H. Autorité-de-santé-dépistage-néonatal-de-la-drépanocytose-en-france, depistage-neonatal-de-la-drepanocytose-en-france 2. OMS | Epidémiologie mondiale des troubles de l'hémoglobine et indicateurs de service dérivés, Bulletin épidémiologique hebdomadaire / Publications et outils, 2012.

I. Akinsheye, A. Alsultan, N. Solovieff, D. Ngo, C. Baldwin et al., Fetal hemoglobin in sickle cell anemia, Blood, vol.118, issue.1, pp.19-27, 2011.
DOI : 10.1182/blood-2011-03-325258

R. Agrawal, R. Patel, V. Shah, L. Nainiwal, and B. Trivedi, Hydroxyurea in Sickle Cell Disease: Drug Review, Indian Journal of Hematology and Blood Transfusion, vol.101, issue.2, pp.91-97, 2014.
DOI : 10.1016/S0027-9684(15)31072-5

URL : https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4022916/pdf

G. Article-bulletin-Épidémiologique-hebdomadaire-galarneau, C. Palmer, V. Sankaran, S. Orkin, J. Hirschhorn et al., Fine-mapping at three loci known to affect fetal hemoglobin levels explains additional genetic variation, Nat Genet. déc, vol.842, issue.12, pp.1049-51, 2010.

D. Labie, Les mécanismes de régulation de l'hémoglobine foetale. Hématologie. 1 mai 2010, pp.235-278

J. Gilman and T. Huisman, DNA sequence variation associated with elevated fetal G gamma globin production, Blood. oct, vol.66, issue.4, pp.783-790, 1985.

G. Lettre, V. Sankaran, M. Bezerra, A. Araújo, M. Uda et al., DNA polymorphisms at the BCL11A, HBS1L-MYB, and ??-globin loci associate with fetal hemoglobin levels and pain crises in sickle cell disease, Proceedings of the National Academy of Sciences, vol.81, issue.3, pp.11869-74, 2008.
DOI : 10.1086/519795

G. Galarneau, C. Palmer, V. Sankaran, S. Orkin, J. Hirschhorn et al., Fine-mapping at three loci known to affect fetal hemoglobin levels explains additional genetic variation, Nature Genetics, vol.42, issue.12, pp.1049-51, 2010.
DOI : 10.1371/journal.pgen.0020132

URL : https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3740938/pdf

M. Uda, R. Galanello, S. Sanna, G. Lettre, V. Sankaran et al., Genome-wide association study shows BCL11A associated with persistent fetal hemoglobin and amelioration of the phenotype of ??-thalassemia, Proceedings of the National Academy of Sciences, vol.55, issue.4, pp.1620-1625, 2008.
DOI : 10.1111/j.0006-341X.1999.00997.x

D. Labie, BCL11A contrôle l'expression de l'hémoglobine foetale. médecine/sciences. 1 nov 2012, pp.923-928

D. Labie, expression de l'hémoglobine foetale est sous le contrôle du répresseur BCL11A. médecine/sciences, pp.457-60, 2009.

A. Wonkam, N. Bitoungui, V. Vorster, A. Ramesar, R. Cooper et al., Association of Variants at BCL11A and HBS1L-MYB with Hemoglobin F and Hospitalization Rates among Sickle Cell Patients in Cameroon, PLoS ONE, vol.122, issue.6, p.92506, 2014.
DOI : 10.1371/journal.pone.0092506.s004

Y. Lai, Y. Chen, B. Chen, H. Zheng, S. Yi et al., -Thalassemia Intermedia Patients, Hemoglobin, vol.79, issue.6, pp.405-415, 2016.
DOI : 10.1002/ajh.22221

URL : https://hal.archives-ouvertes.fr/in2p3-01415342

S. Thein, S. Menzel, X. Peng, S. Best, J. Jiang et al., Intergenic variants of HBS1L-MYB are responsible for a major quantitative trait locus on chromosome 6q23 influencing fetal hemoglobin levels in adults, Proceedings of the National Academy of Sciences, vol.66, issue.1, pp.11346-51, 2007.
DOI : 10.1086/302698

R. Stadhouders, S. Aktuna, S. Thongjuea, A. Aghajanirefah, F. Pourfarzad et al., HBS1L-MYB intergenic variants modulate fetal hemoglobin via long-range MYB enhancers, Journal of Clinical Investigation, vol.124, issue.4, pp.1699-710, 2014.
DOI : 10.1172/JCI71520DS1

URL : http://www.jci.org/articles/view/71520/files/pdf

M. Steinberg and P. Sebastiani, Genetic modifiers of sickle cell disease, American Journal of Hematology, vol.97, issue.8, pp.795-803, 2012.
DOI : 10.1007/BF02185763

P. Joly, C. Pondarré, C. Bardel, F. A. Martin, and C. , The alpha-globin genotype does not influence sickle cell disease severity in a retrospective cross-validation study of the pediatric severity score, European Journal of Haematology, vol.339, issue.1, pp.61-68, 2012.
DOI : 10.1056/NEJM199807023390102

URL : https://hal.archives-ouvertes.fr/hal-00697964

M. Rumaney, V. Bitoungui, A. Vorster, R. Ramesar, A. Kengne et al., The Co-Inheritance of Alpha-Thalassemia and Sickle Cell Anemia Is Associated with Better Hematological Indices and Lower Consultations Rate in Cameroonian Patients and Could Improve Their Survival, PLoS ONE, vol.7, issue.6, p.100516, 2014.
DOI : 10.1371/journal.pone.0100516.t004

M. Steinberg and R. Hebbel, Clinical diversity of sickle cell anemia: Genetic and cellular modulation of disease severity, American Journal of Hematology, vol.306, issue.4, pp.405-421, 1983.
DOI : 10.7326/0003-4819-88-1-1

G. Kato, M. Gladwin, and M. Steinberg, Deconstructing sickle cell disease: Reappraisal of the role of hemolysis in the development of clinical subphenotypes, Blood Reviews, vol.21, issue.1, pp.37-47, 2007.
DOI : 10.1016/j.blre.2006.07.001

M. Steinberg, Genetic Etiologies for Phenotypic Diversity in Sickle Cell Anemia, The Scientific World JOURNAL, vol.9, pp.46-67, 2009.
DOI : 10.1100/tsw.2009.10

J. Flanagan, D. Frohlich, T. Howard, W. Schultz, C. Driscoll et al., Genetic predictors for stroke in children with sickle cell anemia, Blood, vol.117, issue.24, pp.6681-6685, 2011.
DOI : 10.1182/blood-2011-01-332205

S. Embury, M. Clark, G. Monroy, and N. Mohandas, Concurrent sickle cell anemia and alpha-thalassemia. Effect on pathological properties of sickle erythrocytes., Journal of Clinical Investigation, vol.73, issue.1, pp.116-139, 1984.
DOI : 10.1172/JCI111181

L. Chaouch, M. Kalai, D. Chaouachi, F. Mallouli, R. Hafsia et al., Gilbert syndrome acts as a risk factor of developing gallstone among ? hemoglobinopathy Tunisian patients, Tunis Med. avr, vol.93, issue.4, pp.237-278, 2015.

E. Haverfield, C. Mckenzie, T. Forrester, N. Bouzekri, R. Harding et al., UGT1A1 variation and gallstone formation in sickle cell disease, Blood, vol.105, issue.3, pp.968-72, 2005.
DOI : 10.1182/blood-2004-02-0521

URL : http://www.bloodjournal.org/content/bloodjournal/105/3/968.full.pdf

S. Diop, A. Sene, M. Cisse, A. Toure, O. Sow et al., Prevalence and morbidity of G6PD deficiency in sickle cell disease in the homozygote], Dakar Méd, vol.50, issue.2, pp.56-60, 2005.

J. Bouanga, R. Mouélé, C. Préhu, H. Wajcman, J. Feingold et al., Glucose-6-Phosphate Dehydrogenase Deficiency and Homozygous Sickle Cell Disease in Congo, Human Heredity, vol.48, issue.4, pp.192-199, 1998.
DOI : 10.1159/000022801

M. El-hazmi, A. Warsy, H. Bahakim, and A. Swailem, Glucose-6-Phosphate Dehydrogenase Deficiency and the Sickle Cell Gene in Makkah, Saudi Arabia, Journal of Tropical Pediatrics, vol.40, issue.1, pp.12-18, 1994.
DOI : 10.1093/tropej/40.1.12

P. Joly, N. Garnier, K. Kebaili, C. Renoux, A. Dony et al., G6PD deficiency and absence of ??-thalassemia increase the risk for cerebral vasculopathy in children with sickle cell anemia, European Journal of Haematology, vol.163, issue.Suppl 1, pp.404-412, 2016.
DOI : 10.1111/bjh.12590

A. Belisário, R. Sales, R. , E. Toledo, N. Velloso-rodrigues et al., Glucose-6-Phosphate Dehydrogenase Deficiency in Brazilian Children With Sickle Cell Anemia is not Associated With Clinical Ischemic Stroke or High-Risk Transcranial Doppler, Pediatric Blood & Cancer, vol.109, issue.6, pp.1046-1055, 2016.
DOI : 10.1590/0074-0276140123

M. Benkerrou, C. Alberti, N. Couque, Z. Haouari, A. Ba et al., Impact of glucose-6-phosphate dehydrogenase deficiency on sickle cell anaemia expression in infancy and early childhood: a prospective study, British Journal of Haematology, vol.88, issue.5, pp.646-54, 2013.
DOI : 10.1073/pnas.88.19.8568

H. Autorité-de-santé-ald, 10 -Syndromes drépanocytaires majeurs de l'enfant et de l'adolescent. https://www.has-sante.fr/portail/jcms/c_938890/fr/ald-n-10-syndromes- drepanocytaires-majeurs-de-l-enfant-et-de-l-adolescent

E. Vichinsky, D. Hurst, A. Earles, K. Kleman, and B. Lubin, Newborn screening for sickle cell disease: effect on mortality, Pediatrics. juin, vol.81, issue.6, pp.749-55, 1988.

D. Bauer and S. Orkin, Update on fetal hemoglobin gene regulation in hemoglobinopathies, Current Opinion in Pediatrics, vol.23, issue.1, pp.1-8, 2011.
DOI : 10.1097/MOP.0b013e3283420fd0

URL : https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3092400/pdf

M. Odièvre, C. Lapouméroulie, and J. Elion, Hydroxyur??e et dr??panocytose??: r??le des prot??ines d???adh??rence, Archives de P??diatrie, vol.16, issue.2, 2009.
DOI : 10.1016/j.arcped.2008.11.003

C. Et-génétique-de-la-drépanocytose, PDF Download Available). https://www.researchgate.net/publication

F. Bernaudin, Greffe dans la drépanocytose : résultats, perspectives. Httpwwwem- Premiumcomdocelecu-Bordeauxfrdatarevues0929693X0015000508718578, p.32, 2008.
DOI : 10.1016/s0929-693x(08)71857-8

E. Gluckman, B. Cappelli, F. Bernaudin, M. Labopin, F. Volt et al., Sickle cell disease: an international survey of results of HLA-identical sibling hematopoietic stem cell transplantation, Blood, vol.129, issue.11, pp.1548-56, 2017.
DOI : 10.1182/blood-2016-10-745711

J. Ribeil, S. Hacein-bey-abina, E. Payen, A. Magnani, M. Semeraro et al., Gene Therapy in a Patient with Sickle Cell Disease, New England Journal of Medicine, vol.376, issue.9, pp.848-55, 2017.
DOI : 10.1056/NEJMoa1609677

S. Ballas, Sickle cell anemia with few painful crises is characterized by decreased red cell deformability and increased number of dense cells, American Journal of Hematology, vol.74, issue.2, pp.122-152, 1991.
DOI : 10.1002/ajh.2830360211

G. Kato, R. Hebbel, M. Steinberg, and M. Gladwin, Vasculopathy in sickle cell disease: Biology, pathophysiology, genetics, translational medicine, and new research directions, American Journal of Hematology, vol.37, issue.Pt 3, pp.618-643, 2009.
DOI : 10.1042/bj2810627

URL : https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3209715/pdf

G. Kato, V. Mcgowan, R. Machado, J. Little, J. Taylor et al., Lactate dehydrogenase as a biomarker of hemolysis-associated nitric oxide resistance, priapism, leg ulceration, pulmonary hypertension, and death in patients with sickle cell disease, Blood, vol.107, issue.6, pp.2279-85, 2006.
DOI : 10.1182/blood-2005-06-2373

E. Vichinsky, L. Neumayr, J. Gold, M. Weiner, R. Rule et al., Neuropsychological Dysfunction and Neuroimaging Abnormalities in Neurologically Intact Adults With Sickle Cell Anemia, JAMA, vol.303, issue.18, pp.1823-1854, 2010.
DOI : 10.1001/jama.2010.562

M. Kossorotoff, D. Grevent, and M. De-montalembert, Drépanocytose et atteinte vasculaire cérébrale chez l'enfant. Arch Pédiatrie, pp.404-418, 2014.
DOI : 10.1016/j.arcped.2014.01.005

C. Morris, Vascular risk assessment in patients with sickle cell disease, Haematologica, vol.96, issue.1, pp.1-5, 2011.
DOI : 10.3324/haematol.2010.035097

V. Land, . Van-der, M. Peters, B. Biemond, H. Heijboer et al., Markers of endothelial dysfunction differ between subphenotypes in children with sickle cell disease

M. Muszlak, S. Pissard, C. Badens, A. Chamouine, O. Maillard et al., Genetic Modifiers of Sickle Cell Disease: A Genotype-Phenotype Relationship Study in a Cohort of 82 Children on Mayotte Island, Hemoglobin, vol.35, issue.5, pp.156-61, 2015.
DOI : 10.1002/ajh.23457

H. Rosenberg, R. Markowitz, H. Kolberg, C. Park, A. Hubbard et al., Normal splenic size in infants and children: sonographic measurements., American Journal of Roentgenology, vol.157, issue.1, pp.119-140, 1991.
DOI : 10.2214/ajr.157.1.2048509

URL : http://www.ajronline.org/doi/pdf/10.2214/ajr.157.1.2048509

S. Mtatiro, T. Singh, H. Rooks, J. Mgaya, H. Mariki et al., Genome Wide Association Study of Fetal Hemoglobin in Sickle Cell Anemia in Tanzania, PLoS ONE, vol.5, issue.11, p.111464, 2014.
DOI : 10.1371/journal.pone.0111464.t003

M. Montalembert, . De, and R. Girot, Drépanocytose chez l'enfant. Httpwwwem- Premiumcomdocelecu-Bordeauxfrdatatraitespem04-61616

S. Allali, T. Peyrard, D. Amiranoff, J. Cohen, M. Chalumeau et al., Prevalence and risk factors for red blood cell alloimmunization in 175 children with sickle cell disease in a French university hospital reference centre, British Journal of Haematology, vol.26, issue.4, pp.641-648, 2017.
DOI : 10.1111/tme.12314

K. Yazdanbakhsh, R. Ware, and F. Noizat-pirenne, Red blood cell alloimmunization in sickle cell disease: pathophysiology, risk factors, and transfusion management, Blood, vol.120, issue.3, pp.528-565, 2012.
DOI : 10.1182/blood-2011-11-327361

URL : https://hal.archives-ouvertes.fr/inserm-00696264

P. Telfer, P. Coen, S. Chakravorty, O. Wilkey, J. Evans et al., Clinical outcomes in children with sickle cell disease living in England: a neonatal cohort in East London, Haematologica, vol.92, issue.7, pp.905-917, 2007.
DOI : 10.3324/haematol.10937

K. Ohene-frempong, S. Weiner, L. Sleeper, S. Miller, S. Embury et al., Cerebrovascular Accidents in Sickle Cell Disease: Rates and Risk Factors, Blood. 1 janv, vol.91, issue.1, pp.288-94, 1998.

F. Bernaudin, S. Verlhac, C. Arnaud, A. Kamdem, S. Chevret et al., Impact of early transcranial Doppler screening and intensive therapy on cerebral vasculopathy outcome in a newborn sickle cell anemia cohort, Blood, vol.117, issue.4, pp.1130-1170, 2011.
DOI : 10.1182/blood-2010-06-293514

J. Sommet, C. Alberti, N. Couque, S. Verlhac, Z. Haouari et al., Clinical and haematological risk factors for cerebral macrovasculopathy in a sickle cell disease newborn cohort: a prospective study, British Journal of Haematology, vol.120, issue.6, pp.966-77, 2016.
DOI : 10.1182/blood-2011-11-327361

J. Makani, S. Menzel, S. Nkya, S. Cox, E. Drasar et al., Genetics of fetal hemoglobin in Tanzanian and British patients with sickle cell anemia, Blood, vol.117, issue.4, pp.1390-1392, 2011.
DOI : 10.1182/blood-2010-08-302703

G. Cardoso, I. Diniz, A. Silva, . Da, D. Cunha et al., DNA polymorphisms at BCL11A, HBS1L-MYB and Xmn1-HBG2 site loci associated with fetal hemoglobin levels in sickle cell anemia patients from Northern Brazil, Blood Cells, Molecules, and Diseases, vol.53, issue.4, pp.176-185, 2014.
DOI : 10.1016/j.bcmd.2014.07.006

J. Farrell, R. Sherva, Z. Chen, H. Luo, B. Chu et al., A 3-bp deletion in the HBS1L-MYB intergenic region on chromosome 6q23 is associated with HbF expression, Blood, vol.117, issue.18, pp.4935-4980, 2011.
DOI : 10.1182/blood-2010-11-317081

V. Van-der-land, H. Mutsaerts, M. Engelen, H. Heijboer, M. Roest et al., Risk factor analysis of cerebral white matter hyperintensities in children with sickle cell disease, British Journal of Haematology, vol.31, issue.2, pp.274-84, 2016.
DOI : 10.1016/j.neuroimage.2006.01.015

M. Steinberg, D. Chui, G. Dover, P. Sebastiani, and A. Alsultan, Sickle Cell Anemia and Fetal Hemoglobin, The American Journal of the Medical Sciences, vol.308, issue.5, pp.481-486, 2014.
DOI : 10.1097/00000441-199411000-00001

N. Green and S. Barral, Emerging science of hydroxyurea therapy for pediatric sickle cell disease, Pediatric Research, vol.101, issue.1-2, pp.196-204, 2014.
DOI : 10.1016/j.bbmt.2012.05.013

J. Friedrisch, V. Sheehan, J. Flanagan, A. Baldan, C. Summarell et al., The role of BCL11A and HMIP-2 polymorphisms on endogenous and hydroxyurea induced levels of fetal hemoglobin in sickle cell anemia patients from southern Brazil, Blood Cells, Molecules, and Diseases, vol.62, pp.32-39, 2016.
DOI : 10.1016/j.bcmd.2016.11.002

L. Creary, P. Ulug, S. Menzel, C. Mckenzie, N. Hanchard et al., Genetic Variation on Chromosome 6 Influences F Cell Levels in Healthy Individuals of African Descent and HbF Levels in Sickle Cell Patients, PLoS ONE, vol.10, issue.1, 2621086.
DOI : 10.1371/journal.pone.0004218.t002

F. Danjou, M. Francavilla, A. F. Satta, S. Demartis, F. Perseu et al., A genetic score for the prediction of beta-thalassemia severity, Haematologica, vol.100, issue.4, pp.452-459, 2015.
DOI : 10.3324/haematol.2014.113886

C. Ndugwa, D. Higgs, C. Fisher, I. Hambleton, K. Mason et al., Homozygous sickle cell disease in Uganda and Jamaica a comparison of Bantu and Benin haplotypes, West Indian Med J. oct, vol.61, issue.7, pp.684-91, 2012.

C. Santos, B. Dias-elias, D. Da-silva-rocha, L. Cavalcante-barbosa, M. Pinheiro-gonçalves et al., Impact of ??S-Globin Haplotypes on Oxidative Stress in Patients with Sickle Cell Anemia in Steady State, Archives of Medical Research, vol.43, issue.7, pp.536-576, 2012.
DOI : 10.1016/j.arcmed.2012.08.014