Open access

Down Syndrome: Clinical and Genetic Aspects, Genetic Counseling and Prenatal Screening and Diagnosis

Written By

Érika Cristina Pavarino, Joice Matos Biselli, Walter Pinto Junior and Eny Maria Goloni Bertollo

Submitted: 24 April 2012 Published: 06 March 2013

DOI: 10.5772/52950

From the Edited Volume

Down Syndrome

Edited by Subrata Kumar Dey

Chapter metrics overview

3,284 Chapter Downloads

View Full Metrics

1. Introduction

1.1. Clinical and genetic aspects

Down syndrome (DS) or trisomy 21 is the most common genetic disorder with a prevalence of 1 in 660 live births [1]. In 1959, Lejeune and colleagues discovered the genetic basis of DS and named as trisomy of chromosome 21, which is the smallest human autosomal chromosome [2]. Trisomy 21 can occur as three types of chromosomal abnormalities: free trisomy 21, translocation or mosaicism. Free trisomy 21 is characterized by the presence of three complete copies of chromosome 21, occurring in about 90-95% of DS cases [3-5]. More than 90% of the cases of chromosomal nondisjunction are of maternal origin, mainly during meiosis I, about 5% involve an additional paternal extra chromosome and a small proportion (2%) is consequence of post-zygotic mitotic non-disjunction [6]. Translocations are attributed to 1-7% of the cases, with Robertsonian translocation involving chromosomes 14 and 21 being the most common type. Mosaicism, characterized by some cells containing 46 chromosomes and others with 47 chromosomes (with an extra chromosome 1), is reported in 1-7% of DS cases [3-5].

DS phenotype is complex and varies among individuals, who may present a combination of dysmorphic features and developmental delay [7]. The intellectual disability is a characteristic observed in all cases and the most frequent clinical features include muscular hypotonia (99%), diastasis of the muscle rectus of abdomen (90%), upslanted palpebral fissures (90%), microcephaly (85%), flat occipital (80%), joint hyperextension (80%), broad hands with short fingers (70%), short stature (60%), clinodactyly of fifth finger (50%), epicanthal fold (40%), low-set ears (50%), single palmar crease (40%), atlantoaxial instability (15%) and label-femoral instability (10%) [8]. On average, 50-70% of children with DS have congenital heart defects, such as ventricular septal defect, atrial septal defect, tetralogy of Fallot, patent ductus arteriosus and atrioventricular septal defect [3,4,9]. There are also ocular problems, such as refractive errors, nystagmus, abnormalities of the retina, among others [10]. About 80% of cases present hearing loss, which can be conductive, sensorineural, or mixed [11]. Thyroid dysfunction, particularly hypothyroidism [9], periodontal diseases [10], upper airway obstruction [12] and hypogonadism [14] are more frequent in individuals with DS than in the general population. Other important clinical aspects of DS include immunodeficiency [15], increased risk for hematological disorders and leukemia [16] and early onset of Alzheimer's disease [17].

The development of secondary sexual characteristics in DS is similar to other adolescents. The fetal oogenesis of women with the syndrome appears to be normal and, therefore, they are capable of reproduction [18]. On the other hand, men have diminished reproductive capacity, showing testicular histology compatible with oligospermia and, frequently, hypogonadism [19]. However, there have been reports of men with Down syndrome who have fathered pregnancies [20].

Advertisement

2. Genetic counseling

Genetic counseling can be defined as a communication process that takes care of the human problems associated with the occurrence or recurrence of a genetic disease in a family with the purpose of providing individuals and families comprehensive understanding of all the implications related to genetic disease under discussion, the options that the current medicine offers for therapy or for reducing the risk of occurrence or recurrence of the disease and psychotherapeutic support [21,22].

For DS, a well-established risk factor is advanced maternal age at conception [23,24]. The estimated risk for fetal trisomy 21 for a woman aged 20 years at 12 weeks of gestation is about 1 in 1000, and the risk of such woman delivering an affected baby at term is 1 in 1500. The risk for this aneuploidy for a woman aged 35 years at 12 weeks of gestation is about 1 in 250 and the risk of delivering an affected baby at term is 1 in 350 [25].

Although there is considerable variation in the physical features of individuals with DS, most individuals present with a range of characteristics that enable clinical diagnosis of the syndrome [3,4,7]. However, cytogenetic investigation of individuals who present with clinical characteristics of DS is fundamental to establish a precise diagnosis, which may have implications in the genetic counseling process, once it is very important in determining the recurrence risk of the syndrome. In addition, the karyotype analysis of affected individuals identifies cases that may have been inherited making necessary the investigation of the parents' karyotypes. In this case, the cytogenetic investigation of the genitors is essential to establish the risk of recurrence of the syndrome in future generations. Thus, all individuals with a diagnosis suggestive of DS should be referred to a genetic counseling service.

Accurate estimation of recurrence risks depends upon the verification of the individual’s karyotype. Cases of free trisomy 21 and mosaicism generally do not recur in siblings of individuals with DS. For women with maternal age <35 at previous trisomy 21, the revised risk is the age-related risk times 3.5. For those with maternal age ≥35 at previous trisomy 21, the revised risk is the age-related risk times 1.7 [26]. So, these risk times implies that other factors might influence the risk for DS in young mothers [27]. On the other hand, translocation may be recurrent. If neither parent carries a balanced translocation, the DS recurrence risk is low, probably similar to that of free trisomy 21. However, if one of the parents is the carrier of a balanced translocation, the risk of recurrence is dependent on the type of translocation and the sex of the carrier parent. In the case of Robertsonian translocations involving chromosome 13, 14, 15 or 22 and the chromosome 21, the recurrence risk at time of amniocentesis is of up to 17% when the mother is the carrier and of up to 1.4% when the carrier of this balanced translocation is the father. On the other hand, if one of the parents is the carrier of a balanced translocation involving two chromosomes 21, the recurrence risk of DS is 100% [26]. Thus, once diagnosed as a case of DS due to a translocation, a karyotype analysis of both parents is recommended.

For an individual with DS, the theoretical chance to have a child with DS is 50%, and 66% when both partners have DS. However, empiric risks are difficult to estimate, once the reproduction rates are low. Empiric data indicate a 30–50% chance of a woman with DS have a child with DS [26]. However, considering that the rate of fetal death between 11 weeks and term is about 43% for trisomy 21 [28], the chance of birth of a child with DS decreases. For individuals with mosaicism, the maximum theoretical recurrence risk is as high as 50%, but is dependent upon the proportion of trisomic gonadal cells and whether the other partner has DS as well [26].

Genetic counseling is also important to guide the parents about caring for the child with DS. Because individuals with DS often experience delays in reaching various developmental milestones, early intervention with speech therapy, occupational therapy, and physical therapy is recommended as it maximizes long-term outcomes [29]. As healthcare has improved for individuals with DS, the average life expectancy has increased by more than 30 years, from an average of 25 years of age in 1983 to almost 60 years of age in 2000 [30]. A study performed between 1985–2004 in England showed that the one-year survival of live births with DS increased, especially in babies with cardiovascular malformations, reaching almost 100% [31], and a more recent study showed that the 25-year survival of DS individuals is about 87.5% [32].

Genetic counselors should balance the negative aspects of DS, such as birth defects, medical complications, and developmental delay, with positive aspects like available treatments, therapies, and the ability for people with DS and their families to enjoy a high quality of life [33].

Advertisement

3. Prenatal screening and diagnosis

There are several methods that allow the early detection of DS in prenatal phase. At this point, it is not possible avoid congenital malformations or genetic diseases, but the objective is its early detection, looking for emotional and psychological preparation for parents and family and adequate medical support and monitoring for the child’s birth. Furthermore, early detection allows treatment of malformations of the complications that may occur, preventing or attenuating their evolution through surgical correction in utero.

There are some methods used to screen fetus with DS that allow the prenatal diagnosis of the syndrome. Among the screening methods are the nuchal translucency test, the measurement of maternal serum concentrations of various fetoplacental products and fetal ultrasound. The nuchal translucency (NT) test is the measurement of the fluid filled fold at the back of the fetal neck in the first trimester of pregnancy, performed through transabdominal or transvaginal sonography. The test is performed between the 11th and 13th weeks of gestation and the minimum fetal crown–rump length (CRL) should be 45 mm and the maximum 84 mm. Fetal NT increases with CRL and therefore it is essential to take gestation into account when determining whether a given NT thickness is increased [25]. The excess skin in the fetus may be the consequence of excessive accumulation of subcutaneous fluid behind the fetal neck which could be visualized by ultrasonography as increased NT in the third month of intrauterine life [34]. Nowadays, it is well established that the measurement of fetal NT thickness provides effective and early screening for trisomy 21 and other major aneuploidies, such as Edwards syndrome (trisomy 18) and Patau syndrome (trisomy 13) [34-36] besides for screening of congenital heart disease [37]. In case of abnormality in NT measurement, additional tests are needed to elucidate the cause of increased nuchal fold.

Pregnancies with fetal aneuploidies are associated with altered maternal serum concentrations of various fetoplacental products, including alpha-fetoprotein (AFP), free chorionic gonadotropin (β-hCG), unconjugated estriol (uE3), inhibin A (INH-A) and pregnancy associated plasma protein-A (PAPP-A) [38-42]. The measurement of concentrations of maternal serum AFP, β-hCG and uE3, the triple test, is one of a range of screening tests that are used to identify pregnant women whose fetus is likely to be affected by trisomy 21 and who should then be offered a diagnostic test. AFP is produced in the yolk sac and fetal liver, while uE3 and hCG are produced by the placenta. Elevated β-hCG concentration and low levels of AFP and uE3 suggests the presence of a fetus with DS [38-40]. The test is performed in second trimester of pregnancy and the values should be adjusted to gestational age. The expected detection rate and false-positive rate are about 73 - 78% and 7.5 - 9%, respectively [43].

The incorporation of INH-A into maternal serum DS screening in the second trimester, along with AFP, hCG and uE3, is named quadruple test. INH-A is a glycoprotein mainly secreted from the corpus luteum and the placenta [44] and its concentration is raised in the serum of pregnant women carrying a fetus with DS [42]. The quadruple test presents expected detection rate and false-positive rate about 79 - 82% and 6.5 - 7.8%, respectively [43]. The measurement of PAPP-A is also used as a screening gestations of fetus with DS in the first trimester, once the maternal serum concentration of this protein are reduced in these women [41]. The measurement of PAPP-A at 10–14 weeks of pregnancy is used to screen for fetal DS during the first trimester of pregnancy [45,47].

The fetal ultrasound is also considered a method of screening for DS, once any change in the development of organs or structures is easily visualized. The objective is the detection of major and soft markers of aneuploidy, including alterations in central nervous system, face, neck, heart, gastrointestinal tract, genitourinary tract among others [47]. Besides increased nuchal translucency in the first trimester, alterations commonly detected in DS in the second trimester of gestation include lack of visualization of the nasal bone [48], reduced femur and humerus, mild pyelectasis, hyperechoic bowel and echogenic intracardiac focus [47,49].

Importantly, any suspect result of the markers mentioned implies the genetic analysis of the fetus, the only way to accurate diagnosis. The methods for obtaining fetal cells for analysis vary with gestational age. Among the invasive methods for obtaining fetal cells, chorionic villus sampling (CVS) allows diagnosis in the first trimester of pregnancy (between the 10th and 13th weeks of gestation) [50]. The procedure involves aspiration of trophoblastic tissue under continuous ultrasound monitoring, performed via trans-cervical or trans-abdominal. Studies have showed that the risk miscarriage associated to this procedure is about 0.6-1.1% [51,52] and the procedure is not recommended for pregnant women that present bleeding due to an increase in the procedure-related fetal loss rate [51].

The amniocentesis is the method indicated for obtaining fetal cells after 15 weeks of gestation [53]. This requires taking a small sample of amniotic fluid transabdominally under ultrasound guidance. The procedure-related fetal loss rate is about 0.4-0.8 % [51,52]. After 20th week of gestation, the option is percutaneous umbilical blood sampling or cordocentesis, which involves direct sampling of fetal blood from the umbilical cord. The procedure-related loss rate is about 1.0-1.5% and cordocentesis with placenta penetration had a significantly higher rate of fetal loss [54-56].

Considering the risks which accompany invasive methods for obtaining fetal cells [51,52,56], the use of noninvasive methods could be a good option. Several methods to develop a noninvasive prenatal test for trisomy 21 and other aneuploidies have been investigated, including the use of cell-free fetal nucleic acids [57-60] and nucleated red blood fetal cells present in maternal peripheral blood [61,62]. Although studies have showed that noninvasive methods for obtaining fetal cells allow noninvasive prenatal diagnosis for a variety of genetic conditions and may in future form part of national antenatal screening programs for DS and other common genetic disorders, a major obstacle in the widespread application of noninvasive methods for obtaining fetal cells in clinical diagnostics is still that fetal cells / DNA constitutes a small percentage of total cell / DNA in maternal blood and the inconsistencies in enrichment strategies of these fetal samples [62,63].

After obtaining fetal cells, conventional karyotype analysis has been used for the past few decades as the gold standard for the prenatal diagnosis of numerical and major structural chromosomal abnormalities. Nevertheless, it is labor intensive and requires skilled chromosomal analysis with an average reporting time of 14 days. However, the availability of molecular techniques such as fluorescence in situ hybridization (FISH) has allowed the prenatal diagnosis of most frequent trisomies (21, 13, 18) and aneuploidy of sex chromosomes quickly and accurately, obtaining result from one to two days [64,65]. In addition, the technique of polymerase chain reaction quantitative fluorescent (QF-PCR), besides other molecular techniques such as the multiplex ligation-dependent probe amplification (MLPA) test and DNA sequencing, can also be used for a rapid diagnosis of aneuploidies [66-68]. It has been showed that QF-PCR technique presents 95.4% sensitivity, 100% specificity, 99.5% efficiency and is less laborious than the FISH technique, less time consuming, and some results were obtained in eight hours. The sensitivity, specificity, and efficiency of the assay for detecting DS using this technique are about 95.4%, 100%, and 99.5%, respectively [69]. Molecular techniques also enable the diagnosis of pre-implantation embryos in assisted reproduction [70].

It is important to note that the examinations of prenatal diagnosis should not be offered without the guidance of a geneticist to explain the risks to the parents and especially the implications of possible results. Early diagnosis helps couples to program for the treatment of the consequences of the syndrome diagnosed, preventing further damage and making possible the early stimulation of the patients, aiming their better integration into society.

Advertisement

4. Gene expression and DS phenotype

In a recent review of chromosome 21 content, 552 genes were identified in the long arm of the chromosome (21q) [71], including 161 protein-coding genes cataloged in the Reference Sequence database of the National Center for Biotechnology Information (NCBI). The remaining 391 gene models are referred to as novel genes or non-cataloged genes, which could be protein-coding genes or functional RNA genes. Considering that the genetic basis of DS is the presence of three copies of chromosome 21, the first and most commonly accepted hypothesis for DS phenotype is that the genes in triplicate are overexpressed and, thus, the dosage imbalance of genes on chromosome 21 is responsible for the molecular dysfunctions in DS [72]. Among the genes present in chromosome 21, may be highlighted some described in the literature with overexpression associated with phenotypes of DS, most influencing the structure or function of the central nervous system (Table 1). Location of these genes on chromosome 21 is presented in Figure 1.

Gene symbol* Gene location* Candidate gene for Reference
APP 21q21.3 Neurodegeneration [73,74]
BACH1 21q22.11 Alzheimer's disease-like neuropathological changes [75]
DOPEY2 21q22.2 Functional brain alterations and mental retardation [76]
DSCAM 21q22.2 Mental retardation and the precocious dementia [77]
DYRK1A 21q22.13 Leukemogenesis [78]
Impaired brain development [79]
Early onset of neurofibrillary degeneration [80]
ERG 21q22.3 Alzheimer's disease-like neuropathological changes [75]
OLIG2 21q22.11 Developmental brain defects [81]
SIM2 21q22.13 Impairment of learning and memory [82]
Pathogenesis of mental retardation [83]
SOD1 21q22.11 Neurodegeneration [84]
PCP4 21q22.2 Abnormal neuronal development [85]

Table 1.

Chromosome 21 gene-located with overexpression in DS influencing the structure or function of the central nervous system.

* http://www.ncbi.nlm.nih.gov/gene


Figure 1.

Location of genes overexpressed in DS influencing the structure or function of the central nervous system. Figure adapted from the NCBI Map Viewer database (http://www.ncbi.nlm.nih.gov/mapview/).

However, although elevated levels of gene expression on chromosome 21 in trisomy 21 tissues have been reported in several studies, there are evidences that increased copy number does not always correspond with increased gene expression level or even less with increased gene function [86,87]. In addition, studies have showed up- or downregulation of genes located on disomic chromosomes, indicating that the phenotype is due to an unstable environment resulting from the dosage imbalance of the hundreds of genes on chromosome 21 which determines a non-specific disturbance of genomic regulation and expression [88-90].

Besides altered pattern of gene expression, regulatory mechanisms are also altered in trisomy 21. Individuals with DS present altered pattern of DNA methylation in genes present in two or three copies with functional consequences in gene expression [91,92]. More recent studies have shown that trisomy 21 results in altered expression of microRNAs, small molecules of noncoding RNA involved in post-transcriptional gene regulation, which could result in abnormal expression of specific proteins and contribute to the DS phenotype [93-97]

The complete sequencing of chromosome 21 provided basis for the identification of candidate genes for DS phenotype manifestations. Currently, there are several genes located on chromosome 21 associated to DS phenotype and the involvement of other genes still will be elucidated with advances of genomics and proteomics. The knowing of these gene functions and their contribution for DS phenotype are fundamental for the understanding of the syndrome and for providing basis for the planning of therapeutic strategies that could contribute to improve the quality of life of DS individuals.

Advertisement

5. Conclusion

Although individuals with trisomy 21 present several characteristics that make possible the clinical diagnosis of DS, the confirmation of the diagnosis by cytogenetic analysis is essential to establish the recurrence risks of the syndrome. We highlight the importance of the prenatal diagnosis of DS to provide the needed healthcare for the child, to prepare the family emotional and psychologically and to plan early intervention therapies. The successful control of pharmacological and clinical problems of patients with DS is the biggest medical challenge and depends on the understanding of unbalanced metabolism induced by high expression of the genes located on chromosome 21.

Advertisement

Acknowledgments

The authors acknowledge support from FAPESP (São Paulo Research Foundation), CNPq (National Council for Scientific and Technological Development) and CAPES (Coordination for the Improvement of Higher Education Personnel).

References

  1. 1. Jones, K.L. Smith's recognizable patterns of human malformation. 6th ed. Philadelphia: Elsevier Saunders, 2006.
  2. 2. Neri G, Opitz JM. Down syndrome: comments and reflections on the 50th anniversary of Lejeune's discovery. Am J Med Genet A. 2009 Dec;149A(12):2647-54. http://onlinelibrary.wiley.com/doi/10.1002/ajmg.a.33138/abstract (accessed 20 July 2012).
  3. 3. Ahmed I, Ghafoor T, Samore NA, Chattha MN. Down syndrome: clinical and cytogenetic analysis. J Coll Physicians Surg Pak. 2005 Jul;15(7):426-9.
  4. 4. Azman BZ, Ankathil R, Siti Mariam I, Suhaida MA, Norhashimah M, Tarmizi AB, et al. Cytogenetic and clinical profile of Down syndrome in Northeast Malaysia. Singapore Med J. 2007 Jun;48(6):550-4. http://smj.sma.org.sg/4806/4806a10.pdf (accessed 7 June 2012).
  5. 5. Biselli JM, Goloni-Bertollo EM, Ruiz MT, Pavarino-Bertelli EC. Cytogenetic profile of Down syndrome cases seen by a general genetics outpatient service in Brazil. Down's syndrome, research and practice, 2008 Feb; 12(3). http://www.down-syndrome.org/reports/2010/ (accessed 5 June 2012).
  6. 6. Vraneković J, Božović IB, Grubić Z, Wagner J, Pavlinić D, Dahoun S, et al. Down syndrome: parental origin, recombination, and maternal age. Genet Test Mol Biomarkers. 2012 Jan;16(1):70-3.
  7. 7. Pavarino Bertelli EC, Biselli JM, Bonfim D, Goloni-Bertollo EM. Clinical profile of children with Down syndrome treated in a genetics outpatient service in the southeast of Brazil. Rev Assoc Med Bras. 2009 Sep-Oct;55(5):547-52. http://www.scielo.br/scielo.php?script=sci_arttext&pid=S0104-42302009000500017&lng=en&nrm=iso&tlng=en (accessed 5 June 2012)
  8. 8. Mustacchi Z. Síndrome de Down. In: Mustacchi Z, Peres S. Genética Baseada em Evidências Síndromes e Heranças. São Paulo: CID Editora, 2000. p817-894.
  9. 9. Mıhçı E, Akçurin G, Eren E, Kardelen F, Akçurin S, Keser I, et al. Evaluation of congenital heart diseases and thyroid abnormalities in children with Down syndrome. Anadolu Kardiyol Derg. 2010 Oct;10(5):440-5. http://www.anakarder.com/yazilar.asp?yaziid=1739&sayiid= (accessed 20 June 2012).
  10. 10. Morgan J. Why is periodontal disease more prevalent and more severe in people with Down syndrome? Spec Care Dentist. 2007 Sep-Oct;27(5):196-201.
  11. 11. Stirn Kranjc B. Ocular abnormalities and systemic disease in down syndrome. Strabismus. 2012 Jun;20(2):74-7. http://informahealthcare.com/doi/abs/10.3109/09273972.2012.680234 (accessed 20 June 2012).
  12. 12. Raut P, Sriram B, Yeoh A, Hee KY, Lim SB, Daniel ML. High prevalence of hearing loss in Down syndrome at first year of life. Ann Acad Med Singapore. 2011 Nov;40(11):493-8. http://www.annals.edu.sg/pdf/40VolNo11Nov2011/V40N11p493.pdf (accessed 20 June 2012).
  13. 13. Mitchell RB, Call E, Kelly J. Ear, nose and throat disorders in children with Down syndrome. Laryngoscope. 2003 Feb;113(2):259-63. http://onlinelibrary.wiley.com/doi/10.1097/00005537-200302000-00012/abstract (accessed 15 June 2012).
  14. 14. Grinspon RP, Bedecarrás P, Ballerini MG, Iñiguez G, Rocha A, Mantovani Rodrigues Resende EA, et al. Early onset of primary hypogonadism revealed by serum anti-Müllerian hormone determination during infancy and childhood in trisomy 21. Int J Androl. 2011 Oct;34(5 Pt 2):e487-98. http://onlinelibrary.wiley.com/doi/10.1111/j.1365-2605.2011.01210.x/abstract (accessed 5 July 2012).
  15. 15. Ram G, Chinen J. Infections and immunodeficiency in Down syndrome. Clin Exp Immunol. 2011 Apr;164(1):9-16. http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3074212/?tool=pubmed (accessed July 2012).
  16. 16. Bruwier A, Chantrain CF. Hematological disorders and leukemia in children with Down syndrome. Eur J Pediatr. 2011 Nov 24. DOI: 10.1007/s00431-011-1624-1. http://www.springerlink.com/content/a8730m24l57uu14g/?MUD=MP (accessed 3 June 2012).
  17. 17. Lockrow JP, Fortress AM, Granholm AC. Age-related neurodegeneration and memory loss in down syndrome. Curr Gerontol Geriatr Res. 2012;2012:463909. http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3318235/?tool=pubmed (accessed 20 June 2012).
  18. 18. Jagiello GM, Fang JS, Nogawa T, Sung WK, Ducayen MB, Bowne W. Chromosome 21 behavior during fetal oogenesis in Down's syndrome. Obstet Gynecol. 1987 Dec;70(6):878-83.
  19. 19. Mercer ES, Broecker B, Smith EA, Kirsch AJ, Scherz HC, Massad CA. Urological manifestations of Down syndrome. J Urol. 2004 Mar;171(3):1250-3.
  20. 20. Pradhan, M., Dalal, A., Khan, F., & Agrawal, S. Fertility in men with Down syndrome: A case report. Fertil Steril. 2006 Dec;86(6):1765.e1-3. http://www.fertstert.org/article/S0015-0282(06)03067-6/abstract (accessed 20 June 2012).
  21. 21. Epstein CJ. Genetic Counseling: Statement of the American Society of Human GeneticsAd Hoc Committee on Genetic Counseling. Am J Hum Genet. 1975; 27(2): 241-242.
  22. 22. Pinto Junior, W. Diagnóstico pré-natal. Ciênc. saúde coletiva. 2002; 7(1):139-157. http://redalyc.uaemex.mx/pdf/630/63070113.pdf. (accessed 18 June 2012).
  23. 23. Jyothy A, Kumar KS, Mallikarjuna GN, Babu Rao V, Uma Devi B, Sujatha M, et al. Parental age and the origin of extra chromosome 21 in Down syndrome. J Hum Genet. 2001;46(6):347-50. http://www.nature.com/jhg/journal/v46/n6/full/jhg200164a.html. (accessed 20 June 2012).
  24. 24. Allen EG, Freeman SB, Druschel C, Hobbs CA, O'Leary LA, Romitti PA, et al. Maternal age and risk for trisomy 21 assessed by the origin of chromosome nondisjunction: a report from the Atlanta and National Down Syndrome Projects. Hum Genet. 2009 Feb;125(1):41-52. http://www.ncbi.nlm.nih.gov/pmc/articles/PMC2833410/?tool=pubmed (accessed 6 June 2012).
  25. 25. Nicolaides KH. Screening for fetal aneuploidies at 11 to 13 weeks. Prenat Diagn. 2011 Jan;31(1):7-15. http://onlinelibrary.wiley.com/doi/10.1002/pd.2637/abstract (accessed 18 June 2012).
  26. 26. Sheets KB, Crissman BG, Feist CD, Sell SL, Johnson LR, Donahue KC, et al. Practice guidelines for communicating a prenatal or postnatal diagnosis of Down syndrome: recommendations of the national society of genetic counselors. J Genet Couns. 2011 Oct;20(5):432-41. http://www.springerlink.com/content/a238112117611644/ (accessed 18 June 2012).
  27. 27. Pavarino EC, Zampieri BL, Biselli JM, Goloni-Bertollo EM. Abnormal Folate Metabolism and Maternal Risk for Down Syndrome. In: Dey S., editor. Genetics and Etiology of Down Syndrome. Rijeka, Croácia: InTech; 2011. p. 97-120. Available from http://www.intechopen.com/books/genetics-and-etiology-of-down-syndrome/abnormal-folate-metabolism-and-maternal-risk-for-down-syndrome (accessed 15 June 2012).
  28. 28. Morris JK, Wald NJ, Watt HC. Fetal loss in Down syndrome pregnancies. Prenat Diagn. 1999 Feb;19(2):142-5. http://onlinelibrary.wiley.com/doi/10.1002/(SICI)1097-0223(199902)19:2% 3C142::AID-PD486% 3E3.0.CO;2-7/abstract (accessed 30 July 2012).
  29. 29. Rihtman T, Tekuzener E, Parush S, Tenenbaum A, Bachrach SJ, Ornoy A. Are the cognitive functions of children with Down syndrome related to their participation? Dev Med Child Neurol. 2010 Jan;52(1):72-8. http://onlinelibrary.wiley.com/doi/10.1111/j.1469-8749.2009.03356.x/abstract (accessed 20 June 2012).
  30. 30. Glasson EJ, Sullivan SG, Hussain R, Petterson BA, Montgomery PD, Bittles AH. The changing survival profile of people with Down’s syndrome: Implications for genetic counselling. Clin Genet. 2002 Nov;62(5):390-3. http://onlinelibrary.wiley.com/doi/10.1034/j.1399-0004.2002.620506.x/abstract (accessed 12 July 2012).
  31. 31. Irving C, Basu A, Richmond S, Burn J, Wren C. Twenty-year trends in prevalence and survival of Down syndrome. Eur J Hum Genet. 2008 Nov;16(11):1336-40. http://www.nature.com/ejhg/journal/v16/n11/full/ejhg2008122a.html (accessed 15 July 2012).
  32. 32. Wang Y, Hu J, Druschel CM, Kirby RS. Twenty-five-year survival of children with birth defects in New York State: a population-based study. Birth Defects Res A Clin Mol Teratol. 2011 Dec;91(12):995-1003. http://onlinelibrary.wiley.com/doi/10.1002/bdra.22858/abstract (accessed 18 June 2012).
  33. 33. Bryant LD, Murray J, Green JM, Hewison J, Sehmi I, Ellis A. Descriptive information about Down syndrome: A content analysis of serum screening leaflets. Prenat Diagn. 2001 Dec;21(12):1057-63. http://onlinelibrary.wiley.com/doi/10.1002/pd.179/abstract (accessed 5 July 2012).
  34. 34. Nicolaides KH, Azar G, Byrne D, Mansur C, Marks K. Fetal nuchal translucency: ultrasound screening for chromosomal defects in first trimester of pregnancy. BMJ. 1992 Apr 4;304(6831):867-9. http://www.ncbi.nlm.nih.gov/pmc/articles/PMC1882788/?tool=pubmed (accessed 18 June 2012).
  35. 35. Pandya PP, Kondylios A, Hilbert L, Snijders RJ, Nicolaides KH. Chromosomal defects and outcome in 1015 fetuses with increased nuchal translucency. Ultrasound Obstet Gynecol. 1995 Jan;5(1):15-9. http://onlinelibrary.wiley.com/doi/10.1046/j.1469-0705.1995.05010015.x/abstract (accessed 18 June 2012).
  36. 36. Mendoza-Caamal EC, Grether-González P, Hernández-Gómez M, Guzmán-Huerta M, Aguinaga-Ríos M. Birth defects associated with increased nuchal translucency. Ginecol Obstet Mex. 2010 Oct;78(10):533-9.
  37. 37. Mogra R, Alabbad N, Hyett J. Increased nuchal translucency and congenital heart disease. Early Hum Dev. 2012 May;88(5):261-7. http://www.earlyhumandevelopment.com/article/S0378-3782(12)00063-1/abstract (accessed 18 June 2012).
  38. 38. Merkatz IR, Nitowsky HM, Macri JN, Johnson WE. An association between low m7ternal serum alpha-fetoprotein and fetal chromosomal abnormalities. Am J Obstet Gynecol. 1984 Apr 1;148(7):886-94.
  39. 39. Bogart MH, Pandian MR, Jones OW. Abnormal maternal serum chorionic gonadotropin levels in pregnancies with fetal chromosome abnormalities. Prenat Diagn. 1987 Nov;7(9):623-30.
  40. 40. Canick JA, Knight GJ, Palomaki GE, Haddow JE, Cuckle HS, Wald NJ. Low second trimester maternal serum unconjugated oestriol in pregnancies with Down’s syndrome. Br J Obstet Gynaecol. 1988 Apr;95(4):330-3.
  41. 41. Brambati B, Macintosh MC, Teisner B, Maguiness S, Shrimanker K, Lanzani A, et al. Low maternal serum levels of pregnancy associated plasma protein A (PAPP-A) in the first trimester in association with abnormal fetal karyotype. Br J Obstet Gynaecol. 1993 Apr;100(4):324-6.
  42. 42. Aitken DA, Wallace EM, Crossley JA, Swanston IA, van Pareren Y, van Maarle M, et al. 1996. Dimeric inhibin A as a marker for Down’s syndrome in early pregnancy. N Engl J Med. 1996 May 9;334(19):1231-6. http://www.nejm.org/doi/full/10.1056/NEJM199605093341904 (accessed 1 July 2012).
  43. 43. Benn PA. Advances in prenatal screening for Down syndrome: I. General principles and second trimester testing. Clin Chim Acta. 2002 Sep;323(1-2):1-16. http://www.sciencedirect.com/science/article/pii/S0009898102001869 (accessed 26 July 2012).
  44. 44. Minami S, Yamoto M, Nakano R. Sources of inhibin in early pregnancy. Early Pregnancy. 1995 Mar;1(1):62-6.
  45. 45. Malone FD, Canick JA, Ball RH, Nyberg DA, Comstock CH, Bukowski R, et al. First-trimester or second-trimester screening, or both, for Down's syndrome. N Engl J Med. 2005 Nov 10;353(19):2001-11. http://www.nejm.org/doi/full/10.1056/NEJMoa043693 (accessed 18 June 2012).
  46. 46. Kagan KO, Wright D, Baker A, Sahota D, Nicolaides KH. Screening for trisomy 21 by maternal age, fetal nuchal translucency thickness, free beta-human chorionic gonadotropin and pregnancy-associated plasma protein-A. Ultrasound Obstet Gynecol. 2008 Jun;31(6):618-24. http://onlinelibrary.wiley.com/doi/10.1002/uog.5331/abstract (accessed 16 July 2012).
  47. 47. Raniga S, Desai PD, Parikh H. Ultrasonographic soft markers of aneuploidy in second trimester: are we lost? MedGenMed. 2006 Jan 11;8(1):9. http://www.ncbi.nlm.nih.gov/pmc/articles/PMC1681991/?tool=pubmed (accessed 18 June 2012).
  48. 48. Mazzoni GT Jr, Cabral AC, de Lima Faria MM, Castro MJ, de Carvalho Pires M, Johnson DS, et al. Ultrasound evaluation of the fetal nasal bone: what is the most appropriate first-trimester cut-off point for aneuploidy screening? Arch Gynecol Obstet. 2012 May;285(5):1263-70. http://www.springerlink.com/content/w04724766836n9x2/ (accessed 18 June 2012).
  49. 49. Bromley B, Lieberman E, Shipp TD, Benacerraf BR. The genetic sonogram: a method of risk assessment for Down syndrome in the second trimester. J Ultrasound Med. 2002 Oct;21(10):1087-96; quiz 1097-8. http://www.jultrasoundmed.org/content/21/10/1087.long (accessed 6 July 2012).
  50. 50. Simpson JL. Invasive procedures for prenatal diagnosis: Any future left? Best Pract Res Clin Obstet Gynaecol. 2012 Jun 30. DOI: 10.1016/j.bpobgyn.2012.05.007. http://www.sciencedirect.com/science/article/pii/S1521693412000934 (accessed 18 June 2012).
  51. 51. Enzensberger C, Pulvermacher C, Degenhardt J, Kawacki A, Germer U, Gembruch U, et al. Fetal Loss Rate and Associated Risk Factors After Amniocentesis, Chorionic Villus Sampling and Fetal Blood Sampling. Ultraschall Med. 2012 May 23. DOI: 10.1055/s-0031-1299388. https://www.thieme-connect.com/DOI/DOI?10.1055/s-0031-1299388 (accessed 15 July 2012).
  52. 52. Dhaifalah I, Zapletalova J. Safety and risks associated with screening for chromosomal abnormalities during pregnancy Ceska Gynekol. 2012 Summer;77(3):236-241.
  53. 53. Alfirevic Z, Sundberg K, Brigham S. Amniocentesis and chorionic villus sampling for prenatal diagnosis. Cochrane Database Syst Rev. 2003;(3):CD003252. http://onlinelibrary.wiley.com/doi/10.1002/14651858.CD003252/abstract;jsessionid=9599AB64C84E30EF63AFAB4D48359C0F.d03t02 (accessed 7 July 2012).
  54. 54. Tongsong T, Wanapirak C, Kunavikatikul C, Sirirchotiyakul S, Piyamongkol W, Chanprapaph P. Fetal loss rate associated with cordocentesis at midgestation. Am J Obstet Gynecol. 2001 Mar;184(4):719-23. http://www.ajog.org/article/S0002-9378(01)74471-7/abstract (accessed 18 June 2012).
  55. 55. Liao C, Wei J, Li Q, Li L, Li J, Li D. Efficacy and safety of cordocentesis for prenatal diagnosis. Int J Gynaecol Obstet. 2006 Apr;93(1):13-7. http://www.ijgo.org/article/S0020-7292(06)00013-0/abstract (accessed 18 June 2012).
  56. 56. Boupaijit K, Wanapirak C, Piyamongkol W, Sirichotiyakul S, Tongsong T. Effect of placenta penetration during cordocentesis at mid-pregnancy on fetal outcomes. Prenat Diagn. 2012 Jan;32(1):83-7. http://onlinelibrary.wiley.com/doi/10.1002/pd.2916/abstract (accessed 5 June 2012).
  57. 57. Wright CF, Burton H. The use of cell-free fetal nucleic acids in maternal blood for non-invasive prenatal diagnosis. Hum Reprod Update. 2009 Jan-Feb;15(1):139-51. http://humupd.oxfordjournals.org/content/15/1/139.long (accessed 18 June 2012).
  58. 58. Chiu RHK, Akolekar R, Zheng YWL, Leung TY, Sun H, Chan KCA, et al. Non-invasive prenatal assessment of trisomy 21 by multiplexed maternal plasma DNA sequencing: large scale validity study. BMJ. 2011 Jan 11;342:c7401. http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3019239/?tool=pubmed (accessed 7 June 2012).
  59. 59. Canick JA, Kloza EM, Lambert-Messerlian GM, Haddow JE, Ehrich M, van den Boom D, et al. DNA sequencing of maternal plasma to identify Down syndrome and other trisomies in multiple gestations. Prenat Diagn. 2012 Aug;32(8):730-4. http://onlinelibrary.wiley.com/doi/10.1002/pd.3892/abstract (accessed 7 June 2012).
  60. 60. Ashoor G, Syngelaki A, Wagner M, Birdir C, Nicolaides KH. Chromosome-selective sequencing of maternal plasma cell-free DNA for first-trimester detection of trisomy 21 and trisomy 18. Am J Obstet Gynecol. 2012 Apr;206(4):322.e1-5. http://www.ajog.org/article/S0002-9378(12)00060-9/abstract (accessed 1 July 2012).
  61. 61. Yang YH, Kim SH, Yang ES, Kim SK, Kim IK, Park YW, et al. Prenatal diagnosis of fetal trisomy 21 from maternal peripheral blood. Yonsei Med J. 2003 Apr 30;44(2):181-6. http://www.eymj.org/DOIx.php?id=10.3349/ymj.2003.44.2.181 (accessed 18 June 2012).
  62. 62. Sifakis S, Papantoniou N, Kappou D, Antsaklis A. Noninvasive prenatal diagnosis of Down syndrome: current knowledge and novel insights. J Perinat Med. 2012 Feb 13;40(4):319-27. http://www.degruyter.com/view/j/jpme.2012.40.issue-4/jpm-2011-0282/jpm-2011-0282.xml;jsessionid=0E0E90A7F0EAF0618F024D6215251415 (accessed 18 June 2012).
  63. 63. Choolani M, Mahyuddin AP, Hahn S. The promise of fetal cells in maternal blood. Best Pract Res Clin Obstet Gynaecol. 2012 Jul 12. DOI: 10.1016/j.bbr.2011.03.031. http://www.sciencedirect.com/science/article/pii/S152169341200106X (accessed 6 June 2012).
  64. 64. Jia CW, Wang SY, Ma YM, Lan YL, Si YM, Yu L, et al. Fluorescence in situ hybridization in uncultured amniocytes for detection of aneuploidy in 4210 prenatal cases. Chin Med J (Engl). 2011 Apr;124(8):1164-8. http://www.cmj.org/Periodical/paperlist.asp?id=LW2011420367338008724&linkintype=pubmed (accessed 20 June 2012).
  65. 65. Ho SS, Chua C, Gole L, Biswas A, Koay E, Choolani M. Same-day prenatal diagnosis of common chromosomal aneuploidies using microfluidics-fluorescence in situ hybridization. Prenat Diagn. 2012 Apr;32(4):321-8. http://onlinelibrary.wiley.com/doi/10.1002/pd.2946/abstract (accessed 5 July 2012).
  66. 66. Hills A, Donaghue C, Waters J, Waters K, Sullivan C, Kulkarni A, et al. QF-PCR as a stand-alone test for prenatal samples: the first 2 years’ experience in the London region. Prenat Diagn. 2010 Jun;30(6):509-17. http://onlinelibrary.wiley.com/doi/10.1002/pd.2503/abstract (accessed 29 July 2012).
  67. 67. Faas BHW, Cirigliano V, Bui TH. Rapid methods for targeted prenatal diagnosis of common chromosome aneuploidies. Semin Fetal Neonatal Med. 2011 Apr;16(2):81-7. http://www.sfnmjournal.com/article/S1744-165X(11)00004-7/abstract (accessed 12 July 2012).
  68. 68. Canick JA, Kloza EM, Lambert-Messerlian GM, Haddow JE, Ehrich M, van den Boom D, et al. DNA sequencing of maternal plasma to identify Down syndrome and other trisomies in multiple gestations. Prenat Diagn. 2012 Aug;32(8):730-4. http://onlinelibrary.wiley.com/doi/10.1002/pd.3892/abstract;jsessionid=8345A09565F7EB0EE80CE1EE242D45CA.d03t01 (accessed 18 June 2012).
  69. 69. Lee MH, Ryu HM, Kim DJ, Lee BY, Cho EH, Yang JH, et al. Rapid prenatal diagnosis of Down syndrome using quantitative fluorescent PCR in uncultured amniocytes. J Korean Med Sci. 2004 Jun;19(3):341-4. http://www.ncbi.nlm.nih.gov/pmc/articles/PMC2816832/?tool=pubmed (accessed 18 June 2012).
  70. 70. Zhang Y, Xu CM, Zhu YM, Dong MY, Qian YL, Jin F, et al. Preimplantation genetic diagnosis for Down syndrome pregnancy. J Zhejiang Univ Sci B. 2007 Jul;8(7):515-21. http://www.ncbi.nlm.nih.gov/pmc/articles/PMC1906599/?tool=pubmed (accessed 18 June 2012).
  71. 71. Sturgeon X, Gardiner KJ. Transcript catalogs of human chromosome 21 and orthologous chimpanzee and mouse regions. Mamm Genome. 2011 Jun;22(5-6):261-71. http://www.springerlink.com/content/q35t330n314v6107/ (accessed 15 June 2012).
  72. 72. Antonarakis SE, Lyle R, Chrast R, Scott HS. Differential gene expression studies to explore the molecular pathophysiology of Down syndrome. Brain Res Brain Res Rev. 2001 Oct;36(2-3):265-74. http://www.sciencedirect.com/science/article/pii/S0165017301001035 (accessed 15 July 2012).
  73. 73. Millan Sanchez M, Heyn SN, Das D, Moghadam S, Martin KJ, Salehi A. Neurobiological elements of cognitive dysfunction in down syndrome: exploring the role of APP. Biol Psychiatry. 2012 Mar 1;71(5):403-9. http://www.biologicalpsychiatryjournal.com/article/S0006-3223(11)00822-5/abstract (accessed 18 June 2012).
  74. 74. Moncaster JA, Pineda R, Moir RD, Lu S, Burton MA, Ghosh JG, et al. Alzheimer's disease amyloid-beta links lens and brain pathology in Down syndrome. PLoS One. 2010 May 20;5(5):e10659. http://www.ncbi.nlm.nih.gov/pmc/articles/PMC2873949/?tool=pubmed (accessed 20 June 2012).
  75. 75. Shim KS, Ferrando-Miguel R, Lubec G. Aberrant protein expression of transcription factors BACH1 and ERG, both encoded on chromosome 21, in brains of patients with Down syndrome and Alzheimer's disease. J Neural Transm Suppl. 2003;(67):39-49.
  76. 76. Rachidi M, Delezoide AL, Delabar JM, Lopes C. A quantitative assessment of gene expression (QAGE) reveals differential overexpression of DOPEY2, a candidate gene for mental retardation, in Down syndrome brain regions. Int J Dev Neurosci. 2009 Jun;27(4):393-8. http://www.sciencedirect.com/science/article/pii/S0736574809000197 (accessed 20 July 2012).
  77. 77. Saito Y, Oka A, Mizuguchi M, Motonaga K, Mori Y, Becker LE, et al. The developmental and aging changes of Down's syndrome cell adhesion molecule expression in normal and Down's syndrome brains. Acta Neuropathol. 2000 Dec;100(6):654-64. http://www.springerlink.com/content/gbjxfuyf3d4lk98p/ (accessed 20 July 2012).
  78. 78. Malinge S, Bliss-Moreau M, Kirsammer G, Diebold L, Chlon T, Gurbuxani S, et al. Increased dosage of the chromosome 21 ortholog Dyrk1a promotes megakaryoblastic leukemia in a murine model of Down syndrome. J Clin Invest. 2012 Mar 1;122(3):948-62. http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3287382/?tool=pubmed (accessed 15 June 2012).
  79. 79. Mazur-Kolecka B, Golabek A, Kida E, Rabe A, Hwang YW, Adayev T, et al. Effect of DYRK1A activity inhibition on development of neuronal progenitors isolated from Ts65Dn mice. J Neurosci Res. 2012 May;90(5):999-1010. http://onlinelibrary.wiley.com/doi/10.1002/jnr.23007/abstract. (accessed 15 June 2012).
  80. 80. Wegiel J, Kaczmarski W, Barua M, Kuchna I, Nowicki K, Wang KC, et al. Link between DYRK1A overexpression and several-fold enhancement of neurofibrillary degeneration with 3-repeat tau protein in Down syndrome. J Neuropathol Exp Neurol. 2011 Jan;70(1):36-50. http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3083064/?tool=pubmed (accessed 20 June 2012).
  81. 81. Chakrabarti L, Best TK, Cramer NP, Carney RS, Isaac JT, Galdzicki Z, et al. Olig1 and Olig2 triplication causes developmental brain defects in Down syndrome. Nat Neurosci. 2010 Aug;13(8):927-34. http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3249618/?tool=pubmed (accessed 3 July 2012).
  82. 82. Meng X, Peng B, Shi J, Zheng Y, Chen H, Zhang J, et al. Effects of overexpression of Sim2 on spatial memory and expression of synapsin I in rat hippocampus. Cell Biol Int. 2006 Oct;30(10):841-7. http://www.cellbiolint.org/cbi/030/0841/cbi0300841.htm (accessed 20 July 2012).
  83. 83. Rachidi M, Lopes C, Charron G, Delezoide AL, Paly E, Bloch B, et al. Spatial and temporal localization during embryonic and fetal human development of the transcription factor SIM2 in brain regions altered in Down syndrome. Int J Dev Neurosci. 2005 Aug;23(5):475-84. http://www.sciencedirect.com/science/article/pii/S0736574805000638 (accessed 20 July 2012).
  84. 84. Shin JH, London J, Le Pecheur M, Weitzdoerfer R, Hoeger H, Lubec G. Proteome analysis in hippocampus of mice overexpressing human Cu/Zn-superoxide dismutase 1. Neurochem Int. 2005 Jun;46(8):641-53. http://www.sciencedirect.com/science/article/pii/S0197018605000434 (accessed 20 July 2012).
  85. 85. Mouton-Liger F, Thomas S, Rattenbach R, Magnol L, Larigaldie V, Ledru A, et al. PCP4 (PEP19) overexpression induces premature neuronal differentiation associated with Ca(2+) /calmodulin-dependent kinase II-δ activation in mouse models of Down syndrome. J Comp Neurol. 2011 Oct 1;519(14):2779-802. http://onlinelibrary.wiley.com/doi/10.1002/cne.22651/abstract (accessed 20 July 2012).
  86. 86. Aït Yahya-Graison E, Aubert J, Dauphinot L, Rivals I, Prieur M, Golfier G, et al. Classification of human chromosome 21 gene-expression variations in Down syndrome: impact on disease phenotypes. Am J Hum Genet. 2007 Sep;81(3):475-91. http://www.ncbi.nlm.nih.gov/pmc/articles/PMC1950826/?tool=pubmed (accessed 9 July 2012).
  87. 87. Kahlem P, Sultan M, Herwig R, Steinfath M, Balzereit D, Eppens B, et al. Transcript level alterations reflect gene dosage effects across multiple tissues in a mouse model of down syndrome. Genome Res. 2004 Jul;14(7):1258-67. http://www.ncbi.nlm.nih.gov/pmc/articles/PMC442140/?tool=pubmed (accessed 29 July 2012).
  88. 88. Saran NG, Pletcher MT, Natale JE, Cheng Y, Reeves RH. Global disruption of the cerebellar transcriptome in a Down syndrome mouse model. Hum Mol Genet. 2003 Aug 15;12(16):2013-9. http://hmg.oxfordjournals.org/content/12/16/2013.long (accessed 20 July 2012).
  89. 89. FitzPatrick DR. Transcriptional consequences of autosomal trisomy: primary gene dosage with complex downstream effects. Trends Genet. 2005 May;21(5):249-53. http://www.sciencedirect.com/science/article/pii/S0168952505000582 (accessed 29 June 2012).
  90. 90. Dauphinot L, Lyle R, Rivals I, Dang MT, Moldrich RX, Golfier G, et al. The cerebellar transcriptome during postnatal development of the Ts1Cje mouse, a segmental trisomy model for Down syndrome. Hum Mol Genet. 2005 Feb 1;14(3):373-84. http://hmg.oxfordjournals.org/content/14/3/373.long (accessed 29 June 2012).
  91. 91. Kuromitsu J, Yamashita H, Kataoka H, Takahara T, Muramatsu M, Sekine T, et al. A unique downregulation of h2-calponin gene expression in Down syndrome: a possible attenuation mechanism for fetal survival by methylation at the CpG island in the trisomic chromosome 21. Mol Cell Biol. 1997 Feb;17(2):707-12. http://www.ncbi.nlm.nih.gov/pmc/articles/PMC231796/?tool=pubmed (accessed 29 June 2012).
  92. 92. Kerkel K, Schupf N, Hatta K, Pang D, Salas M, Kratz A, et al. Altered DNA methylation in leukocytes with trisomy 21. PLoS Genet. 2010 Nov 18;6(11):e1001212. http://www.ncbi.nlm.nih.gov/pmc/articles/PMC2987931/?tool=pubmed (accessed 29 June 2012).
  93. 93. Sethupathy P, Borel C, Gagnebin M, Grant GR, Deutsch S, Elton TS, et al. Human microRNA-155 on chromosome 21 differentially interacts with its polymorphic target in the AGTR1 3' untranslated region: a mechanism for functional single-nucleotide polymorphisms related to phenotypes. Am J Hum Genet. 2007 Aug;81(2):405-13. http://www.ncbi.nlm.nih.gov/pmc/articles/PMC1950808/?tool=pubmed (accessed 6 June 2012).
  94. 94. Kuhn DE, Nuovo GJ, Martin MM, Malana GE, Pleister AP, Jiang J, et al. Human chromosome 21-derived miRNAs are overexpressed in down syndrome brains and hearts. Biochem Biophys Res Commun. 2008 Jun 6;370(3):473-7. http://www.ncbi.nlm.nih.gov/pmc/articles/PMC2585520/?tool=pubmed (accessed 15 June 2012).
  95. 95. Elton TS, Sansom SE, Martin MM. Trisomy-21 gene dosage over-expression of miRNAs results in the haploinsufficiency of specific target proteins. RNA Biol. 2010 Sep-Oct;7(5):540-7. http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3073250/?tool=pubmed (accessed 26 June 2012).
  96. 96. Kuhn DE, Nuovo GJ, Terry AV Jr, Martin MM, Malana GE, Sansom SE, et al. Chromosome 21-derived microRNAs provide an etiological basis for aberrant protein expression in human Down syndrome brains. J Biol Chem. 2010 Jan 8;285(2):1529-43. http://www.ncbi.nlm.nih.gov/pmc/articles/PMC2801278/?tool=pubmed (accessed 15 June 2012).
  97. 97. Keck-Wherley J, Grover D, Bhattacharyya S, Xu X, Holman D, Lombardini ED, et al. Abnormal microRNA expression in Ts65Dn hippocampus and whole blood: contributions to Down syndrome phenotypes. Dev Neurosci. 2011;33(5):451-67. http://content.karger.com/produktedb/produkte.asp?DOI=10.1159/000330884 (accessed 15 June 2012).

Written By

Érika Cristina Pavarino, Joice Matos Biselli, Walter Pinto Junior and Eny Maria Goloni Bertollo

Submitted: 24 April 2012 Published: 06 March 2013