Glycosaminoglycan levels in dried blood spots of patients with mucopolysaccharidoses and mucolipidoses

https://doi.org/10.1016/j.ymgme.2016.12.010Get rights and content

Highlights

  • DBS is feasible for GAG measurement by tandem mass spectrometry.

  • Untreated patients had higher GAG levels compared to control samples.

  • Treated patients had lower GAG levels compared to untreated patients.

  • GAG in DBS seem to be feasible for diagnosis and therapeutic monitoring.

Abstract

Mucopolysaccharidoses (MPSs) and mucolipidoses (ML) are groups of lysosomal storage disorders in which lysosomal hydrolases are deficient leading to accumulation of undegraded glycosaminoglycans (GAGs), throughout the body, subsequently resulting in progressive damage to multiple tissues and organs. Assays using tandem mass spectrometry (MS/MS) have been established to measure GAGs in serum or plasma from MPS and ML patients, but few studies were performed to determine whether these assays are sufficiently robust to measure GAG levels in dried blood spots (DBS) of patients with MPS and ML.

Material and methods

In this study, we evaluated GAG levels in DBS samples from 124 MPS and ML patients (MPS I = 16; MPS II = 21; MPS III = 40; MPS IV = 32; MPS VI = 10; MPS VII = 1; ML = 4), and compared them with 115 age-matched controls. Disaccharides were produced from polymer GAGs by digestion with chondroitinase B, heparitinase, and keratanase II. Subsequently, dermatan sulfate (DS), heparan sulfate (HS-0S, HS-NS), and keratan sulfate (mono-sulfated KS, di-sulfated KS, and ratio of di-sulfated KS in total KS) were measured by MS/MS.

Results

Untreated patients with MPS I, II, VI, and ML had higher levels of DS compared to control samples. Untreated patients with MPS I, II, III, VI, and ML had higher levels of HS-0S; and untreated patients with MPS II, III and VI and ML had higher levels of HS-NS. Levels of KS were age dependent, so although levels of both mono-sulfated KS and di-sulfated KS were generally higher in patients, particularly for MPS II and MPS IV, age group numbers were not sufficient to determine significance of such changes. However, the ratio of di-sulfated KS in total KS was significantly higher in all MPS patients younger than 5 years old, compared to age-matched controls. MPS I and VI patients treated with HSCT had normal levels of DS, and MPS I, VI, and VII treated with ERT or HSCT had normal levels of HS-0S and HS-NS, indicating that both treatments are effective in decreasing blood GAG levels.

Conclusion

Measurement of GAG levels in DBS is useful for diagnosis and potentially for monitoring the therapeutic efficacy in MPS.

Introduction

Mucopolysaccharidoses (MPSs) are a group of lysosomal storage disorders (LSDs) caused by a deficiency of lysosomal hydrolases responsible for the catabolism of glycosaminoglycans (GAGs) [1], [2]. Mucolipidoses (ML) are related diseases caused by a deficiency of N-acetylglucosaminyl-1-phosphotransferase. This enzyme deficiency produces unphosphorylated lysosomal enzymes, which leads to inhibition of reuptake of enzymes and accumulation of GAGs and lipids. MPSs and ML are classified according to the enzyme deficiency (Table 1).

The MPSs and ML are progressive LSDs that share many clinical features such as: coarse faces, neurological impairment (MPS I, II, III, VII and ML II), skeletal dysplasia (all, but maybe mild in MPS III), hepatosplenomegaly, joint rigidity, and heart valvular disease [3]. MPSs are usually asymptomatic at birth, and the initial signs and symptoms appear with progression of the disease during the first one or two years of age. Mucolipidoses II (ML II; I-cell disease) is fatal during childhood or the first decade of life, and can even produce intra-uterine fractures, while ML III has a milder somatic phenotype with slower progression throughout childhood but leads to severe neurodegeneration with a fatal outcome during adulthood [2], [4].

ML II and III are caused by impaired trafficking of several lysosomal enzymes [2], [5]. The prevalence of ML is variable among different populations: 0.3 cases per 100,000 live births in Australia, 0.16 per 100,000 live births in Portugal, and 0.08 per 100,000 live births in the Netherlands [6], [7]. The incidence in Quebec, Canada is very high, 1:6184, due to a founder effect [8]. The combined incidence of MPSs is 1:25,000 live births, and therefore more common than ML [9].

Enzyme replacement therapy (ERT) is available commercially in practice for MPS I, II, VI, and IVA [10], [11], [12], [13]. Hematopoietic stem cell transplantation (HSCT) is recommended for MPS I [14], [15]. Several studies indicate that HSCT will also improve outcomes for MPS II [16], [17], [18], MPS IVA [19], [20], MPS VI [21] and MPS VII [22].

Levels of GAGs in patients with MPS have been studied for several decades. Initially, total urinary GAGs were measured using a variety of dye methods [23], [24], [25], [26], [27], [28], [29], [30], [31], [32], [33], [34]. Although these methods were useful and cost-effective; they gave high false-positive rates [35], could not be easily applied to measure GAGs in blood and/or tissues due to the presence of proteins and other interferent molecules, and could not distinguish specific GAG(s) [36]. Measurement of total urinary GAG using a dimethylmethylene blue (DMMB) method did not distinguish a substantial number of MPS IVA patients from age-matched controls [37], [38], [39], [40], [41]. The development of ELISA methods in early 90's made it possible to measure HS and KS in blood and urine of MPS and ML patients [40], [42], [43], [44]. We used an ELISA method to show that KS levels in blood are elevated not only in MPS IV, but also in other types of MPS and ML [44]. However, ELISA assays are expensive and cannot distinguish subclasses of HS and KS. Since 2001, protocols have been developed for GAG analysis using tandem mass spectrometry (MS/MS). Two main branches of GAG detection methods by MS/MS have been developed: detection of digested disaccharides (direct or labeled with aniline) [45], [46], [47], [48], [49], [50] and chemically depolymerized GAGs by methanolysis and/or butanolysis [51], [52], [53], [54], [55], [56]. Such MS/MS methods have been used to measure specific GAGs in blood and urine of MPS and ML patients [51], [54], [55], [57], [58], [59], [60], [61], [62], [63], [64], [65]. MS/MS provides a sensitive, specific, and reproducible GAG analysis and allows measurement of several GAGs simultaneously, indicating its potential for use in mass screening, prognosis, and monitoring therapeutic effect in patients with MPS and ML. More recently, MS/MS methods have been developed to measure GAGs in dried blood spots (DBS) [57], [61], [66], [67].

In this study, we have simultaneously determined levels of dermatan sulfate (DS), heparan sulfate (HS-0S, HS-NS), and keratan sulfate (mono, di-sulfated, and ratio di-sulfated in total KS) in DBS of control subjects and patients with MPS I, II, III, IV, VI, VII; and ML II and III by liquid chromatography tandem mass spectrometry (LC/MS/MS). We have also evaluated GAG levels in ERT and HSCT treated patients with some types of MPS.

Section snippets

Enzymes and standards

Chondroitinase B, heparitinase, keratanase II, chondrosine (internal standard-IS), and the unsaturated disaccharides: heparan ΔDi-0S [2-acetamido-2-deoxy-4-O-(4-deoxy-a-L-threo-hex-4-enopyranosyluronic acid)-d-glucose] (HS-0S), heparan ΔDi-NS [2-deoxy-2-sulfamino 4-O-(4-deoxy-α-L-threo-hex-4-enopyranosyluronic acid)-d-glucose] (HS-NS), chondro ΔDi-4S [2-acetamido-2-deoxy 3-O-(β-D-gluco-4-enepyranosyluronic acid)-4-O-D-sulfo-galactose] (Di-4S), mono-sulfated KS [Galβ1-4GlcNAc(6S)], and

Results

Ages of the patients with MPS were as follows: untreated MPS I (mean: 3 ± 2 years; range: 3 months to 5 years; n = 7), MPS I with ERT (mean: 26 ± 18 years; range: 1.1 to 37.9 years; n = 6), MPS I with HSCT (12.8 years; transplant age = 2.5 years, n = 1), MPS I with ERT + HSCT (ages 4 and 15 years; transplant age: 2.3 and 3.8 years; n = 2); untreated MPS II (mean: 9 ± 7 years; range: 1.1 to 29 years; n = 21); untreated MPS III (mean: 13 ± 8 years; range: 1 to 34.2 years; n = 37), MPS IIIA with HSCT (ages 8 and 17 years, transplant

Discussion

We have demonstrated the usefulness and significance of assay of disaccharides from DBS by LC/MS/MS as a diagnostic approach and therapeutic monitoring tool for MPS and ML. We measured several GAGs (DS, HS, and KS) simultaneously and found that the majority of untreated MPS and ML patients had higher levels of at least one GAG compared to age-matched controls. We have also shown a reduction of GAGs in patients treated with ERT and/or HSCT in MPS I, VI, and VII when compared to other untreated

Conflict of interest

Francyne Kubaski, Yasuyuki Suzuki, Kenji Orii, Roberto Giugliani, Heather J. Church, Robert W. Mason, Vũ Chí Dũng, Can Thi Bich Ngoc, Seiji Yamaguchi, Hironori Kobayashi, Katta M. Girisha, Toshiyuki Fukao, Tadao Orii, and Shunji Tomatsu declare that they have no conflict of interests.

Contributions to the project

Francyne Kubaski has contributed to the concept and planning of the project, collection of data, data analysis, the draft of the manuscript, and reporting of the work described.

Yasuyuki Suzuki has contributed to the concept and planning of the project, collection of samples, and reporting of the work described.

Kenji Orii has contributed to the concept and planning of the project, collection of samples, and reporting of the work described.

Roberto Giugliani has contributed to the concept and

Acknowledgements

This work was supported by grants from the Japanese MPS Society, the Austrian MPS Society, The Bennett Foundation, and International Morquio Organization (Carol Ann Foundation). R.W.M. and S.T. were supported by an Institutional Development Award (IDeA) from the National Institute of General Medical Sciences of NIH under grant numbers P20GM103464 and P30GM114736. S.T. was supported by National Institutes of Health grant 1R01HD065767. F.K. was supported by INAGEMP and Conselho Nacional de

References (88)

  • E.W. Gold

    The quantitative spectrophotometric estimation of total sulfated glycosaminoglycan levels. Formation of soluble alcian blue complexes

    Biochim. Biophys. Acta

    (1981)
  • R.W. Farndale et al.

    Improved quantitation and discrimination of sulphated glycosaminoglycans by use of dimethylmethylene blue

    Biochim. Biophys. Acta

    (1986)
  • H.K. Berry

    Screening for mucopolysaccharide disorders with the Berry spot test

    Clin. Biochem.

    (1987)
  • S. Björnsson

    Quantitation of proteoglycans as glycosaminoglycans in biological fluids using an alcian blue dot blot analysis

    Anal. Biochem.

    (1998)
  • C.B. Whitley et al.

    Urinary glycosaminoglycan excretion quantified by an automated semimicro method in specimens conveniently transported from around the globe

    Mol. Genet. Metab.

    (2002)
  • P. Mabe et al.

    Evaluation of reliability for urine mucopolysaccharidosis screening by dimethylmethylene blue and Berry spot tests

    Clin. Chim. Acta

    (2004)
  • C.B. Whitley et al.

    Maroteaux-Lamy syndrome (mucopolysaccharidosis type VI): a single dose of galsulfase further reduces urine glycosaminoglycans after hematopoietic stem cell transplantation

    Mol. Genet. Metab.

    (2010)
  • S. Tomatsu et al.

    Newborn screening and diagnosis of mucopolysaccharidoses

    Mol. Genet. Metab.

    (2013)
  • T. Oguma et al.

    Analytical method for keratan sulfates by high-performance liquid chromatography/turbo-ionspray tandem mass spectrometry

    Anal. Biochem.

    (2001)
  • T. Oguma et al.

    Analytical method of heparan sulfates using high-performance liquid chromatography turbo-ionspray ionization tandem mass spectrometry

    J. Chromatogr. B Biomed. Sci. Appl.

    (2001)
  • T. Oguma et al.

    Analytical method for the determination of disaccharides derived from keratan, heparan, and dermatan sulfates in human serum and plasma by high-performance liquid chromatography/turbo ionspray ionization tandem mass spectrometry

    Anal. Biochem.

    (2007)
  • C. Auray-Blais et al.

    Efficient analysis of urinary glycosaminoglycans by LC-MS/MS in mucopolysaccharidoses type I, II and VI

    Mol. Genet. Metab.

    (2011)
  • H. Zhang et al.

    A straightforward, quantitative ultra-performance liquid chromatography-tandem mass spectrometric method for heparan sulfate, dermatan sulfate and chondroitin sulfate in urine: an improved clinical screening test for the mucopolysaccharidoses

    Mol. Genet. Metab.

    (2015)
  • C. Auray-Blais et al.

    UPLC-MS/MS detection of disaccharides derived from glycosaminoglycans as biomarkers of mucopolysaccharidoses

    Anal. Chim. Acta

    (2016)
  • S. Tomatsu et al.

    Validation of disaccharide compositions derived from dermatan sulfate and heparan sulfate in mucopolysaccharidoses and mucolipidoses II and III by tandem mass spectrometry

    Mol. Genet. Metab.

    (2010)
  • T. Shimada et al.

    Novel heparan sulfate assay by using automated high-throughput mass spectrometry: application to monitoring and screening for mucopolysaccharidoses

    Mol. Genet. Metab.

    (2014)
  • C. Auray-Blais et al.

    An improved method for glycosaminoglycan analysis by LC-MS/MS of urine samples collected on filter paper

    Clin. Chim. Acta

    (2012)
  • J. de Ruijter et al.

    Heparan sulfate and dermatan sulfate derived disaccharides are sensitive markers for newborn screening for mucopolysaccharidoses types I, II and III

    Mol. Genet. Metab.

    (2012)
  • G. Baldo et al.

    Placenta analysis of prenatally diagnosed patients reveals early GAG storage in mucopolysaccharidoses II and VI

    Mol. Genet. Metab.

    (2011)
  • D.J. Rowan et al.

    Long circulating enzyme replacement therapy rescues bone pathology in mucopolysaccharidosis VII murine model

    Mol. Genet. Metab.

    (2012)
  • S. Tomatsu et al.

    Therapies for the bone in mucopolysaccharidoses

    Mol. Genet. Metab.

    (2015)
  • T. Shimada et al.

    Novel heparan sulfate assay by using automated high-throughput mass spectrometry: application to monitoring and screening for mucopolysaccharidoses

    Mol. Genet. Metab.

    (2014)
  • N. Elizabeht et al.

    The Mucopolysaccharidoses

    (2001)
  • T. Braulke et al.

    I-cell disease, pseudo-Hurler polydystrophy: disorders of lysosomal enzyme phosphorylation, localization

  • S.S. Cathey et al.

    Phenotype and genotype in mucolipidoses II and III alpha/beta: a study of 61 probands

    J. Med. Genet.

    (2010)
  • S. Tomatsu et al.

    Heparan sulfate levels in mucopolysaccharidoses and mucolipidoses

    J. Inherit. Metab. Dis.

    (2005)
  • R. Pinto et al.

    Prevalence of lysosomal storage diseases in Portugal

    Eur. J. Hum. Genet.

    (2004)
  • M.F. Coutinho et al.

    Origin and spread of a common deletion causing mucolipidosis type II: insights from patterns of haplotypic diversity

    Clin. Genet.

    (2011)
  • M. De Braekeleer

    Hereditary disorders in Saguenay-Lac-St-Jean (Quebec, Canada)

    Hum. Hered.

    (1991)
  • P. Harmatz et al.

    Enzyme replacement therapy for mucopolysaccharidosis VI: a phase 3, randomized, double-blind, placebo-controlled, multinational study of recombinant human N-acetylgalactosamine 4-sulfatase (recombinant human arylsulfatase B or rhASB) and follow-on, open-label extension study

    J. Pediatr.

    (2006)
  • C.J. Hendriksz et al.

    Efficacy and safety of enzyme replacement therapy with BMN 110 (elosulfase alfa) for Morquio A syndrome (mucopolysaccharidosis IVA): a phase 3 randomised placebo-controlled study

    J. Inherit. Metab. Dis.

    (2014)
  • J. Muenzer et al.

    Mucopolysaccharidosis I: management and treatment guidelines

    Pediatrics

    (2009)
  • R. Giugliani et al.

    Mucopolysaccharidosis I, II, and VI: brief review and guidelines for treatment

    Genet. Mol. Biol.

    (2010)
  • E. Herskhovitz et al.

    Bone marrow transplantation for Maroteaux-Lamy syndrome (MPS VI): long-term follow-up

    J. Inherit. Metab. Dis.

    (1999)
  • Cited by (0)

    View full text