Hostname: page-component-848d4c4894-x24gv Total loading time: 0 Render date: 2024-05-07T21:48:27.140Z Has data issue: false hasContentIssue false

Smith–Lemli–Opitz syndrome

Published online by Cambridge University Press:  22 July 2011

Andrea E. DeBarber
Affiliation:
Department of Physiology and Pharmacology, Oregon Health & Science University (OHSU), Portland, OR, USA
Yasemen Eroglu
Affiliation:
Pediatric Gastroenterology, Oregon Health & Science University (OHSU), Portland, OR, USA Department of Pediatrics, Oregon Health & Science University (OHSU), Portland, OR, USA
Louise S. Merkens
Affiliation:
Department of Pediatrics, Oregon Health & Science University (OHSU), Portland, OR, USA
Anuradha S. Pappu
Affiliation:
Department of Physiology and Pharmacology, Oregon Health & Science University (OHSU), Portland, OR, USA Department of Pediatrics, Oregon Health & Science University (OHSU), Portland, OR, USA
Robert D. Steiner*
Affiliation:
Department of Pediatrics, Oregon Health & Science University (OHSU), Portland, OR, USA Department of Molecular & Medical Genetics, Child Development and Rehabilitation Center, Doernbecher Children's Hospital, OHSU, Portland, OR, USA
*
*Corresponding author: Robert D. Steiner, Department of Pediatrics, OHSU, Portland, OR 97239, USA. E-mail: steinerr@ohsu.edu

Abstract

Smith–Lemli–Opitz syndrome (SLOS) is an autosomal recessive, multiple congenital malformation and intellectual disability syndrome, with clinical characteristics that encompass a wide spectrum and great variability. Elucidation of the biochemical and genetic basis for SLOS, specifically understanding SLOS as a cholesterol deficiency syndrome caused by mutation in DHCR7, opened up enormous possibilities for therapeutic intervention. When cholesterol was discovered to be the activator of sonic hedgehog, cholesterol deficiency with inactivation of this developmental patterning gene was thought to be the cause of SLOS malformations, yet this explanation is overly simplistic. Despite these important research breakthroughs, there is no proven treatment for SLOS. Better animal models are needed to allow potential treatment testing and the study of disease pathophysiology, which is incompletely understood. Creation of human cellular models, especially models of brain cells, would be useful, and in vivo human studies are also essential. Biomarker development will be crucial in facilitating clinical trials in this rare condition, because the clinical phenotype can change over many years. Additional research in these and other areas is critical if we are to make headway towards ameliorating the effects of this devastating condition.

Type
Review Article
Copyright
Copyright © Cambridge University Press 2011

Access options

Get access to the full version of this content by using one of the access options below. (Log in options will check for institutional or personal access. Content may require purchase if you do not have access.)

References

References

1Smith, D.W., Lemli, L. and Opitz, J.M. (1964) A newly recognized syndrome of multiple congenital anomalies. Journal of Pediatrics 64, 210-217CrossRefGoogle ScholarPubMed
2Opitz, J.M. et al. (1969) The RSH syndrome. Birth Defects 2, 43-52Google Scholar
3Dallaire, L. (1969) Syndrome of retardation with urogenital and skeletal anomalies (Smith-Lemli-Opitz Syndrome): clinical features and mode of inheritance. Journal of Medical Genetics 6, 113-120CrossRefGoogle ScholarPubMed
4Irons, M. et al. (1993) Defective cholesterol biosynthesis in Smith-Lemli-Opitz syndrome. Lancet 341, 1414CrossRefGoogle ScholarPubMed
5Kandutsch, A.A. and Russell, A.E. (1960) Preputial gland tumor sterols. 3 A metabolic pathway from lanosterol to cholesterol. Journal of Biological Chemistry 235, 2256-2261CrossRefGoogle ScholarPubMed
6Tint, G.S. et al. (1994) Defective cholesterol biosynthesis associated with the Smith-Lemli-Opitz syndrome. New England Journal of Medicine 330, 107-113CrossRefGoogle ScholarPubMed
7Shefer, S. et al. (1995) Markedly inhibited 7-dehydrocholesterol-delta 7-reductase activity in liver microsomes from Smith-Lemli-Opitz homozygotes. Journal of Clinical Investigation 96, 1779-1785CrossRefGoogle ScholarPubMed
8Moebius, F.F. et al. (1998) Molecular cloning and expression of the human delta7-sterol reductase. Proceedings of the National Academy of Sciences of the United States of America 95, 1899-1902CrossRefGoogle ScholarPubMed
9Fitzky, B.U. et al. (1998) Mutations in the Delta7-sterol reductase gene in patients with the Smith-Lemli-Opitz syndrome. Proceedings of the National Academy of Sciences of the United States of America 95, 8181-8186CrossRefGoogle ScholarPubMed
10Wassif, C.A. et al. (1998) Mutations in the human sterol delta7-reductase gene at 11q12–13 cause Smith-Lemli-Opitz syndrome. American Journal of Human Genetics 63, 55-62CrossRefGoogle ScholarPubMed
11Waterham, H.R. et al. (1998) Smith-Lemli-Opitz syndrome is caused by mutations in the 7-dehydrocholesterol reductase gene. American Journal of Human Genetics 63, 329-338CrossRefGoogle ScholarPubMed
12Wassif, C.A. et al. (2001) Biochemical, phenotypic and neurophysiological characterization of a genetic mouse model of RSH/Smith-Lemli–Opitz syndrome. Human Molecular Genetics 10, 555-564CrossRefGoogle ScholarPubMed
13Fitzky, B.U. et al. (2001) 7-Dehydrocholesterol-dependent proteolysis of HMG-CoA reductase suppresses sterol biosynthesis in a mouse model of Smith-Lemli-Opitz/RSH syndrome. Journal of Clinical Investigation 108, 905-915CrossRefGoogle Scholar
14Correa-Cerro, L.S. et al. (2006) Development and characterization of a hypomorphic Smith-Lemli-Opitz syndrome mouse model and efficacy of simvastatin therapy. Human Molecular Genetics 15, 839-851CrossRefGoogle ScholarPubMed
15Xu, G. et al. (1995) Reproducing abnormal cholesterol biosynthesis as seen in the Smith-Lemli-Opitz syndrome by inhibiting the conversion of 7-dehydrocholesterol to cholesterol in rats. Journal of Clinical Investigation 95, 76-81CrossRefGoogle ScholarPubMed
16Fliesler, S.J. et al. (2004) Retinal degeneration in a rodent model of Smith-Lemli-Opitz syndrome: electrophysiologic, biochemical, and morphologic features. Archives of Ophthalmology 122, 1190-1200CrossRefGoogle Scholar
17Cunniff, C. et al. (1997) Clinical and biochemical spectrum of patients with RSH/Smith-Lemli-Opitz syndrome and abnormal cholesterol metabolism. American Journal of Medical Genetics 68, 263-2693.0.CO;2-N>CrossRefGoogle ScholarPubMed
18Cooper, M.K. et al. (2003) A defective response to Hedgehog signaling in disorders of cholesterol biosynthesis. Nature Genetics 33, 508-513CrossRefGoogle ScholarPubMed
19Porter, F.D. and Herman, G.E. (2010) Malformation syndromes caused by disorders of cholesterol synthesis. Journal of Lipid Research 52, 6-34CrossRefGoogle ScholarPubMed
20Xu, L., Korade, Z. and Porter, N.A. (2010) Oxysterols from free radical chain oxidation of 7-dehydrocholesterol: product and mechanistic studies. Journal of American Chemical Society 132, 2222-2232CrossRefGoogle ScholarPubMed
21Korade, Z. et al. (2010) Biological activities of 7-dehydrocholesterol-derived oxysterols: implications for Smith-Lemli-Opitz syndrome. Journal of Lipid Research 51, 3259-3269CrossRefGoogle ScholarPubMed
22Chignell, C.F. et al. (2006) Ultraviolet A sensitivity in Smith-Lemli-Opitz syndrome: possible involvement of cholesta-5, 7, 9(11)-trien-3 beta-ol. Free Radical Biology and Medicine 41, 339-346CrossRefGoogle Scholar
23De, F.E. et al. (1996) Cholesta-5, 7, 9(11)-trien-3 beta-ol found in plasma of patients with Smith-Lemli-Opitz syndrome indicates formation of sterol hydroperoxide. Journal of Lipid Research 37, 2280-2287Google Scholar
24Ryan, A.K. et al. (1998) Smith-Lemli-Opitz syndrome: a variable clinical and biochemical phenotype. Journal of Medical Genetics 35, 558-565CrossRefGoogle ScholarPubMed
25Ginat, S. et al. (2000) Smith-Lemli-Opitz syndrome: a multiple malformation/mental retardation syndrome caused by defective cholesterol synthesis. Endocrinologist 10, 300-313CrossRefGoogle Scholar
26Porter, F.D. (2000) RSH/Smith-Lemli-Opitz syndrome: a multiple congenital anomaly/mental retardation syndrome due to an inborn error of cholesterol biosynthesis. Molecular Genetics and Metabolism 71, 163-174CrossRefGoogle Scholar
27Kelley, R.I. and Hennekam, R.C. (2000) The Smith-Lemli-Opitz syndrome. Journal of Medical Genetics 37, 321-335CrossRefGoogle ScholarPubMed
28Battaile, K.P. and Steiner, R.D. (2000) Smith-Lemli-Opitz syndrome: the first malformation syndrome associated with defective cholesterol synthesis. Molecular Genetics and Metabolism 71, 154-162CrossRefGoogle ScholarPubMed
29Yu, H. and Patel, S.B. (2005) Recent insights into the Smith-Lemli-Opitz syndrome. Clinical Genetics 68, 383-391CrossRefGoogle ScholarPubMed
30Porter, F.D. (2008) Smith-Lemli-Opitz syndrome: pathogenesis, diagnosis and management. European Journal of Human Genetics 16, 535-541CrossRefGoogle ScholarPubMed
31Phillipi, C.A. and Steiner, R.D. (2008) Disorders of cholesterol biosynthesis, genetics of. Encyclopedia of Life Sciences. [DOI: 10.1002/9780470015902.a0020223]Google Scholar
32Steiner, R.D., Martin, L.S. and Hume, R.F. (2007) ‘Smith Lemli Opitz syndrome’. eMedicine from WebMD. Updated. [http://www.emedicine.com/ped/topiclist.htm]Google Scholar
33Kelley, R.I. (1995) Diagnosis of Smith-Lemli-Opitz syndrome by gas chromatography/mass spectrometry of 7-dehydrocholesterol in plasma, amniotic fluid and cultured skin fibroblasts. Clinica Chimica Acta 236, 45-58CrossRefGoogle ScholarPubMed
34Kratz, L.E. and Kelley, R.I. (1999) Prenatal diagnosis of the RSH/Smith-Lemli-Opitz syndrome. American Journal of Medical Genetics 82, 376-3813.0.CO;2-B>CrossRefGoogle ScholarPubMed
35Tint, G.S. et al. (1998) Fetal Smith-Lemli-Opitz syndrome can be detected accurately and reliably by measuring amniotic fluid dehydrocholesterols. Prenatal Diagnostics 18, 651-6583.0.CO;2-V>CrossRefGoogle ScholarPubMed
36Nowaczyk, M.J. et al. (2000) Difficult prenatal diagnosis in mild Smith-Lemli-Opitz syndrome. American Journal of Medical Genetics 95, 396-3983.0.CO;2-L>CrossRefGoogle ScholarPubMed
37Linck, L.M. et al. (2000) Fetal demise with Smith-Lemli-Opitz syndrome confirmed by tissue sterol analysis and the absence of measurable 7-dehydrocholesterol Delta(7)-reductase activity in chorionic villi. Prenatal Diagnostics 20, 238-2403.0.CO;2-W>CrossRefGoogle ScholarPubMed
38Andersson, H.C. et al. (1999) Adrenal insufficiency in Smith-Lemli-Opitz syndrome. American Journal of Medical Genetics 82, 382-3843.0.CO;2-9>CrossRefGoogle ScholarPubMed
39Shackleton, C. et al. (2002) Identification of 7(8) and 8(9) unsaturated adrenal steroid metabolites produced by patients with 7-dehydrosterol-delta7-reductase deficiency (Smith-Lemli-Opitz syndrome). Journal of Steroid Biochemistry and Molecular Biology 82, 225-232CrossRefGoogle Scholar
40Pitt, J.J. (2007) High-throughput urine screening for Smith-Lemli-Opitz syndrome and cerebrotendinous xanthomatosis using negative electrospray tandem mass spectrometry. Clinica Chimica Acta 380, 81-88CrossRefGoogle ScholarPubMed
41Marcos, J. et al. (2004) The implications of 7-dehydrosterol-7-reductase deficiency (Smith-Lemli-Opitz syndrome) to neurosteroid production. Steroids 69, 51-60CrossRefGoogle ScholarPubMed
42Shackleton, C.H. et al. (2001) Dehydro-oestriol and dehydropregnanetriol are candidate analytes for prenatal diagnosis of Smith-Lemli-Opitz syndrome. Prenatal Diagnostics 21, 207-2123.0.CO;2-I>CrossRefGoogle ScholarPubMed
43Shackleton, C.H. et al. (2007) Dehydrosteroid measurements in maternal urine or serum for the prenatal diagnosis of Smith-Lemli-Opitz syndrome (SLOS). American Journal of Medical Genetics A 143A, 2129-2136CrossRefGoogle ScholarPubMed
44Matabosch, X. et al. (2009) Steroid production and excretion by the pregnant mouse, particularly in relation to pregnancies with fetuses deficient in Delta7-sterol reductase (Dhcr7), the enzyme associated with Smith-Lemli-Opitz syndrome. Journal of Steroid Biochemistry and Molecular Biology 116, 61-70CrossRefGoogle ScholarPubMed
45Irons, M. et al. (1994) Abnormal cholesterol metabolism in the Smith-Lemli-Opitz syndrome: report of clinical and biochemical findings in four patients and treatment in one patient. American Journal of Medical Genetics 50, 347-352CrossRefGoogle ScholarPubMed
46Steiner, R.D. et al. (2000) Sterol balance in the Smith-Lemli-Opitz syndrome. Reduction in whole body cholesterol synthesis and normal bile acid production. Journal of Lipid Research 41, 1437-1447CrossRefGoogle ScholarPubMed
47Natowicz, M.R. and Evans, J.E. (1994) Abnormal bile acids in the Smith-Lemli-Opitz syndrome. American Journal of Medical Genetics 50, 364-367CrossRefGoogle ScholarPubMed
48Batta, A.K. et al. (1994) Identification of 7-dehydrocholesterol and related sterols in patients with Smith-Lemli-Opitz syndrome. American Journal of Medical Genetics 50, 334Google Scholar
49Honda, A. et al. (1999) Bile acid synthesis in the Smith-Lemli-Opitz syndrome: effects of dehydrocholesterols on cholesterol 7alpha-hydroxylase and 27-hydroxylase activities in rat liver. Journal of Lipid Research 40, 1520-1528CrossRefGoogle ScholarPubMed
50Charman, C.R. et al. (1998) Photosensitivity associated with the Smith-Lemli-Opitz syndrome. British Journal of Dermatology 138, 885-888CrossRefGoogle ScholarPubMed
51Valencia, A. et al. (2006) 7-Dehydrocholesterol enhances ultraviolet A-induced oxidative stress in keratinocytes: roles of NADPH oxidase, mitochondria, and lipid rafts. Free Radical Biology and Medicine 41, 1704-1718CrossRefGoogle ScholarPubMed
52Vaughan, D.K. et al. (2006) Light-induced exacerbation of retinal degeneration in a rat model of Smith-Lemli-Opitz syndrome. Experimental Eye Research 82, 496-504CrossRefGoogle Scholar
53Wassif, C.A. et al. (2003) 27-Hydroxylation of 7- and 8-dehydrocholesterol in Smith-Lemli-Opitz syndrome: a novel metabolic pathway. Steroids 68, 497-502CrossRefGoogle ScholarPubMed
54Bjorkhem, I. et al. (2001) Oxysterols in the circulation of patients with the Smith-Lemli-Opitz syndrome: abnormal levels of 24S- and 27-hydroxycholesterol. Journal of Lipid Research 42, 366-371CrossRefGoogle ScholarPubMed
55Rossi, M. et al. (2005) Vitamin D status in patients affected by Smith-Lemli-Opitz syndrome. Journal of Inherited Metabolic Disease 28, 69-80CrossRefGoogle ScholarPubMed
56Zimmerman, P.A., Hercules, D.M. and Naylor, E.W. (1997) Direct analysis of filter paper blood specimens for identification of Smith-Lemli-Opitz syndrome using time-of-flight secondary ion mass spectrometry. American Journal of Medical Genetics 68, 300-3043.0.CO;2-X>CrossRefGoogle ScholarPubMed
57Griffiths, W.J. et al. (2008) Potential of sterol analysis by liquid chromatography-tandem mass spectrometry for the prenatal diagnosis of Smith-Lemli-Opitz syndrome. Clinical Chemistry 54, 1317-1324CrossRefGoogle ScholarPubMed
58Honda, A. et al. (2008) Highly sensitive analysis of sterol profiles in human serum by LC-ESI-MS/MS. Journal of Lipid Research 49, 2063-2073CrossRefGoogle ScholarPubMed
59Johnson, D.W., ten Brink, H.J. and Jakobs, C. (2001) A rapid screening procedure for cholesterol and dehydrocholesterol by electrospray ionization tandem mass spectrometry. Journal of Lipid Research 42, 1699-1705CrossRefGoogle ScholarPubMed
60Paglia, G. et al. (2010) Direct analysis of sterols from dried plasma/blood spots by an atmospheric pressure thermal desorption chemical ionization mass spectrometry (APTDCI-MS) method for a rapid screening of Smith-Lemli-Opitz syndrome. Analyst 135, 789-796CrossRefGoogle ScholarPubMed
61Nwokoro, N.A. and Mulvihill, J.J. (1997) Cholesterol and bile acid replacement therapy in children and adults with Smith-Lemli-Opitz (SLO/RSH) syndrome. American Journal of Medical Genetics 68, 315-3213.0.CO;2-W>CrossRefGoogle ScholarPubMed
62Tierney, E., Nwokoro, N.A. and Kelley, R.I. (2000) Behavioral phenotype of RSH/Smith-Lemli-Opitz syndrome. Mental Retardation and Developmental Disabilities Research Reviews 6, 131-1343.0.CO;2-R>CrossRefGoogle ScholarPubMed
63Mueller, C. et al. (2003) Normal cognition and behavior in a Smith-Lemli-Opitz syndrome patient who presented with Hirschsprung disease. American Journal of Medical Genetics A 123A, 100-106CrossRefGoogle Scholar
64Tierney, E. et al. (2001) Behavior phenotype in the RSH/Smith-Lemli-Opitz syndrome. American Journal of Medical Genetics 98, 191-2003.0.CO;2-M>CrossRefGoogle ScholarPubMed
65Kogan, M.D. et al. (2009) Prevalence of parent-reported diagnosis of autism spectrum disorder among children in the US 2007. Pediatrics 124, 1395-1403CrossRefGoogle ScholarPubMed
66Bukelis, I. et al. (2007) Smith-Lemli-Opitz syndrome and autism spectrum disorder. American Journal of Psychiatry 164, 1655-1661CrossRefGoogle ScholarPubMed
67Sikora, D.M. et al. (2006) The near universal presence of autism spectrum disorders in children with Smith-Lemli-Opitz syndrome. American Journal of Medical Genetics A 140, 1511-1518CrossRefGoogle ScholarPubMed
68Tierney, E. et al. (2006) Abnormalities of cholesterol metabolism in autism spectrum disorders. American Journal of Medical Genetics. Part B, Neuropsychiatric Genetics 141B, 666-668CrossRefGoogle ScholarPubMed
69Arndt, T.L., Stodgell, C.J. and Rodier, P.M. (2005) The teratology of autism. International Journal of Developmental Neuroscience 23, 189-199CrossRefGoogle ScholarPubMed
70Johnson, C.P. and Myers, S.M. (2007) Identification and evaluation of children with autism spectrum disorders. Pediatrics 120, 1183-1215CrossRefGoogle ScholarPubMed
71Bolanos, J.P., Medina, J.M. and Williamson, D.H. (1990) Inhibition of sterol but not fatty acid synthesis by valproate in developing rat brain in vivo. Biochemical Journal 272, 251-253CrossRefGoogle Scholar
72Kuwagata, M. et al. (2009) Observation of fetal brain in a rat valproate-induced autism model: a developmental neurotoxicity study. International Journal of Developmental Neuroscience 27, 399-405CrossRefGoogle Scholar
73Chevy, F. et al. (2002) Limb malformations of rat fetuses exposed to a distal inhibitor of cholesterol biosynthesis. Journal of Lipid Research 43, 1192-1200CrossRefGoogle ScholarPubMed
74Linck, L.M. et al. (2000) Cholesterol supplementation with egg yolk increases plasma cholesterol and decreases plasma 7-dehydrocholesterol in Smith-Lemli-Opitz syndrome. American Journal of Medical Genetics 93, 360-3653.0.CO;2-P>CrossRefGoogle ScholarPubMed
75Elias, E.R. et al. (1997) Clinical effects of cholesterol supplementation in six patients with the Smith-Lemli-Opitz syndrome (SLOS). American Journal of Medical Genetics 68, 305-3103.0.CO;2-X>CrossRefGoogle ScholarPubMed
76Martin, A. et al. (2001) Smith-Lemli-Opitz syndrome. Journal of the American Academy of Child and Adolescent Psychiatry 40, 506-507CrossRefGoogle ScholarPubMed
77Sikora, D.M. et al. (2004) Cholesterol supplementation does not improve developmental progress in Smith-Lemli-Opitz syndrome. Journal of Pediatrics 144, 783-791Google Scholar
78Tierney, E. et al. (2010) Analysis of short-term behavioral effects of dietary cholesterol supplementation in Smith-Lemli-Opitz syndrome. American Journal of Medical Genetics A 152A, 91-95CrossRefGoogle ScholarPubMed
79Azurdia, R.M., Anstey, A.V. and Rhodes, L.E. (2001) Cholesterol supplementation objectively reduces photosensitivity in the Smith-Lemli-Opitz syndrome. British Journal of Dermatology 144, 143-145CrossRefGoogle ScholarPubMed
80Starck, L., Lovgren-Sandblom, A. and Bjorkhem, I. (2002) Cholesterol treatment forever? The first Scandinavian trial of cholesterol supplementation in the cholesterol-synthesis defect Smith-Lemli-Opitz syndrome. Journal of Internal Medicine 252, 314-321CrossRefGoogle ScholarPubMed
81Jira, P.E. et al. (2000) Simvastatin. A new therapeutic approach for Smith-Lemli-Opitz syndrome. Journal of Lipid Research 41, 1339-1346CrossRefGoogle ScholarPubMed
82Starck, L., Lovgren-Sandblom, A. and Bjorkhem, I. (2002) Simvastatin treatment in the SLO syndrome: a safe approach? American Journal of Medical Genetics 113, 183-189CrossRefGoogle ScholarPubMed
83Haas, D. et al. (2007) Effects of cholesterol and simvastatin treatment in patients with Smith-Lemli-Opitz syndrome (SLOS). Journal of Inherited Metabolic Disease 30, 375-387CrossRefGoogle ScholarPubMed
84Saheki, A. et al. (1994) In vivo and in vitro blood-brain barrier transport of 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase inhibitors. Pharmaceutical Research 11, 305-311CrossRefGoogle Scholar
85Chan, Y.M. et al. (2009) Effects of dietary cholesterol and simvastatin on cholesterol synthesis in Smith-Lemli-Opitz syndrome. Pediatric Research 65, 681-685CrossRefGoogle ScholarPubMed
86Wassif, C.A. et al. (2005) Residual cholesterol synthesis and simvastatin induction of cholesterol synthesis in Smith-Lemli-Opitz syndrome fibroblasts. Molecular Genetics and Metabolism 85, 96-107CrossRefGoogle ScholarPubMed
87Vega, G.L. et al. (2003) Reduction in levels of 24S-hydroxycholesterol by statin treatment in patients with Alzheimer disease. Archives of Neurology 60, 510–505CrossRefGoogle ScholarPubMed
88Matabosch, X. et al. (2010) Increasing cholesterol synthesis in 7-dehydrosterol reductase (DHCR7) deficient mouse models through gene transfer. Journal of Steroid Biochemistry and Molecular Biology 122, 303-309CrossRefGoogle ScholarPubMed
89Lindegaard, M.L. et al. (2008) Characterization of placental cholesterol transport: ABCA1 is a potential target for in utero therapy of Smith-Lemli-Opitz syndrome. Human Molecular Genetics 17, 3806-3813CrossRefGoogle ScholarPubMed
90Irons, M.B. et al. (1999) Antenatal therapy of Smith-Lemli-Opitz syndrome. Fetal Diagnosis and Therapy 14, 133-137CrossRefGoogle ScholarPubMed
91Correa-Cerro, L.S. et al. (2005) DHCR7 nonsense mutations and characterisation of mRNA nonsense mediated decay in Smith-Lemli-Opitz syndrome. Journal of Medical Genetics 42, 350-357CrossRefGoogle ScholarPubMed
92Kim, J.H., Lee, J.N. and Paik, Y.K. (2001) Cholesterol biosynthesis from lanosterol. A concerted role for Sp1 and NF-Y-binding sites for sterol-mediated regulation of rat 7-dehydrocholesterol reductase gene expression. Journal of Biological Chemistry 276, 18153-18160CrossRefGoogle ScholarPubMed
93Lee, J.N., Bae, S.H. and Paik, Y.K. (2002) Structure and alternative splicing of the rat 7-dehydrocholesterol reductase gene. Biochimica et Biophysica Acta 1576, 148-156CrossRefGoogle ScholarPubMed
94Nishino, H. and Ishibash, T. (2000) Evidence for requirement of NADPH-cytochome P450 oxidaoreductase in the microsomal NADPH-sterol Δ7-reductase system. Archives of Biochemistry and Biophysics 374, 293-298CrossRefGoogle Scholar
95Bae, S.H. et al. (1999) Cholesterol biosynthesis from lanosterol. Molecular cloning, tissue distribution, expression, chromosomal localization, and regulation of rat 7-dehydrocholesterol reductase, a Smith-Lemli-Opitz syndrome-related protein. Journal of Biological Chemistry 274, 14624-14631CrossRefGoogle Scholar
96Waterham, H.R. and Wanders, R.J. (2000) Biochemical and genetic aspects of 7-dehydrocholesterol reductase and Smith-Lemli-Opitz syndrome. Biochimica et Biophysica Acta 1529, 340-356CrossRefGoogle ScholarPubMed
97Honda, M. et al. (1998) 7-Dehydrocholesterol down-regulates cholesterol biosynthesis in cultured Smith-Lemli-Opitz syndrome skin fibroblasts. Journal of Lipid Research 39, 647-657CrossRefGoogle ScholarPubMed
98Honda, A. et al. (1995) Defective conversion of 7-dehydrocholesterol to cholesterol in cultured skin fibroblasts from Smith-Lemli-Opitz syndrome homozygotes. Journal of Lipid Research 36, 1595-1601CrossRefGoogle ScholarPubMed
99Ginat, S. et al. (2004) Lowered DHCR7 activity measured by ergosterol conversion in multiple cell types in Smith-Lemli-Opitz syndrome. Molecular Genetics and Metabolism 83, 175-183CrossRefGoogle ScholarPubMed
100Witsch-Baumgartner, M. et al. (2004) Maternal apo E genotype is a modifier of the Smith-Lemli-Opitz syndrome. Journal of Medical Genetics 41, 577-584CrossRefGoogle ScholarPubMed
101Jenkins, K.T. et al. (2008) Enhanced placental cholesterol efflux by fetal HDL in Smith-Lemli-Opitz syndrome. Molecular Genetics and Metabolism 94, 240-247CrossRefGoogle ScholarPubMed
102Jira, P.E. et al. (2001) Novel mutations in the 7-dehydrocholesterol reductase gene of 13 patients with Smith–Lemli–Opitz syndrome. Annals of Hum Genetics 65, 229-236CrossRefGoogle ScholarPubMed
103Battaile, K.P. et al. (2001) Carrier frequency of the common mutation IVS8–1G > C in DHCR7 and estimate of the expected incidence of Smith-Lemli-Opitz syndrome. Molecular Genetics and Metabolism 72, 67-71CrossRefGoogle ScholarPubMed
104Nowaczyk, M.J. et al. (2004) Incidence of Smith-Lemli-Opitz syndrome in Canada: results of three-year population surveillance. Journal of Pediatrics 145, 530-535CrossRefGoogle ScholarPubMed
105Waye, J.S. et al. (2002) Smith-Lemli-Opitz syndrome: carrier frequency and spectrum of DHCR7 mutations in Canada. Journal of Medical Genetics 39, E31CrossRefGoogle ScholarPubMed
106Witsch-Baumgartner, M. et al. (2000) Mutational spectrum in the Delta7-sterol reductase gene and genotype-phenotype correlation in 84 patients with Smith-Lemli-Opitz syndrome. American Journal of Human Genetics 66, 402-412CrossRefGoogle Scholar
107Ciara, E. et al. (2004) DHCR7 mutations and genotype-phenotype correlation in 37 Polish patients with Smith-Lemli-Opitz syndrome. Clinical Genetics 66, 517-524CrossRefGoogle ScholarPubMed
108Goluszko, P. and Nowicki, B. (2005) Membrane cholesterol: a crucial molecule affecting interactions of microbial pathogens with mammalian cells. Infection and Immunity 73, 7791-7796CrossRefGoogle ScholarPubMed
109Langius, F.A. et al. (2003) Identification of three patients with a very mild form of Smith-Lemli-Opitz syndrome. American Journal of Medical Genetics A 122A, 24-29CrossRefGoogle ScholarPubMed
110Waye, J.S., Eng, B. and Nowaczyk, M.J. (2007) Prenatal diagnosis of Smith-Lemli-Opitz syndrome (SLOS) by DHCR7 mutation analysis. Prenatal Diagnostics 27, 638-640CrossRefGoogle ScholarPubMed
111Jezela-Stanek, A. et al. (2006) Maternal urinary steroid profiles in prenatal diagnosis of Smith-Lemli-Opitz syndrome: first patient series comparing biochemical and molecular studies. Clinical Genetics 69, 77-85CrossRefGoogle ScholarPubMed
112Liss, J. et al. (2008) Pregnancy and life after preimplantation genetic diagnosis of Smith-Lemli-Opitz syndrome. Fertility and Sterility 90, 2011-2016CrossRefGoogle ScholarPubMed
113Roux, C., Horvath, C. and Dupuis, R. (1979) Teratogenic action and embryo lethality of AY9944R prevention by a hypercholesterolemia provoking diet. Teratology 19, 35-38CrossRefGoogle ScholarPubMed
114Roux, C. et al. (1980) Teratogenic effect of an inhibitor of cholesterol synthesis (AY 9944) in rats: correlation with maternal cholesterolemia. Journal of Nutrition 110, 2310-2312CrossRefGoogle ScholarPubMed
115Barbu, V. et al. (1988) Cholesterol prevents the teratogenic action of AY 9944: importance of the timing of cholesterol supplementation to rats. Journal of Nutrition 118, 774-779CrossRefGoogle ScholarPubMed
116Wolf, C. et al. (1996) Changes in serum sterols of rats treated with 7-dehydrocholesterol-delta 7-reductase inhibitors: comparison to levels in humans with Smith-Lemli-Opitz syndrome. Journal of Lipid Research 37, 1325-1333CrossRefGoogle ScholarPubMed
117Gaoua, W. et al. (1999) Oxidized derivatives of 7-dehydrocholesterol induce growth retardation in cultured rat embryos: a model for antenatal growth retardation in the Smith-Lemli-Opitz syndrome. Journal of Lipid Research 40, 456-463CrossRefGoogle Scholar
118Roux, C. et al. (2000) Role of cholesterol in embryonic development. American Journal of Clinical Nutrition 71, 1270S-1279SCrossRefGoogle ScholarPubMed
119Gaoua, W. et al. (2000) Cholesterol deficit but not accumulation of aberrant sterols is the major cause of the teratogenic activity in the Smith-Lemli-Opitz syndrome animal model. Journal of Lipid Research 41, 637-646CrossRefGoogle Scholar
120Krakowiak, P.A. et al. (2003) Lathosterolosis: an inborn error of human and murine cholesterol synthesis due to lathosterol 5-desaturase deficiency. Human Molecular Genetics 12, 1631-1641CrossRefGoogle ScholarPubMed
121Marcos, J. et al. (2007) Cholesterol biosynthesis from birth to adulthood in a mouse model for 7-dehydrosterol reductase deficiency (Smith-Lemli-Opitz syndrome). Steroids 72, 802-808CrossRefGoogle Scholar
122Patti, G.J. et al. (2010) Nanostructure-initiator mass spectrometry (NIMS) imaging of brain cholesterol metabolites in Smith-Lemli-Opitz syndrome. Neuroscience 170, 858-864CrossRefGoogle ScholarPubMed
123Yu, H. et al. (2007) Selective reconstitution of liver cholesterol biosynthesis promotes lung maturation but does not prevent neonatal lethality in Dhcr7 null mice. BMC Developmental Biology 7, 27CrossRefGoogle Scholar
124Yu, H. et al. (2005) Partial rescue of neonatal lethality of Dhcr7 null mice by a nestin promoter-driven DHCR7 transgene expression. Brain Research. BMC Developmental Biology 156, 46-60CrossRefGoogle ScholarPubMed
125Jiang, X.S. et al. (2010) Quantitative proteomics analysis of inborn errors of cholesterol synthesis: identification of altered metabolic pathways in DHCR7 and SC5D deficiency. Molecular and Cellular Proteomics 9, 1461-1475CrossRefGoogle ScholarPubMed
126Ford, D.A. et al. (2008) Lipidomic analysis of the retina in a rat model of Smith-Lemli-Opitz syndrome: alterations in docosahexaenoic acid content of phospholipid molecular species. Journal of Neurochemistry 105, 1032-1047CrossRefGoogle Scholar
127Boesze-Battaglia, K. et al. (2008) Alteration of retinal rod outer segment membrane fluidity in a rat model of Smith-Lemli-Opitz syndrome. Journal of Lipid Research 49, 1488-1499CrossRefGoogle Scholar
128Caruso, P.A. et al. (2004) MRI and 1H MRS findings in Smith-Lemli-Opitz syndrome. Neuroradiology 46, 3-14CrossRefGoogle ScholarPubMed
129Kroll, R.A. and Neuwelt, E.A. (1998) Outwitting the blood-brain barrier for therapeutic purposes: osmotic opening and other means. Neurosurgery 42, 1083-1099CrossRefGoogle ScholarPubMed
130Cartier, N. et al. (2009) Hematopoietic stem cell gene therapy with a lentiviral vector in X-linked adrenoleukodystrophy. Science 326, 818-823CrossRefGoogle ScholarPubMed
131Woollett, L.A. (2008) Where does fetal and embryonic cholesterol originate and what does it do? Annual Review of Nutrition 28, 97-114CrossRefGoogle Scholar
132Ullrich, K. et al. (1996) Smith-Lemli-Opitz syndrome: treatment with cholesterol and bile acids. Neuropediatrics 27, 111-112CrossRefGoogle ScholarPubMed
133Rodrigues, C.M. et al. (2002) Neuroprotection by a bile acid in an acute stroke model in the rat. Journal of Cerebral Blood Flow and Metabolism 22, 463-471CrossRefGoogle Scholar

Further reading, resources and contacts

There are several websites of interest for families of patients and researchers of SLOS.

The Smith-Lemli-Opitz/RSH Foundation website: