Recent progress in understanding and manipulating haemoglobin switching for the haemoglobinopathies

The major β‐haemoglobinopathies, sickle cell disease and β‐thalassaemia, represent the most common monogenic disorders worldwide and a steadily increasing global disease burden. Allogeneic haematopoietic stem cell transplantation, the only curative therapy, is only applied to a small minority of patients. Common clinical management strategies act mainly downstream of the root causes of disease. The observation that elevated fetal haemoglobin expression ameliorates these disorders has motivated longstanding investigations into the mechanisms of haemoglobin switching. Landmark studies over the last decade have led to the identification of two potent transcriptional repressors of γ‐globin, BCL11A and ZBTB7A. These regulators act with additional trans‐acting epigenetic repressive complexes, lineage‐defining factors and developmental programs to silence fetal haemoglobin by working on cis‐acting sequences at the globin gene loci. Rapidly advancing genetic technology is enabling researchers to probe deeply the interplay between the molecular players required for γ‐globin (HBG1/HBG2) silencing. Gene therapies may enable permanent cures with autologous modified haematopoietic stem cells that generate persistent fetal haemoglobin expression. Ultimately rational small molecule pharmacotherapies to reactivate HbF could extend benefits widely to patients.

[1]  Beta Thalassemia , 2020, Definitions.

[2]  N. Matsumoto,et al.  Identification of novel BCL11A variants in patients with epileptic encephalopathy: Expanding the phenotypic spectrum , 2018, Clinical genetics.

[3]  Chad A. Cowan,et al.  Utility of CRISPR/Cas9 systems in hematology research. , 2017, Experimental hematology.

[4]  K. Quinlan,et al.  KLF1 drives the expression of fetal hemoglobin in British HPFH. , 2017, Blood.

[5]  R. Proia,et al.  IGF2BP1 overexpression causes fetal-like hemoglobin expression patterns in cultured human adult erythroblasts , 2017, Proceedings of the National Academy of Sciences.

[6]  S. Thein Molecular basis of β thalassemia and potential therapeutic targets. , 2017, Blood cells, molecules & diseases.

[7]  Ryan K. Dale,et al.  The LDB1 complex co-opts CTCF for erythroid lineage specific long-range enhancer interactions , 2017, bioRxiv.

[8]  W. Eaton,et al.  Treating sickle cell disease by targeting HbS polymerization. , 2017, Blood.

[9]  K. Quinlan,et al.  The regulation of human globin promoters by CCAAT box elements and the recruitment of NF-Y. , 2017, Biochimica et biophysica acta. Gene regulatory mechanisms.

[10]  Yukio Nakamura,et al.  An immortalized adult human erythroid line facilitates sustainable and scalable generation of functional red cells , 2017, Nature Communications.

[11]  Matthew C. Canver,et al.  Variant-aware saturating mutagenesis using multiple Cas9 nucleases identifies regulatory elements at trait-associated loci , 2017, Nature Genetics.

[12]  X. Yang,et al.  Long-Term Engraftment and Fetal Globin Induction upon BCL11A Gene Editing in Bone-Marrow-Derived CD34+ Hematopoietic Stem and Progenitor Cells , 2017, Molecular therapy. Methods & clinical development.

[13]  Y. T. Lee,et al.  HMGA2 Moderately Increases Fetal Hemoglobin Expression in Human Adult Erythroblasts , 2016, PloS one.

[14]  J. D. Macklis,et al.  Strict in vivo specificity of the Bcl11a erythroid enhancer. , 2016, Blood.

[15]  Sruthi Mantri,et al.  CRISPR/Cas9 β-globin gene targeting in human haematopoietic stem cells , 2016, Nature.

[16]  J. Vadolas,et al.  Animal models of β-hemoglobinopathies: utility and limitations , 2016, Journal of blood medicine.

[17]  Dana Carroll,et al.  Selection-free genome editing of the sickle mutation in human adult hematopoietic stem/progenitor cells , 2016, Science Translational Medicine.

[18]  Matthew C. Canver,et al.  Lineage-specific BCL11A knockdown circumvents toxicities and reverses sickle phenotype. , 2016, The Journal of clinical investigation.

[19]  S. Orkin,et al.  Bcl11a Deficiency Leads to Hematopoietic Stem Cell Defects with an Aging-like Phenotype. , 2016, Cell reports.

[20]  Yuting Tan,et al.  Genome editing using CRISPR-Cas9 to create the HPFH genotype in HSPCs: An approach for treating sickle cell disease and β-thalassemia , 2016, Proceedings of the National Academy of Sciences.

[21]  Aaron R Cooper,et al.  CRISPR/Cas9-Mediated Correction of the Sickle Mutation in Human CD34+ cells. , 2016, Molecular therapy : the journal of the American Society of Gene Therapy.

[22]  S. Rivella,et al.  Forced chromatin looping raises fetal hemoglobin in adult sickle cells to higher levels than pharmacologic inducers. , 2016, Blood.

[23]  R. Hardison,et al.  A genome-editing strategy to treat β-hemoglobinopathies that recapitulates a mutation associated with a benign genetic condition , 2016, Nature Medicine.

[24]  S. Sawiak,et al.  BCL11A Haploinsufficiency Causes an Intellectual Disability Syndrome and Dysregulates Transcription , 2016, American journal of human genetics.

[25]  G. Lettre,et al.  Fetal haemoglobin in sickle-cell disease: from genetic epidemiology to new therapeutic strategies , 2016, The Lancet.

[26]  T. Maeda,et al.  Regulation of hematopoietic development by ZBTB transcription factors , 2016, International Journal of Hematology.

[27]  J. D. Engel,et al.  The LSD1 inhibitor RN-1 recapitulates the fetal pattern of hemoglobin synthesis in baboons (P. anubis) , 2016, Haematologica.

[28]  S. Orkin,et al.  Paying for future success in gene therapy , 2016, Science.

[29]  J. D. Macklis,et al.  Ctip1 Controls Acquisition of Sensory Area Identity and Establishment of Sensory Input Fields in the Developing Neocortex , 2016, Neuron.

[30]  A. Perkins,et al.  Krüppeling erythropoiesis: an unexpected broad spectrum of human red blood cell disorders due to KLF1 variants. , 2016, Blood.

[31]  J. Shearstone,et al.  Chemical Inhibition of Histone Deacetylases 1 and 2 Induces Fetal Hemoglobin through Activation of GATA2 , 2016, PloS one.

[32]  S. Borwornpinyo,et al.  Gene Therapy of the β-Hemoglobinopathies by Lentiviral Transfer of the βA(T87Q)-Globin Gene , 2016, Human gene therapy.

[33]  Matthew C. Canver,et al.  Transcription factors LRF and BCL11A independently repress expression of fetal hemoglobin , 2016, Science.

[34]  Matthew C. Canver,et al.  EHMT1 and EHMT2 inhibition induces fetal hemoglobin expression. , 2015, Blood.

[35]  Luigi Naldini,et al.  Gene therapy returns to centre stage , 2015, Nature.

[36]  Aleksandra A. Kolodziejczyk,et al.  Single-cell transcriptomic reconstruction reveals cell cycle and multi-lineage differentiation defects in Bcl11a-deficient hematopoietic stem cells , 2015, Genome Biology.

[37]  Matthew C. Canver,et al.  BCL11A enhancer dissection by Cas9-mediated in situ saturating mutagenesis , 2015, Nature.

[38]  Yuri R. Bendaña,et al.  Functional footprinting of regulatory DNA , 2015, Nature Methods.

[39]  S. Orkin,et al.  Hemoglobin switching's surprise: the versatile transcription factor BCL11A is a master repressor of fetal hemoglobin. , 2015, Current opinion in genetics & development.

[40]  Y. T. Lee,et al.  Inhibition of G9a methyltransferase stimulates fetal hemoglobin production by facilitating LCR/γ-globin looping. , 2015, Blood.

[41]  J. D. Engel,et al.  The LSD1 inhibitor RN-1 induces fetal hemoglobin synthesis and reduces disease pathology in sickle cell mice. , 2015, Blood.

[42]  J. Stamatoyannopoulos,et al.  2p15-p16.1 microdeletions encompassing and proximal to BCL11A are associated with elevated HbF in addition to neurologic impairment. , 2015, Blood.

[43]  Jacob C. Ulirsch,et al.  BCL11A deletions result in fetal hemoglobin persistence and neurodevelopmental alterations. , 2015, The Journal of clinical investigation.

[44]  Laura J. Norton,et al.  Editing the genome to introduce a beneficial naturally occurring mutation associated with increased fetal globin , 2015, Nature Communications.

[45]  Lei Zhang,et al.  Correction of the sickle cell disease mutation in human hematopoietic stem/progenitor cells. , 2015, Blood.

[46]  Michael R. Tallack,et al.  KLF1-null neonates display hydrops fetalis and a deranged erythroid transcriptome. , 2014, Blood.

[47]  Philip D. Gregory,et al.  Reactivation of Developmentally Silenced Globin Genes by Forced Chromatin Looping , 2014, Cell.

[48]  Tizhen Yan,et al.  KLF1 mutations are relatively more common in a thalassemia endemic region and ameliorate the severity of β-thalassemia. , 2014, Blood.

[49]  I. Yaniv,et al.  Hematopoietic stem cell transplantation in thalassemia major and sickle cell disease: indications and management recommendations from an international expert panel , 2014, Haematologica.

[50]  Haley O. Tucker,et al.  Dendritic cell fate is determined by BCL11A , 2014, Proceedings of the National Academy of Sciences.

[51]  A. Dean,et al.  The hematopoietic regulator TAL1 is required for chromatin looping between the β-globin LCR and human γ-globin genes to activate transcription , 2014, Nucleic acids research.

[52]  Shondra M. Pruett-Miller,et al.  Nuclease-mediated gene editing by homologous recombination of the human globin locus , 2013, Nucleic acids research.

[53]  Matthew C. Canver,et al.  An Erythroid Enhancer of BCL11A Subject to Genetic Variation Determines Fetal Hemoglobin Level , 2013, Science.

[54]  Y. T. Lee,et al.  LIN28B-mediated expression of fetal hemoglobin and production of fetal-like erythrocytes from adult human erythroblasts ex vivo. , 2013, Blood.

[55]  Ryan K. Dale,et al.  Ldb1-nucleated transcription complexes function as primary mediators of global erythroid gene activation. , 2013, Blood.

[56]  S. Orkin,et al.  Corepressor-dependent silencing of fetal hemoglobin expression by BCL11A , 2013, Proceedings of the National Academy of Sciences.

[57]  Anand P. Patil,et al.  Global epidemiology of sickle haemoglobin in neonates: a contemporary geostatistical model-based map and population estimates , 2013, The Lancet.

[58]  R. Neades,et al.  Mi2β Is Required for γ-Globin Gene Silencing: Temporal Assembly of a GATA-1-FOG-1-Mi2 Repressor Complex in β-YAC Transgenic Mice , 2012, PLoS genetics.

[59]  David C Williams,et al.  Mi2β-mediated silencing of the fetal γ-globin gene in adult erythroid cells. , 2012, Blood.

[60]  Yukio Nakamura,et al.  Establishment of Immortalized Human Erythroid Progenitor Cell Lines Able to Produce Enucleated Red Blood Cells , 2012, PloS one.

[61]  P. Gregory,et al.  Controlling Long-Range Genomic Interactions at a Native Locus by Targeted Tethering of a Looping Factor , 2012, Cell.

[62]  Y. Saunthararajah,et al.  Effects of tetrahydrouridine on pharmacokinetics and pharmacodynamics of oral decitabine. , 2012, Blood.

[63]  Ryan K. Dale,et al.  Distinct Ldb1/NLI complexes orchestrate γ-globin repression and reactivation through ETO2 in human adult erythroid cells. , 2011, Blood.

[64]  Cong Peng,et al.  Correction of Sickle Cell Disease in Adult Mice by Interference with Fetal Hemoglobin Silencing , 2011, Science.

[65]  Hélène Rouard,et al.  Proof of principle for transfusion of in vitro-generated red blood cells. , 2011, Blood.

[66]  Morgan L. Maeder,et al.  In Situ Genetic Correction of the Sickle Cell Anemia Mutation in Human Induced Pluripotent Stem Cells Using Engineered Zinc Finger Nucleases , 2011, Stem cells.

[67]  Ninad M Walavalkar,et al.  p66α–MBD2 coiled-coil interaction and recruitment of Mi-2 are critical for globin gene silencing by the MBD2–NuRD complex , 2011, Proceedings of the National Academy of Sciences.

[68]  E. Lander,et al.  MicroRNA-15a and -16-1 act via MYB to elevate fetal hemoglobin expression in human trisomy 13 , 2011, Proceedings of the National Academy of Sciences.

[69]  Joel N Hirschhorn,et al.  Fine-mapping at three loci known to affect fetal hemoglobin levels explains additional genetic variation , 2010, Nature Genetics.

[70]  A. D. de Brevern,et al.  A dominant mutation in the gene encoding the erythroid transcription factor KLF1 causes a congenital dyserythropoietic anemia. , 2010, American journal of human genetics.

[71]  Jérôme Larghero,et al.  Transfusion independence and HMGA2 activation after gene therapy of human β-thalassaemia , 2010, Nature.

[72]  M. Warr,et al.  Hematopoietic stem cell quiescence promotes error-prone DNA repair and mutagenesis. , 2010, Cell stem cell.

[73]  Timothy L Bailey,et al.  A global role for KLF1 in erythropoiesis revealed by ChIP-seq in primary erythroid cells. , 2010, Genome research.

[74]  F. Grosveld,et al.  Haploinsufficiency for the erythroid transcription factor KLF1 causes Hereditary Persistence of Fetal Hemoglobin , 2010, Nature Genetics.

[75]  Christine Steinhoff,et al.  The genome-wide dynamics of the binding of Ldb1 complexes during erythroid differentiation. , 2010, Genes & development.

[76]  E. Reddy,et al.  Conditional c-myb knockout in adult hematopoietic stem cells leads to loss of self-renewal due to impaired proliferation and accelerated differentiation , 2009, Proceedings of the National Academy of Sciences.

[77]  F. Marincola,et al.  Let-7 microRNAs are developmentally regulated in circulating human erythroid cells , 2009, Journal of Translational Medicine.

[78]  Christian Gieger,et al.  A genome-wide meta-analysis identifies 22 loci associated with eight hematological parameters in the HaemGen consortium , 2009, Nature Genetics.

[79]  C. Palii,et al.  Dual role for the methyltransferase G9a in the maintenance of β-globin gene transcription in adult erythroid cells , 2009, Proceedings of the National Academy of Sciences.

[80]  P. Pandolfi,et al.  LRF is an essential downstream target of GATA1 in erythroid development and regulates BIM-dependent apoptosis. , 2009, Developmental cell.

[81]  Christian Gieger,et al.  Multiple loci influence erythrocyte phenotypes in the CHARGE Consortium , 2009, Nature Genetics.

[82]  Stuart H. Orkin,et al.  Developmental and species-divergent globin switching are driven by BCL11A , 2009, Nature.

[83]  S. Hurtley Editing the Genome , 2009 .

[84]  Robert L Moritz,et al.  PRMT5-mediated methylation of histone H4R3 recruits DNMT3A, coupling histone and DNA methylation in gene silencing , 2009, Nature Structural &Molecular Biology.

[85]  J. Hirschhorn,et al.  Supporting Online Material Materials and Methods Figs. S1 to S10 Tables S1 to S7 References Human Fetal Hemoglobin Expression Is Regulated by the Developmental Stage-specific Repressor Bcl11a , 2022 .

[86]  W. de Laat,et al.  Joining the loops: β‐Globin gene regulation , 2008, IUBMB life.

[87]  N. Burton,et al.  Mutations in EKLF/KLF1 form the molecular basis of the rare blood group In(Lu) phenotype. , 2008, Blood.

[88]  J. Hirschhorn,et al.  DNA polymorphisms at the BCL11A, HBS1L-MYB, and β-globin loci associate with fetal hemoglobin levels and pain crises in sickle cell disease , 2008, Proceedings of the National Academy of Sciences.

[89]  Matthew Darlison,et al.  Global epidemiology of haemoglobin disorders and derived service indicators. , 2008, Bulletin of the World Health Organization.

[90]  Gonçalo R. Abecasis,et al.  Genome-wide association study shows BCL11A associated with persistent fetal hemoglobin and amelioration of the phenotype of β-thalassemia , 2008, Proceedings of the National Academy of Sciences.

[91]  Chunhui Hou,et al.  A positive role for NLI/Ldb1 in long-range beta-globin locus control region function. , 2007, Molecular cell.

[92]  Simon Heath,et al.  A QTL influencing F cell production maps to a gene encoding a zinc-finger protein on chromosome 2p15 , 2007, Nature Genetics.

[93]  G. Abecasis,et al.  Heritability of Cardiovascular and Personality Traits in 6,148 Sardinians , 2006, PLoS genetics.

[94]  J. Frampton,et al.  Coordination of erythropoiesis by the transcription factor c-Myb. , 2006, Blood.

[95]  M. Nefedov,et al.  Humanized β-Thalassemia Mouse Model Containing the Common IVSI-110 Splicing Mutation* , 2006, Journal of Biological Chemistry.

[96]  M. Porteus,et al.  Mammalian gene targeting with designed zinc finger nucleases. , 2006, Molecular therapy : the journal of the American Society of Gene Therapy.

[97]  A. McDowall,et al.  A global role for EKLF in definitive and primitive erythropoiesis. , 2005, Blood.

[98]  F. Grosveld,et al.  The Erythroid Phenotype of EKLF-Null Mice: Defects in Hemoglobin Metabolism and Membrane Stability , 2005, Molecular and Cellular Biology.

[99]  M. Groudine,et al.  Proximity among distant regulatory elements at the beta-globin locus requires GATA-1 and FOG-1. , 2005, Molecular cell.

[100]  F. Grosveld,et al.  The active spatial organization of the beta-globin locus requires the transcription factor EKLF. , 2004, Genes & development.

[101]  Takuro Nakamura,et al.  Bcl11a is essential for normal lymphoid development , 2003, Nature Immunology.

[102]  Erik Splinter,et al.  Looping and interaction between hypersensitive sites in the active beta-globin locus. , 2002, Molecular cell.

[103]  D. J. Weatherall,et al.  Phenotype—genotype relationships in monogenic disease: lessons from the thalassaemias , 2001, Nature Reviews Genetics.

[104]  B. Reinhart,et al.  Conservation of the sequence and temporal expression of let-7 heterochronic regulatory RNA , 2000, Nature.

[105]  S. Orkin,et al.  Fetal expression of a human Agamma globin transgene rescues globin chain imbalance but not hemolysis in EKLF null mouse embryos. , 2000, Blood.

[106]  D. Leprince,et al.  Novel BTB/POZ domain zinc-finger protein, LRF, is a potential target of the LAZ-3/BCL-6 oncogene , 1999, Oncogene.

[107]  B. Forget Molecular Basis of Hereditary Persistence of Fetal Hemoglobin , 1998, Annals of the New York Academy of Sciences.

[108]  R. Kole,et al.  A common human beta globin splicing mutation modeled in mice. , 1998, Blood.

[109]  A. Mutero,et al.  The human beta globin locus introduced by YAC transfer exhibits a specific and reproducible pattern of developmental regulation in transgenic mice. , 1997, Blood.

[110]  E. Rubin,et al.  Transgenic knockout mice with exclusively human sickle hemoglobin and sickle cell disease. , 1997, Science.

[111]  T. Townes,et al.  Knockout-transgenic mouse model of sickle cell disease. , 1997, Science.

[112]  T. Rabbitts,et al.  The LIM‐only protein Lmo2 is a bridging molecule assembling an erythroid, DNA‐binding complex which includes the TAL1, E47, GATA‐1 and Ldb1/NLI proteins , 1997, The EMBO journal.

[113]  S. Orkin,et al.  Lethal β-thalassaemia in mice lacking the erythroid CACCC-transcription factor EKLF , 1995, Nature.

[114]  F. Grosveld,et al.  Defective haematopoiesis in fetal liver resulting from inactivation of the EKLF gene , 1995, Nature.

[115]  M L Terrin,et al.  Effect of hydroxyurea on the frequency of painful crises in sickle cell anemia. Investigators of the Multicenter Study of Hydroxyurea in Sickle Cell Anemia. , 1995, The New England journal of medicine.

[116]  O. Platt,et al.  Mortality in sickle cell disease. Life expectancy and risk factors for early death. , 1994, The New England journal of medicine.

[117]  Y. Kan,et al.  Germ-line transmission and developmental regulation of a 150-kb yeast artificial chromosome containing the human beta-globin locus in transgenic mice. , 1993, Proceedings of the National Academy of Sciences of the United States of America.

[118]  J. Bieker,et al.  A novel, erythroid cell-specific murine transcription factor that binds to the CACCC element and is related to the Krüppel family of nuclear proteins , 1993, Molecular and cellular biology.

[119]  T. Huisman,et al.  Distal CCAAT box deletion in the A gamma globin gene of two black adolescents with elevated fetal A gamma globin. , 1988, Nucleic acids research.

[120]  S. Orkin,et al.  Hydroxyurea enhances fetal hemoglobin production in sickle cell anemia. , 1984, The Journal of clinical investigation.

[121]  T. Ley,et al.  5-azacytidine selectively increases gamma-globin synthesis in a patient with beta+ thalassemia. , 1982, The New England journal of medicine.

[122]  D. Zwiers,et al.  5-Azacytidine stimulates fetal hemoglobin synthesis in anemic baboons. , 1982, Proceedings of the National Academy of Sciences of the United States of America.

[123]  J. Clegg,et al.  The British type of non‐deletion HPFH: characterization of developmental changes in vivo and erythroid growth in vitro , 1982, British journal of haematology.

[124]  H. Koeffler,et al.  Human myeloid leukemia cell lines: a review. , 1980, Blood.

[125]  R. Flavell,et al.  DNA methylation in the human γδβ-globin locus in erythroid and nonerythroid tissues , 1980, Cell.

[126]  G. Serjeant,et al.  HETEROCELLULAR HEREDITARY PERSISTENCE OF FETAL HÆMOGLOBIN AND HOMOZYGOUS SICKLE-CELL DISEASE , 1977, The Lancet.

[127]  H. Aviv,et al.  Messenger RNA population analysis during erythroid differentiation: a kinetical approach. , 1977, Journal of molecular biology.

[128]  T. Huisman,et al.  A G gamma type of the hereditary persistence of fetal hemoglobin with beta chain production in cis. , 1975, American journal of human genetics.

[129]  F. J. Ensell,et al.  ATMOSPHERIC OZONE AND FEMORAL FRACTURES , 1975, The Lancet.

[130]  C. Lozzio,et al.  Human chronic myelogenous leukemia cell-line with positive Philadelphia chromosome. , 1975, Blood.

[131]  J. Clegg,et al.  A Form of Hereditary Persistence of Fetal Haemoglobin Characterized by Uneven Cellular Distribution of Haemoglobin F and the Production of Haemoglobins A and A2 in Homozygotes , 1975, British journal of haematology.

[132]  G. Stamatoyannopoulos,et al.  HEREDITARY PERSISTENCE OF FETAL HEMOGLOBIN IN GREECE. A STUDY AND A COMPARISON. , 1964, Blood.

[133]  W. Schroeder,et al.  THE AMINO ACID SEQUENCE OF THE GAMMA CHAIN OF HUMAN FETAL HEMOGLOBIN. , 1963, Biochemistry.

[134]  Charlotte Friend,et al.  CELL-FREE TRANSMISSION IN ADULT SWISS MICE OF A DISEASE HAVING THE CHARACTER OF A LEUKEMIA , 1957, The Journal of experimental medicine.

[135]  Watson Cj Fundamental and clinical aspects of hemoglobin and bile pigment metabolism. , 1948 .

[136]  E. Asmussen,et al.  SOME PROPERTIES OF HUMAN FETAL AND MATERNAL BLOOD. , 1941, The Journal of clinical investigation.

[137]  J. Davies,et al.  Gene Therapy in a Patient with Sickle Cell Disease. , 2017, The New England journal of medicine.

[138]  J. Stockman Proof of principle for transfusion of in vitro–generated red blood cells , 2013 .

[139]  R. Ware,et al.  Advances in the use of hydroxyurea. , 2009, Hematology. American Society of Hematology. Education Program.

[140]  G. Stamatoyannopoulos Hereditary Persistence of Fetal Hemoglobin in Greece , 2005 .

[141]  A. Migliaccio,et al.  In vitro mass production of human erythroid cells from the blood of normal donors and of thalassemic patients. , 2002, Blood cells, molecules & diseases.

[142]  M Farrall,et al.  Genetic influences on F cells and other hematologic variables: a twin heritability study. , 2000, Blood.

[143]  G. Lathrop,et al.  Dissecting the loci controlling fetal haemoglobin production on chromosomes 11p and 6q by the regressive approach , 1996, Nature Genetics.

[144]  Peter Fraser,et al.  Transcription complex stability and chromatin dynamics in vivo , 1995, Nature.

[145]  M. Antoniou Induction of Erythroid-Specific Expression in Murine Erythroleukemia (MEL) Cell Lines. , 1991, Methods in molecular biology.

[146]  R. Flavell,et al.  DNA methylation in the human gamma delta beta-globin locus in erythroid and nonerythroid tissues. , 1980, Cell.

[147]  C. J. Watson Fundamental and clinical aspects of hemoglobin and bile pigment metabolism. , 1948, Boletin de la Asociacion Medica de Puerto Rico.