Genetic aspects of congenital and idiopathic scoliosis

Philip F Giampietro, Philip F Giampietro

Abstract

Congenital and idiopathic scoliosis represent disabling conditions of the spine. While congenital scoliosis (CS) is caused by morphogenic abnormalities in vertebral development, the cause(s) for idiopathic scoliosis is (are) likely to be varied, representing alterations in skeletal growth, neuromuscular imbalances, disturbances involving communication between the brain and spine, and others. Both conditions are characterized by phenotypic and genetic heterogeneities, which contribute to the difficulties in understanding their genetic basis that investigators face. Despite the differences between these two conditions there is observational and experimental evidence supporting common genetic mechanisms. This paper focuses on the clinical features of both CS and IS and highlights genetic and environmental factors which contribute to their occurrence. It is anticipated that emerging genetic technologies and improvements in phenotypic stratification of both conditions will facilitate improved understanding of the genetic basis for these conditions and enable targeted prevention and treatment strategies.

Figures

Figure 1
Figure 1
Diagram of spine illustrating defects of formation (wedge and hemivertebrae) and segmentation (vertebral bar and block vertebrae). Reprinted with permission from McMaster [6].
Figure 2
Figure 2
3D reconstruction illustrating congenital scoliosis. Left T4 hemivertebrae displayed. Courtesy of Dr. Kenneth Noonan.
Figure 3
Figure 3
Algorithm for International Consortium for Vertebral Anomalies and Scoliosis (ICVAS) classification of congenital vertebral malformation. Reproduced with permission Expert opinion in [7]. Reproduced from Expert Opinion in Expert Opin. Med. Diagn. (2008) 2(10):1107-1121 with permission of Informa UK Ltd.
Figure 4
Figure 4
Illustration of somite formation from the presomitic mesoderm (PSM) in the chick embryo. Paired somites are formed every 90 minutes in a periodic fashion every 90 minutes shown in (a). (b).A molecular clocked linked to segmentation by dynamic and periodic expression of the cyclic genes in the PSM. Top: Lunatic Fringe mRNA expression appears as a wave sweeping across the whole PSM once during each somite formation as illustrated by in situ hybridization in this 17-somite-old chick embryo. During each somite formations, PSM cells illustrated by the dot undergo a phase of upregulation of the cycling genes followed by a phase of down regulation of these genes. Bottom: As shown in this schematic representation of the progression of somitogenesis in the embryo, the cycles of expression of the cyclic genes will last while the cells remain in the PSM, which corresponds approximately to the time to form 12 somites in the chick embryo. These PSM cells undergo 12 oscillations of the expression of the cycling genes. Reproduced with permission Expert Opinion in [9]. Reproduced from Expert Opinion in Expert Opin. Med. Diagn. (2008) 2(10):1107-1121 with permission of Informa UK Ltd.
Figure 5
Figure 5
Radiographic features of spondylocostal dystostosis including contiguous vertebral malformations with asymmetric rib malformations. Photograph courtesy of Peter D. Turnpenny M.D., Royal Devon and Exeter Hospital.
Figure 6
Figure 6
Radiograph features of spondylothoracic dysostosis demonstrating contiguous vertebral malformations with symmetric posterior rib fusion. Reproduced with permission Expert Opinion in [9].

References

    1. Riseborough EJ, Wynne Davies R. A genetic survey of idiopathic scoliosis in Boston, Massachusetts. Journal of Bone and Joint Surgery—Series A. 1973;55(5):974–982.
    1. Shands AR, Eisberg HB. The incidence of scoliosis in the state of Delaware; a study of 50,000 minifilms of the chest made during a survey for tuberculosis. Journal of Bone and Joint Surgery— Series A. 1955;37(6):1243–1249.
    1. Rogala EJ, Drummond DS, Gurr J. Scoliosis: incidence and natural history. A prospective epidemiological study. Journal of Bone and Joint Surgery—Series A. 1978;60(2):173–176.
    1. Brand MC. Examination of the newborn with congenital scoliosis: focus on the physical. Advances in Neonatal Care. 2008;8(5):265–273.
    1. Davies BR, Durán M. Malformations of the cranium, vertebral column, and related central nervous system: morphologic heterogeneity may indicate biological diversity. Birth Defects Research Part A—Clinical and Molecular Teratology. 2003;67(8):563–571.
    1. McMaster MJ. Congenital deformities of the spine. Journal of the Royal College of Surgeons of Edinburgh. 2002;47(2):475–480.
    1. Giampietro PF, Dunwoodie SL, Kusumi K, et al. Molecular diagnosis of vertebral segmentation disorders in humans. Expert Opinion on Medical Diagnostics. 2008;2(10):1107–1121.
    1. Pourquié O. Vertebrate segmentation: from cyclic gene networks to scoliosis. Cell. 2011;145(5):650–663.
    1. Cooke J, Zeeman EC. A clock and wavefront model for control of the number of repeated structures during animal morphogenesis. Journal of Theoretical Biology. 1976;58(2):455–476.
    1. Bessho Y, Hirata H, Masamizu Y, Kageyama R. Periodic repression by the bHLH factor Hes7 is an essential mechanism for the somite segmentation clock. Genes and Development. 2003;17(12):1451–1456.
    1. Dunwoodie SL, Clements M, Sparrow DB, Sa X, Conlon RA, Beddington RSP. Axial skeletal defects caused by mutation in the spondylocostal dysplasia/pudgy gene Dll3 are associated with disruption of the segmentation clock within the presomitic mesoderm. Development. 2002;129(7):1795–1806.
    1. Holley SA, Geisler R, Nüsslein-Volhard C. Control of her1 expression during zebrafish somitogenesis by a Delta- dependent oscillator and an independent wave-front activity. Genes and Development. 2000;14(13):1678–1690.
    1. Jouve C, Palmeirim I, Henrique D, et al. Notch signalling is required for cyclic expression of the hairy-like gene HES1 in the presomitic mesoderm. Development. 2000;127(7):1421–1429.
    1. Dequéant ML, Glynn E, Gaudenz K, et al. A complex oscillating network of signaling genes underlies the mouse segmentation clock. Science. 2006;314(5805):1595–1598.
    1. Aulehla A, Wehrle C, Brand-Saberi B, et al. Wnt3a plays a major role in the segmentation clock controlling somitogenesis. Developmental Cell. 2003;4(3):395–406.
    1. Cole SE, Levorse JM, Tilghman SM, Vogt TF. Clock regulatory elements control cyclic expression of Lunatic fringe during somitogenesis. Developmental Cell. 2002;3(1):75–84.
    1. Sewell W, Sparrow DB, Smith AJ, et al. Cyclical expression of the Notch/Wnt regulator Nrarp requires modulation by Dll3 in somitogenesis. Developmental Biology. 2009;329(2):400–409.
    1. Dubrulle J, McGrew MJ, Pourquié O. FGF signaling controls somite boundary position and regulates segmentation clock control of spatiotemporal Hox gene activation. Cell. 2001;106(2):219–232.
    1. Sawada A, Shinya M, Jiang YJ, Kawakami A, Kuroiwa A, Takeda H. Fgf/MAPK signalling is a crucial positional cue in somite boundary formation. Development. 2001;128(23):4873–4880.
    1. Aulehla A, Pourquié O. Oscillating signaling pathways during embryonic development. Current Opinion in Cell Biology. 2008;20(6):632–637.
    1. Dunty WC, Jr., Biris KK, Chalamalasetty RB, Taketo MM, Lewandoski M, Yamaguchi TP. Wnt3a/β-catenin signaling controls posterior body development by coordinating mesoderm formation and segmentation. Development. 2008;135(1):85–94.
    1. Biris KK, Dunty WC, Jr., Yamaguchi TP. Mouse Ripply2 is downstream of Wnt3a and is dynamically expressed during somitogenesis. Developmental Dynamics. 2007;236(11):3167–3172.
    1. Moritomo Y, Koga O, Miyamoto H, Tsuda T. Congenital anophthalmia with caudal vertebral anomalies in Japanese Brown cattle. The Journal of Veterinary Medical Science. 1995;57(4):693–696.
    1. Chalamalasetty RB, Dunty WC, Jr., Biris KK, et al. The Wnt3a/beta-catenin target gene Mesogenin1 controls the segmentation clock by activating a Notch signalling program. Nature Communications. 2011;2(1, article 390)
    1. Vermot J, Pourquié O. Retinoic acid coordinates somitogenesis and left-right patterning in vertebrate embryos. Nature. 2005;435(7039):215–220.
    1. Ahn UM, Ahn NU, Nallamshetty L, et al. The etiology of adolescent idiopathic scoliosis. American Journal of Orthopedics. 2002;31(7):387–395.
    1. Tredwell SJ, Smith DF, Macleod PJ, Wood BJ. Cervical spine anomalies in fetal alcohol syndrome. Spine. 1982;7(4):331–334.
    1. Holmes LB, Harvey EA, Coull BA, et al. The teratogenicity of anticonvulsant drugs. The New England Journal of Medicine. 2001;344(15):1132–1138.
    1. Menegola E, Broccia ML, Nau H, Prati M, Ricolfi R, Giavini E. Teratogenic effects of sodium valproate in mice and rats at midgestation and at term. Teratogenesis Carcinogenesis and Mutagenesis. 1996;16(2):97–108.
    1. Vorhees CV. Teratogenicity and developmental toxicity of valproic acid in rats. Teratology. 1987;35(2):195–202.
    1. Breen JG, Claggett TW, Kimmel GL, Kimmel CA. Heat shock during rat embryo development in vitro results in decreased mitosis and abundant cell death. Reproductive Toxicology. 1999;13(1):31–39.
    1. Åberg A, Westbom L, Källén B. Congenital malformations among infants whose mothers had gestational diabetes or preexisting diabetes. Early Human Development. 2001;61(2):85–95.
    1. Martinez-Frias ML, Bermejo E, Rodriguez-Pinilla E, Prieto L, Frias JL. Epidemiological analysis of outcomes of pregnancy in gestational diabetic mothers. American Journal of Medical Genetics. 1998;78:140–145.
    1. Passarge E, Lenz W. Syndrome of caudal regression in infants of diabetic mothers: observations of further cases. Pediatrics. 1966;37(4):672–675.
    1. Alexander PG, Tuan RS. Role of environmental factors in axial skeletal dysmorphogenesis. Birth Defects Research Part C—Embryo Today. 2010;90(2):118–132.
    1. Allache R, De Marco P, Merello E, Capra V, Kibar Z. Role of the planar cell polarity gene CELSR1 in neural tube defects and caudal agenesis. Birth Defects Research Part A—Clinical and Molecular Teratology. 2012;94(3):176–181.
    1. Simons M, Mlodzik M. Planar cell polarity signaling: from fly development to human disease. Annual Review of Genetics. 2008;42:517–540.
    1. Hickory W, Nanda R, Catalanotto FA. Fetal skeletal malformations associated with moderate zinc deficiency during pregnancy. Journal of Nutrition. 1979;109(5):883–891.
    1. Sköld AC, Wellfelt K, Danielsson BR. Stage-specific skeletal and visceral defects of the IKr-blocker almokalant: further evidence for teratogenicity via a hypoxia-related mechanism. Teratology. 2001;64(6):292–300.
    1. Tian Y, Ishikawa H, Yamaguchi T, Yamauchi T, Yokoyama K. Teratogenicity and developmental toxicity of chlorpyrifos: maternal exposure during organogenesis in mice. Reproductive Toxicology. 2005;20(2):267–271.
    1. Bengtsson BE, Larsson A, Bengtsson A, Renberg L. Sublethal effects of tetrachloro-1,2-benzoquinone—a component in bleachery effluents from pulp mills—on vertebral quality and physiological parameters in fourhorn sculpin. Ecotoxicology and Environmental Safety. 1988;15(1):62–71.
    1. Farley FA, Loder RT, Nolan BT, et al. Mouse model for thoracic congenital scoliosis. Journal of Pediatric Orthopaedics. 2001;21(4):537–540.
    1. Wéry N, Narotsky MG, Pacico N, Kavlock RJ, Picard JJ, Gofflot F. Defects in cervical vertebrae in boric acid-exposed rat embryos are associated with anterior shifts of box gene expression domains. Birth Defects Research Part A—Clinical and Molecular Teratology. 2003;67(1):59–67.
    1. Owen MH, Ryan LM, Holmes LB. Effects of retinoic acid on Dominant hemimelia expression in mice. Birth Defects Research Part A—Clinical and Molecular Teratology. 2009;85(1):36–41.
    1. Alexander PG, Chau L, Tuan RS. Role of nitric oxide in chick embryonic organogenesis and dysmorphogenesis. Birth Defects Research Part A—Clinical and Molecular Teratology. 2007;79(8):581–594.
    1. Bnait KS, Seller MJ. Ultrastructural changes in 9-day old mouse embryos following maternal tobacco smoke inhalation. Experimental and Toxicologic Pathology. 1995;47(6):453–461.
    1. Fichtner RR, Sullivan KM, Zyrkowski CL, Trowbridge FL. Racial/ethnic differences in smoking, other risk factors, and low birth weight among low-income pregnant women, 1978–1988. MMWR. CDC Surveillance Summaries. 1990;39(3):13–21.
    1. Kaspiris A, Grivas TB, Weiss HR. Congenital scoliosis in monozygotic twins: case report and review of possible factors contributing to its development. Scoliosis. 2008;3(1, article 17)
    1. Corsello G, Piro E. The world of twins: an update. Journal of Maternal-Fetal and Neonatal Medicine. 2010;23(supplement 3):59–62.
    1. Niemitz EL, Feinberg AP. Epigenetics and assisted reproductive technology: a call for investigation. American Journal of Human Genetics. 2004;74(4):599–609.
    1. Bennett GD. Hyperthermia: malformations to chaperones. Birth Defects Research Part B. 2010;89(4):279–288.
    1. Holmes LB. Common Malformations. Oxford University Press; 2012. Vertebral anomalies: hemivertebrae; pp. 283–289.
    1. Aburakawa K, Harada M, Otake S. Clinical evaluations of the treatment of scoliosis. Trauma and Orthopaedic Surgery. 1996;39:55–62.
    1. Takikawa K, Haga N, Maruyama T, et al. Spine and rib abnormalities and stature in spondylocostal dysostosis. Spine. 2006;31(7):E192–E197.
    1. Klippel M, Feil A. Un cas d'absence des vertebres cervicales. Nouvelle Iconog. de la Salpêtrière. 1912;25:223–250.
    1. Thomsen MN, Schneider U, Weber M, Johannisson R, Niethard FU. Scoliosis and congenital anomalies associated with Klippel-Feil syndrome types I-III. Spine. 1997;22(4):396–401.
    1. Mortier GR, Lachman RS, Bocian M, Rimoin DL. Multiple vertebral segmentation defects: analysis of 26 new patients and review of the literature. American Journal of Medical Genetics. 1996;61(4):310–319.
    1. Offiah A, Alman B, Cornier AS, et al. Pilot assessment of a radiologic classification system for segmentation defects of the vertebrae. American Journal of Medical Genetics, Part A. 2010;152(6):1357–1371.
    1. Bulman MP, Kusumi K, Frayling TM, et al. Mutations in the human Delta homologue, DLL3, cause axial skeletal defects in spondylocostal dysostosis. Nature Genetics. 2000;24(4):438–441.
    1. Whittock NV, Sparrow DB, Wouters MA, et al. Mutated/MESP2 causes spondylocostal dysostosis in humans. American Journal of Human Genetics. 2004;74(6):1249–1254.
    1. Sparrow DB, Chapman G, Wouters MA, et al. Mutation of the LUNATIC FRINGE gene in humans causes spondylocostal dysostosis with a severe vertebral phenotype. American Journal of Human Genetics. 2006;78(1):28–37.
    1. Sparrow DB, Sillence D, Wouters MA, Turnpenny PD, Dunwoodie SL. Two novel missense mutations in HAIRY-AND-ENHANCER-OF-SPLIT-7 in a family with spondylocostal dysostosis. European Journal of Human Genetics. 2010;18(6):674–679.
    1. Turnpenny PD, Whittock N, Duncan J, Dunwoodie S, Kusumi K, Ellard S. Novel mutations in DLL3, a somitogenesis gene encoding a ligand for the Notch signalling pathway, cause a consistent pattern of abnormal vertebral segmentation in spondylocostal dysostosis. Journal of Medical Genetics. 2003;40(5):333–339.
    1. Moseley JE, Bonforte RJ. Spondylothoracic dysplasia—a syndrome of congenital anomalies. American Journal of Roentgenology, Radium Therapy, and Nuclear Medicine. 1969;106(1):166–169.
    1. Cornier AS, Staehling-Hampton K, Delventhal KM, et al. Mutations in the MESP2 gene cause spondylothoracic dysostosis/Jarcho-Levin syndrome. American Journal of Human Genetics. 2008;82(6):1334–1341.
    1. Ghebranious N, Blank RD, Raggio CL, et al. A missense T(Brachyury) mutation contributes to vertebral malformations. Journal of Bone and Mineral Research. 2008;23(10):1576–1583.
    1. Ghebranious N, Burmester JK, Glurich I, et al. Evaluation, of SLC35A3 as a candidate gene for human vertebral malformations. American Journal of Medical Genetics, Part A. 2006;140(12):1346–1348.
    1. Ghebranious N, Raggio CL, Blank RD, et al. Lack of evidence of WNT3A as a candidate gene for congenital vertebral malformations. Scoliosis. 2007;2(1, article 13)
    1. Giampietro PF, Raggio CL, Reynolds C, et al. DLL3 as a candidate gene for vertebral malformations. American Journal of Medical Genetics, Part A. 2006;140(22):2447–2453.
    1. Giampietro PF, Raggio CL, Reynolds CE, et al. An analysis of PAX1 in the development of vertebral malformations. Clinical Genetics. 2005;68(5):448–453.
    1. Papapetrou C, Drummond F, Reardon W, Winter R, Spitz L, Edwards YH. A genetic study of the human T gene and its exclusion as a major candidate gene for sacral agenesis with anorectal atresia. Journal of Medical Genetics. 1999;36(3):208–213.
    1. Fei Q, Wu Z, Wang H, et al. The association analysis of TBX6 polymorphism with susceptibility to congenital scoliosis in a Chinese han population. Spine. 2010;35(9):983–988.
    1. Sparrow DB, Chapman G, Smith AJ, et al. A mechanism for gene-environment interaction in the etiology of congenital scoliosis. Cell. 2012;149(2):295–306.
    1. Waterland RA, Dolinoy DC, Lin JR, Smith CA, Shi X, Tahiliani KG. Maternal methyl supplements increase offspring DNA methylation at Axin fused. Genesis. 2006;44(9):401–406.
    1. Gonzaga-Jauregui C, Lupski JR, Gibbs RA. Human genome sequencing in health and disease. Annual Review of Medicine. 2012;63:35–61.
    1. Giampietro PF, Raggio CL, Blank RD. Use of synteny conversion in identification of candidate genes for somitogenesis in humans. Open Journal of Orthopedics. 2012;2:62–68.
    1. Cohen MM, Rollnick BR, Kaye CI. Oculoauriculovertebral spectrum: an updated critique. Cleft Palate Journal. 1989;26(4):276–286.
    1. Cousley R, Naora H, Yokoyama M, Kimura M, Otani H. Validity of the Hfm transgenic mouse as a model for hemifacial microsomia. Cleft Palate-Craniofacial Journal. 2002;39(1):81–92.
    1. Wang R, Martínez-Frías ML, Graham JM., Jr. Infants of diabetic mothers are at increased risk for the oculo-auriculo-vertebral sequence: a case-based and case-control approach. Journal of Pediatrics. 2002;141(5):611–617.
    1. Lammer EJ, Chen DT, Hoar RM, et al. Retinoic acid embryopathy. The New England Journal of Medicine. 1985;313(14):837–841.
    1. Smithells RW, Leck I. The incidence of limb and ear defects since the withdrawal of thalidomide. The Lancet. 1963;281(7290):1095–1097.
    1. Rooryck C, Souakri N, Cailley D, et al. Array-CGH analysis of a cohort of 86 patients with oculoauriculovertebral spectrum. American Journal of Medical Genetics, Part A. 2010;152(8):1984–1989.
    1. Clarke RA, Singh S, McKenzie H, Kearsley JH, Yip MY. Familial Klippel-Feil syndrome and paracentric inversion inv(8)(q22.2q23.3) American Journal of Human Genetics. 1995;57(6):1364–1370.
    1. Wildervank L. The cerrvico-oculo-acusticus syndrome. In: Vinken P, Bruyn G, Myrianthopoulous N, editors. Congenital Malformations of the Spine and Spinal Cord Handbook of Clinical Neurology. New York, NY, USA: North Holland; 1978.
    1. Gardner WJ. Klippel-Feil syndrome, iniencephalus, anencephalus, hindbrain hernia and mirror movements. Overdistention of the neural tube. Child’s Brain. 1979;5(4):361–379.
    1. Gunderson CH, Solitare GB. Mirror movements in patients with the Klippel-Feil syndrome. Neuropathologic observations. Archives of Neurology. 1968;18(6):675–679.
    1. Rasmussen P. Persistent mirror movements: a clinical study of 17 children, adolescents and young adults. Developmental Medicine and Child Neurology. 1993;35(8):699–707.
    1. Royal SA, Tubbs RS, D’Antonio MG, Rauzzino MJ, Oakes WJ. Investigations into the association between cervicomedullary neuroschisis and mirror movements in patients with Klippel-Feil syndrome. American Journal of Neuroradiology. 2002;23(4):724–729.
    1. Högen T, Chan W-M, Riedel E, et al. Wildervanck's syndrome and mirror movements: a congenital disorder of axon migration? Journal of Neurology. 2012;259(4):761–763.
    1. Tassabehji M, Zhi MF, Hilton EN, et al. Mutations in GDF6 are associated with vertebral segmentation defects in Klippel-Feil syndrome. Human Mutation. 2008;29(8):1017–1027.
    1. Ye M, Berry-Wynne KM, Asai-Coakwell M, et al. Mutation of the bone morphogenetic protein GDF3 causes ocular and skeletal anomalies. Human Molecular Genetics. 2009;19(2):287–298.
    1. Weinstein S. The thoracolumbar spine. In: Weinstein S, Buckwalter J, editors. Turek's Orthopedics: Principles and Their Application. Philadelphia, Pa, USA: Lippincott Company; 1994. pp. 447–484.
    1. Nachemson AL, Peterson LE, Bradford DS, et al. Effectiveness of treatment with a brace in girls who have adolescent idiopathic scoliosis. A prospective, controlled study based on data from the Brace Study of the Scoliosis Research Society. Journal of Bone and Joint Surgery—Series A. 1995;77(6):815–822.
    1. Bradford DS, Tay BKB, Hu SS. Adult scoliosis: surgical indications, operative management, complications, and outcomes. Spine. 1999;24(24):2617–2629.
    1. Garland H. Hereditary scoliosis. British Medical Journal. 1934;1(3816, article 328)
    1. Enneking WF, Harrington P. Pathological changes in scoliosis. Journal of Bone and Joint Surgery—Series A. 1969;51(1):165–184.
    1. Miller NH. Cause and natural history of adolescent idiopathic scoliosis. Orthopedic Clinics of North America. 1999;30(3):343–352.
    1. Miller NH, Mims B, Child A, Milewicz DM, Sponseller P, Blanton SH. Genetic analysis of structural elastic fiber and collagen genes in familial adolescent idiopathic scoliosis. Journal of Orthopaedic Research. 1996;14(6):994–999.
    1. Robin GC, Cohen T. Familial scoliosis. A clinical report. Journal of Bone and Joint Surgery—Series B. 1975;57(2):146–147.
    1. Weinstein SL, Dolan LA, Cheng JC, Danielsson A, Morcuende JA. Adolescent idiopathic scoliosis. The Lancet. 2008;371(9623):1527–1537.
    1. Wise CA, Barnes R, Gillum J, Herring JA, Bowcock AM, Lovett M. Localization of susceptibility to familial idiopathic scoliosis. Spine. 2000;25(18):2372–2380.
    1. Chan V, Fong GCY, Luk KDK, et al. A genetic locus for adolescent idiopathic scoliosis linked to chromosome 19p13.3. American Journal of Human Genetics. 2002;71(2):401–406.
    1. Salehi LB, Mangino M, De Serio S, et al. Assignment of a locus for autosomal dominant idiopathic scoliosis (IS) to ohuman chromosome 17p11. Human Genetics. 2002;111(4-5):401–404.
    1. Justice CM, Miller NH, Marosy B, Zhang J, Wilson AF. Familial idiopathic scoliosis: evidence of an X-linked susceptibility locus. Spine. 2003;28(6):589–594.
    1. Morcuende JA, Minhas R, Dolan L, et al. Allelic variants of human melatonin 1A receptor in patients with familial adolescent idiopathic scoliosis. Spine. 2003;28(17):2025–2029.
    1. Bashiardes S, Veile R, Allen M, et al. SNTG1, the gene encoding γ1-syntrophin: a candidate gene for idiopathic scoliosis. Human Genetics. 2004;115(1):81–89.
    1. Miller NH, Justice CM, Marosy B, et al. Identification of candidate regions for familial idiopathic scoliosis. Spine. 2005;30(10):1181–1187.
    1. Alden KJ, Marosy B, Nzegwu N, Justice CM, Wilson AF, Miller NH. Idiopathic scoliosis: identification of candidate regions on chromosome 19p13. Spine. 2006;31(16):1815–1819.
    1. Gao X, Gordon D, Zhang D, et al. CHD7 gene polymorphisms are associated with susceptibility to idiopathic scoliosis. American Journal of Human Genetics. 2007;80(5):957–965.
    1. Ocaka L, Zhao C, Reed JA, et al. Assignment of two loci for autosomal dominant adolescent idiopathic scoliosis to chromosomes 9q31.2-q34.2 and 17q25.3-qtel. Journal of Medical Genetics. 2008;45(2):87–92.
    1. Raggio CL, Giampietro PF, Dobrin S, et al. A novel locus for adolescent idiopathic scoliosis on chromosome 12p. Journal of Orthopaedic Research. 2009;27(10):1366–1372.
    1. Gurnett CA, Alaee F, Bowcock A, et al. Genetic linkage localizes an adolescent idiopathic scoliosis and pectus excavatum gene to chromosome 18 q. Spine. 2009;34(2):E94–E100.
    1. Sharma S, Gao X, Londono D, et al. Genome-wide association studies of adolescent idiopathic scoliosis suggest candidate susceptibility genes. Human Molecular Genetics. 2011;20(7):1456–1466.
    1. Takahashi Y, Kou I, Takahashi A, et al. A genome-wide association study identifies common variants near LBX1 associated with adolescent idiopathic scoliosis. Nature Genetics. 2011;43(12):1237–1240.
    1. Webb JK. Reviewer’s comment. European Spine Journal. 1999;8(2):p. 117.
    1. Carr AJ, Ogilvie DJ, Wordsworth BP, Priestly LM, Smith R, Sykes B. Segregation of structural collagen genes in adolescent idiopathic scoliosis. Clinical Orthopaedics and Related Research. 1992;(274):305–310.
    1. Dubousset J, Queneau P, Thillard M. Experimental scoliosis induced by pineal gland and dienephalic lesions in young chickens: its relation with clinical findings. Orthopaedic Transactions. 1983;7, article 7
    1. Moreau A, Wang DS, Forget S, et al. Melatonin signaling dysfunction in adolescent idiopathic scoliosis. Spine. 2004;29(16):1772–1781.
    1. Wang R, Qiu Y, Rui B. Neurotrophin-3 mRNA expression in paravertebral muscles of patients with idiopathic scoliosis. Chinese Journal of Spine and Spinal Cord. 2007;15:532–534.
    1. Kindsfater K, Lowe T, Lawellin D, Weinstein D, Akmakjian J. Levels of platelet calmodulin for the prediction of progression and severity of adolescent idiopathic scoliosis. Journal of Bone and Joint Surgery—Series A. 1994;76(8):1186–1192.
    1. Floman Y, Liebergall M, Robin GC, Eldor A. Abnormalities of aggregation, thromboxane A2 synthesis, and 14C serotonin release in platelets of patients with idiopathic scoliosis. Spine. 1983;8(3):236–241.
    1. Inoue M, Minami S, Nakata Y, et al. Prediction of curve progression in idiopathic scoliosis from gene polymorphic analysis. Studies in Health Technology and Informatics. 2002;91:90–96.
    1. Lowe T, Lawellin D, Smith D, et al. Platelet calmodulin levels in adolescent idiopathic scoliosis: do the levels correlate with curve progression and severity? Spine. 2002;27(7):768–775.
    1. Ward K, Ogilvie JW, Singleton MV, Chettier R, Engler G, Nelson LM. Validation of DNA-based prognostic testing to predict spinal curve progression in adolescent idiopathic scoliosis. Spine. 2010;35(25):E1455–E1464.
    1. Jiang J, Qian B, Mao S, et al. A promoter polymorphism of tissue inhibitor of metalloproteinase-2 gene is associated with severity of thoracic adolescent idiopathic scoliosis. Spine. 2012;37(1):41–47.
    1. Qiu Y, Mao S-H, Qian B-P, et al. A promoter polymorphism of neurotrophin 3 gene is associated with curve severity and bracing effectiveness in adolescent idiopathic scoliosis. Spine. 2012;37(2):127–133.
    1. Tourtellotte WG, Milbrandt J. Sensory ataxia and muscle spindle agenesis in mice lacking the transcription factor Egr3. Nature Genetics. 1998;20(1):87–91.
    1. Purkiss SB, Driscoll B, Cole WG, Alman B. Idiopathic scoliosis in families of children with congenital scoliosis. Clinical Orthopaedics and Related Research. 2002;(401):27–31.
    1. Vissers LELM, Van Ravenswaaij CMA, Admiraal R, et al. Mutations in a new member of the chromodomain gene family cause CHARGE syndrome. Nature Genetics. 2004;36(9):955–957.
    1. Jacobs-Mcdaniels NL, Albertson RC. Chd7 plays a critical role in controlling left-right symmetry during zebrafish somitogenesis. Developmental Dynamics. 2011;240(10):2272–2280.
    1. Patten SA, Jacobs-McDaniels NL, Zaouter C, Drapeau P, Albertson RC, Moldovan F. Role of Chd7 in zebrafish: a model for CHARGE syndrome. PLoS ONE. 2012;7(2)e31650

Source: PubMed

Подписаться