Mini Review
J Blood Disord. 2014;1(2): 6.
Hematopoietic Stem Cell Therapy and Novel Approaches for Mucopolysaccharidoses
Edwards TN1 and Louboutin JP2*
1Faculty of Medical Sciences, University of the West Indies, Jamaical
2Department of Basic Medical Sciences, University of the West Indies, Jamaica
*Corresponding author: Louboutin JP, Department of Basic Medical Sciences, University of the West Indies, Mona Campus, Kingston, Jamaica.
Received: September 01, 2014; Accepted: September 22, 2014; Published: September 23, 2014
Abstract
Mucopolysaccharidoses (MPS) are a group of inherited genetic linked enzyme deficiency lysosomal storage disorders. The deficiency of the enzymeis caused by a gene mutation that leads to the accumulation of harmful Glycosaminoglycans (GAGs) in the cells, tissues and blood. Disease-mutation leads to abnormal accumulation of GAGs: Dermatan Sulfate (DS), Heparan Sulfate (HS), chondroitin sulfate, and keratan sulfate resulting in clinical manifestations of varying severity. Recent advances have shown abnormal HS involvement in Central Nervous System (CNS) dysfunction. Therapeutic options vary according to the type of MPS. In severe MPS I patients, the best therapeutic approach is Hematopoietic Stem Cell Transplantation (HSCT). Unlike in other MPS, when it is performed in MPS I patients, HSCT has the ability to continuously produce the deficient enzyme that crosses the blood-brain barrier, a useful effect for CNS defects and in fine for mitigating cognitive and developmental delays. Pharmacological options include Enzyme Replacement Therapy (ERT). Three recombinant human enzymes have so far been approved by the US Food and Drug Administration (FDA) for the treatment of MPS: laronidase for MPS I, idursulfase for MPS II, and galsulfase for MPS VI. However, some of the results of ERT are still controversial. Finally, novel therapeutic avenues (i.e., substrate reduction therapy) have been recently proposed. We review here the different therapeutic approaches proposed in MPS, focusing on MPS I, the MPS that is benefiting most from HSCT so far.
Keywords: Mucopolysaccharidosis; Stem cell therapy; Bone marrow transplantation
Introduction and Epidemiology
Mucopolysaccharidoses (MPS) are a group of genetic-linkedmetabolic diseases that lead to significant morbidity and fatality in children and adults. MPS are a category of lysosomal storage disorder and comprises of seven clinical types (I, II, III, IV and VI, VII, IX) with specific gene mutations in eleven different lysosomal enzymes. The incidence of MPS has an estimate of 1:50,000 to 1:250,000 with MPSIII (Sanfillippo Syndrome), the most common and MPS VII (Sly disease) the rarest [1]. MPS are autosomal recessive with the exception for MPS II (Hunter Syndrome), which is X-linked. They are due to enzymatic deficit resulting in cellular damage with clinical features of varying degrees of severity [2,3]. In the case of MPS I patients, genetic mutations in the alpha-L-Iduronidase (IDUA) gene (located on chromosome 4p16.3) can be homozygous or heterozygous and are the causative mutation in 95% of cases [4]. According to the Human Gene Mutation Database, MPS I has 110 mutations in the IDUA gene with the majority of nonsense, missense mutations, splicing or small deletions/insertions [5,6]. In MPS I patients, there are more than 30 polymorphisms or nonpathogenic sequence variants within the iduronidase gene that modify the severity of the clinical disease presentation with a pathogenic allele [6,7]. MPS I alleles have variations among different ethnic populations with two nonsense mutations W402X and Q70X alleles found in majority of Europeans (61 to 72% mutations). The mutations found in the European population are rare among Japanese, Koreans, or Moroccan patients[6,8]. Specifically, in the Japanese population, there are two types of missense mutations that are conserved in patients displaying mild MPS I (Scheie) phenotype, while in India, other mutations are responsible for mild forms of MPS I [2]. Patients displaying the more severe phenotype of MPS I (Hurler), are usually homozygous for a nonsense allele or two different nonsense alleles, while milder MPS I forms (Hurler-Scheie and Scheie) have around 1 missense or null mutations [6,7]. Gene locations of other MPS are indicated in Table 1.
MPS are progressive in nature with clinical presentations of multisystemic, chronic and severe complications [3]. MPS features are presented in Table 1. For example, MPS I is separated into three different diseases based on their clinical presentation: Hurler (severe), Hurler-Scheie syndrome (intermediate) and Scheie syndrome (milder) [6]. According to their type, MPS I can be apparent in utero, in children or adults. MPS I am caused by the deficiency of lysosomal enzyme, IDUA, with subsequent improper degradation of Dermatan Sulfate (DS) and Heparan Sulfate (HS) [6]. The clinical manifestations of MPS I include progressive cardiac and pulmonary disease, inguinal and umbilical hernias, corneal clouding and musculoskeletal disease. Patients with MPS I (Hurler), the most severe phenotype, suffer from a progressive intellectual decline with CNS involvement, cognitive abnormalities and limited life expectancy [6]. Diagnoses are based on the accumulation of GAGs in the urine, a sensitive, but nonspecific, test. A more precise diagnosis involves the analysis of the deficient IDUA activity in the fibroblasts, leukocytes, serum or blood spots[6,9-11]. Prenatal screening is done through enzyme and DNA testing and early diagnosis such as newborn screening can improves disease outcome [6]. Diagnosis of MPS I patients are made between 4 and 18 months based on orthopedic and visceral signs. Births are usually normal but infants can present a giant mongoloid spot; cognitive dysfunction is the hallmark, but can be normal or slightly impaired according to the MPS I phenotypes [1]. Clinical manifestations and types of accumulated products of other MPS are featured in Table 1.
Type
Common name
Incidence
Gene
Locus
Deficient enzyme
Accumulated products
Symptoms
MPS IH
Hurler syndrome
1:100,000
IDUA
4p16.3
α-L-iduronidase
Heparan sulfate
Dermatan sulfate
Cardiomyopathy,mental retardation, coarse facial features, macroglossia, corneal clouding, micrognathia
MPS IH/S
Hurler-Scheie syndrome
MPS IS
Scheie syndrome or MPS type V
MPS II
Hunter syndrome
1:250,000
IDS
Xq28
Iduronate sulfatase
Heparan sulfate
Dermatan sulfate
Mental retardation
MPS IIIA
Sanfilippo syndrome A
Sulfamidase deficiency
1:280,000-1:50,000
SGSH
17q25.3
Heparansulfamidase
Heparan sulfate
Motor dysfunction, developmental delay, spasticity, hyperactivity, deathy by second decade
MPS IIIB
Sanfilippo syndrome B
NAGLU deficiency
NAGLU
17q21.2
N-acetylglucosaminidase
MPS IIIC
Sanfilippo syndrome C
HGSNAT
8p11.21
Heparan-?-glucosaminide N-acetyltransferase
MPS IIID
Sanfilippo syndrome D
GNS
12q14.3
N-acetylglucosamine 6-sulfatase
MPS IVA
Morquio syndrome A
1:75,000
GALNS
16q24.3
Galactose-6-sulfate sulfatase
Keratan sulfate
Chondroitin 6-sulfate
Motor dysfunction, skeletal dysplasia, short stature
MPS IVB
Morquio syndrome B
GLB1
3p22.3
β-galactosidase
Keratan sulfate
MPS V
MPS IS
IDUA
4p16.3
α-L-iduronidase
Heparan sulfate
Dermatan sulfate
See MPS I
MPS VI
Maroteaux-Lamy syndrome
ARSB deficiency
ARSB
5q14.1
N-acetylgalactosamine-4-sulfatase
Dermatan sulfate
Kyphosis, motor dysfunction, heart defects, skeletal dysplasia, short stature
MPS VII
Sly syndrome
GUSB deficiency
1:250,000
GUSB
7q11.21
β-glucuronidase
Heparan sulfate
Dermatan sulfate
Chondroitin
4,6-sulfate
Developmental delay, short stature, corneal clouding, hepatomegaly, skeletal dysplasia
MPS IX
Natowicz syndrome
Hyaluronidase deficiency
HYAL1
3p21.31
Hyaluronidase
Hyaluronic acid
Painful swelling of nodular soft-tissue masses around joints
Table 1: Mucopolysaccharidosis: clinical signs, genetic and molecular abnormalities.
GAGs
The different GAGs involved in MPS, with their corresponding diseases, are featured in Table 1. GAGs are large protein polymers consisting of multiple branches of carbohydrates. They are components of proteoglycans that provide the structural support to the Extracellular Matrix (EM) and structures that are cartilaginous in nature, such as heart valves and joints. GAGs are also involved in the communication and regulation of cellular components. In MPS, inborn defects in the production of lysosomal enzymes result in a progressive accumulation of undegraded or partially degraded GAGs. Consequently, a destabilization of the lysosome occurs, leading to the disturbances of normal cell function and metabolism in the peripheral organs and CNS. Disease-mutation causes harmful accumulation of GAGs in cells, connective tissue and blood [1,3,11].
The main GAGs found in MPS are Heparan Sulfate (HS),Dermatan Sulfate (DS), Keratan Sulfate (KS) or Chondroitin Sulfate (CS). The lysosomal enzyme, alpha-L-iduronidase is involved in the degradation of certain GAGs such as Heparan Sulfate (HS) and Dermatan Sulfate (DS) [2]. It is not completely understood how the accumulation of GAGs that is due to specific enzyme deficiency leads to clinical symptoms. Both primary and secondary effects of the deficiency of HS/DS breakdown and altered cellular function respectively underlie disease manifestations. DS is a component of both connective tissue and skin and HS is involved in angiogenesis and associated with CNS dysfunction [6]. HS and DS are involved in MPS I, II, III, V, VI, VII, and we will describe them in further details.
HS, abundantly present on the cell surface and in the EM, interacts with growth factors, growth factor binding proteins, extracellular proteases, protease inhibitors, chemokines, and adhesive proteins. HS regulates the activity, gradient formation and the stability of ligand-receptor interactions in different cells [12-16]. HS is also involved with the formation of new blood vessels during embryonic development and facilitates the action of pro- and anti angiogenic factors during vascular formation as shown in studies using zebra fish and mouse models. Studies in animal models showed a HS-based modulation of the signaling of growth factors like VEGF-A [16,17], PDGF-B [16,18-20] and TGF-β [16,21,22] in order to promote steps in angiogenesis [16].
Excess accumulation of HS severely affects the CNS by inducing cerebral dysfunctions, deregulating major cell migrations, axonguidance, neuritogenesis, synaptogenesis, and structural plasticity of neuronal cells [1,3,23]. Studies in MPS I mice showed an increase in HS accumulation, lysosomal storage, neuroinflammation, astrocytosis, microgliosis [23]. Lipid accumulation is present in the brains of Hurler patients with mental retardation but is not found in Scheie patients with normal intelligence. The accumulation of GAGs may be associated with the infiltration of macrophages of the arachnoid villi and thickening of the leptomeninges in the brain. The increased thickness of leptomeninges may be a result of defective pacchionian granulations, causing slowly progressive communicating hydrocephalus. MRIs in MPS I patients show abnormal thickening of the leptomeninges in the brain, enlargement of ventricles, subarachnoid space and Perivascular Spaces (PVS). The enlargement of the PVS is due to a meshwork of fibrous tissue and cells with abnormal storage of GAGs which may result to a disorder of the movement and absorption of cerebrospinal fluid [1,24].
Dermatan Sulfate (DS) is a GAG mainly found in tendons, blood vessels, heart valves, skin and lungs. DS has different roles in fibrosis, coagulation, infection, wound repair, cardiovascular disease and carcinogenesis. An accumulation of DS in the mitral valve can lead to a myxomatous degeneration of the leaflets and finally to mitral valve prolapse into the left atrium and cardiac insufficiency (mainly by left heart failure).
Keratan Sulfate (KS) can be found in cartilage, bone and cornea. KS can also be found in the CNS where it is involved in brain development and glial scar formation following an injury. KS are large molecules acting as cushions to absorb mechanical shocks. The amount of KS depends on the tissue where it is found: KS is 10 times more abundant in cornea than in cartilage and 2-4 times more in cornea than in other tissues.
Chondroitin Sulfate (CS) is part of cartilage and provides resistance to compression. CS interacts with proteins of EM. In the brain, CS levels are increased after injury and act to prevent regeneration of damaged nerve endings. CS is also involved in cortical development where it acts as a stop signal for neurons migrating from the ventricular zone. In chondrocytes, CS reduces IL-1beta-induced-NF kappa B translocation. Role of proteoglycans of CS are not yet as well understood compared to the ones HS.
Treatments of MPS
Non-pharmacological therapies: Hematopoietic stem celltransplantation (HSCT)
Transplantation of Hematopoietic Stem Cells (HSCs) can significantly increase survival rates in MPS patients, more specifically in severe MPS I (Hurler) patients [6,25]. HSCT therapy has been performed for years in both animal models and human patients. HSCT is based on the transplantation of Bone Marrow (BM), Peripheral Blood Stem Cells (PBSC) or Umbilical Cord Blood (UCB). HSCT has had major challenges involving finding compatible stem cells, HLA match for bone marrow transplantation and high morbidity and mortality rates associated with the failure to achieve complete engraftment. Graft-Versus-Host Disease (GVHD) is another complication related to stem cell or BM transplantation. To address HSCT challenges, umbilical cord blood uses safe and effective source of stem cells [26,27]. Umbilical cord blood is more readilyavailable and requires less strict HLA matching as bone marrow transplantation. Better tissue matching techniques involving graftversus- host prophylaxis have been a major improvement in reducing transplant-related morbidity and mortality rates.
HSCT provides normal and bone-derived cells that allow the continuous release of the deficient enzyme available for uptake by cells, which consequently reduces excess accumulation of GAGs in tissues [11,28,29]. The success of monocyte-derived cells is associated with the ability to cross the blood-brain barrier, incorporate in microglia and secrete enzyme in neurons [29-34]. The first successful treatment of allogeneic HSCT was performed on a patient with MPS I in 1980 [26,27]. Thirteen months after treatment, there was a decrease in IDUA in plasma, reduction of hepatosplenomegaly and corneal clouding and an arrest in the developmental deterioration. Twenty years later, the patient had full engraftment and an improved intelligence [35]. Since then, hundreds of severe MPS I (Hurlersyndrome) patients have undergone HSCT treatment, primarily from bone marrow, but increasingly from umbilical cord blood [6,36].
To optimize successful engraftment in MPS I patients, pretransplantation preparation involves sufficient immunosuppression and myeloablation [11]. HSCT is effective, but there are limits to the treatment of bone involvement due to cartilage cells deriving from mesenchymal stem cells and HSCT does not provide sufficient amounts of enzymes to cartilage. This occurs because cartilage is a vascular and it is difficult for circulating enzymes to reach growth plates and cartilaginous tissues. Moreover, the rate of diffusion is affected because of the large molecular weight of lysosomal hydrolases [37-41].
Overall, HSCT is useful in significantly improving progressive degeneration of the CNS and in reducing cognitive dysfunction in patients with severe MPS I [25]. The treatment is most effective during early administration at beginning of disease onset and developmental deterioration. Efficacy of treatment at the CNS level is associated with early treatment and with graft-cells producing deficient enzyme partly in microglia cells [6,11]. HSCT also significantly reduces complications in the peripheral organs of MPS I patients. The effectiveness of HSCT may be due to the local presence of graftderived cells (Kupffer cells in the liver) or the uptake of the deficient enzymes [11].
In severely affected MPS I (Hurler) patients under 2 years with developmental quotient of less than 70% normal, the recommended treatment option is HSCT. However, HSCT is not recommended for patients above 2, due to the lack of compatible donors, graft rejection, and GVHD [11]. Successful HSCT therapies significantly improve survival years from 6.8 years to 20 years and beyond. Importantly, when HSCT is performed early in Hurler patients, neurocognition stops declining and improves overtime [25-27,36]. There are also improvements in the resolution of hepatosplenomegaly, reduction in sleep apnea and upper airway disease, preservation of hearing, upper extremities movements and a reduction of GAGs in the urine. However, MPS I patients treated with HSCT continued to have a decline in musculoskeletal disease, worsened vision, cardiac valve disease and stunted growth. [27,39]. In other milder MPS I types, HSCT results are not as effective in neurocognitive involvement as in severe MPS I (Hurler) patients [25,27,35,42-45].Unlike in Hurler syndrome, HSCT may not be effective in the other MPS diseases in which diagnosis is made in children older than 2 years. At that time, CNS disease may be irreversible, especially in MPS III patients, who show developmental delay and motor dysfunction around age 3 with very little somatic disease [27,45]. The efficacy of HSCT in MPS II is still debated [3]. Other MPS disorders including MPS IV (Morquio) and VI (Maroteaux-Lamy) present normal cognitive function and development but present somatic disorders. In these MPS, HSCT would not be the best treatment option because of the increased morbidity and mortality rates due to this procedure, which outweighs the benefits of treating the somatic complications [3,27].
Pharmacological therapies
Partly because of the side effects of HSCT, Enzyme Replacement Therapy (ERT) has been proposed for the treatment of MPS. Three recombinant human enzymes have been approved so far by the US Food and Drug Administration (FDA) for the treatment of MPS: laronidase (Aldurazyme®) for MPS I, idursulfase (Elaprase®) for MPS II, and galsulfase (Naglazyme®) for MPS VI. Once injected intravenously, these enzymes are internalized to reach their target in lysosomes to replace the defective enzymes. ERT can control somatic manifestations of MPS, including joints abnormalities, organ enlargement and pulmonary insufficiency. However, there are currently two limitations of ERT: first, enzymes cannot cross the blood-brain barrier and consequently, ERT cannot correct brain abnormalities; secondly, heart valves and bones are relatively resistant to ERT.
+ MPS I
Recently, Enzyme Replacement Therapy (ERT) was used alone or in conjunction with HSCT. Laronidase is an ERT drug that is an analogue of human alpha-L-iduronidase produced in the CHO (Chinese hamster Ovary) cells [25,27]. In the treatment of MPS I, laronidase can be used in conjunction with HSCT for a short term during transplant therapy typically up to 6 weeks, leading up to engraftment [27,35,42,44,45]. This particular recombinant enzyme demonstrated in clinical trials [46-49] to be effective in the reduction of GAGs accumulation in the liver and the urine [27,36]. The use of iduronidase drug therapy has limitations due to the inability to cross the blood brain barrier, which limits its effectiveness in the CNS of patients with Hurler syndrome. The use of HSCT and laronidase is used in combination to improve treatment and clinical status for MPS I patients and is effective during transplant improving the likelihood of the success of the HSCT treatment [27,43,50,51].
ERT should be offered to MPS I patients at the time of diagnosis and continued until severe neuropathy or any other condition appears that may affects prognosis. Laronidase should be given if the patient fulfills one of the following criteria: patient with age > 2 years; patient with age < 2 years but who is expected to have attenuated phenotype; patient with age < 2 years and who is expected to have a severe phenotype with intelligence quotient of less than 70 [6].
+ MPS II
Idursulfase (Elaprase®) is a recombinant human iduronate-2- sulphatase approved for the treatment of MPS II. Clinical trials showed prolonged improvements in forced vital capacity (particularly when ERT is given weekly). Spleen and liver size, and GAGs levels in urine[52,53] were reduced under treatment by idursulfase. Guidelines are still discussed [3].
+ MPS VI
Galsulfase (Naglazyme®) is a recombinant human N-acetylgalactosamine 4-sulfatase (rhASB). It is approved for the treatment of MPS VI. Galfulfase has to be initiated at the beginning of the course of the disease, and continued until neuropathy compromises its efficacy.
Some positive results were noticed in Phase 1/2 and Phase 2 clinical trials [54,55], leading to a Phase 3 study. Primary and secondary endpoints (both involving locomotion) were improved in the Phase 3 trial [56,57]. Beneficial effects were observed during at least 240 weeks. Respiratory function is also improved with galsulfase [58]. Side effects were negligible.
Novel Therapeutic Approaches
+ Substrate reduction therapy
The goal of Substrate Reduction Therapy (SRT) is to reduce and possibly inhibit the synthesis of GAGs using small molecules
Among the molecules used, Genistein, a plant isoflavone, has demonstrated potential for blocking the Epidermal Growth Factor (EGF)-mediated signal transduction responsible for the expression of genes coding for the enzymes synthesizing GAGs. Genistein can reduce GAGs accumulation and neuroinflammation in brains of MPS II [59] and MPS III mice [60]. Clinical trials in small groups of patients with MPS II and III showed improvements in joint movements and cognition respectively [61], but the long term efficacy of Genistein is not totally established yet [3].
The fluorescent dye Rhoda mine B can inhibit the elongation of GAGs chain. Rhoda mine B has been shown to interrupt GAGs synthesis in cell cultures of MPS III and VI. It can also improve cognition in MPS IIIA mice [62]. Possible side effects of Rhoda mine B need further investigations to assess its toxicity [3].
Miglustat (Zavesca®) has been primarily used for the treatment of Niemann-Pick type C disease and Gaucher's disease [63]. Miglustat inhibits the synthesis of glucosylceramide synthase, a precursor of GM2 ganglioside. Miglustat may play a role in improving neurological deterioration seen in MPS [64]. However, its efficacy has not been fully shown in MPS to date [3].
+ Gene therapy
Gene therapy provides the transfer of relevant cDNA to produce and secrete the deficient enzyme for uptake in cells. Different vectors carrying the missing gene were used in animal models of MPS [65]. Similarly, different ways of gene delivery have been tested so far [66-69].
+ Other therapies
Other treatments include small-molecules therapies such as chaperone molecules, and stop cod on read-through therapies [29]. All available therapeutic approaches use the phenomena of cross reference correction. This allows the reuptake of exogenous enzyme and deliver it to the lysosome via mannose-6-phosphate receptormediated process but most are limited by their inability to cross the blood-brain barrier [29,70].
However, other treatment options such as exogenous enzyme replacement therapy, have challenges with severe MPS I patients (Hurler), due to their failure to correct neurocognitive disorder, because of their inability to cross the blood brain barrier [25].
Conclusion
MPS are diseases leading to the harmful accumulation of GAGs in cells, tissues and blood. The accumulation of GAGs has adverse effects with somatic and clinical manifestation, particularly involving the CNS. HSCT has been the most successful approach in treating MPS I, especially Hurler, the more severe phenotype. Even though challenges are present, HSCT efficacy is due to its ability to provide enzymes able to cross the blood-brain barrier, mitigating the decline of cognitive functions and developmental delays, manifestations that were a major challenge to many of the other treatments for MPS patients.
References
- Heron B. [Neurologic manifestations in mucopolysaccharidoses]. Arch Pediatr. 2014; 21: 14-21.
- Lacombe D, Germain DP. [Genetic aspects of mucopolysaccharidoses]. Arch Pediatr. 2014; 21: 22-26.
- Noh H, Lee JI. Current and potential therapeutic strategies for mucopolysaccharidoses. J Clin Pharm Ther. 2014; 39: 215-224.
- Scott HS, Litjens T, Hopwood JJ, Morris CP. A common mutation for mucopolysaccharidosis type I associated with a severe Hurler syndrome phenotype. Hum Mutat. 1992; 1: 103-108.
- Stenson PD, Ball EV, Mort M, Phillips AD, Shiel JA, Thomas NS, et al. Human Gene Mutation Database (HGMD): Hum Mutat. 2003; 21: 577-581.
- Muenzer J, Wraith JE, Clarke LA. International Consensus Panel on Management and Treatment of Mucopolysaccharidosis I. Mucopolysaccharidosis I: management and treatment guidelines. Pediatrics. 2009; 123: 19-29.
- Scott HS, Nelson PV, Litjens T, Hopwood JJ, Morris CP. Multiple polymorphisms within the alpha-L-iduronidase gene (IDUA): implications for a role in modification of MPS-I disease phenotype. Hum Mol Genet. 1993; 2: 1471-1473.
- Terlato NJ, Cox GF. Can mucopolysaccharidosis type I disease severity be predicted based on a patient's genotype? A comprehensive review of the literature. Genet Med. 2003; 5: 286-294.
- Hall CW, Liebaers I, Di Natale P, Neufeld EF. Enzymic diagnosis of the genetic mucopolysaccharide storage disorders. Methods Enzymol. 1978; 50: 439-456.
- Chamoles NA, Blanco MB, Gaggioli D, Casentini C. Hurler-like phenotype: enzymatic diagnosis in dried blood spots on filter paper. Clin Chem. 2001; 47: 2098-2102.
- Caillaud C. [Principles of therapeutic approaches for mucopolysaccharidoses]. Arch Pediatr. 2014; 21: 39-45.
- Bernfield M, Gotte M, Park PW, Reizes O, Fitzgerald ML, Lincecum J, et al. Functions of cell surface heparan sulfate proteoglycans. Annu Rev Biochem. 1999; 68: 729-777.
- Bishop JR, Schuksz M, Esko JD. Heparan sulphate proteoglycans fine-tune mammalian physiology. Nature. 2007; 446: 1030-1037.
- Esko JD, Selleck SB. Order out of chaos: assembly of ligand binding sites in heparan sulfate. Annu Rev Biochem. 2002; 71: 435-471.
- Forsberg E, Kjellen L. Heparan sulfate: lessons from knockout mice. J Clin Invest. 2001; 108: 175-180.
- Fuster MM, Wang L. Endothelial heparan sulfate in angiogenesis. Prog Mol Biol Transl Sci. 2010; 93: 179-212.
- Chen E, Stringer SE, Rusch MA, Selleck SB, Ekker SC. A unique role for 6-O sulfation modification in zebra fish vascular development. Dev Biol. 2005; 284: 364-376.
- Lindblom P, Gerhardt H, Liebner S, Abramsson A, Enge M, Hellstrom M, et al. Endothelial PDGF-B retention is required for proper investment of pericytes in the microvessel wall. Genes Dev. 2003; 17: 1835-1840.
- Lindahl P, Johansson BR, Levéen P, Betsholtz C. Pericyte loss and microaneurysm formation in PDGF-B-deficient mice. Science. 1997; 277: 242-245.
- Abramsson A, Lindblom P, Betsholtz C. Endothelial and nonendothelial sources of PDGF-B regulate pericyte recruitment and influence vascular pattern formation in tumors. J Clin Invest. 2003; 112: 1142-1151.
- Hirschi KK, Rohovsky SA, D'Amore PA. PDGF, TGF-beta, and heterotypic cell-cell interactions mediate endothelial cell-induced recruitment of 10T1/2 cells and their differentiation to a smooth muscle fate. J Cell Biol. 1998; 141: 805-814.
- Hirschi KK, Burt JM, Hirschi KD, Dai C. Gap junction communication mediates transforming growth factor-beta activation and endothelial-induced mural cell differentiation. Circ Res. 2003; 93: 429-437.
- Wilkinson FL, Holley RJ, Langford-Smith KJ, Badrinath S, Liao A, Langford-Smith A, et al. Neuropathology in mouse models of mucopolysaccharidosis type I, IIIA and IIIB. PLoS One. 2012; 7: 35787.
- Matheus MG, Castillo M, Smith JK, Armao D, Towle D, Muenzer J. Brain MRI findings in patients with mucopolysaccharidosis types I and II and mild clinical presentation. Neuroradiology. 2004; 46: 666-672.
- Prasad VK, Kurtzberg J. Transplant outcomes in mucopolysaccharidoses. Semin Hematol. 2010; 47: 59-69.
- Staba SL, Escolar ML, Poe M, Kim Y, Martin PL, Szabolcs P, et al. Cord-blood transplants from unrelated donors in patients with Hurler's syndrome. N Engl J Med. 2004; 350: 1960-1969.
- Valayannopoulos V, Wijburg FA. Therapy for the mucopolysaccharidoses. Rheumatology (Oxford). 2011; 50: 49-59.
- 28. Haskins ME, Giger U. Lysosomal storage diseases. Academic Press; New York: 1997.
- Haskins ME. Animal models for mucopolysaccharidosis disorders and their clinical relevance. Acta Paediatr Suppl. 2007; 96: 56-62.
- Dial SM, Byrne T, Haskins M, Gasper PW, Rose B, Wenger DA, et al. Urine glycosaminoglycan concentrations in mucopolysaccharidosis VI-affected cats following bone marrow transplantation or leukocyte infusion. Clin Chim Acta. 1997; 263: 1-14.
- Simonaro CM, Haskins ME, Kunieda T, Evans SM, Visser JW, Schuchman EH. Bone marrow transplantation in newborn rats with mucopolysaccharidosis type VI: biochemical, pathological, and clinical findings. Transplantation. 1997; 63: 1386-1393.
- Norrdin RW, Simske SJ, Gaarde S, Schwardt JD, Thrall MA. Bone changes in mucopolysaccharidosis VI in cats and the effects of bone marrow transplantation: mechanical testing of long bones. Bone. 1995; 17: 485-489.
- Gasper PW, Thrall MA, Wenger DA, Macy DW, Ham L, Dornsife RE, et al. Correction of feline arylsulphatase B deficiency (mucopolysaccharidosis VI) by bone marrow transplantation. Nature. 1984; 312: 467-469.
- Turner AS, Norrdin RW, Gaarde S, Connally HE, Thrall MA. Bone mineral density in feline mucopolysaccharidosis VI measured using dual-energy X-ray absorptiometry. Calcif Tissue Int. 1995; 57: 191-195.
- Lee V, Li CK, Shing MM, Chik KW, Lam CW, Tsang KS, et al. Umbilical cord blood transplantation for Maroteaux-Lamy syndrome (mucopolysaccharidosis type VI). Bone Marrow Transplant. 2000; 26: 455-458.
- Peters C, Shapiro EG, Krivit W. Hurler syndrome: past, present, and future. J Pediatr. 1998; 133: 7-9.
- Koç ON, Day J, Nieder M, Gerson SL, Lazarus HM, Krivit W. Allogeneic mesenchymal stem cell infusion for treatment of metachromatic leukodystrophy (MLD) and Hurler syndrome (MPS-IH). Bone Marrow Transplant. 2002; 30: 215-222.
- Kingma SD, Wagemans T, IJlst L, Wijburg FA, van Vlies N. Genistein increases glycosaminoglycan levels in mucopolysaccharidosis type I cell models. J Inherit Metab Dis. 2014; 37: 813-821.
- Aldenhoven M, Boelens JJ, de Koning TJ. The clinical outcome of Hurler syndrome after stem cell transplantation. Biol Blood Marrow Transplant. 2008; 14: 485-498.
- Baldo G, Mayer FQ, Martinelli BZ, de Carvalho TG, Meyer FS, de Oliveira PG, et al. Enzyme replacement therapy started at birth improves outcome in difficult-to-treat organs in mucopolysaccharidosis I mice. Mol Genet Metab. 2013; 109: 33-40.
- Oussoren E, Brands MM, Ruijter GJ, der Ploeg AT, Reuser AJ. Bone, joint and tooth development in mucopolysaccharidoses: relevance to therapeutic options. Biochim Biophys Acta. 2011; 1812: 1542-1556.
- Guffon N, Bertrand Y, Forest I, Fouilhoux A, Froissart R. Bone marrow transplantation in children with Hunter syndrome: outcome after 7 to 17 years. J Pediatr. 2009; 154: 733-737.
- Boelens JJ, Rocha V, Aldenhoven M, Wynn R, O'Meara A, Michel G, et al. Risk factor analysis of outcomes after unrelated cord blood transplantation in patients with hurler syndrome. Biol Blood Marrow Transplant. 2009; 15: 618-625.
- Yamada Y, Kato K, Sukegawa K, Tomatsu S, Fukuda S, Emura S, et al. Treatment of MPS VII (Sly disease) by allogeneic BMT in a female with homozygous A619V mutation. Bone Marrow Transplant. 1998; 21: 629-634.
- Valstar MJ, Ruijter GJ, van Diggelen OP, Poorthuis BJ, Wijburg FA. Sanfilippo syndrome: a mini-review. J Inherit Metab Dis. 2008; 31: 240-252.
- Wraith JE, Clarke LA, Beck M, Kolodny EH, Pastores GM, Muenzer J, et al. Enzyme replacement therapy for mucopolysaccharidosis type I: a randomized, double-blinded, placebo-controlled. Multinational study of recombinant human alpha-L-iduronidase (laronidase). J Pediatr. 2004; 144: 581-588.
- Wraith JE, Beck M, Lane R, van der Ploeg A, Shapiro E, Xue Y, et al. Enzyme replacement therapy in patients who have mucopolysaccharidosis I and are younger than 5 years: results of a multinational study of recombinant human alpha-L-iduronidase (laronidase). Pediatrics. 2007; 120: 37-46.
- Clarke LA, Wraith JE, Beck M, Kolodny EH, Pastores GM, Muenzer J, et al. Long-term efficacy and safety of laronidase in the treatment of mucopolysaccharidosis I. Pediatrics. 2009; 123: 229-240.
- Sifuentes M, Doroshow R, Hoft R, Mason G, Walot I, Diament M, et al. A follow-up study of MPS I patients treated with laronidase enzyme replacement therapy for 6 years. Mol Genet Metab. 2007; 90: 171-180.
- Tolar J, Grewal SS, Bjoraker KJ, Whitley CB, Shapiro EG, Charnas L, et al. Combination of enzyme replacement and hematopoietic stem cell transplantation as therapy for Hurler syndrome. Bone Marrow Transplant. 2008; 41: 531-535.
- Wynn RF, Mercer J, Page J, Carr TF, Jones S, Wraith JE. Use of enzyme replacement therapy (Laronidase) before hematopoietic stem cell transplantation for mucopolysaccharidosis I: experience in 18 patients. J Pediatr. 2009; 154: 135-139.
- Muenzer J, Beck M, Eng CM, Giugliani R, Harmatz P, Martin R, et al. Long-term, open-labeled extension study of idursulfase in the treatment of Hunter syndrome. Genet Med. 2011; 13: 95-101.
- Muenzer J, Beck M, Giugliani R, Suzuki Y, Tylki-Szymanska A, Valayannopoulos V, et al. Idursulfase treatment of Hunter syndrome in children younger than 6 years: results from the Hunter Outcome Survey. Genet Med. 2011; 13: 102-109.
- Harmatz P, Whitley CB, Waber L, Pais R, Steiner R, Plecko B, et al. Enzyme replacement therapy in mucopolysaccharidosis VI (Maroteaux-Lamy syndrome). J Pediatr. 2004; 144: 574-580.
- Harmatz P, Ketteridge D, Giugliani R, Guffon N, Teles EL, Miranda MC, et al. Direct comparison of measures of endurance, mobility, and joint function during enzyme-replacement therapy of mucopolysaccharidosis VI (Maroteaux-Lamy syndrome): results after 48 weeks in a phase 2 open-label clinical study of recombinant human N-acetylgalactosamine 4-sulfatase. Pediatrics. 2005; 115: 681-689.
- Harmatz P, Giugliani R, Schwartz I, Guffon N, Teles EL, Miranda MC, 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; 148: 533-539.
- Harmatz P, Giugliani R, Schwartz IV, Guffon N, Teles EL, Miranda MC, et al. Long-term follow-up of endurance and safety outcomes during enzyme replacement therapy for mucopolysaccharidosis VI: Final results of three clinical studies of recombinant human N-acetylgalactosamine 4-sulfatase. Mol Genet Metab. 2008; 94: 469-475.
- Harmatz P, Yu ZF, Giugliani R, Schwartz IV, Guffon N, Teles EL, et al. Enzyme replacement therapy for mucopolysaccharidosis VI: evaluation of long-term pulmonary function in patients treated with recombinant human N-acetylgalactosamine 4-sulfatase. J Inherit Metab Dis. 2010; 33: 51-60.
- Friso A, Tomanin R, Salvalaio M, Scarpa M. Genistein reduces glycosaminoglycan levels in a mouse model of mucopolysaccharidosis type II. Br J Pharmacol. 2010; 159: 1082-1091.
- Malinowska M, Wilkinson FL, Bennett W, Langford-Smith KJ, O'Leary HA, Jakobkiewicz-Banecka J, et al. Genistein reduces lysosomal storage in peripheral tissues of mucopolysaccharide IIIB mice. Mol Genet Metab. 2009; 98: 235-242.
- Marucha J, Tylki-Szymanska A, Jakobkiewicz-Banecka J, Piotrowska A, Kloska A, Czartoryska B, et al. Improvement in the range of joint motion in seven patients with mucopolysaccharidosis type II during experimental gene expression-targeted isoflavone therapy. Am J Med Genet A. 2011; 155: 2257-2262.
- Roberts AL, Thomas BJ, Wilkinson AS, Fletcher JM, Byers S. Inhibition of glycosaminoglycan synthesis using rhodamine B in a mouse model of mucopolysaccharidosis type IIIA. Pediatr Res. 2006; 60: 309-314.
- Patterson MC, Vecchio D, Prady H, Abel L, Wraith JE. Miglustat for treatment of Niemann-Pick C disease: a randomized controlled study. Lancet Neurol. 2007; 6: 765-772.
- Guffon N, Bin-Dorel S, Decullier E, Paillet C, Guitton J, Fouilhoux A. Evaluation of miglustat treatment in patients with type III mucopolysaccharidosis: a randomized, double-blind, placebo-controlled study. J Pediatr. 2011; 159: 838-844.
- Ponder KP, Haskins ME. Gene therapy for mucopolysaccharidosis. Expert Opin Biol Ther. 2007; 7: 1333-1345.
- Richard M, Arfi A, Seguin J, Gandolphe C, Scherman D. Widespread biochemical correction of murine mucopolysaccharidosis type VII pathology by liver hydrodynamic plasmid delivery. Gene Ther. 2009; 16: 746-756.
- Cressant A, Desmaris N, Verot L, Bréjot T, Froissart R, Vanier MT, et al. Improved behavior and neuropathology in the mouse model of Sanfilippo type IIIB disease after adeno-associated virus-mediated gene transfer in the striatum. J Neurosci. 2004; 24: 10229-10239.
- Ruzo A, Marco S, García M, Villacampa P, Ribera A, Ayuso E, et al. Correction of pathological accumulation of glycosaminoglycans in central nervous system and peripheral tissues of MPSIIIA mice through systemic AAV9 gene transfer. Hum Gene Ther. 2012; 23: 1237-1246.
- Fu H, Dirosario J, Killedar S, Zaraspe K, McCarty DM. Correction of neurological disease of mucopolysaccharidosis IIIB in adult mice by rAAV9 trans-blood-brain barrier gene delivery. Mol Ther. 2011; 19: 1025-1033.
- Natowicz MR, Chi MM, Lowry OH, Sly WS. Enzymatic identification of mannose 6-phosphate on the recognition marker for receptor-mediated pinocytosis of beta-glucuronidase by human fibroblasts. Proc Natl Acad Sci USA. 1979; 76: 4322-4326.