Identification of Hematopoietic Stem Cell–Specific miRNAs Enables Gene Therapy of Globoid Cell Leukodystrophy

Hematopoietic stem cell–specific microRNAs allow regulation of therapeutic transgene expression and enable effective gene therapy of globoid cell leukodystrophy. Scratching the Surface of the Holy Grail In Monty Python and the Holy Grail, when King Arthur cuts off one of the arms of the Black Knight, he claims it is only a scratch. Similarly, gene therapy—the insertion of genes into cells to reverse a condition or repair a biological process—has been heralded as a Holy Grail for the treatment of genetic diseases for nearly 40 years. Yet, the complications of gene therapy, including immune responses to the viral vector and cancers that result from insertional mutagenesis, are more comparable to a severed arm than a surface wound. However, researchers with the resiliency of the Black Knight have presided over recent successes, most notably in metastatic melanoma and immune cells, and have reignited the quest for gene therapy solutions to otherwise untreatable diseases. Gentner et al. build on these successes by identifying new microRNAs that can restrict gene therapy vectors to particular immune cell types and thus be used to safely treat globoid cell leukodystrophy (also known as Krabbe disease). Globoid cell leukodystrophy is a rare metabolic disorder caused by a mutation in a lysosomal enzyme called galactocerebrosidase (GALC). In patients who carry the mutation in both copies of the GALC gene, unmetabolized lipids accumulate in myelin-secreting glial cells, rendering them unable to produce the myelin sheath that normally wraps and protects nerves. This aberration results in severe and often fatal degeneration of motor skills. Bone marrow transplantation has been shown to benefit these patients if the disease is caught early enough. Genetic manipulation of the hematopoietic stem and progenitor cells (HSPCs) found in bone marrow may improve this therapy; however, high-level GALC expression in HSPCs, but not in more differentiated immune cells, is toxic. To address this issue, Gentner et al. identified miRNAs—short RNA sequences that often silence gene expression—that were specifically expressed in HSPCs but not in more differentiated cells. They then used these miRNAs in a GALC/HSPC gene therapy system to suppress GALC function in HSPCs upon transfer into a mouse model of globoid cell leukodystrophy. As these cells matured, amounts of HSPC-specific miRNA decreased and GALC expression increased. This approach protected the HSPCs from GALC toxicity, but allowed for successful gene therapy of the disease. In addition, these hematopoietic stem cell–specific miRNAs could be used as simple markers with which to isolate HSPCs for study and transplantation. This work thus provides a basis for improvements in HSPC-mediated gene therapy and may offer globoid cell leukodystrophy patients a new therapeutic option that resembles a scratch more than a chop. Globoid cell leukodystrophy (GLD; also known as Krabbe disease) is an invariably fatal lysosomal storage disorder caused by mutations in the galactocerebrosidase (GALC) gene. Hematopoietic stem cell (HSC)–based gene therapy is being explored for GLD; however, we found that forced GALC expression was toxic to HSCs and early progenitors, highlighting the need for improved regulation of vector expression. We used a genetic reporter strategy based on lentiviral vectors to detect microRNA activity in hematopoietic cells at single-cell resolution. We report that miR-126 and miR-130a were expressed in HSCs and early progenitors from both mice and humans, but not in differentiated progeny. Moreover, repopulating HSCs could be purified solely on the basis of miRNA expression, providing a new method relevant for human HSC isolation. By incorporating miR-126 target sequences into a GALC-expressing vector, we suppressed GALC expression in HSCs while maintaining robust expression in mature hematopoietic cells. This approach protected HSCs from GALC toxicity and allowed successful treatment of a mouse GLD model, providing a rationale to explore HSC-based gene therapy for GLD.

[1]  L. Naldini,et al.  Endogenous microRNA regulation suppresses transgene expression in hematopoietic lineages and enables stable gene transfer , 2006, Nature Medicine.

[2]  C. Croce,et al.  CD34+ hematopoietic stem-progenitor cell microRNA expression and function: A circuit diagram of differentiation control , 2006, Proceedings of the National Academy of Sciences.

[3]  I. Verma,et al.  Gene therapy: Therapeutic gene causing lymphoma , 2006, Nature.

[4]  L. Naldini,et al.  Gene therapy augments the efficacy of hematopoietic cell transplantation and fully corrects mucopolysaccharidosis type I phenotype in the mouse model. , 2010, Blood.

[5]  D. Gorski,et al.  Regulation of angiogenesis through a microRNA (miR-130a) that down-regulates antiangiogenic homeobox genes GAX and HOXA5. , 2008, Blood.

[6]  D. Bartel,et al.  MicroRNAs in the Hox network: an apparent link to posterior prevalence , 2008, Nature Reviews Genetics.

[7]  R. Humphries,et al.  Hox genes in hematopoiesis and leukemogenesis , 2007, Oncogene.

[8]  E. Passegué,et al.  MicroRNA-126 Regulates HOXA9 by Binding to the Homeobox , 2008, Molecular and Cellular Biology.

[9]  P. Lásló,et al.  Multilineage Transcriptional Priming and Determination of Alternate Hematopoietic Cell Fates , 2006, Cell.

[10]  J. Dick,et al.  Expansion of human cord blood CD34(+)CD38(-) cells in ex vivo culture during retroviral transduction without a corresponding increase in SCID repopulating cell (SRC) frequency: dissociation of SRC phenotype and function. , 2000, Blood.

[11]  A. Orlacchio,et al.  The galactocerebrosidase enzyme contributes to the maintenance of a functional hematopoietic stem cell niche. , 2010, Blood.

[12]  Hans Martin,et al.  Genomic instability and myelodysplasia with monosomy 7 consequent to EVI1 activation after gene therapy for chronic granulomatous disease , 2010, Nature Medicine.

[13]  C. Sander,et al.  A Mammalian microRNA Expression Atlas Based on Small RNA Library Sequencing , 2007, Cell.

[14]  George A Calin,et al.  MicroRNA fingerprints during human megakaryocytopoiesis. , 2006, Proceedings of the National Academy of Sciences of the United States of America.

[15]  F. Deist,et al.  Gene therapy of human severe combined immunodeficiency (SCID)-X1 disease. , 2000, Science.

[16]  I. Weissman,et al.  Identification of a hierarchy of multipotent hematopoietic progenitors in human cord blood. , 2007, Cell stem cell.

[17]  Luigi Naldini,et al.  Endogenous microRNA can be broadly exploited to regulate transgene expression according to tissue, lineage and differentiation state , 2007, Nature Biotechnology.

[18]  J. Dick,et al.  Stem cell concepts renew cancer research. , 2008, Blood.

[19]  Christine Kinnon,et al.  Mutations in TNFRSF13B Encoding TACI Are Associated With Common Variable Immunodeficiency in Humans , 2006, Pediatrics.

[20]  D. Wenger,et al.  Molecular genetics of Krabbe disease (globoid cell leukodystrophy): Diagnostic and clinical implications , 1997, Human mutation.

[21]  J. Lieberman,et al.  Small RNAs Guide Hematopoietic Cell Differentiation and Function , 2010, The Journal of Immunology.

[22]  Allen D. Delaney,et al.  Comprehensive microRNA expression profiling of the hematopoietic hierarchy , 2010, Proceedings of the National Academy of Sciences.

[23]  L. Lockman,et al.  Microglia: The Effector Cell for Reconstitution of the Central Nervous System following Bone Marrow Transplantation for Lysosomal and Peroxisomal Storage Diseases , 1995, Cell transplantation.

[24]  Ru-Fang Yeh,et al.  miR-126 regulates angiogenic signaling and vascular integrity. , 2008, Developmental cell.

[25]  D. Bartel MicroRNAs: Target Recognition and Regulatory Functions , 2009, Cell.

[26]  Manfred Schmidt,et al.  Hematopoietic Stem Cell Gene Therapy with a Lentiviral Vector in X-Linked Adrenoleukodystrophy , 2009, Science.

[27]  Mark Ende,et al.  Hematopoietic stem-cell transplantation. , 2006, The New England journal of medicine.

[28]  O. Kirak,et al.  Regulation of progenitor cell proliferation and granulocyte function by microRNA-223 , 2008, Nature.

[29]  H. W. Lee,et al.  Dissociation between stem cell phenotype and NOD/SCID repopulating activity in human peripheral blood CD34(+) cells after ex vivo expansion. , 2001, Experimental hematology.

[30]  Luigi Naldini,et al.  Stable knockdown of microRNA in vivo by lentiviral vectors , 2009, Nature Methods.

[31]  L. Naldini,et al.  Correction of metachromatic leukodystrophy in the mouse model by transplantation of genetically modified hematopoietic stem cells. , 2004, The Journal of clinical investigation.

[32]  M. Belickova,et al.  MicroRNA expression profiles in umbilical cord blood cell lineages. , 2010, Stem cells and development.

[33]  Alessandro Aiuti,et al.  Gene therapy for immunodeficiency due to adenosine deaminase deficiency. , 2009, The New England journal of medicine.

[34]  John McAnally,et al.  The endothelial-specific microRNA miR-126 governs vascular integrity and angiogenesis. , 2008, Developmental cell.

[35]  L. Naldini,et al.  Gene therapy of metachromatic leukodystrophy reverses neurological damage and deficits in mice. , 2006, The Journal of clinical investigation.