Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 58
Filter
2.
Mol Genet Metab Rep ; 22: 100549, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31890590

ABSTRACT

In neuronopathic Hunter syndrome, neurobehavioral symptoms are known to be serious but have been incompletely described. While families face significant stress stemming from this complex and far-reaching array of symptoms, neither caregiver burden nor the neurobehavioral symptoms have been measured comprehensively. We delineated these neurobehavioral characteristics and their impact on the caregiver using multiple approaches. Methods: As part of the initial phase of developing a Hunter-specific behavioral assessment tool, we used multiple methods to obtain data on patient behaviors and caregiver burden, with the intention of drafting item sets for the tool. We utilized 1) caregiver descriptions from focus groups and individual interviews, 2) observations from video-recorded play of affected children, 3) descriptions from historic chart review, 4) consultation with patient advocacy groups and international experts, 5) reports from a caregiver advisory board, and 6) literature review. Results: Neurobehavioral symptoms were diverse and categorized as focus/attention, impulsivity/heightened activity, sensation seeking, emotional/behavioral function, social interaction, and sleep. A significant reported challenge was susceptibility to misinterpretation of some behaviors as defiant or aggressive, particularly if physical. Caregiver burden involved social isolation, exhaustion, stress, and financial and vocational strain. These new descriptions will aid in developing quantitative measures of change in neurobehavioral symptoms and family burden. These descriptions will be the foundation of a neurobehavioral rating scale, which is very much needed to aid in patient management and assess interventions for individuals with neuronopathic Hunter syndrome.

3.
Clin Genet ; 93(5): 1008-1014, 2018 05.
Article in English | MEDLINE | ID: mdl-29396849

ABSTRACT

Lysosomal storage diseases (LSDs) are a group of genetic disorders, resulting from deficiencies of lysosomal enzyme. Genotype-phenotype correlation is essential for timely and proper treatment allocation. Recently, by integrating prediction outcomes of 7 bioinformatics tools, we developed a SAAMP algorithm to predict the impact of individual amino-acid substitution. To optimize this approach, we evaluated the performance of these bioinformatics tools in a broad array of genes. PolyPhen and PROVEAN had the best performances, while SNP&GOs, PANTHER and I-Mutant had the worst performances. Therefore, SAAMP 2.0 was developed by excluding 3 tools with worst performance, yielding a sensitivity of 94% and a specificity of 90%. To generalize the guideline to proteins without known structures, we built the three-dimensional model of iduronate-2-sulfatase by homology modeling. Further, we investigated the phenotype severity of known disease-causing mutations of the GLB1 gene, which lead to 2 LSDs (GM1 gangliosidosis and Morquio disease type B). Based on the previous literature and structural analysis, we associated these mutations with disease subtypes and proposed a theory to explain the complicated genotype-phenotype correlation. Collectively, an updated guideline for phenotype prediction with SAAMP 2.0 was proposed, which will provide essential information for early diagnosis and proper treatment allocation, and they may be generalized to many monogenic diseases.


Subject(s)
Genetic Association Studies , Iduronate Sulfatase/chemistry , Lysosomal Storage Diseases/genetics , beta-Galactosidase/chemistry , Algorithms , Amino Acid Substitution/genetics , Computational Biology , Gangliosidosis, GM1/genetics , Gangliosidosis, GM1/pathology , Genetic Predisposition to Disease , Genotype , Humans , Iduronate Sulfatase/genetics , Lysosomal Storage Diseases/pathology , Mucopolysaccharidosis IV/genetics , Mucopolysaccharidosis IV/pathology , Mutant Proteins/chemistry , Mutant Proteins/genetics , Mutation , Protein Conformation , beta-Galactosidase/genetics
4.
JIMD Rep ; 10: 87-94, 2013.
Article in English | MEDLINE | ID: mdl-23430808

ABSTRACT

BACKGROUND: Serious cardiac valve disease and left ventricular hypertrophy occur in most untreated older children with severe mucopolysaccharidosis type I. Although it is assumed that early intervention prevents these processes, evaluation of cardiac findings in these infants has not yet been reported. METHODS: We reviewed echocardiograms of 13 untreated infants < 1 year of age with severe mucopolysaccharidosis type I who had undergone evaluation for hematopoietic cell transplantation. We recorded left ventricular chamber dimensions, septal and posterior wall thicknesses, ventricular function, and aortic sinus diameters. We evaluated mitral and aortic valves for increased thickness, regurgitation, and stenosis. RESULTS: Average age (7M, 6F) was 221 (range 25-347) days. Left ventricular chamber dimension was ≥2 SD of normal in 3/13; wall thicknesses were ≥2 SD of normal in 2/13 infants. Systolic function was normal. Mitral valves were thickened in all infants; mitral regurgitation was present in 9/13, but significant in only three infants. Aortic valves were thickened in 10/13, but no infant had significant aortic regurgitation. Neither mitral nor aortic stenosis occurred. Aortic roots were dilated to ≥2 SD of normal in 5/13. CONCLUSIONS: Characteristic cardiac features of severe mucopolysaccharidosis type I can be seen in infancy. Mitral and aortic valve thickening are nearly universally present, even in the youngest infants. In 20-30 % of infants, other abnormalities such as left ventricular dilation, increased wall thickness, and mild mitral/aortic regurgitation may occur. Aortic root dilation is a frequent finding. Early intervention with enzyme replacement therapy may minimize the incidence and severity of cardiac findings in these infants. SUMMARY: Serious cardiac valve disease and left ventricular hypertrophy occur in most untreated older children with severe mucopolysaccharidosis type I. Although it is assumed that early intervention prevents these processes, evaluation of cardiac findings in these infants has not yet been reported. In our study of 13 infants with severe untreated MPS I < 1 year of age, mitral and aortic valve thickening was nearly universally present and aortic root dilation was frequent. Despite this, we found a lower incidence of left ventricular hypertrophy and both a lower incidence and milder expression of mitral and aortic valve dysfunction than previously reported in older children. These findings suggest that earlier intervention, including neonatal screening, may be of benefit to children with severe MPS I.

5.
Bone Marrow Transplant ; 47(9): 1235-40, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22179554

ABSTRACT

Mucopolysaccharidosis type I (MPS I) is an autosomal recessive inherited disease caused by deficiency of the glycosidase α-L-iduronidase (IDUA). Deficiency of IDUA leads to lysosomal accumulation of glycosaminoglycans (GAG) heparan and dermatan sulfate and associated multi-systemic disease, the most severe form of which is known as Hurler syndrome. Since 1981, the treatment of Hurler patients has often included allogeneic BMT from a matched donor. However, mouse models of the disease were not developed until 1997. To further characterize the MPS-I mouse model and to study the effectiveness of BMT in these animals, we engrafted a cohort (n=33) of 4-8-week-old Idua(-/-) animals with high levels (88.4±10.3%) of wild-type donor marrow. Engrafted animals displayed an increased lifespan, preserved cardiac function, partially restored IDUA activity in peripheral organs and decreased GAG accumulation in both peripheral organs and in the brain. However, levels of GAG and GM3 ganglioside in the brain remained elevated in comparison to unaffected animals. As these results are similar to those observed in Hurler patients following BMT, this murine-transplantation model can be used to evaluate the effects of novel, more effective methods of delivering IDUA to the brain as an adjunct to BMT.


Subject(s)
Bone Marrow Transplantation/methods , Mucopolysaccharidosis I/metabolism , Mucopolysaccharidosis I/surgery , Animals , Cohort Studies , Disease Models, Animal , Iduronidase/genetics , Iduronidase/metabolism , Longevity , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mucopolysaccharidosis I/enzymology
6.
Bone Marrow Transplant ; 41(12): 1005-11, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18278070

ABSTRACT

Short stature is characteristic of Hurler syndrome, or mucopolysaccharidosis type IH (MPS IH). Hematopoietic stem cell transplantation (HSCT) is used to treat children with MPS IH. While HSCT corrects some of the metabolic features of MPS IH, its effects on growth are not well delineated. We investigated growth in patients with MPS IH after HSCT and described accompanying endocrine abnormalities. A cohort of 48 patients with MPS IH who had received HSCT between 1983 and 2005 were included. The prevalence of short stature (height <-2 s.d. score, SDS) before HSCT was 9%, and increased to 71% at last follow-up (6.9+/-5.1 years after HSCT). Short stature was positively associated with increased age at HSCT (P=0.002) and TBI (P=0.009). In total, 23% had growth hormone deficiency and/or low insulin-like growth factor-1, one female patient had premature adrenarche, one precocious puberty and 27% had clinical or subclinical hypothyroidism. Growth failure is highly prevalent in children with MPS IH after HSCT. Children who had no TBI exposure and were younger at the time of HSCT had a better height outcome.


Subject(s)
Adolescent Development/radiation effects , Child Development/radiation effects , Hematopoietic Stem Cell Transplantation , Mucopolysaccharidosis I/therapy , Transplantation Conditioning/adverse effects , Whole-Body Irradiation/adverse effects , Adolescent , Child , Child, Preschool , Female , Follow-Up Studies , Humans , Infant , Male , Retrospective Studies , Transplantation Conditioning/methods
7.
Bone Marrow Transplant ; 41(6): 531-5, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18037941

ABSTRACT

Hurler syndrome (mucopolysaccharidosis type I, MPS IH) is characterized by a deficiency of alpha-L-iduronidase resulting in progressive multiorgan dysfunction. We sought to determine whether enzyme replacement therapy (ERT) with iduronidase in the peritransplant period affects outcome of hematopoietic stem cell transplantation (HSCT) for MPS IH. Seven children with MPS IH at a median age of 1.5 years at the time of myeloablative HSCT were eligible. All patients had null mutations in IDUA gene. Iduronidase (0.58 mg/kg per dose) was administered intravenously in 11-14 weekly doses before HSCT and 8 weekly doses after HSCT. The infusions were well tolerated. All patients developed antibodies to iduronidase but all engrafted with >90% donor hematopoiesis. A majority of patients had significant pulmonary complications before ERT and HSCT but all are alive and well with a median follow-up of more than 1 year after HSCT. This suggests that ERT prior to HSCT is unlikely to alter engraftment. In addition, morbidity was acceptable, despite a previous history of pulmonary difficulties that suggested that these patients were high risk for these complications. Therefore, we recommend treatment of MPS IH patients with combination of ERT and HSCT therapy to further investigate its potential to enhance outcomes with HSCT.


Subject(s)
Hematopoietic Stem Cell Transplantation , Iduronidase/administration & dosage , Mucopolysaccharidosis I/therapy , Combined Modality Therapy , Graft Survival , Humans , Iduronidase/blood , Infant , Infusion Pumps , Lung Diseases/etiology , Mucopolysaccharidosis I/complications
8.
Genomics ; 74(3): 299-305, 2001 Jun 15.
Article in English | MEDLINE | ID: mdl-11414757

ABSTRACT

Sanfilippo syndrome type B, or mucopolysaccharidosis (MPS) IIIB, is an autosomal recessive disease caused by a deficiency of lysosomal alpha-N-acetylglucosaminidase (NAGLU). In Dromaius novaehollandiae (emu), a progressive neurologic disease was recently discovered, which was characterized by NAGLU deficiency and heparan sulfate accumulation. To define the molecular basis, the sequences of the normal emu NAGLU cDNA and gene were determined by PCR-based approaches using primers for highly conserved regions of evolutionarily distant NAGLU homologues. It was observed that the emu NAGLU gene is structurally similar to that of human and mouse, but the introns are considerably shorter. The cDNA had an open reading frame (ORF) of 2259 bp. The deduced amino acid sequence is estimated to share 64% identity with human, 63% with mouse, 41% with Drosophila, 39% with tobacco, and 35% with the Caenorhabditis elegans enzyme. Three normal and two affected emus were studied for nucleotide sequence covering the entire coding region and exon-intron boundaries. Unlike the human gene, emu NAGLU appeared to be highly polymorphic: 19 variations were found in the coding region alone. The two affected emus were found to be homozygous for a 2-bp deletion, 1098-1099delGG, in exon 6. The resulting frameshift predicts a longer ORF of 2370 bp encoding a polypeptide with 37 additional amino acids and 387 altered amino acids. The availability of mutation screening in emus now permits early detection of MPS IIIB in breeding stocks and is an important step in characterizing this unique, naturally occurring avian model for the development of gene transfer studies.


Subject(s)
Acetylglucosaminidase/genetics , Dromaiidae/genetics , Mucopolysaccharidosis III/genetics , Amino Acid Sequence , Animals , DNA Mutational Analysis , DNA, Complementary/chemistry , DNA, Complementary/genetics , Disease Models, Animal , Exons , Frameshift Mutation , Introns , Molecular Sequence Data , Mucopolysaccharidosis III/enzymology , Mucopolysaccharidosis III/pathology , Mutation , Sequence Alignment , Sequence Analysis, DNA , Sequence Deletion , Sequence Homology, Amino Acid
9.
Gene Ther ; 7(21): 1875-83, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11110421

ABSTRACT

To optimize a gene transfer system for hematopoietic stem cell gene therapy of patients with mucopolysaccharidosis (MPS) type I, 10 retroviral vectors were constructed to express the human alpha-L-iduronidase (IDUA) cDNA. These vectors were designed to evaluate the potential effects of specific promoters, the addition of selectable markers, and the use of multiple promoters versus an internal ribosome entry site for expression of IDUA and selectable maker genes. The effect of vector design was investigated in primary patient fibroblasts (F(MPS)) or murine fibroblast cell lines; while overall comparison of transgene expression was determined in patients' peripheral blood lymphocytes (PBL(MPS)) and CD34+ progenitors (PBPC(MPS)). We observed that the human PGK promoter introduced the highest IDUA activity per 1% relative transgene frequency in F(MPS). Use of the same promoter to separately regulate both the therapeutic gene and a drug-resistance gene resulted in decreased expression of the unselected gene. Co-selection using bicistronic vectors not only increased the number of transductants, but also elevated transgene expression under selective pressure in transgene-positive progenitors. Bicistronic vector LP1CD overcame down-regulation and practically introduced the highest IDUA level in unselected PBL(MPS) and an intermediate level in PBPC(MPS). These studies provide a better understanding of factors contributing to efficient gene expression in hematopoietic cells.


Subject(s)
Genetic Therapy/methods , Genetic Vectors/genetics , Hematopoietic Stem Cells/enzymology , Iduronidase/genetics , Mucopolysaccharidosis I/therapy , Animals , Cell Line , Drug Resistance/genetics , Fibroblasts/enzymology , Gene Expression , Genetic Engineering , Humans , Iduronidase/analysis , Methotrexate , Mice , Phosphoglycerate Kinase/genetics , Polymerase Chain Reaction , Promoter Regions, Genetic , Retroviridae/genetics , Transfection
10.
Genomics ; 68(1): 80-4, 2000 Aug 15.
Article in English | MEDLINE | ID: mdl-10950929

ABSTRACT

Heparan sulfate sulfamidase (HSS) is a lysosomal exohydrolase that, when deficient, results in intralysosomal accumulation of heparan sulfate and the clinical phenotype of Sanfilippo syndrome type A. The first animal disease homolog of human Sanfilippo syndrome type A has been recently indentified in Dachshund littermates. To identify the molecular defect, the nucleotide sequences of the normal canine HSS gene and cDNA were determined using PCR-based approaches. The coding region showed 87% nucleotide homology, and 89% amino acid sequence homology, with human HSS. All exon-intron borders were conserved. Sequence analysis of the entire coding region with exon-intron boundaries was performed in the propositus, a healthy littermate, and six unrelated normal dogs. Comparison revealed a 3-bp deletion, 737-739delCCA, resulting in the loss of threonine at position 246 in both alleles of the propositus and in one allele of a healthy littermate. Prediction of the three-dimensional structure of canine HSS, based on homology with human arylsulfatases A and B, suggested the pathogenic effect of this deletion. Six other sequence variations in exons, and 10 in introns, appear to be benign polymorphisms. This study supports the potential development of a canine model of Sanfilippo syndrome type A to evaluate gene therapy for this disorder.


Subject(s)
Hydrolases/genetics , Mucopolysaccharidosis III/genetics , Amino Acid Sequence , Animals , Arylsulfatases/genetics , DNA Mutational Analysis , DNA, Complementary/chemistry , DNA, Complementary/genetics , Disease Models, Animal , Dogs , Humans , Hydrolases/chemistry , Molecular Sequence Data , Molecular Structure , Mucopolysaccharidosis III/enzymology , Mucopolysaccharidosis III/pathology , Mutation , Polymorphism, Genetic , Sequence Alignment , Sequence Deletion , Sequence Homology, Amino Acid
11.
Mol Genet Metab ; 70(3): 170-8, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10924271

ABSTRACT

Expression of iduronate-2-sulfatase (IDS) from three different promoters in four retroviral vectors was studied in peripheral blood lymphocytes from patients with Hunter syndrome (PBL(MPS)), i.e., the LTR in vectors L2SN and L2, avian beta-actin promoter in LB2, and the CMV early promoter in LNC2. PBL(MPS) were exposed to packaging cell supernatant resulting in transduction frequencies ranging 10-fold from 5 to 49%. Surprisingly, IDS activities were equally high in all transduced lymphocyte populations: 515 U/mg/h in PBL(MPS)-L2SN, 734 in PBL(MPS)-LB2, 352 in PBL(MPS)-L2, and 389 in PBL(MPS)-LNC2 compared to controls (<10 in PBL(MPS)-LXSN or PBL(MPS)). The half-life of endocytosed IDS in PBL(MPS) was 1.9 days. However, the level of lymphocyte IDS activity from proviral expression was found to be only a fraction of the total, a large portion being derived from reuptake of enzyme from murine packaging cells, i.e., a "second source" of enzyme. Therefore, measurement of transgene lysosomal enzyme soon after exposure of target cells to vector supernatant may yield a gross overestimate of long-term transgene expression by transduced cells. Nevertheless, patient fibroblasts cocultured with transduced PBL(MPS) had reduced (35)SO(4)-GAG accumulation, levels similar to those of normal fibroblasts. These studies revealed a broadly applicable phenomenon: cells can be charged with a lysosomal enzyme to levels much higher than those found in nature. By "supercharging" cells with a lysosomal protein (or other molecule bearing the mannose-6-phosphate ligand), such cells may be exploited as vehicles for systemic delivery of therapeutic or diagnostic agents.


Subject(s)
Iduronate Sulfatase/genetics , Retroviridae/genetics , T-Lymphocytes/enzymology , Transfection , Cells, Cultured , Coculture Techniques , DNA Primers/chemistry , Fibroblasts/cytology , Fibroblasts/enzymology , Fibroblasts/virology , Gene Expression , Genetic Therapy/methods , Genetic Vectors , Glycosaminoglycans/metabolism , Humans , Iduronate Sulfatase/metabolism , Mucopolysaccharidosis II/blood , Polymerase Chain Reaction/methods , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Skin/cytology , Skin/enzymology , Skin/virology , T-Lymphocytes/cytology , T-Lymphocytes/virology
13.
Hum Gene Ther ; 10(17): 2799-810, 1999 Nov 20.
Article in English | MEDLINE | ID: mdl-10584926

ABSTRACT

The process of growing and transducing large quantities of human primary peripheral blood lymphocytes (PBLs) with high gene transfer efficiency continues to be one of the major challenges for clinical and experimental gene therapy. Toward developing a clinical trial of lymphocyte gene therapy for mucopolysaccharidosis type II (i.e., Hunter syndrome), we investigated a novel method that exploited the innate capability of a hollow-fiber bioreactor system to filter large quantities of vector supernatant and facilitate transduction. An aliquot (5 x 10(7)) of PBL apheresis product was precultured in a gas-permeable culture bag or a bioreactor, and then transduced with a retroviral vector L2SN containing the iduronate-2-sulfatase (IDS) and neomycin resistance genes. We observed that the total number of PBLs could be expanded up to 187-fold, yielding up to 10(10) cells at the end of a 7-day culture period. The multiplicity of infection could be increased (up to 20-fold) by ultrafiltrating a large volume of vector supernatant through the semipermeable membrane of this system. A high level of transduction efficiency (up to 57%) was achieved, resulting in IDS enzyme activity as high as 1250 U/mg/hr in transduced PBL(MPS) 15 days after transduction. This level was markedly increased from that of nontransduced cells (<3 U/mg/hr) and was even greater than that of normal PBLs (mean, 809; n = 10). After 12 days of G418 selection, PBL(MPS) transductants exhibited a proviral IDS enzyme level approximately threefold higher than that in normal PBLs. These results indicated that the hollow-fiber bioreactor could be used to culture and transduce human primary PBLs in clinically useful quantities with relatively high gene transfer efficiency and transgene expression.


Subject(s)
Gene Transfer Techniques , Genetic Therapy/methods , Genetic Vectors , Leukocytes, Mononuclear , Mucopolysaccharidosis II/therapy , Retroviridae/genetics , Transduction, Genetic , Ultrafiltration/methods , Bioreactors , Cell Division , Humans , Iduronate Sulfatase/biosynthesis , Iduronate Sulfatase/genetics , Mucopolysaccharidosis II/blood
14.
Hum Gene Ther ; 10(15): 2559-66, 1999 Oct 10.
Article in English | MEDLINE | ID: mdl-10543620

ABSTRACT

Accurate quantification of gene transfer (or gene correction) is a universal challenge in the field of gene therapy. In developing a clinical trial of lymphocyte gene therapy for Hunter syndrome (mucopolysaccharidosis type II), methods using Southern blot or automated DNA sequencing technology were employed, but found to be laborious and subject to considerable variation. As an alternative approach, we explored a real-time kinetic PCR assay appropriate to new instrumentation (PE Biosystems model 7700). A TaqMan probe was designed to hybridize directly across the exon 2-exon 3 junction of the iduronate-2-sulfatase transgene cDNA. In this assay system, cDNA from the retroviral vector L2SN generates a PCR product that is 84 nucleotides long and readily quantified by TaqMan probe binding and subsequent cleavage. Evaluation of this method demonstrated sensitivity over at least 5 logs with respect to the standard (vector plasmid pL2SN). There was no detectable signal from genomic DNA from nontransduced cells, thus indicating the specificity of this assay. The sample preparation method used to prepare specimens was a relatively simple cell lysis procedure, without DNA extraction, and represents a significant advancement over the more complex methods of DNA extraction that are typically used for such assays. This specific assay, and comparison to previous methods, illustrates the utility of a new method that is readily generalized to many gene therapy studies, and that has the potential to be extended to measure gene expression by means of quantitative RT-PCR.


Subject(s)
Gene Transfer Techniques/standards , Polymerase Chain Reaction/methods , Base Sequence , Cell Line , DNA Primers , Humans , Kinetics , Reproducibility of Results , Sensitivity and Specificity
15.
Hum Gene Ther ; 10(13): 2163-72, 1999 Sep 01.
Article in English | MEDLINE | ID: mdl-10498248

ABSTRACT

Mucopolysaccharidosis type I (MPS I), a deficiency in the lysosomal enzyme alpha-L-iduronidase (IDUA), is characterized by skeletal abnormalities, hepatosplenomegaly and neurological dysfunction. To evaluate the potential for treatment of the disease using a gene delivery approach, recombinant adeno-associated virus (rAAV) vectors were constructed and evaluated for expression of the human IDUA cDNA in transduced cells. 293 cells transduced with these AAV vectors contained IDUA activity at 0.5 to 1.4 micromol/mg x hr, 50- to 140-fold above background (control-transduced) levels. In time course studies of transduced 293 cells, IDUA activity levels peaked 1 week after transduction and persisted at 50% of the peak level for at least 6 weeks. Transduced MPS I fibroblasts also expressed high levels of IDUA activity (114-290 nmol/mg x hr), which persisted for at least 3 weeks in the absence of selection. In addition, transduced MPS I fibroblasts were capable of clearing intracellular radiolabeled glycosaminoglycan (GAG). As a test of the ability of these vectors to mediate metabolic cross-correction, transduced HuH7 human hepatoma cells were demonstrated to release enzyme that was subsequently taken up by nontransduced MPS I fibroblasts. These results illustrate the effectiveness of AAV vectors for delivery and expression of human IDUA gene sequences and for potential treatment of MPS I.


Subject(s)
Gene Transfer Techniques , Iduronidase/metabolism , Mucopolysaccharidosis I/enzymology , Cell Line , Dependovirus/genetics , Fibroblasts , Gene Expression , Genetic Vectors , Humans , Iduronidase/genetics , Mucopolysaccharidosis I/pathology
16.
Transfusion ; 39(4): 343-50, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10220258

ABSTRACT

BACKGROUND: Gene therapy using autologous peripheral blood lymphocytes (PBLs) has been used to produce adenosine deaminase with which to treat patients with severe combined immunodeficiency. Patients with mucopolysaccharidosis type II (MPS II) lack iduronate-2-sulfatase (IDS), and serial PBL gene therapy may benefit these patients. STUDY DESIGN AND METHODS: The purpose of these studies was to develop a method to transduce PBLs from a patient with MPS II by using a retroviral vector, LS2N, containing the IDS gene. PBLs were collected by apheresis and cryopreserved in aliquots for the performance of multiple transductions and expansions. The PBLs were expanded in number and then transduced in a hollow-fiber bioreactor (HFBR). Additional culture allowed for further expansion. RESULTS: Fresh PBLs (6.2 x 10(7)) from a patient with MPS II were transduced with L2SN and expanded in an HFBR with an extracapillary space of 11 mL. After 10 days of culture, 4.1 x 10(9) cells were harvested. Cryopreserved MPS II PBLs could not be reliably expanded if they were placed in the HFBR immediately after being thawed; however, cells were successfully transduced and expanded in the HFBR if they were first cultured in a bag. To increase the cell yield, PBLs were expanded in a 60-mL HFBR after transduction and expansion in an 11-mL HFBR. In four separate experiments, 2 x 10(8) cryopreserved PBL were cultured for 3 days in a bag and transferred to an 11-mL HFBR, where they were transduced daily with L2SN for 3 days and then expanded for 4 additional days. Cells were then transferred into a 60-mL HFBR and expanded for an additional 7 days. In the four experiments, 5.5 x 10(9), 7.4 x 10(9), 1.12 x 10(9), and 19.4 x 1(9) cells were produced. The vector was detected in the harvested cells, but the proportion of cells transduced was less than 2.5 percent, the lowest standard used in the assay. In two of the experiments, cells harvested from the HFBR were used in a gene therapy clinical trial. CONCLUSION: Autologous cryopreserved PBLs can be transduced and expanded to produce >1 x 10(10) cells. This procedure is being used for a Phase I/II clinical trial of lymphocyte gene therapy.


Subject(s)
Genetic Therapy , Iduronate Sulfatase/genetics , Lymphocytes/metabolism , Mucopolysaccharidosis II/therapy , Retroviridae/genetics , Cells, Cultured , Cryopreservation , Humans , Male , Middle Aged
17.
J Gene Med ; 1(6): 433-40, 1999.
Article in English | MEDLINE | ID: mdl-10753069

ABSTRACT

BACKGROUND: The ability to obtain high-titer and large quantities of retroviral vector production in a 'closed' system would have profound implications in clinical and experimental gene therapy. METHODS: We studied the cell growth and vector production of three retroviral packaging cell lines in a variety of conditions using hollow-fiber bioreactors designed as an 'artificial capillary system' (ACS) and enhanced with the application of a hermetically sealing device for sterile welding of connecting plastic tubings. Vector titer, fetal bovine serum (FBS) concentration, volume and the duration of productivity were assessed to optimize vector production. RESULTS: In this pilot study, we observed that retroviral vector production (frozen-and-thawed) from cultures containing as low as 2.5% FBS yielded titers up to 2.2 x 10(7) cfu/ml, 14.4-fold higher than titers obtained from control dish cultures. Up to 3 liters of vector supernatant were generated during a 2-month large-scale production run. There was a potential to double this volume of higher-titer supernatant by increasing the frequency of harvest. It seemed that a lower metabolic rate (i.e. lactate production) in the packaging cell culture was associated with higher vector producing ability. CONCLUSIONS: These data demonstrated the feasibility of producing retroviral vector with enhanced titers and clinically useful quantities in a 'closed' ACS. Thus a new approach for large-scale retroviral vector production is developed.


Subject(s)
Bioreactors , Cell Culture Techniques/instrumentation , Genetic Vectors/isolation & purification , Retroviridae/growth & development , Virus Cultivation/methods , Animals , Cattle , Cell Survival , Culture Media , Culture Media, Conditioned , Disinfection , Equipment Design , Feasibility Studies , Fetal Blood , Pilot Projects , Retroviridae/isolation & purification , Virus Cultivation/instrumentation
18.
Am J Hum Genet ; 62(1): 53-63, 1998 Jan.
Article in English | MEDLINE | ID: mdl-9443875

ABSTRACT

Sanfilippo syndrome type B, or mucopolysaccharidosis type IIIB, results from defects in the gene for alpha-N-acetylglucosaminidase (NAGLU); only a few mutations have been described. To rapidly identify most NAGLU mutations, an automated sequencing procedure was developed for analysis of the entire coding region, including exon-intron borders. By this method, eight affected families were studied, and the mutations in all 16 alleles were identified, more than doubling the number of published mutations for this gene. Eight mutations were described for the first time: five missense mutations (Y140C, Y455C, P521L, S612G, and R674C), two nonsense mutations (W675X and Q706X), and one 24-nucleotide insertion. Currently, 36% of all point mutations (8 of 22 alleles) involve R674, a codon having a CpG dinucleotide in the critical initial position. Other mutations were found in more than one family, raising the possibility that some may be relatively common and, possibly, ancient mutations. Six new nonpathological mutations were also identified and likely represent polymorphic variants of the NAGLU gene, two of which might alter enzyme level. Establishing genotype-phenotype relationships will be vital in the evaluation of experimental treatments such as gene therapy.


Subject(s)
Acetylglucosaminidase/genetics , Mucopolysaccharidosis III/genetics , Mutation , Acetylglucosaminidase/metabolism , DNA/analysis , DNA Mutational Analysis , DNA Primers , Female , Genotype , Humans , Male , Mucopolysaccharidosis III/enzymology , Phenotype , Polymerase Chain Reaction
19.
Am J Hum Genet ; 62(1): 64-9, 1998 Jan.
Article in English | MEDLINE | ID: mdl-9443878

ABSTRACT

Sanfilippo syndrome type B (mucopolysaccharidosis III B) is a rare autosomal recessive disease caused by deficiency of alpha-N-acetylglucosaminidase, one of the enzymes required for the lysosomal degradation of heparan sulfate. The gene for this enzyme, NAGLU, recently was isolated, and several mutations were characterized. We have identified, in amplified exons from nine fibroblast cell lines derived from Sanfilippo syndrome type B patients, 10 additional mutations: Y92H, P115S, Y140C, E153K, R203X, 650insC, 901delAA, P358L, A664V, and L682R. Four of these mutations were found in homozygosity, and only two were seen in more than one cell line. Thus, Sanfilippo syndrome type B shows extensive molecular heterogeneity. Stable transfection of Chinese hamster ovary cells, by cDNA mutagenized to correspond to the NAGLU missense mutations, did not yield active enzyme, demonstrating the deleterious nature of the mutations. Nine of the 10 amino acid substitutions identified to date are clustered near the amino or the carboxyl end of alpha-N-acetylglucosaminidase, suggesting a role for these regions in the transport or function of the enzyme.


Subject(s)
Acetylglucosaminidase/genetics , Mucopolysaccharidosis III/genetics , Mutation , Acetylglucosaminidase/metabolism , Animals , CHO Cells , Cell Line , Cricetinae , DNA, Complementary , Humans , Mucopolysaccharidosis III/enzymology , Mutagenesis , Oligonucleotides , Transfection
20.
J Hematother ; 7(6): 505-14, 1998 Dec.
Article in English | MEDLINE | ID: mdl-9919944

ABSTRACT

Mucopolysaccharidosis type I (MPS I) results from a deficiency of alpha-L-iduronidase enzyme (IDUA), an enzyme responsible for the catabolism of glycosaminoglycans. Genetically modified progenitor cells may permit a therapeutic effect similar to that obtained from allogeneic BMT without the associated risks. To that end, CD34+ peripheral blood hematopoietic progenitor cells from patients with MPS I were mobilized using G-CSF, collected by apheresis, and enriched using avidin-biotin separation techniques. These cells were cultured in a hollow fiber bioreactor and transduced with a retroviral vector (LP1CD) containing the cDNA for human IDUA and a murine dihydrofolate reductase (DHFR) enzyme. Approximately 4%-16% of the colonies expressed methotrexate drug resistance. Expression of the IDUA enzyme in the progenitor cells was initially high and declined after approximately 10 days of culture. These results indicate that PBPC from patients with MPS I can be mobilized, isolated, enriched, and transduced with a therapeutic gene.


Subject(s)
Cell- and Tissue-Based Therapy , Genetic Therapy , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/physiology , Iduronidase/genetics , Mucopolysaccharidosis I/therapy , Adult , Animals , Gene Transfer Techniques , Genetic Vectors , Hematopoietic Stem Cell Mobilization , Humans , Mice , Mucopolysaccharidosis I/genetics , Retroviridae
SELECTION OF CITATIONS
SEARCH DETAIL
...