Rev Esp Endocrinol Pediatr

 View / Download PDF
Rev Esp Endocrinol Pediatr 2013;4 Suppl(1):51-56 | Doi. 10.3266/RevEspEndocrinolPediatr.pre2013.Mar.162
New aspects in endocrine dysfunction in Prader-Willi syndrome

Sent for review: 21 Mar. 2013 | Accepted: 22 Mar. 2013  | Published: 2 May. 2013
Maithe Tauber1, Jean Pierre Salles 1, Gwenaelle Diene2, Emmanuelle Mimoun 2, Catherine Molinas2, Sophie Çabal-Berthoumieu 2, Carine Mantoulan2, Francoise Lorenzini3, Florence Labrousse 3, Muriel Coupaye4, Christine Poitou 4
1Centre de référence du syndrome de Prader-Willi. Unité d’Endocrinologie, Hôpital des Enfants, CHU de Toulouse. Centre de Physiopathologie de Toulouse. Toulouse (France)
2Centre de référence du syndrome de Prader-Willi. Unité d’Endocrinologie, Hôpital des Enfants, CHU de Toulouse . Toulouse (France)
3Centre de référence du syndrome de Prader-Willi. Service d’Endocrinologie, Maladies Métaboliques et Nutrition, hôpital Rangueil, CHU de Toulouse. Toulouse (France)
4Centre de référence du syndrome de Prader-Willi. Unité de Nutrition et d’Endocrinologie, Hôpital de la Pitié Salpêtrière. Toulouse, Paris (France)
Correspondence:Maithe Tauber, Centre de référence du syndrome de Prader-Willi. Unité d’Endocrinologie, Hôpital des Enfants, CHU de Toulouse. Centre de Physiopathologie de Toulouse, Toulouse, France
E-mail: tauber.mt@chu-toulouse.fr
Table 1 - Characteristics of the Whole Cohort and of the Two Groups According to Receiving or not Receiving GH treatment in childhood (modified from Coupaye et al)
Figure 1 - Organization of the PWS locus
Figure 2 - Comparison between mean IGF-1 levels expressed in SDS between children with PWS (n=47) in darked grey bars and GHD children (n=32) in light grey bars (2A)
Abstract

Prader-Willi syndrome (PWS) first described in 1956 is considered as a paradigm of a neurodevelopmental disorder with endocrine dysfunction resulting from a yet unknown hypothalamic trouble. This article reviews some new aspects of the syndrome and focus on GH treatment, ghrelin and oxytocin (OT) dysregulation.

The natural history of the disease has been described due to the early diagnosis during the first months of life and the nutritional phases have been precised. In addition, it is now become apparent that eating behaviour troubles are part of general behavioural problems. Brain imaging studies have shown that some brain regions may be important in PWS.

The pathophysiology of the disease is discussed. The role of SNORD116 gene cluster is suggested by the description of patients with microdeletions encompassing this gene cluster. The pathophysiology of the abnormal ghrelin and OT pathways is important to describe and understand. Administration of OT has been shown to improve some of the behaviour problems in adults.

These new aspects offer an opportunity for therapeutical use and possible early intervention.

Key Words: PWS, GH, Oxytocin, ghrelin, SNORD116 gene cluster Palabras clave:

Introduction

PWS is a relatively common contiguous gene syndrome (estimated prevalence 1/15 000) resulting from the lack of expression of paternal alleles within the chromosome region 15q11-q13 which includes SNURF-SNRPN and several small nucleolar RNAs (snoRNAs) genes 1, 2 and is shown in Figure 1. It is the best known example of a human disorder involving genomic imprinting3. The phenotype, firstly described by Prader, Labhart and Willi in 19564 includes neonatal hypotonia, early onset of hyperphagia and development of morbid obesity, short stature, hypogonadism, learning disabilities, behavioural problems and psychiatric phenotypes with severe consequences and difficult management issues for patients, families and carers5.

Natural history of the syndrome

Recent progress in early diagnosis has shed light on the natural history of this syndrome and has revealed more complex nutritional phases than the 2 classically described emphasizing that excessive weight gain start before the increase of calorie intake6, 7. In addition eating behavior in individuals with PWS have been well characterized including a permanent obsession with food and excessive food searching, food storage, foraging and hoarding and deficit in satiation (longer meal duration and higher food intake to fill full) and satiety (early return of hunger after meal)8, 9. Their overeating leads to extremely morbid obesity with major consequences on morbidity and mortality.

Behavioural and psychiatric problems

It is now become apparent that eating behaviour troubles are part of general behavioural problems with learning difficulties, deficit of social cues and psychiatric disorders. Indeed individuals with PWS show pronounced emotional lability and a striking inability to control their emotions, which results in frequent temper outbursts. They also show poor social adjustment, with poor peer relationships, tendencies towards social withdrawal, and few attributions of feeling in social relationships compared with others with the same level of intellectual disabilities10.

Endocrine dysfunctions with a focus on growth hormone (GH)

PWS is a genetic paradigm for hypothalamic-pituitary endocrine dysfunction whose mechanism remains poorly understood. The prevalence of endocrine dysfunctions has been poorly studied and we reported it in a publication involving the first 142 children included in the French national data base11. Indeed 87 % of the children presented with a low peak of GH and almost 100% have low IGF-1 values. The label for GH treatment in children with PWS was obtained in 2000 both in Europe and USA with different criteria i) treatment of growth retardation in USA and ii) optimizing body composition and/or growth retardation in Europe. Early diagnosis performed in most countries around 1 month of life has opened the opportunity to start treatment early in life. In our experience in France, infants are now starting GH treatment at 6 months of life. This early treatment has been shown to increase head circumference12 and significantly ameliorated psychomotor development particularly in those infants with lower motor development13. We recently published that children with PWS displayed higher IGF-1 levels compared with children with GH deficiency suggesting a higher GH sensitivity14. In order to demonstrate this hypothesis, we implemented a prospective national clinical trial which ended last year and confirmed the same results (see figure 2). The pathophysiology of this observation is under investigation in our laboratory using cultured dermal fibroblasts. The implications of these data is that start of GH treatment must be done with escalating dose regarding the clinical tolerance (importance to search for snoring and ENT investigation prior to and on GH treatment) in order to avoid a rapid and excessive IGF-1 levels. In our review on the deaths observed in children with PWS treated or not-treated with GH, we proposed that the cluster of sudden deaths observed during the first months after the start of GH treatment may result from a too rapid and excessive increase of IGF-1 levels which may in turn stimulate adenoids enlargement thus worsening the possibility of obstructive apnoea15.

In adults GH treatment is not indicated in those patients re-evaluated at completion of growth and who are not GHD using the adolescent definition, a situation found in 50% of the patients. Interestingly we recently reported that the beneficial effect of GH treatment in childhood in body composition and metabolic modifications remains after the interruption of treatment in adults with PWS16 and is shown in Table 1.

Pathophysiology of PWS

Is there a unique gene responsible for the entire syndrome or complementary effects of some of the imprinted genes?

The chromosomal region involved in PWS is very complex encompassing imprinted and non-imprinted genes. Figure 1 shows a schematic representation of the region. To date there is no animal model that completely reproduces the human PWS phenotype and particularly the nutritional phases which start with deficit of suckling and failure to thrive and later on lead to early morbid obesity with hyperphagia and deficit of satiety. The hypothetical minimal paternal deletion region associated with PWS phenotype deduced from clinical cases with chromosomal translocations removes SNORD109A, the SNORD116 cluster (30 snoRNAs copies) and IPW17 and is shown in figure 1 by the dotted frame. Increasing evidence from animal and clinical data suggests that the loss of the paternally expressed snoRNA SNORD116 gene cluster plays a significant role in many of the features of the PWS phenotype18. Indeed, 3 human cases have been reported showing that the deletion of the SNORD116 gene cluster leads to PWS features including hyperphagia and obesity19-21. We also reported a new case with the shortest microdeletion (manuscript submitted) confirming the role of the critical minimal region for PWS.

Knockout mice with deletion of only SNORD116 genes showed the phenotype of hyperphagia without the occurrence of obesity on regular and high-fat diets17. However these data do not argue against a role for the SNORD115 which may act in a complementary manner to the SNORD116 in the pathophysiology of PWS.

SnoRNAs are a group of small RNAs ranging from 80 to 300 nucleotides in length that are located primarily in nucleoli and are required for the chemical modification and maturation of rRNAs. Animal studies have recently shown that the SNORD116 cluster is expressed in the hypothalamus22. In addition, very recently, it has been shown that the enhanced expression of SNORD116 in hypothalamic nuclei was observed at weaning and young adult stages, but was less obvious postnatally when expression was significantly more widespread suggesting that the expression of the SNORD116 is regulated developmentally and may play a regulatory role on hypothalamic development and function.

What are the role of oxytocin and ghrelin in the pathophysiology?

A significant decrease in the number and volume of OT-producing neurons of the hypothalamic paraventricular nucleus (PVN) was reported in PW patients23. Similarly, an alteration in the OT system was described in 2 PWS mouse models (Necdin KO and Magel2 KO)24, 25. Besides its well-known effects on uterine contractions and milk secretion, OT is known as an anorexigenic hormone which effects on feeding control and regulation of AgRP26-28 have been recently reported. Moreover individuals with PWS, unlike those with other known causes of obesity, have high circulating levels of the orexigenic hormone ghrelin, which may explain obesity29-31.Ghrelin acts on various feeding circuits such as hypothalamic centers driving energy intake and hunger, on reward/dopaminergic circuits32 and in the hind brain. We showed that hyperghrelinemia is present very early in life in infants with Prader-Willi syndrome and precedes obesity31. Of note, ghrelin is now considered as a major signal mediating perinatally acquired predispositions to adult diseases33.

Interestingly OT requires intact ghrelin to regulate satiety and conversely ghrelin modulates OT neurons, this crosstalk between these two hormones may be a key feature in the pathophysiology of this neurodevelopmental disease.

Very recently, we demonstrated in Magel 2 KO pups that a single OT injection before the first five hours of life completely rescued the newborn mice from early death by the recovery of normal suckling25. For all these reasons we performed a proof-of-concept study with OT administration to PWS babies to see if it is well tolerated and may change early phenotype. Preliminary data are currently analysed.

OT was recently identified as a key neuropeptide in the social interactions of numerous species by enhancing peer recognition and bonding behaviour. In humans, nasal OT administration improves emotion recognition and face processing in healthy and autistic individuals. We therefore hypothesised that the possible dysfunction in OT may also explain, at least in part, the poor social adjustment of patients with PWS and their inability to control emotions, which in turn might explain their unpredictable disruptive behaviours and frequent temper outbursts. We were able to demonstrate in a double-blind, randomised, placebo-controlled study of 24 adult patients with PWS, that the group that received a single intranasal administration of 24 UI of OT displayed significantly increased trust in others and decreased sadness tendencies, with less disruptive behaviour and a trend towards decreased food intake, than did patients who received placebo34. We also demonstrated in a brain imaging study using PET scan that the anterior cingulum, a brain region involved in the control of emotion, anticipation of movement, empathy, theory of mind and in feeding behaviour, is relatively poorly perfused in PWS, suggesting that OT pathways may be impaired35. It would be then of great interest to see if the administration of OT may change the brain perfusion pathway in PWS.

The mechanism of the abnormal OT pathway is not known in PWS. In the MAGEL2 KO PWS mouse model a restricted production of bioactive oxytocin was detected specifically in the hypothalamus of the mutant new-borns, although the prohormone was normally produced25.

Further animal and humans studies bench to bed studies are need to answer these questions. We are going to start two clinical trials to try to get more insights in the mechanism of actions of OT.

Conclusions

PWS is clearly a paradigm of endocrine dysfunction from a complex hypothalamic origin. It is a neurodevelopmental disorder whose pathophysiology is still unknown. The role of the SNORD116 gene seems to be crucial in the development of the hypothalamus and the early oxytocin-ghrelin dysfunction may be important to understand and possibly treat this very complex disease.

References

1. Cavaille J, Buiting K, Kiefmann M, Lalande M, Brannan CI, Horsthemke B, Bachellerie JP, Brosius J, Huttenhofer A. Identification of brain-specific and imprinted small nucleolar RNA genes exhibiting an unusual genomic organization. Proc Natl Acad Sci U S A 2000, 97(26):14311-14316.

2. de los Santos T, Schweizer J, Rees CA, Francke U. Small evolutionarily conserved RNA, resembling C/D box small nucleolar RNA, is transcribed from PWCR1, a novel imprinted gene in the Prader-Willi deletion region, which Is highly expressed in brain. Am J Hum Genet 2000, 67(5):1067-1082.

3. Cassidy SB, Driscoll DJ. Prader-Willi syndrome. Eur J Hum Genet 2009, 17(1):3-13.

4. Prader A: [Adrenogenital syndrome, adrenogenital salt deficiency syndrome and Cushing's syndrome in childhood.]. Schweiz Med Wochenschr 1956, 86(12):289-299.

5. Goldstone AP, Holland AJ, Hauffa BP, Hokken-Koelega AC, Tauber M: Recommendations for the diagnosis and management of Prader-Willi syndrome. J Clin Endocrinol Metab 2008, 93(11):4183-4197.

6. Miller JL, Lynn CH, Driscoll DC, Goldstone AP, Gold JA, Kimonis V, Dykens E, Butler MG, Shuster JJ, Driscoll DJ: Nutritional phases in Prader-Willi syndrome. Am J Med Genet A 2011, 155A(5):1040-1049.

7. Butler JV, Whittington JE, Holland AJ, McAllister CJ, Goldstone AP: The transition between the phenotypes of Prader-Willi syndrome during infancy and early childhood. Dev Med Child Neurol 2009.

8. Lindgren AC, Barkeling B, Hagg A, Ritzen EM, Marcus C, Rossner S: Eating behavior in Prader-Willi syndrome, normal weight, and obese control groups. J Pediatr 2000, 137(1):50-55.

9. McAllister CJ, Whittington JE, Holland AJ: Development of the eating behaviour in Prader-Willi Syndrome: advances in our understanding. Int J Obes (Lond) 2011, 35(2):188-197.

10. Koenig K, Klin A, Schultz R: Deficits in social attribution ability in Prader-Willi syndrome. J Autism Dev Disord 2004, 34(5):573-582.

11. Diene G, Mimoun E, Feigerlova E, Caula S, Molinas C, Grandjean H, Tauber M: Endocrine disorders in children with Prader-Willi syndrome--data from 142 children of the French database. Horm Res Paediatr 2010, 74(2):121-128.

12. Festen DA, de Lind van Wijngaarden R, van Eekelen M, Otten BJ, Wit JM, Duivenvoorden HJ, Hokken-Koelega AC: Randomized controlled GH trial: effects on anthropometry, body composition and body proportions in a large group of children with Prader-Willi syndrome. Clin Endocrinol (Oxf) 2008, 69(3):443-451.

13. Festen DA, Wevers M, de Weerd AW, van den Bossche RA, Duivenvoorden HJ, Otten BJ, Wit JM, Hokken-Koelega AC: Psychomotor development in infants with Prader-Willi syndrome and associations with sleep-related breathing disorders. Pediatr Res 2007, 62(2):221-224.

14. Feigerlova E, Diene G, Oliver I, Gennero I, Salles JP, Arnaud C, Tauber M: Elevated Insulin-Like Growth Factor-I Values in Children with Prader-Willi Syndrome Compared with Growth Hormone (GH) Deficiency Children over Two Years of GH Treatment. J Clin Endocrinol Metab 2010, 95(10):4600-4608.

15. Tauber M, Diene G, Molinas C, Hebert M: Review of 64 cases of death in children with Prader-Willi syndrome (PWS). Am J Med Genet A 2008, 146(7):881-887.

16. Coupaye M, Lorenzini F, Lloret-Linares C, Molinas C, Pinto G, Diene G, Mimoun E, Demeer G, Labrousse F, Jauregi J et al: Growth Hormone Therapy for Children and Adolescents with Prader-Willi Syndrome Is Associated with Improved Body Composition and Metabolic Status in Adulthood. J Clin Endocrinol Metab 2013.

17. Ding F, Prints Y, Dhar MS, Johnson DK, Garnacho-Montero C, Nicholls RD, Francke U: Lack of Pwcr1/MBII-85 snoRNA is critical for neonatal lethality in Prader-Willi syndrome mouse models. Mamm Genome 2005, 16(6):424-431.

18. Skryabin BV, Gubar LV, Seeger B, Pfeiffer J, Handel S, Robeck T, Karpova E, Rozhdestvensky TS, Brosius J: Deletion of the MBII-85 snoRNA gene cluster in mice results in postnatal growth retardation. PLoS Genet 2007, 3(12):e235.

19. Duker AL, Ballif BC, Bawle EV, Person RE, Mahadevan S, Alliman S, Thompson R, Traylor R, Bejjani BA, Shaffer LG et al: Paternally inherited microdeletion at 15q11.2 confirms a significant role for the SNORD116 C/D box snoRNA cluster in Prader-Willi syndrome. Eur J Hum Genet 2010, 18(11):1196-1201.

20. Sahoo T, del Gaudio D, German JR, Shinawi M, Peters SU, Person RE, Garnica A, Cheung SW, Beaudet AL: Prader-Willi phenotype caused by paternal deficiency for the HBII-85 C/D box small nucleolar RNA cluster. Nat Genet 2008, 40(6):719-721.

21. de Smith AJ, Purmann C, Walters RG, Ellis RJ, Holder SE, Van Haelst MM, Brady AF, Fairbrother UL, Dattani M, Keogh JM et al: A deletion of the HBII-85 class of small nucleolar RNAs (snoRNAs) is associated with hyperphagia, obesity and hypogonadism. Hum Mol Genet 2009, 18(17):3257-3265.

22. Zhang Q, Bouma GJ, McClellan K, Tobet S: Hypothalamic Expression of SnoRNA Snord116 is Consistent with a Link to the hyperphagia and obesity symptoms of Prader-Willi Syndrome. Int J Dev Neurosci 2012.

23. Swaab DF, Purba JS, Hofman MA: Alterations in the hypothalamic paraventricular nucleus and its oxytocin neurons (putative satiety cells) in Prader-Willi syndrome: a study of five cases. J Clin Endocrinol Metab 1995, 80(2):573-579.

24. Muscatelli F, Abrous DN, Massacrier A, Boccaccio I, Le Moal M, Cau P, Cremer H: Disruption of the mouse Necdin gene results in hypothalamic and behavioral alterations reminiscent of the human Prader-Willi syndrome. Hum Mol Genet 2000, 9(20):3101-3110.

25. Schaller F, Watrin F, Sturny R, Massacrier A, Szepetowski P, Muscatelli F: A single postnatal injection of oxytocin rescues the lethal feeding behaviour in mouse newborns deficient for the imprinted Magel2 gene. Hum Mol Genet 2010, 19(24):4895-4905.

26. Olszewski PK, Klockars A, Schioth HB, Levine AS: Oxytocin as feeding inhibitor: maintaining homeostasis in consummatory behavior. Pharmacol Biochem Behav 2010, 97(1):47-54.

27. Baskerville TA, Douglas AJ: Dopamine and oxytocin interactions underlying behaviors: potential contributions to behavioral disorders. CNS Neurosci Ther 2010, 16(3):e92-e123.

28. Atasoy D, Betley JN, Su HH, Sternson SM: Deconstruction of a neural circuit for hunger. Nature 2012, 488(7410):172-177.

29. Cummings DE, Clement K, Purnell JQ, Vaisse C, Foster KE, Frayo RS, Schwartz MW, Basdevant A, Weigle DS: Elevated plasma ghrelin levels in Prader Willi syndrome. Nat Med 2002, 8(7):643-644.

30. Tauber M, Conte Auriol F, Moulin P, Molinas C, Delagnes V, Salles JP: Hyperghrelinemia is a common feature of Prader-Willi syndrome and pituitary stalk interruption: a pathophysiological hypothesis. Horm Res 2004, 62(1):49-54.

31. Feigerlova E, Diene G, Conte-Auriol F, Molinas C, Gennero I, Salles JP, Arnaud C, Tauber M: Hyperghrelinemia precedes obesity in Prader-Willi syndrome. J Clin Endocrinol Metab 2008, 93(7):2800-2805.

32. Abizaid A: Ghrelin and Dopamine: New Insights on the Peripheral Regulation of Appetite. J Neuroendocrinol 2009.

33. Steculorum SM, Bouret SG: Developmental effects of ghrelin. Peptides 2011, 32(11):2362-2366.

34. Tauber M, Mantoulan C, Copet P, Jauregui J, Demeer G, Diene G, Roge B, Laurier V, Ehlinger V, Arnaud C et al: Oxytocin may be useful to increase trust in others and decrease disruptive behaviours in patients with Prader-Willi syndrome: a randomised placebo-controlled trial in 24 patients. Orphanet J Rare Dis 2011, 6(1):47.

35. Mantoulan C, Payoux P, Diene G, Glattard M, Roge B, Molinas C, Sevely A, Zilbovicius M, Celsis P, Tauber M: PET scan perfusion imaging in the Prader-Willi syndrome: new insights into the psychiatric and social disturbances. J Cereb Blood Flow Metab 2011, 31(1):275-282.



Comments
Name*: Surname*:
E-mail*:
Hospital*:
Address:
C.P.: Country:
Comments*:
(450 Words)
Security code*:
* Required Fields
Send
Send Send Send
Send