)
1 Structures of protein-protein complexes, with constituent subunits annotated by * and †
2 Structure of mouse MMADHC determined
As the field of structural biology gears towards the study of larger, more complex, and more heterogeneous biological systems, the methods of x-ray crystallography and EM, traditionally known for the respective strengths in the resolution and size of samples, will likely evolve towards the middle ground and dominate the playing field for next-generation protein structure determination. On the one hand, x-ray crystallography breaks new grounds on the size of macromolecular complexes it can resolve, as evidenced in the recent 3.6 Å crystal structure of yeast respiratory chain complex I, a mitochondrial membrane-spanning machinery comprising >14 protein subunits (Zickermann et al 2015). On the other hand, an equally impressive, 2.2 Å cryo-EM structure of an inhibitor-bound human galactosidase (Bartesaghi et al 2015) demonstrated the capability of EM to resolve ‘smaller’ complexes, but with the necessary atomic resolution to reveal ligand binding interactions.

An integrated (structural, biochemical, computational) approach to characterize disease causing mutations for IEMs

Inborn errors of metabolism are model subjects to be studied by structural biology, due to two distinctive features. First, the predominant majority of disease-causing mutations are within the exonic region of the affected gene, rather than in the non-coding introns. Hence they are expected to affect the integrity (e.g. stability) and function (e.g. catalysis) of the encoded enzyme (Yue et al 2014), a largely globular conformation well suited for protein structure determination. Second, a large proportion of these mutations (e.g. ~65 % of all genetic variants for the metabolic disorders phenylketonuria, classical homocystinuria and ornithine transcarbamylase deficiency) are of the missense type that result in non-synonymous changes of a single amino acid, as opposed to insertion or deletion of larger peptide fragments. Missense changes are therefore more ‘subtle’ in character, context-dependent in function, and dictated by the physico-chemical properties (e.g. size and hydrophobicity) of the mutated amino acid. Structures of the wild-type and mutant metabolic enzymes determined at atomic resolution (i.e. with the details to resolve individual amino acid residues) prove useful in ascertaining the structural and molecular principles for the amino acid substitution in the local environment, as well as detecting mutation trends and hotspot regions on the protein polypeptide as a whole (Froese et al 2013; Shafqat et al 2013; Riemersma et al 2015).
Nevertheless, structural studies of disease-associated mutant proteins are often more intractable than their wild-type counterparts (Kang and Stevens 2009). This is because the mutation defects that impact on the native integrity and function of the enzyme often precludes its recombinant production in the quantity (milligrams) and quality (purity, homogeneity) that are required for the in vitro biophysical, biochemical or structural experiments. To complement this gap, a plethora of in silico prediction methods have been developed (e.g. FoldX, PolyPhen-2, SIFT and SNPs3d), adopting different algorithms of sequence, physico-chemical and structural information, to interpret the molecular effect and pathogenicity of amino acid substitutions (For reviews, see Stefl et al 2013; Studer et al 2013 and references therein). For the modern-day high-throughput pipeline in characterizing sequence variants, in silico approaches are more preferable to experimental approaches, as the latter are time-consuming and require significant resource for the design and setup.
In silico methods also prove particularly useful in characterizing those metabolic disorders with broad genotypes (i.e. large numbers of disease-causing alleles), such as phenylketonuria (MIM 261600), the most common inborn errors of amino acid metabolism, where more than 400 out of 834 disease alleles annotated in the locus-specific database PAHvdb (http://​www.​biopku.​org/​home/​pah.​asp) are due to missense changes. A general lesson from surveying such a large catalogue of mutations (Wettstein et al 2015) is that disease-causing mutations, as opposed to the harmless variants, are often accompanied by drastic changes in the physico-chemical properties (polarity, hydrophobicity, charge, side-chain geometry) and interaction networks (hydrogen bonds, salt bridges, hydrophobic interactions) of the amino acid involved. The mutated residues are also more commonly localized within the protein core and conserved functional sites, as reflected by their strong amino acid sequence conservation. The utility of in silico prediction tools is also exemplified in its application to triage a subset of all known disease mutations as a starting point for experimental characterization of mutant proteins. This is illustrated in the example of methylmalonyl CoA mutase (MUT), one of the two destination enzymes requiring the vitamin B12 cofactor for catalysis, where to date >130 missense mutations are found on this 750-amino acid polypeptide to cause methylmalonic aciduria (MIM 251000). From a mapping of all known MUT mutations onto the protein crystal structure (Froese et al 2010), we selected 23 of them representative of diverse exonic regions, clinical phenotypes and ethnic populations, and performed a series of biochemical and cellular studies that characterize the protein stability (recombinant expression level, differential scanning fluorimetry) and enzyme catalysis (activity assay in patient cells) (Forny et al 2014) (Fig. 1). Among the different molecular defects catalogued from these mutations, we found that a subset of mutants are thermally less stable than wild-type in vitro, and can be partly rescued in vitro and in vivo by exogenous supplementation of chemical chaperones (CC) such as glycerol, proline, betaine and trimethylamine N-oxide. These CCs are low-molecular-weight osmolytes that stabilise proteins, without directly interacting with them, by preferential exclusion at the protein surface, thereby altering the thermodynamic free energy of the protein in the solvent environment (Arakawa et al 2006). Together, this study provides a proof of concept that stabilizing mutant protein could be one therapeutic strategy to rescue its defective function.

Small molecule drug development for loss-of-function disorders is challenging

The availability of high-resolution protein structures has since facilitated greatly the process of small molecule drug development in human medicine (Yue et al 2014). Even before its high-throughput advances, structural biology already played a role in the lead optimization step of drug development, in which structures of an established therapeutic target in complex with a drug candidate can direct the chemical alterations of the drug compounds to improve their affinity, potency and selectivity. Nowadays, the timeframe of protein structure determination has become sufficiently short, hence allowing its application in other stages of the conventional drug discovery pipeline. For example, protein structures can reveal distinguishing features across different members of an enzyme/domain family to explore specificity, detect binding pockets and evaluate their ‘druggability’ (i.e. how amenable a small molecule can bind to these pockets), guide compound screening campaign using biophysical/in silico assays, and develop structure-activity relationship for a chemical series of compounds.
When considering therapeutic possibilities, inborn errors of metabolism, like other genetic diseases, can be seen as a dysregulation of dosage for the associated protein (Beaulieu et al 2012). Depending on the particular disease, or even the disease allele, a genetic lesion can theoretically lead to a higher- or lower-than-physiological level of the associated protein that signify a gain-of-function (GOF) or loss-of-function (LOF) phenotype, respectively. Our systematic study of PAH and MUT mutations, as well as of other enzymes (McCorvie and Timson 2013; Balmer et al 2014; Burda et al 2015), all conform to the general concept that IEMs are by and large LOF diseases due to pathogenic mechanisms that effect the structure (e.g. misfolding, aggregation) and function (loss of catalysis, loss of interactions) of the enzyme. This therefore poses a conceptual challenge for drug discovery, since the most intuitive therapeutic target for an IEM (i.e. the metabolic enzyme harbouring a LOF mutation itself) implies an imperative to develop an activator of the deficient or defective enzyme (Segalat 2007). While the drug development industry has more traction in GOF diseases, by means of small molecule inhibitors aimed at reducing the mRNA, protein or activity levels, the road is much less travelled for developing a therapy to activate or upregulate the levels of mRNA, protein or activity as treatment for LOF diseases. With the explosion of genomic data and disease linkage from the advent of next-generation sequencing, novel therapy design and principles are urgently required for LOF diseases (Boycott et al 2013).

Allosteric activators as next generation pharmacological chaperones?

One emerging therapeutic approach for LOF diseases involves the use of small molecule ligands known as pharmacological chaperones (PCs) to stabilize and activate the mutant enzyme (Muntau et al 2014), with the rationale that a moderate increase in the mutant enzyme activity beyond a certain threshold level (e.g. 10 % of many lysosomal storage enzymes) could suffice to delay disease onset and ameliorate phenotypes (Suzuki et al 2009). To date, PCs have promising potentials for several IEMs, including the Fabry, Gaucher and Pompe Diseases, which have reached early-stage clinical trials (Boyd et al 2013; Parenti et al 2015); while others are gaining proof of concept (Santos-Sierra et al 2012; Jorge-Finnigan et al 2013; Makley et al 2015). These first-generation PC molecules often originate as substrate-mimetics or cofactor-mimetics of the target enzymes, and the field of structural biology has been useful in characterizing how the PC molecule interacts with the enzyme active site (Bateman et al 2011; Guce et al 2011; Torreblanca et al 2012; Suzuki et al 2014). However, active-site-directed PCs create a conundrum in which by binding to the active site, these ligands could potentially compete with the native substrate or cofactor of the target enzyme. Hence the counter-intuitive mode of action for these PCs could restrict their application to certain sub-inhibitory concentration and dosage window.
We propose that the next generation of PC molecules should target an enzyme’s domains or pockets that are distant from its active site, and therefore would not compete with the enzyme’s native ligands. These allosteric, non-catalytic binding sites are often specific to an enzyme’s unique structure and function, and necessitate structural and chemical biology approaches to help identification and validation. As a first lesson to design allosteric PCs, my group has the vested interest in understanding the molecular mechanism of naturally-occurring enzymes built with a modality for ligand-binding allosteric regulation. An example for such enzyme is cystathionine-β-synthase (CBS) (Fig. 2a), which catalyzes the conversion of homocysteine to cystathionine, a reaction up-regulated by the native ligand of CBS, S-adenosyl-L-methionine (SAM). Inherited mutations of the cbs gene lead to classical homocystinuria (MIM 236200), in which protein misfolding is the major cause of malfunction among the predominant alleles. In addition to a core catalytic domain harbouring the enzyme active site, CBS contains a regulator domain at its C-terminus which binds SAM in order to activate the catalytic domain (Pey et al 2013). We have applied crystallography, biochemical and biophysical methods to explain how CBS is allosterically activated by SAM, by showing that binding of SAM to the regulatory domain results in large conformational change that relieves its steric inhibition of the enzyme catalytic domain (McCorvie et al 2014). We therefore posit that targeting the CBS regulatory domain with a small molecule could be a strategy to activate mutant CBS enzyme using the same native mechanism, thereby rescuing disease alleles with defective enzyme activity.
CBS is not a lone example of allosteric activation via a multi-domain protein architecture (Jaffe and Lawrence 2012; Jaffe et al 2013). Similarly, the enzyme phenylalanine hydroxylase (PAH), of which genetic mutations lead to PKU, is activated by its own substrate phenylalanine (Phe), postulated to bind not only to the enzyme active site in the catalytic domain, but also to a separate regulatory domain (Zhang et al 2014) (Fig. 2b). We recently provided the first structural evidence that binding of Phe to the PAH regulatory domain causes this domain to dimerize and, like CBS, relieves its steric inhibition on the catalytic domain (Patel et al 2016). We therefore believe that screening for small molecule binders in allosteric sites such as the regulatory domains of CBS and PAH could be a common principle for drug discovery of LOF inborn errors of metabolism, at least in those enzymes with allosteric sites. There are emerging studies identifying non-inhibitory ligands in allosteric sites and cryptic pockets of metabolic enzymes, either by means of high throughput screening of large compound libraries (Marugan et al 2010; Patnaik et al 2012; Porto et al 2012) or by the more chemistry-efficient small molecule fragment approach (Landon et al 2009).

Multiple intervention avenues within a metabolic pathway

When determining the appropriate therapeutic targets for a metabolic disorder, it is often beneficial to look at the different reaction steps around the affected enzyme of a metabolic pathway, in order to maximize intervention possibilities (Fig. 3). As an example, normal glycogen synthesis is catalysed by the concerted action of three enzymes in humans: glycogenin (GYG), glycogen synthase (GYS) and branching enzyme (GBE). Andersen disease (MIM 232500) and adult polyglucosan body disease (APBD; MIM 263570) caused by GBE mutations, as well as Lafora disease due to mutations in malin or laforin proteins (MIM 254780), are all neurological diseases sharing a common neuropathology of malformed glycogen (‘polyglucosan’) accumulation, although their genetic defects affect different aspects of glycogen metabolism. We have recently applied structural biology to characterize GBE disease-causing mutations, in particular the prevalent allele causing APBD, p.Y329S. A homology model of the GBE mutant, based on the experimental wild-type crystal structure (Froese et al 2015), shows that the Tyr329-to-Ser substitution creates a cavity at the protein surface away from the active site, and results in lower protein expression in vitro and in vivo, and reduced protein stability. We reasoned that a small molecule specifically targeting this surface-accessible cavity could function as a ‘molecular strut’ to stabilize the local defect of this mutant enzyme. As proof of therapeutic principle, a tetra-peptide (Leu-Thr-Lys-Glu) designed computationally to fill this cavity can be taken up in patient cells, binds specifically to mutant GBE protein and rescues mutant enzyme activity to a moderate level (Fig. 3a).
The peptide approach above can be considered an example of pharmacological chaperone that is targeted directly at the site of mutation defect, distant from the enzyme active site. An alternative therapeutic avenue, however, can be based on substrate inhibition, with the rationale that the same biosynthesis enzymes for normal glycogen production are also responsible for polyglucosan formation. Recently, a gene knockdown of muscle glycogen synthase (GYS1) eliminated polyglucosan formation and restored neurological functions in a mouse model of Lafora disease (Pederson et al 2013), as well as a neuronal model for APBD (Kakhlon et al 2013). These clinical findings are further supported by the natural existence of very rare GYS mutations which lead to its enzyme deficiency (MIM 611556) with mild phenotypes in humans. Together, they provide proof of principle that downregulation of glycogen synthesis eliminates neurological abnormalities due to polyglucosan formation, and lend support to the inhibition of GYS1 enzyme activity, or its interaction with the functional partner GYG necessary for glycogen synthesis, as potential molecular therapy for Lafora and Andersen diseases, as well as for the lysosomal storage disorder Pombe (GAA mutations) whose hallmark is muscle glycogen accumulation. The inhibition approach is particularly attractive to the drug development industry, and can be considered as an example of ‘substrate reduction therapy’ (Fig. 3b) where there is precedence in IEMs (e.g. Miglustat for the treatment of Gaucher disease (Platt and Jeyakumar 2008)) to reduce the consequence of toxic metabolite accumulation due to defective enzymes (Schiffmann 2015). We anticipate that progress in the structural biology of GYS and GYG (Chaikuad et al 2011; Zeqiraj et al 2014) should pave the next step forward in developing novel inhibitors for these enzymes. This example therefore illustrates the principle of targeting different components of a metabolic pathway for the treatment of a genetic disease. As we enter the post-genomic era from the advent of next generation sequencing, with emerging new diseases (Ebrahimi-Fakhari et al 2016) and unmet medical need, there is an urgent call for novel, ‘imaginative’ routes of therapeutic intervention to tackle IEMs, and rare diseases as a whole. To this end, a recent example of functionally bypassing a deficient protein by a suppressor mutation (Yoon et al 2014) may represent one of many future therapeutic approaches aimed at activating an alternative gene, isoform or enzymatic mechanism (Fig. 3c) to functionally compensate for the defective gene and product.

Acknowledgments

We are grateful to all current and former colleagues from SGC Oxford and the group of Wyatt Yue, who have contributed to this work. The Structural Genomics Consortium is a registered charity (number 1097737) that receives funds from AbbVie, Boehringer Ingelheim, the Canada Foundation for Innovation, the Canadian Institutes for Health Research, Genome Canada, GlaxoSmithKline, Janssen, Lilly Canada, the Novartis Research Foundation, the Ontario Ministry of Economic Development and Innovation, Pfizer, Takeda, and the Wellcome Trust [092809/Z/10/Z]. W.W.Y. is further supported by a gift donation from the APBD Research Foundation, and by the Pathfinder Award from the Wellcome Trust.
Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

Literatur
Zurück zum Zitat Arakawa T, Ejima D, Kita Y, Tsumoto K (2006) Small molecule pharmacological chaperones: from thermodynamic stabilization to pharmaceutical drugs. Biochim Biophys Acta 1764:1677–1687CrossRefPubMed Arakawa T, Ejima D, Kita Y, Tsumoto K (2006) Small molecule pharmacological chaperones: from thermodynamic stabilization to pharmaceutical drugs. Biochim Biophys Acta 1764:1677–1687CrossRefPubMed
Zurück zum Zitat Balmer C, Pandey AV, Rufenacht V et al (2014) Mutations and polymorphisms in the human argininosuccinate lyase (ASL) gene. Hum Mutat 35:27–35CrossRefPubMed Balmer C, Pandey AV, Rufenacht V et al (2014) Mutations and polymorphisms in the human argininosuccinate lyase (ASL) gene. Hum Mutat 35:27–35CrossRefPubMed
Zurück zum Zitat Bartesaghi A, Merk A, Banerjee S et al (2015) 2.2 A resolution cryo-EM structure of beta-galactosidase in complex with a cell-permeant inhibitor. Science 348:1147–1151CrossRefPubMed Bartesaghi A, Merk A, Banerjee S et al (2015) 2.2 A resolution cryo-EM structure of beta-galactosidase in complex with a cell-permeant inhibitor. Science 348:1147–1151CrossRefPubMed
Zurück zum Zitat Bateman KS, Cherney MM, Mahuran DJ, Tropak M, James MN (2011) Crystal structure of beta-hexosaminidase B in complex with pyrimethamine, a potential pharmacological chaperone. J Med Chem 54:1421–1429CrossRefPubMedPubMedCentral Bateman KS, Cherney MM, Mahuran DJ, Tropak M, James MN (2011) Crystal structure of beta-hexosaminidase B in complex with pyrimethamine, a potential pharmacological chaperone. J Med Chem 54:1421–1429CrossRefPubMedPubMedCentral
Zurück zum Zitat Beaulieu CL, Samuels ME, Ekins S et al (2012) A generalizable pre-clinical research approach for orphan disease therapy. Orphanet J Rare Dis 7:39CrossRefPubMedPubMedCentral Beaulieu CL, Samuels ME, Ekins S et al (2012) A generalizable pre-clinical research approach for orphan disease therapy. Orphanet J Rare Dis 7:39CrossRefPubMedPubMedCentral
Zurück zum Zitat Boycott KM, Vanstone MR, Bulman DE, MacKenzie AE (2013) Rare-disease genetics in the era of next-generation sequencing: discovery to translation. Nat Rev Genet 14:681–691CrossRefPubMed Boycott KM, Vanstone MR, Bulman DE, MacKenzie AE (2013) Rare-disease genetics in the era of next-generation sequencing: discovery to translation. Nat Rev Genet 14:681–691CrossRefPubMed
Zurück zum Zitat Boyd RE, Lee G, Rybczynski P et al (2013) Pharmacological chaperones as therapeutics for lysosomal storage diseases. J Med Chem 56:2705–2725CrossRefPubMed Boyd RE, Lee G, Rybczynski P et al (2013) Pharmacological chaperones as therapeutics for lysosomal storage diseases. J Med Chem 56:2705–2725CrossRefPubMed
Zurück zum Zitat Burda P, Schafer A, Suormala T et al (2015) Insights into severe 5,10-methylenetetrahydrofolate reductase deficiency: molecular genetic and enzymatic characterization of 76 patients. Hum Mutat 36:611–621CrossRefPubMed Burda P, Schafer A, Suormala T et al (2015) Insights into severe 5,10-methylenetetrahydrofolate reductase deficiency: molecular genetic and enzymatic characterization of 76 patients. Hum Mutat 36:611–621CrossRefPubMed
Zurück zum Zitat Chaikuad A, Froese DS, Berridge G, von Delft F, Oppermann U, Yue WW (2011) Conformational plasticity of glycogenin and its maltosaccharide substrate during glycogen biogenesis. Proc Natl Acad Sci U S A 108:21028–21033CrossRefPubMedPubMedCentral Chaikuad A, Froese DS, Berridge G, von Delft F, Oppermann U, Yue WW (2011) Conformational plasticity of glycogenin and its maltosaccharide substrate during glycogen biogenesis. Proc Natl Acad Sci U S A 108:21028–21033CrossRefPubMedPubMedCentral
Zurück zum Zitat Ebrahimi-Fakhari D, Saffari A, Wahlster L, et al (2016) Congenital disorders of autophagy: an emerging novel class of inborn errors of neuro-metabolism. Brain 139:317–337 Ebrahimi-Fakhari D, Saffari A, Wahlster L, et al (2016) Congenital disorders of autophagy: an emerging novel class of inborn errors of neuro-metabolism. Brain 139:317–337
Zurück zum Zitat Forny P, Froese DS, Suormala T, Yue WW, Baumgartner MR (2014) Functional characterization and categorization of missense mutations that cause methylmalonyl-CoA mutase (MUT) deficiency. Hum Mutat 35:1449–1458CrossRefPubMedPubMedCentral Forny P, Froese DS, Suormala T, Yue WW, Baumgartner MR (2014) Functional characterization and categorization of missense mutations that cause methylmalonyl-CoA mutase (MUT) deficiency. Hum Mutat 35:1449–1458CrossRefPubMedPubMedCentral
Zurück zum Zitat Froese DS, Gravel RA (2010) Genetic disorders of vitamin B12 metabolism: eight complementation groups — eight genes. Expert Rev Mol Med 12:e37 Froese DS, Gravel RA (2010) Genetic disorders of vitamin B12 metabolism: eight complementation groups — eight genes. Expert Rev Mol Med 12:e37
Zurück zum Zitat Froese DS, Kochan G, Muniz JR et al (2010) Structures of the human GTPase MMAA and vitamin B12-dependent methylmalonyl-CoA mutase and insight into their complex formation. J Biol Chem 285:38204–38213CrossRefPubMedPubMedCentral Froese DS, Kochan G, Muniz JR et al (2010) Structures of the human GTPase MMAA and vitamin B12-dependent methylmalonyl-CoA mutase and insight into their complex formation. J Biol Chem 285:38204–38213CrossRefPubMedPubMedCentral
Zurück zum Zitat Froese DS, Krojer T, Wu X et al (2012) Structure of MMACHC reveals an arginine-rich pocket and a domain-swapped dimer for its B12 processing function. Biochemistry 51:5083–5090CrossRefPubMed Froese DS, Krojer T, Wu X et al (2012) Structure of MMACHC reveals an arginine-rich pocket and a domain-swapped dimer for its B12 processing function. Biochemistry 51:5083–5090CrossRefPubMed
Zurück zum Zitat Froese DS, Forouhar F, Tran TH et al (2013) Crystal structures of malonyl-coenzyme A decarboxylase provide insights into its catalytic mechanism and disease-causing mutations. Structure 21:1182–1192CrossRefPubMedPubMedCentral Froese DS, Forouhar F, Tran TH et al (2013) Crystal structures of malonyl-coenzyme A decarboxylase provide insights into its catalytic mechanism and disease-causing mutations. Structure 21:1182–1192CrossRefPubMedPubMedCentral
Zurück zum Zitat Froese DS, Kopec J, Fitzpatrick F et al (2015a) Structural insights into the MMACHC-MMADHC protein complex involved in vitamin B12 trafficking. J Biol Chem 290:29167–29177CrossRefPubMedPubMedCentral Froese DS, Kopec J, Fitzpatrick F et al (2015a) Structural insights into the MMACHC-MMADHC protein complex involved in vitamin B12 trafficking. J Biol Chem 290:29167–29177CrossRefPubMedPubMedCentral
Zurück zum Zitat Froese DS, Michaeli A, McCorvie TJ et al (2015b) Structural basis of glycogen branching enzyme deficiency and pharmacologic rescue by rational peptide design. Hum Mol Genet 24:5667–5676CrossRefPubMedPubMedCentral Froese DS, Michaeli A, McCorvie TJ et al (2015b) Structural basis of glycogen branching enzyme deficiency and pharmacologic rescue by rational peptide design. Hum Mol Genet 24:5667–5676CrossRefPubMedPubMedCentral
Zurück zum Zitat Guce AI, Clark NE, Rogich JJ, Garman SC (2011) The molecular basis of pharmacological chaperoning in human alpha-galactosidase. Chem Biol 18:1521–1526CrossRefPubMedPubMedCentral Guce AI, Clark NE, Rogich JJ, Garman SC (2011) The molecular basis of pharmacological chaperoning in human alpha-galactosidase. Chem Biol 18:1521–1526CrossRefPubMedPubMedCentral
Zurück zum Zitat Jaffe EK, Lawrence SH (2012) Allostery and the dynamic oligomerization of porphobilinogen synthase. Arch Biochem Biophys 519:144–153CrossRefPubMed Jaffe EK, Lawrence SH (2012) Allostery and the dynamic oligomerization of porphobilinogen synthase. Arch Biochem Biophys 519:144–153CrossRefPubMed
Zurück zum Zitat Jaffe EK, Stith L, Lawrence SH, Andrake M, Dunbrack RL Jr (2013) A new model for allosteric regulation of phenylalanine hydroxylase: implications for disease and therapeutics. Arch Biochem Biophys 530:73–82CrossRefPubMedPubMedCentral Jaffe EK, Stith L, Lawrence SH, Andrake M, Dunbrack RL Jr (2013) A new model for allosteric regulation of phenylalanine hydroxylase: implications for disease and therapeutics. Arch Biochem Biophys 530:73–82CrossRefPubMedPubMedCentral
Zurück zum Zitat Jorge-Finnigan A, Brasil S, Underhaug J et al (2013) Pharmacological chaperones as a potential therapeutic option in methylmalonic aciduria cblB type. Hum Mol Genet 22:3680–3689CrossRefPubMedPubMedCentral Jorge-Finnigan A, Brasil S, Underhaug J et al (2013) Pharmacological chaperones as a potential therapeutic option in methylmalonic aciduria cblB type. Hum Mol Genet 22:3680–3689CrossRefPubMedPubMedCentral
Zurück zum Zitat Kakhlon O, Glickstein H, Feinstein N et al (2013) Polyglucosan neurotoxicity caused by glycogen branching enzyme deficiency can be reversed by inhibition of glycogen synthase. J Neurochem 127:101–113PubMed Kakhlon O, Glickstein H, Feinstein N et al (2013) Polyglucosan neurotoxicity caused by glycogen branching enzyme deficiency can be reversed by inhibition of glycogen synthase. J Neurochem 127:101–113PubMed
Zurück zum Zitat Kang TS, Stevens RC (2009) Structural aspects of therapeutic enzymes to treat metabolic disorders. Hum Mutat 30:1591–1610CrossRefPubMed Kang TS, Stevens RC (2009) Structural aspects of therapeutic enzymes to treat metabolic disorders. Hum Mutat 30:1591–1610CrossRefPubMed
Zurück zum Zitat Landon MR, Lieberman RL, Hoang QQ et al (2009) Detection of ligand binding hot spots on protein surfaces via fragment-based methods: application to DJ-1 and glucocerebrosidase. J Comput Aided Mol Des 23:491–500CrossRefPubMed Landon MR, Lieberman RL, Hoang QQ et al (2009) Detection of ligand binding hot spots on protein surfaces via fragment-based methods: application to DJ-1 and glucocerebrosidase. J Comput Aided Mol Des 23:491–500CrossRefPubMed
Zurück zum Zitat Makley LN, McMenimen KA, DeVree BT et al (2015) Pharmacological chaperone for alpha-crystallin partially restores transparency in cataract models. Science 350:674–677CrossRefPubMed Makley LN, McMenimen KA, DeVree BT et al (2015) Pharmacological chaperone for alpha-crystallin partially restores transparency in cataract models. Science 350:674–677CrossRefPubMed
Zurück zum Zitat Marugan JJ, Zheng W, Ferrer M et al (2010) Discovery, SAR, and biological evaluation of a non-inhibitory chaperone for acid alpha glucosidase. In Probe Reports from the NIH Molecular Libraries Program Bethesda (MD) Marugan JJ, Zheng W, Ferrer M et al (2010) Discovery, SAR, and biological evaluation of a non-inhibitory chaperone for acid alpha glucosidase. In Probe Reports from the NIH Molecular Libraries Program Bethesda (MD)
Zurück zum Zitat McCorvie TJ, Timson DJ (2013) In silico prediction of the effects of mutations in the human UDP-galactose 4′-epimerase gene: towards a predictive framework for type III galactosemia. Gene 524:95–104CrossRefPubMed McCorvie TJ, Timson DJ (2013) In silico prediction of the effects of mutations in the human UDP-galactose 4′-epimerase gene: towards a predictive framework for type III galactosemia. Gene 524:95–104CrossRefPubMed
Zurück zum Zitat McCorvie TJ, Kopec J, Hyung SJ et al (2014) Inter-domain communication of human cystathionine beta-synthase: structural basis of S-adenosyl-L-methionine activation. J Biol Chem 289:36018–36030CrossRefPubMedPubMedCentral McCorvie TJ, Kopec J, Hyung SJ et al (2014) Inter-domain communication of human cystathionine beta-synthase: structural basis of S-adenosyl-L-methionine activation. J Biol Chem 289:36018–36030CrossRefPubMedPubMedCentral
Zurück zum Zitat Muntau AC, Leandro J, Staudigl M, Mayer F, Gersting SW (2014) Innovative strategies to treat protein misfolding in inborn errors of metabolism: pharmacological chaperones and proteostasis regulators. J Inherit Metab Dis 37:505–523CrossRefPubMed Muntau AC, Leandro J, Staudigl M, Mayer F, Gersting SW (2014) Innovative strategies to treat protein misfolding in inborn errors of metabolism: pharmacological chaperones and proteostasis regulators. J Inherit Metab Dis 37:505–523CrossRefPubMed
Zurück zum Zitat Osman KT, Edwards A (2014) Structural genomics of human proteins. Methods Mol Biol 1140:27–34CrossRefPubMed Osman KT, Edwards A (2014) Structural genomics of human proteins. Methods Mol Biol 1140:27–34CrossRefPubMed
Zurück zum Zitat Parenti G, Andria G, Valenzano KJ (2015) Pharmacological chaperone therapy: preclinical development, clinical translation, and prospects for the treatment of lysosomal storage disorders. Mol Ther 23:1138–1148CrossRefPubMed Parenti G, Andria G, Valenzano KJ (2015) Pharmacological chaperone therapy: preclinical development, clinical translation, and prospects for the treatment of lysosomal storage disorders. Mol Ther 23:1138–1148CrossRefPubMed
Zurück zum Zitat Patel D, Kopec J, Fitzpatrick F, McCorvie TJ, Yue WW (2016) Structural basis for ligand-dependent dimerization of phenylalanine hydroxylase regulatory domain. Sci Rep 6:23748. doi:10.1038/srep23748 Patel D, Kopec J, Fitzpatrick F, McCorvie TJ, Yue WW (2016) Structural basis for ligand-dependent dimerization of phenylalanine hydroxylase regulatory domain. Sci Rep 6:23748. doi:10.​1038/​srep23748
Zurück zum Zitat Patnaik S, Zheng W, Choi JH et al (2012) Discovery, structure-activity relationship, and biological evaluation of noninhibitory small molecule chaperones of glucocerebrosidase. J Med Chem 55:5734–5748CrossRefPubMedPubMedCentral Patnaik S, Zheng W, Choi JH et al (2012) Discovery, structure-activity relationship, and biological evaluation of noninhibitory small molecule chaperones of glucocerebrosidase. J Med Chem 55:5734–5748CrossRefPubMedPubMedCentral
Zurück zum Zitat Pederson BA, Turnbull J, Epp JR et al (2013) Inhibiting glycogen synthesis prevents Lafora disease in a mouse model. Ann Neurol 74:297–300PubMedPubMedCentral Pederson BA, Turnbull J, Epp JR et al (2013) Inhibiting glycogen synthesis prevents Lafora disease in a mouse model. Ann Neurol 74:297–300PubMedPubMedCentral
Zurück zum Zitat Pey AL, Majtan T, Sanchez-Ruiz JM, Kraus JP (2013) Human cystathionine beta-synthase (CBS) contains two classes of binding sites for S-adenosylmethionine (SAM): complex regulation of CBS activity and stability by SAM. Biochem J 449:109–121CrossRefPubMed Pey AL, Majtan T, Sanchez-Ruiz JM, Kraus JP (2013) Human cystathionine beta-synthase (CBS) contains two classes of binding sites for S-adenosylmethionine (SAM): complex regulation of CBS activity and stability by SAM. Biochem J 449:109–121CrossRefPubMed
Zurück zum Zitat Porto C, Ferrara MC, Meli M et al (2012) Pharmacological enhancement of alpha-glucosidase by the allosteric chaperone N-acetylcysteine. Mol Ther 20:2201–2211CrossRefPubMedPubMedCentral Porto C, Ferrara MC, Meli M et al (2012) Pharmacological enhancement of alpha-glucosidase by the allosteric chaperone N-acetylcysteine. Mol Ther 20:2201–2211CrossRefPubMedPubMedCentral
Zurück zum Zitat Riemersma M, Froese DS, van Tol W et al (2015) Human ISPD Is a cytidyltransferase required for dystroglycan o-mannosylation. Chem Biol 22:1643–1652CrossRefPubMed Riemersma M, Froese DS, van Tol W et al (2015) Human ISPD Is a cytidyltransferase required for dystroglycan o-mannosylation. Chem Biol 22:1643–1652CrossRefPubMed
Zurück zum Zitat Santos-Sierra S, Kirchmair J, Perna AM et al (2012) Novel pharmacological chaperones that correct phenylketonuria in mice. Hum Mol Genet 21:1877–1887CrossRefPubMed Santos-Sierra S, Kirchmair J, Perna AM et al (2012) Novel pharmacological chaperones that correct phenylketonuria in mice. Hum Mol Genet 21:1877–1887CrossRefPubMed
Zurück zum Zitat Schiffmann R (2015) The consequences of genetic and pharmacologic reduction in sphingolipid synthesis. J Inherit Metab Dis 38:77–84CrossRefPubMed Schiffmann R (2015) The consequences of genetic and pharmacologic reduction in sphingolipid synthesis. J Inherit Metab Dis 38:77–84CrossRefPubMed
Zurück zum Zitat Shafqat N, Kavanagh KL, Sass JO et al (2013) A structural mapping of mutations causing succinyl-CoA:3-ketoacid CoA transferase (SCOT) deficiency. J Inherit Metab Dis 36:983–987CrossRefPubMedPubMedCentral Shafqat N, Kavanagh KL, Sass JO et al (2013) A structural mapping of mutations causing succinyl-CoA:3-ketoacid CoA transferase (SCOT) deficiency. J Inherit Metab Dis 36:983–987CrossRefPubMedPubMedCentral
Zurück zum Zitat Studer RA, Dessailly BH, Orengo CA (2013) Residue mutations and their impact on protein structure and function: detecting beneficial and pathogenic changes. Biochem J 449:581–594CrossRefPubMed Studer RA, Dessailly BH, Orengo CA (2013) Residue mutations and their impact on protein structure and function: detecting beneficial and pathogenic changes. Biochem J 449:581–594CrossRefPubMed
Zurück zum Zitat Su XD, Zhang H, Terwilliger TC, Liljas A, Xiao J, Dong Y (2015) Protein crystallography from the perspective of technology developments. Crystallogr Rev 21:122–153CrossRefPubMedPubMedCentral Su XD, Zhang H, Terwilliger TC, Liljas A, Xiao J, Dong Y (2015) Protein crystallography from the perspective of technology developments. Crystallogr Rev 21:122–153CrossRefPubMedPubMedCentral
Zurück zum Zitat Suzuki Y, Ogawa S, Sakakibara Y (2009) Chaperone therapy for neuronopathic lysosomal diseases: competitive inhibitors as chemical chaperones for enhancement of mutant enzyme activities. Perspect Med Chem 3:7–19 Suzuki Y, Ogawa S, Sakakibara Y (2009) Chaperone therapy for neuronopathic lysosomal diseases: competitive inhibitors as chemical chaperones for enhancement of mutant enzyme activities. Perspect Med Chem 3:7–19
Zurück zum Zitat Suzuki H, Ohto U, Higaki K et al (2014) Structural basis of pharmacological chaperoning for human beta-galactosidase. J Biol Chem 289:14560–14568CrossRefPubMedPubMedCentral Suzuki H, Ohto U, Higaki K et al (2014) Structural basis of pharmacological chaperoning for human beta-galactosidase. J Biol Chem 289:14560–14568CrossRefPubMedPubMedCentral
Zurück zum Zitat Torreblanca R, Lira-Navarrete E, Sancho J, Hurtado-Guerrero R (2012) Structural and mechanistic basis of the interaction between a pharmacological chaperone and human phenylalanine hydroxylase. Chembiochem 13:1266–1269CrossRefPubMed Torreblanca R, Lira-Navarrete E, Sancho J, Hurtado-Guerrero R (2012) Structural and mechanistic basis of the interaction between a pharmacological chaperone and human phenylalanine hydroxylase. Chembiochem 13:1266–1269CrossRefPubMed
Zurück zum Zitat Wettstein S, Underhaug J, Perez B et al (2015) Linking genotypes database with locus-specific database and genotype-phenotype correlation in phenylketonuria. Eur J Hum Genet 23:302–309CrossRefPubMed Wettstein S, Underhaug J, Perez B et al (2015) Linking genotypes database with locus-specific database and genotype-phenotype correlation in phenylketonuria. Eur J Hum Genet 23:302–309CrossRefPubMed
Zurück zum Zitat Yoon H, Knight SA, Pandey A et al (2014) Frataxin-bypassing Isu1: characterization of the bypass activity in cells and mitochondria. Biochem J 459:71–81CrossRefPubMedPubMedCentral Yoon H, Knight SA, Pandey A et al (2014) Frataxin-bypassing Isu1: characterization of the bypass activity in cells and mitochondria. Biochem J 459:71–81CrossRefPubMedPubMedCentral
Zurück zum Zitat Yue WW, Oppermann U (2011) High-throughput structural biology of metabolic enzymes and its impact on human diseases. J Inherit Metab Dis 34:575–581CrossRefPubMed Yue WW, Oppermann U (2011) High-throughput structural biology of metabolic enzymes and its impact on human diseases. J Inherit Metab Dis 34:575–581CrossRefPubMed
Zurück zum Zitat Yue WW, Froese DS, Brennan PE (2014) The role of protein structural analysis in the next generation sequencing era. Top Curr Chem 336:67–98CrossRefPubMed Yue WW, Froese DS, Brennan PE (2014) The role of protein structural analysis in the next generation sequencing era. Top Curr Chem 336:67–98CrossRefPubMed
Zurück zum Zitat Zeqiraj E, Tang X, Hunter RW et al (2014) Structural basis for the recruitment of glycogen synthase by glycogenin. Proc Natl Acad Sci U S A 111:E2831–2840CrossRefPubMedPubMedCentral Zeqiraj E, Tang X, Hunter RW et al (2014) Structural basis for the recruitment of glycogen synthase by glycogenin. Proc Natl Acad Sci U S A 111:E2831–2840CrossRefPubMedPubMedCentral
Zurück zum Zitat Zhang S, Roberts KM, Fitzpatrick PF (2014) Phenylalanine binding is linked to dimerization of the regulatory domain of phenylalanine hydroxylase. Biochemistry 53:6625–6627CrossRefPubMedPubMedCentral Zhang S, Roberts KM, Fitzpatrick PF (2014) Phenylalanine binding is linked to dimerization of the regulatory domain of phenylalanine hydroxylase. Biochemistry 53:6625–6627CrossRefPubMedPubMedCentral
Zurück zum Zitat Zickermann V, Wirth C, Nasiri H et al (2015) Structural biology. Mechanistic insight from the crystal structure of mitochondrial complex I. Science 347:44–49CrossRefPubMed Zickermann V, Wirth C, Nasiri H et al (2015) Structural biology. Mechanistic insight from the crystal structure of mitochondrial complex I. Science 347:44–49CrossRefPubMed
Metadaten
Titel
From structural biology to designing therapy for inborn errors of metabolism
verfasst von
Wyatt W. Yue
Publikationsdatum
30.05.2016
Verlag
Springer Netherlands
Erschienen in
Journal of Inherited Metabolic Disease / Ausgabe 4/2016
Print ISSN: 0141-8955
Elektronische ISSN: 1573-2665
DOI
https://doi.org/10.1007/s10545-016-9923-3

Weitere Artikel der Ausgabe 4/2016

Journal of Inherited Metabolic Disease 4/2016 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Reizdarmsyndrom: Diäten wirksamer als Medikamente

29.04.2024 Reizdarmsyndrom Nachrichten

Bei Reizdarmsyndrom scheinen Diäten, wie etwa die FODMAP-arme oder die kohlenhydratreduzierte Ernährung, effektiver als eine medikamentöse Therapie zu sein. Das hat eine Studie aus Schweden ergeben, die die drei Therapieoptionen im direkten Vergleich analysierte.

Notfall-TEP der Hüfte ist auch bei 90-Jährigen machbar

26.04.2024 Hüft-TEP Nachrichten

Ob bei einer Notfalloperation nach Schenkelhalsfraktur eine Hemiarthroplastik oder eine totale Endoprothese (TEP) eingebaut wird, sollte nicht allein vom Alter der Patientinnen und Patienten abhängen. Auch über 90-Jährige können von der TEP profitieren.

Niedriger diastolischer Blutdruck erhöht Risiko für schwere kardiovaskuläre Komplikationen

25.04.2024 Hypotonie Nachrichten

Wenn unter einer medikamentösen Hochdrucktherapie der diastolische Blutdruck in den Keller geht, steigt das Risiko für schwere kardiovaskuläre Ereignisse: Darauf deutet eine Sekundäranalyse der SPRINT-Studie hin.

Bei schweren Reaktionen auf Insektenstiche empfiehlt sich eine spezifische Immuntherapie

Insektenstiche sind bei Erwachsenen die häufigsten Auslöser einer Anaphylaxie. Einen wirksamen Schutz vor schweren anaphylaktischen Reaktionen bietet die allergenspezifische Immuntherapie. Jedoch kommt sie noch viel zu selten zum Einsatz.

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.