68
Views
5
CrossRef citations to date
0
Altmetric
Review

Development of mammalian artificial chromosomes for the treatment of genetic diseases: Sandhoff and Krabbe diseases

, , &
Pages 195-206 | Published online: 20 Apr 2005

Bibliography

  • RAPER SE, CHIRMULE N, LEE FS et al.:Fatal systemic inflammatory response syndrome in an ornithine transcarbamylase deficient patient following adenoviral gene transfer. MoL Genet. Metab. (2003) 80:148–158.
  • CHRIST M, LUSKY M, STOECKEL Fet al.: Gene therapy with recombinant adenovirus vectors: evaluation of the host immune response. ImmunoL Lett. (1997) 57:19–25.
  • COUTO LB: Preclinical gene therapy studies for hemophilia using adeno-associated virus (AAV) vectors. Semin. Thromb. Hemost. (2004) 30:161–171.
  • MILLER DG, RUTLEDGE EA, RUSSELL DW: Chromosomal effects of adeno-associated virus vector integration. Nat. Genet. (2002) 30:147–148.
  • MILLER DG, PETEK LM, RUSSELL DW: Adeno-associated virus vectors integrate at chromosome breakage sites. Nat. Genet. (2004) 36:767–773.
  • HACEIN-BEY-ABINA S, VON KALLE C, SCHMIDT M et al.: A serious adverse event after successful gene therapy for X-linked severe combined immunodeficiency. N EngL J. Med. (2003) 348:255–256.
  • HACEIN-BEY-ABINA S, VON KALLE C, SCHMIDT M et al.: LM02-associated clonal T cell proliferation in two patients after gene therapy for SCID-X1. Science (2003) 302:415–419.
  • HADLACZKY G: Satellite DNA-based artificial chromosomes for use in gene therapy. Curr. Opin. MoL Ther. (2001) 3:125–132.
  • LIPPS HJ, JENKE AC, NEHLSEN K et al.: Chromosome-based vectors for gene therapy. Gene (2003) 304:23–33.
  • LIM HN, FARR CJ: Chromosome-based vectors for mammalian cells: an overview. Methods MoL Biol. (2004) 240:167–186.
  • PEREZ CF, VANDERBYL SL, MILLS KA, LEDEBUR JR HC: The ACE System: a versatile chromosome engineering technology with applications for gene-based cell therapy. Bioprocessing (2004) 3:61–68.
  • LINDENBAUM M, PERKINS E, CSONKA E et al.: A mammalian artificial chromosome engineering system (ACE System) applicable to biopharmaceutical protein production, transgenesis and gene-based cell therapy. Nucleic Acids Res. (2004) 32:e172.
  • CALOS MP: The potential of extrachromosomal replicating vectors for gene therapy. Trends Genet. (1996) 12:463–466.
  • KELLEHER ZT, FU H, LIVANOS E et al.:Epstein-Barr-based episomal chromosomes shuttle 100 kb of self-replicating circular human DNA in mouse cells. Nat. BiotechnoL (1998) 16:762–768.
  • VOS JM: Therapeutic mammalian artificialepisomal chromosomes. Curr. Opin. MoL Ther. (1999) 1:204–215.
  • HUERTAS D, HOWE S, MCGUIGAN A, HUXLEY C: Expression of the human CFTR gene from episomal oriP-EBNA1-YACs in mouse cells. Hum. MoL Genet. (2000) 9:617–629.
  • HARRINGTON JJ, VAN BOKKELEN G, MAYS RW, GUSTASHAW K, WILLARD HF: Formation of de novo centromeres and construction of first-generation human artificial microchromosomes. Nat. Genet. (1997) 15:345-355. First demonstration that satellite DNA co-transfected with genomic sequences into human tumour cells leads to artificial chromosome formation.
  • IKENO M, GRIMES B, OKAZAKI T et al.: Construction of YAC-based mammalian artificial chromosomes. Nat. BiotechnoL (1998) 16:431–439.
  • HENNING KA, NOVOTNY EA, COMPTON ST et al.: Human artificial chromosomes generated by modification of a yeast artificial chromosome containing both human alpha satellite and single-copy DNA sequences. Proc. NatL Acad. Sci. USA (1999) 96:592–597.
  • LAS HERAS JI, D'AIUTO L, COOKE H: Mammalian artificial chromosome formation in human cells after lipofection of a PAC precursor. Methods Mol Biol. (2004) 240:187–206.
  • CARINE K, SOLUS J, WALTZER E et al.: Chinese hamster cells with a minichromosome containing the centromere region of human chromosome 1. Somat. Cell Md. Genet. (1986) 12:479–491.
  • FARR CJ, BAYNE RA, KIPLING D et al.: Generation of a human X-derived minichromosome using telomere-associated chromosome fragmentation. EMBO J. (1995) 14:5444–5454.
  • HELLER R, BROWN KE, BURGTORF C, BROWN WR: Mini-chromosomes derived from the human Y chromosome by telomere directed chromosome breakage. Proc. Natl Acad. Sci. USA (1996) 93:7125–7130.
  • KUROIWA Y, TOMIZUKA K, SHINOHARA T et al.: Manipulation of human minichromosomes to carry greater than megabase-sized chromosome inserts. Nat. Biotechnol (2000) 18:1086–1090.
  • KATOH M, AYABE F, NORIKANE S et al.: Construction of a novel human artificial chromosome vector for gene delivery. Biochem. Biophys. Res. Commun. (2004) 321:280–290.
  • SAFFERY R, WONG LH, IRVINE DV et al.: Construction of neocentromere-based human minichromosomes by telomere-associated chromosomal truncation. Proc. Natl Acad. Sci. USA (2001) 98:5705–5710.
  • VOET T, VERMEESCH J, CARENS A et al.: Efficient male and female germline transmission of a human chromosomal vector in mice. Genome Res. (2001) 11:124–136.
  • WONG LH, SAFFERY R, CHOO KH: Construction of neocentromere-based human minichromosomes for gene delivery and centromere studies. Gene Ther. (2002) 9:724–726.
  • PRAZNOVSZKY T, KERESO J, TUBAK V et al.: De novo chromosome formation in rodent cells. Proc. Nail. Acad. Sci. USA (1991) 88:11042–11046.
  • HADLACZKY G, PRAZNOVSZKY T, CSERPAN I et al.: Centromere formation in mouse cells cotransformed with human DNA and a dominant marker gene. Proc. Natl Acad. Sci. USA (1991) 88:8106–8110.
  • HOLLO G, KERESO J, PRAZNOVSZKY T et al.: Evidence for a megareplicon covering megabases of centromeric chromosome segments. Chromosome Res. (1996) 4:240–247.
  • •Provides experimental data on the mechanism of large-scale amplification and de novo chromosome formation in mammalian cells.
  • KERESO J, PRAZNOVSZKY T, CSERPAN I et al.: De novo chromosome formations by large-scale amplification of the centromeric region of mouse chromosomes. Chromosome Res. (1996) 4:226–239.
  • •Describes the basic principles of the generation of SATACs.
  • CSONKA E, CSERPAN I, FODOR K et al.: Novel generation of human satellite DNA- based artificial chromosomes in mammalian cells. J. Cell Sci. (2000) 113:3207–3216.
  • ••This paper describes the generation of ahuman SATAC in vitro.
  • DE JONG G, TELENIUS AH, TELENIUS H et al.: Mammalian artificial chromosome pilot production facility: large-scale isolation of fitnctional satellite DNA-based artificial chromosomes. Cytometry (1999) 35:129–133.
  • DE JONG G, TELENIUS A, VANDERBYL S, MEITZ A. DRAYER J: Efficient in-vitro transfer of a 60-Mb mammalian artificial chromosome into murine and hamster cells using cationic lipids and dendrimers. Chromosome Res. (2001) 9:475–485.
  • •This paper describes the procedures to transfect isolated MACs into mammalian cells using simple procedures and commercially available cationic reagents.
  • VANDERBYL S, MACDONALD N, DE JONG G: A flow cytometry technique for measuring chromosome-mediated gene transfer. Cytometry (2001) 44:100–105.
  • VANDERBYL S, MACDONALD GN, SIDHU S et al.: Transfer and stable transgene expression of a mammalian artificial chromosome into bone marrow-derived human mesenchymal stem cells. Stem Cells (2004) 22:324–333.
  • ••This paper reports the first data for thestable transfer of a MAC into human adult stem cells.
  • CO DO, BOROWSKI AH, LEUNG JD et al.: Generation of transgenic mice and germline transmission of a mammalian artificial chromosome introduced into embryos by pronuclear microinjection. Chromosome Res. (2000) 8:183–191.
  • ••This paper presents the first successful generation of transgenic animals with purified MACs.
  • MONTEITH DP, LEUNG JD, BOROWSKI AH et al.: Pronuclear microinjection of purified artificial chromosomes for generation of transgenic mice: pick-and-inject technique. Methods Mol Biol. (2004) 240:227–242.
  • SANDS VE: Short arm enlargement in acrocentric chromosomes. Am. J. Hum. Genet. (1969) 21:293–304.
  • WARBURTON D, ATWOOD KC, HENDERSON AS: Variation in the number of genes for rRNA among human acrocentric chromosomes: correlation with frequency of satellite association. Cytogenet. Cell Genet. (1976) 17:221–230.
  • VERMA RS, DOSIK H, LUBS HA Size variation polymorphisms of the short arm of human acrocentric chromosomes determined by R-banding by fluorescence using acridine orange (RFA). Hum. Genet. (1977) 38:231–234.
  • SOFUNI T, TANABE K, AWA AA: Chromosome heteromorphisms in the Japanese. II. Nucleolus organizer regions of variant chromosomes in D and G groups. Hum. Genet. (1980) 55:265–270.
  • •Human chromosomes 13, 14 and 15 comprise the D group, while chromosomes 21 and 22 comprise the G group.
  • TRASK B, VAN DEN ENGH G, MAYALL B, GRAY JW: Chromosome heteromorphism quantified by high-resolution bivariate flow karyotyping. Am. J. Hum. Genet. (1989) 45:739–752.
  • EARLE E, DALE S, CHOO KH: Amplification of satellite III DNA in an unusually large chromosome 14p+ variant. Hum. Genet. (1989) 82:187–190.
  • STERGIANOU K, GOULD CP, WATERS JJ, HULTEN MA: A DA/DAPI positive human 14p heteromorphism defined by fluorescence in-situ hybridisation using chromosome 15-specific probes D15Z1 (satellite III) and p-TRA-25 (alphoid). Hereditas (1993) 119:105–110.
  • FRIEDRICH U, CAPRANI M, NIEBUHR E, THERKELSEN AJ, JORGENSEN AL: Extreme variant of the short arm of chromosome 15. Hum. Genet. (1996) 97:710–713.
  • CONTE RA, KLEYMAN SM, LAUNDON C, VERMA RS: Characterization of two extreme variants involving the short arm of chromosome 22: are they identical? Ann. Genet. (1997) 40:145–149.
  • COOPER HL, HIRSCHHORN K: Enlarged satellites as a familial chromosome marker. Am. J. Hum. Genet. (1962) 14:107–124.
  • THERKELSEN AJ: Enlarged short arm of a small acrocentric chromosome in grandfather, mother, and child, the latter with Down's syndrome. Cytogenetics (1964) 10:441–451.
  • YODER FE, BIAS WB, BORGAONKAR DS et al: Cytogenetic and linkage studies of a familial 15p* variant. Am. J. Hum. Genet. (1974) 26:535–548.
  • •An acrocentric chromosome with an amplified short arm is carried in four generations of one family.
  • BERNSTEIN R, DAWSON B, GRIFFITHS J: Human inherited marker chromosome 22 short-arm enlargement: investigation of rDNA gene multiplicity, Ag-band size, and acrocentric association. Hum. Genet. (1981) 58:135–139.
  • TRASK B, VAN DEN ENGH G, GRAY JN(I: Inheritance of chromosome heteromorphisms analyzed by high-resolution bivariate flow karyotyping. Am.J. Hum. Genet. (1989) 45:753–760.
  • SCHMID M, NANDA I, STEINLEIN C, EPPLEN JT: Amplification of (GACA)n simple repeats in an exceptional 14p+ marker chromosome. Hum. Genet. (1994) 93:375–382.
  • MUKERJEE D, BURDETTE WJ: A familial minute isochromosome. Am. J. Hum. Genet. (1966) 18:62–69.
  • •Supernumary chromosome derived from chromosome 21 carried by two generations of one family.
  • HOWARD-PEEBLES PN: A familial bisatellited extra metacentric microchromosome in man. J. Hered. (1979) 70:347–348.
  • •Supernumary chromosome derived from an acrocentric short arm carried in two generations of one family.
  • BLENNOW E, BUT TH, KRISTOFFERSSON U et al: Swedish survey on extra structurally abnormal chromosomes in 39 105 consecutive prenatal diagnoses: prevalence and characterization by fluorescence in situ hybridization. Prenat. Diagn. (1994) 14:1019–1028.
  • •One of a series of reports showing that supernumerary chromosomes derived from the short arms of acrocentric chromosomes produce no ill effects in the human population.
  • BRONDUM-NIELSEN K, MIKKELSEN M: A 10-year survey, 1980–1990, of prenatally diagnosed small supernumerary marker chromosomes, identified by FISH analysis. Outcome and follow-up of 14 cases diagnosed in a series of 12,699 prenatal samples. Prenat. Diagn. (1995) 15:615–619.
  • •One of a series of reports showing that supernumerary chromosomes derived from the short arms of acrocentric chromosomes produce no ill effects in the human population.
  • CROLLA JA, HOWARD P, MITCHELL C, LONG FL, DENNIS NR: A molecular and FISH approach to determining karyotype and phenotype correlations in six patients with supernumerary marker(22) chromosomes. Am. J. Med. Genet. (1997) 72:440–447.
  • •One of a series of reports showing that supernumerary chromosomes derived from the short arms of acrocentric chromosomes produce no ill effects in the human population.
  • ADHVARYU SG, PETERS-BROWN T, LIVINGSTON E, QUMSIYEH MB: Familial supernumerary marker chromosome evolution through three generations. Prenat. Diagn. (1998) 18:178–181.
  • HADLACZKY G, BUJDOSO G, KOLTAI E, SCHWANITZ G, CSONKA E: A natural equivalent to human satellite DNA-based artificial chromosome persists over 140 years, in a three-generation family. Eur. J. Hum. Genet. (2002) 10:143–143.
  • •Supernumerary chromosome derived from chromosome 15 is carried in three generations of one family.
  • GOODELL MA, BROSE K, PARADIS G, CONNER AS, MUULIGAN RC: Isolation and functional properties of murine hematopoietic stem cells that are replicating in vivo. J. Exp. Med. (1996) 183:1797–1806.
  • GOODELL MA, ROSENZWEIG M, KIM H et al.: Dye efflux studies suggest that hematopoietic stem cells expressing low or undetectable levels of CD34 antigen exist in multiple species. Nat. Med. (1997) 3:1337–1345.
  • FERRARI G, CUSELLA-DE ANGELIS G,COLETTA M et al.: Muscle regeneration by bone marrow-derived myogenic progenitors. Science (1998) 279:1528–1530.
  • KRAUSE DS, THEISE ND, COLLECTOR MI et al.: Multi-organ, multi-lineage engraftment by a single bone marrow-derived stem cell. Cell (2001) 105:369–377.
  • BLACK I, WOODBURY D: Adult rat andhuman bone marrow stromal stem cells differentiate into neurons. Blood Cells MoL Dis. (2001) 27:632–636.
  • BRAZELTON TR, ROSSI FM, KESHET GI, BLAU HM: From marrow to brain: expression of neuronal phenotypes in adult mice. Science (2000) 290:1775–1779.
  • MEZEY E, CHANDROSS KJ, HARTA G, MAKI RA, MCKERCHER SR: Turning blood into brain: cells bearing neuronal antigens generated in vivo from bone marrow. Science (2000) 290:1779–1782.
  • TERSKIKH AV, EASTERDAY MC, LI L et al.: From hematopoiesis to neuropoiesis: Evidence of overlapping genetic programs. Proc. Natl Acad. Sci. USA (2001) 98:7934–7939.
  • GUSSONI E, SONEOKA Y, STRICKLAND CD et al: Dystrophin expression in the mdx mouse restored by stem cell transplantation. Nature (1999) 401:390–394.
  • PETERSEN BE, BOWEN WC, PATRENE KD: Bone Marrow as a potential source of hepatic oval cells. Science (1999) 284:1168–1170.
  • ALISON MR, POULSOM R, FORBES SJ: Update on hepatic stem cells. Liver (2001) 21:367–373.
  • THEISE ND, NIMMAKAYALU M, GARDNER R et al: Liver from bone marrow in humans. Hepatology (2000) 32:11–16.
  • THEISE ND, BADVE S, SAXENA R et al: Derivation of hepatocytes from bone marrow cells in mice after radiation-induced myeloablation. Hepato/ogy (2000) 31:235–240.
  • MEZEY E, KEYS, VOGELSANG G, SZALAYOVA I, LANGE GD, CRAIN B: Transplanted bone marrow generates new neurons in human brains. Proc. Natl Acad. Sci. USA (2003) 100:1364–1369.
  • ALISON MR, POULSOUM R, OTTO WR et al: Plastic adult stem cells: 204 will they graduate from the school of hard knocks? J. Cell Sci. (2003) 116:599–603.
  • NEWSOME PN, JOHANNESSEN I, BOYLE S et al.: Human cord blood-derived cells can differentiate into hepatocytes in the mouse liver with no evidence of cellular fusion. Gastroenterology (2003) 124:1891–900.
  • BONDE J, HESS DA, NOLTA JA: Recent advances in hematopoietic stem cell biology. Curr. Opin. HematoL (2004) 11:392–398.
  • COGLE CR, YACHNIS AT, LAYWELL ED et al.: Bone marrow transdifferentiation in brain after transplantation: a retrospective study. Lancet (2004) 363:1432–1437.
  • COVA L, RATTI A, VOLTA M et aL: Stem cell therapy for neurodegenerative diseases: the issue of transdifferentiation. Stem Cells Dev. (2004) 13:121–131.
  • MEZEY E: Commentary: on bone marrow stem cells and openmindedness. Stem Cells Dev. (2004) 13:147–152.
  • NYGREN JM, JOVINGE S, BREITBACH M: Bone marrow-derived hematopoietic cells generate cardiomyocytes at a low frequency through cell fusion, but not transdifferentiation. Nat. Med. (2004) 10:494–501.
  • STADTFELD M, GRAF T: Assessing the role of hematopoietic plasticity for endothelial and hepatocyte development by non-invasive lineage tracing. Development (2005) 132:203–213.
  • DONADONI C, CORTI S, LOCATELLI F et al.: Improvement of combined FISH and immunofluorescence to trace the fate of somatic stem cells after transplantation. J. Histochem. Cytochem. (2004) 52:1333–1339.
  • BECKER SB: Hematopoietic stem cell gene therapy for inherited bone marrow disorders: past accomplishments and continued challenges. J. Cell. Biochem. Suppl (2002) 38:55–64.
  • BUERN JA, GUENECHE G, CASADO JA, LAMANA ML, SEGOVIA JC: Genetic modification of hematopoietic stem cells: recent advances in the gene therapy of inherited diseases. Arch. Med. Res. (2003) 34:589–599.
  • VOLLWEILER JL, ZIELSKE SP, REESE JS, GERSON SL: Hematopoietic stem cell gene therapy: progress toward therapeutic targets. Bone Marrow Transplant. (2003) 32:1–7.
  • FRIEDENSTEIN AJ, DERIGLASOVA UF, KULAGINA NN et al.: Precursors for fibroblasts in different populations of hematopoietic cells as detected by the in vitro colony assay method. Exp. Hematol. (1974) 2:83–92.
  • PITTENGER MF, MACKAY AM, BECK SC et al.: Multilineage potential of adult human mesenchymal stem cells. Science (1999) 284:143–147.
  • PROCKOP D: Marrow stromal cells as stem cells for nonhematopoietic tissues. Science (1997) 276:71–74.
  • WAKITANI S, SAITO T, CAPLAN Al: Myogenic cells derived from rat bone marrow mesenchymal stem cells exposed to 5-azacytidine. Muscle Nerve (1995) 18:1417–1426.
  • KOTTON DN, MA BY, CARDOSO WV et al.: Bone marrow-derived cells as progenitors of lung alveolar epithelium. Development (2001) 128:5181–5188.
  • KOPEN GC, PROCKOP DJ, PHINNEY DG: Marrow stromal cells migrate throughout forebrain and cerebellum, and they differentiate into astrocytes after injection into neonatal mouse brains. Proc. Natl Acad. Sci. USA (1999) 96:10711–10716.
  • SPEES JL, OLSON SD, YLOSTALO J et al.: Differentiation, cell fusion, and nuclear fusion during ex vivo repair of epithelium by human adult stem cells from bone marrow stroma. Proc. Natl Acad. Sci. USA (2003) 100:2397–2402.
  • SANCHEZ-RAMOS J, SONG S, CARDOZO-PELAEZ F et al.: Adult bone marrow stromal cells differentiate into neural cells in vitro. Exp. Neurol (2000) 164:247–256.
  • DENG W, OBROCKA M, FISCHER I, PROCKOP DJ: In vitro differentiation of human marrow stromal cells into early progenitors of neural cells by conditions that increase intracellular cyclic AMP. Biochem. Biophys. Res. Commun. (2001) 282:148–152.
  • FERRARI G, CUSELLA-DE ANGELIS G,COLETTA M et al.: Muscle regeneration by bone marrow-derived myogenic progenitors. Science (1998) 279:1528–1530.
  • LIECHTY KW, MACKENZIE TC, SHAABAN AF et al.: Human mesenchymal stem cells engraft and demonstrate site-specific differentiation after in utero transplantation in sheep. Nat. Med. (2000) 6:1282–1286.
  • KOTTON DN, MA BY, CARDOSO WV et al.: Bone marrow-derived cells as progenitors of lung alveolar epithelium. Development (2001) 128:5181–5188.
  • CHEN J, LI Y, WANG L, LU M, ZHANG X, CHOPP M: Therapeutic benefit of intracerebral transplantation of bone marrow stromal cells after cerebral ischemia in rats. J. Neurol Sci. (2001) 189:49–57.
  • HOFSTETTER CP, SCHWARZ EJ, HESS D et al.: Marrow stromal cells form guiding strands in the injured spinal cord and promote recovery. Proc. Natl Acad. Sci. USA (2002) 99:2199–2204.
  • ANKENY DP, MCTIGUE DM, JAKEMAN LB: Bone marrow transplants provide tissue protection and directional guidance for axons after contusive spinal cord injury in rats. Exp. NeuroL (2004) 190:17–31.
  • HORWITZ EM, PROCKOP DJ, FITZPATRICK LA et al.: Transplantability and therapeutic effects of bone marrow-derived mesenchymal cells in children with osteogenesis imperfecta. Nat. Med. (1999) 5:309–313.
  • CHOPP M, ZHANG XH, LI Y et ell.: Spinal cord injury in rat: treatment with bone marrow stromal cell transplantation. Neuroreport (2000) 11:3001–3005.
  • LI Y, CHEN J, WANG L, ZHANG L, LU M, CHOPP M: Intracerebral transplantation of bone marrow stromal cells in a 1-methy1-4-pheny1-1,2,3,6-tetrahydropyridine mouse model of Parkinson's disease. Neurosci. Lett. (2001) 316:67–70.
  • JIN HK, CARTER JE, HUNTLEY GW, SCHUCHMAN EH: Intracerebral transplantation of mesenchymal stem cells into acid sphingomyelinase-deficient mice delays the onset of neurological abnormalities and extends their life span. Clin. Invest. (2002) 109:1183–1191.
  • TOMA C, PITTENGER MF, CAHILL KS, BYRNE BJ, KESSLER PD: Human mesenchymal stem cells differentiate to a cardiomyocyte phenotype in the adult murine heart. Circulation (2002) 105:93–98.
  • SCHWARZ EJ, REGER RL, ALEXANDER GM et al: Rat marrow stromal cells rapidly transduced with a self-inactivating retrovirus synthesize L-DOPA in vitro. Gene Ther. (2001) 8:1214–1223.
  • ZHANG XY, LA RUSSA VF, BAO L, KOLLS J, SCHWARZENBERGER P, REISER J: Lentiviral vectors for sustained transgene expression in human bone marrow-derived stromal cells. MoL Ther. (2002) 5:555–565.
  • PEISTER A, MELLAD JA, WANG M, TUCKER HA, PROCKOP DJ: Stable transfection of MSCs by electroporation. Gene Ther. (2004) 11:224–228.
  • LAZARUS HM, HAYNESWORTH SE, GERSON SL, ROSENTHAL NS, CAPLAN AT: Ex vivo expansion and subsequent infusion of human bone marrow-derived stromal progenitor cells (mesenchymal progenitor cells): implications for therapeutic use. Bone Marrow Tranplant (1995) 16:557–564.
  • KEATING A, BERKAHN L, FILSHIE R: A Phase I study of the transplantation of genetically marked autologous bone marrow stromal cells. Hum. Gene Ther. (1998) 9:591–600.
  • KOC 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.
  • KOC ON, GERSON SL, COOPER BW et al.: Rapid hematopoietic recovery after coinfusion of autologous-blood stem cells and culture-expanded marrow mesenchymal stem cells in advanced breast cancer patients receiving high-dose chemotherapy. J. Clin. Oncol (2000) 18:307–316.
  • HORWITZ EM, GORDON PL, KOO WK et al.: Isolated allogeneic bone marrow-derived mesenchymal cells engraft and stimulate growth in children with osteogenesis imperfecta: implications for cell therapy of bone. Proc. Nail. Acad. Sci. USA (2002) 99:8932–8937.
  • IOANNOU YA, ENRIQUEZ A, BENJAMIN C: Gene therapy for lysosomal storage disorders. Expert Opin. Biol. Ther. (2003) 3:789–801.
  • SANDHOFF K, ANDREAE U, JATZKEWITZ H: Deficient hexosaminidase activity in an exceptional case of Tay-Sachs disease with additional storage of kidney globoside in visceral organs. Lift Sci. (1968) 7:283–288.
  • PHANEUF D, WAKAMATSU N, HUANG JQ et al: Dramatically different phenotypes in mouse models of human Tay-Sachs and Sandhoff diseases. Hum. Mol Genet. (1996) 5:1–14.
  • SANGO K, MCDONALD MP, CRAWLEY JN et al: Mice lacking both subunits of lysosomal beta-hexosaminidase display gangliosidosis and mucopolysaccharidosis. Nat. Genet. (1996) 14:348–352.
  • HUANG JQ, TRASLER JM, IGDOURA S, MICHAUD J, HANAL N, GRAVEL RA: Apoptotic cell death in mouse models of GM2 gangliosidosis and observations on human Tay-Sachs and Sandhoff diseases. Hum. Mol Genet. (1997) 6:1879–1885.
  • BASKIN GB, ALROY J, LI YT, DAYAL Y, RAGHAVAN SS, SHARER L: Galactosylceramide-lipidosis in rhesus monkeys. Lab. Invest. (1989) 60:7A.
  • BASKIN GB, RATTERREE M, DAVISON BB et al: Genetic galactocerebrosidase deficiency (globoid cell leukodystrophy, Krabbe disease) in rhesus monkeys (Macaca mulatta). Lab. Anim. Sci. (1998) 48:476–482.
  • SUZUKI K, SUZUKI K: Genetic galactosylceramidase deficiency (globoid cell leukodystrophy, Krabbe disease) in different mammalian species. Neurochem. Pathol (1985) 3:53–68.
  • KOBAYASHI T, GOTO I, YAMANAKA T, SUZUKI Y, NAKANO T, SUZUKI K: Infantile and fetal globoid cell leukodystrophy: analysis of galactosylceramide and galactosylsphingosine. Ann. NeuroL (1988) 24:517–522.
  • SVENNERHOLM L, VANIET MT, MANSSON JE: Krabbe disease: a galactosylsphingosine (psychosine) lipidosis. J. Lipid Res. (1980) 21:53–64.
  • MIYATAKI T, SUZUKI K: Globoid cell leukodystrophy: additional deficiency of psychosine galactosidase. Biochem. Biophys. Res. Commun. (1972) 48:538–543.
  • VANIER MT, SVENNERHOLM L: Chemical pathology of Krabbe disease: the occurrence of psychosine and other neutral sphingolipids. Adv. Exp. Med. Biol. (1976) 68:115–126.
  • ZLOTOGORA J, REGEV R, HADAR S, IANCU TC: Growth pattern in Krabbe's disease. Acta Paediatr. Scand. (1986) 75:251–254.
  • SUZUKI K: Globoid cell leukodystrophy (Krabbe's disease): update. J. Child Neurol (2003) 18:595–603.
  • LUZI P, RAFI MA, VICTORIA T, BASKIN GB, WENGER DA: Characterization of the rhesus monkey galactocerebrosidase (GALC) cDNA and gene and identification of the mutation causing globoid cell leukodystrophy in this primate. Genomics (1997) 42:407–409.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.