Genetics of Restless Legs Syndrome (RLS)


Chr

Region (hg19)

Peak marker

Size

Max LOD

Model

Replication

Reference

(Mb)

status

12q12–21

94176800-104264737

D12S1044

10.09

3.59

Auto rec

+

[47]
  
94176800
  
Pseudodominant
  
14q13–21

34459194-47133518

D14S288

12.68

3.23

Auto dom

+

[7]
  
43171519

(1.3)
    
9p24–22

516800-19680020

D9S286

19.18

3.9

Auto dom

+

[17]
  
8043378

(16.60)

3.22

Model-free
  
9p21

22340644-ca. 3225000

D9S147E

9.9

3.6

Auto dom


[48]
  
31044744
     
2q33

197566845-208825061

D2S325

11.26

5.5

Auto dom

(+)

[49]
  
207978881

(0.045)
 
Reduced pen (0.7)
  
20p13

82754-5315186

D20S849

5.2

3.86

Auto dom

(+)

[50]
  
5142034

(4.5)
 
Reduced pen (0.7)
  
16p12

22758479-23312075

Several

1.18

3.5

Auto dom

(+)

[51]
     
Reduced pen (0.8)
  
19p13

0-2518075

D19S878

2.5

3.59

Auto dom


[52]
  
2310697
     

For the size of the linkage region, first the originally reported size is given and, secondly, if pertinent, the best current approximation after additional fine-mapping and replication studies + replicated with significant LOD score, (+) replicated with LOD score suggestive of linkage, − not replicated



Next to the seven linkage regions that were found to have genome-wide significant LOD scores above the conventional threshold of 3.3, a total of 21 linkage regions on 14 chromosomes have also been reported with LOD scores ranging between 1.00 and 2.61 [6, 17, 49, 5456]. For a more in-depth discussion of the RLS linkage loci, please cf. [60].

Despite this plethora of evidence supporting the existence of single genetic variants of strong effect that play a role in familial RLS, it is also important to realize that most of these loci where only found in single or—in the best case—a few families leaving many more families where the underlying genetic factors remain obscure.

The recurrent finding in the family studies was that of genetic heterogeneity and complexity in RLS. Interestingly, a large German RLS family in whom linkage analysis argued for the existence of two independent linkage loci on chromosomes 4 and 17 also exists, possibly reflecting an oligogenic mode of inheritance in this family (Winkelmann J et al., unpublished observation). Also, replication of the above loci has proven very difficult [4952, 57, 59, 61], and the maximum LOD scores found fall short of the maximum attainable scores projected by the pedigree structure. Overall, linkage studies in RLS have failed to the extent that no underlying genetic factor could be identified for any of the above loci, even when the most up-to-date technologies such as targeted next-generation sequencing were employed to resolve the regions [62, 63].



Candidate Gene Association Studies


Over the past two decades, several candidate gene studies have been performed in RLS. Because dopaminergic drugs are one of the mainstays of treatment, an involvement of dopaminergic pathways in the pathophysiology of RLS has long been projected. Accordingly, in one of the first association studies in RLS, SNPs in eight genes playing a role in dopaminergic neurotransmission were examined for possible association with the RLS phenotype in 92 cases and 182 controls. However, no association of any of the tested genetic variants with the RLS phenotype was observed [48]. Next to the dopaminergic system, the iron metabolism has long been implicated in RLS. However, no SNP located within any of 111 iron-related genes ± 4 Mb showed a replicable association with the RLS phenotype in three case/control samples totaling to 2,425 cases and 3,285 general population controls [64]. In summary, candidate association studies—in parallel to the linkage analyses—have been unsuccessful in identifying genetic factors involved in RLS.


Genome-Wide Association Studies


To date, three genome-wide association studies (GWAS) have been performed for RLS and one for RLS and periodic limb movements in sleep (PLMS) (Tables 15.2 and 15.3). The PLMS GWAS was carried out under the deCODE Genetics umbrella and included 306 cases with RLS and PLMS and 15,664 controls from Iceland in the genome-wide phase. An intronic variant in BTBD9 within a linkage disequilibrium (LD) block on chromosome 6p21.2 showed genome-wide significant association (p nominal = 2 × 10−9, OR = 1.8) and was replicated in a second Icelandic and a US-American sample (combined sample (617 cases/17,528 controls): p nominal = 3 × 10−14, OR = 1.7). Moreover, the major allele of the lead SNP (rs3923809) was also associated with an increase in PLMS of approximately 3/h as well as a 13 % decrease in serum iron per allele when tested in 965 individuals [69].


Table 15.2
Study characteristics of GWAS performed for RLS














































































































































































Genome-wide sample (cases/controls)

Origin

SNP array

Replication sample(s) (cases/controls)

Origin

Lead SNPs

Candidate gene

Replication status

Reference

306/15,633

Iceland

Human Hap300 and Hap300-duo+

123/1,233

Iceland

rs3923809

BTBD9

+

[65]
   
Bead, Illumina

188/662

USA
       

401/1,644

Germany

500 K, Affymetrix

903/891

Germany

rs2300478

MEIS1

+

[66]
     
255/287

Canada

rs9296249

BTBD9

+
 
         
rs1026732

MAP2K5/SKOR1

+
 

628/1,644

Germany

500 K, Affymetrix (n = 401 + 1,644)

1,271/1,901

Germany

rs4626664

PTPRD

+

[67]
   
Genome-wide human SNP 5.0

279/368

Czech Republic

rs1975197

PTPRD

+
 
   
Array, Affymetrix (n = 227)

285/842

Canada
       

954/1,814

Germany and Austria

Genome-wide human SNP 5.0

1,236/1,471

Germany and Austria

rs2300478

MEIS1

+

[68]
   
Array, Affymetrix (cases)

1,104/1,065

Germany and Austria

rs9357271

BTBD9

+
 
   
Genome-wide human SNP 6.0

351/597

Czech Republic

rs1975197

PTPRD

+
 
   
Array, Affymetrix (controls)

141/360

Finland

rs12593813

MAP2K5/SKOR1

+
 
     
182/768

France

rs6747972

Intergenic

 
     
285/285

Canada

rs3104767

TOX3/BC034767

 
     
556/1,208

USA
       


For a more detailed description of results, also see Table 15.3

+ statistically significant in independent population, − not (yet) replicated



Table 15.3
RLS GWAS loci [68]












































































Locus

Chr

LD block (Mb)

Lead SNP

Risk allele

Risk allele freq (cases/controls)

Pjoint

Odds ratio (95 % CI)

MEIS1

2

66.57–66.64

rs2300478

G

0.35/0.24

3.40 × 10−49

1.68 (1.57–1.81)

MAP2K5/SKOR1

15

65.25–65.94

rs12593813

G

0.75/0.68

1.37 × 10−22

1.41 (1.32–1.52)

BTBD9

6

37.82–38.79

rs9357271

T

0.82/0.76

7.75 × 10−22

1.47 (1.35–1.47)

TOX3/BC034767

16

51.07–51.21

rs3104767

G

0.65/0.58

9.40 × 10−19

1.35 (1.27–1.43)

Intergenic

2

67.88–68.00

rs6747972

A

0.47/0.44

9.03 × 10−11

1.23 (1.16–1.31)

PTPRD

9

8.80–8.88

rs1975197

A

0.19/0.16

3.49 × 10−10

1.29 (1.19–1.40)

Simultaneously, the first RLS GWAS, which included 401 German cases and 1,644 general population controls in the genome-wide phase as well as 903 German cases and 891 controls and 255 Canadian cases and 287 controls in the replication samples, also showed association to the same SNP and the same 115 kb LD block on chromosome 6p containing intron 5 of BTBD9. However, on chromosome 2p, an association signal located within a 32 kb LD block containing intron 8 and exon 9 of MEIS1 was more strongly associated with the RLS phenotype in all individuals included in the genome-wide phase as well as the combined sample (rs2300478, p nominal = 3.41 × 10−28, OR = 1.74). Fine-mapping and haplotype analysis in the German replication sample revealed a haplotype associated with RLS with an increased OR of up to 2.75 (95 % CI: 2.23–3.41) (p nominal = 5.87 × 10−20, frequency in cases 0.231 vs. 0.102 in controls). A third association signal of genome-wide significance was located within a 48 kb locus on chromosome 15q spanning the 3′ end of MAP2K5 as well as SKOR1 (formerly called LBXCOR1) (combined p nominal = 6.09 × 10−17) [70].

A GWAS-based analysis of the RLS-3 locus encompassing 31 Mb on chromosome 9p23–24 in 628 cases and 1,644 general population controls revealed two independent (r 2 = 0) SNPs within two independent LD blocks in intron 8 (rs4626664) and intron 10 (rs1975197) of the 5′ untranslated region (UTR) of PTPRD which were replicated in a sample of 1,835 cases and 3,111 controls from Germany, the Czech Republic, and Canada. When combined with the genome-wide discovery sample, both SNPs surpassed thresholds for genome-wide significance (rs4626664: p nominal = 5.91 × 10−10, OR = 1.44; rs1975197: p nominal = 5.81 × 10−9, OR = 1.31). No variants in any of the 35 coding and 10 noncoding exons of PTPRD could be identified in nine affected individuals from an RLS-3-linked family, and the common variants in PTPRD only explain a minor portion of the original RLS-3 linkage signal [71].

An increased sample size of 922 cases and 1,526 controls in the genome-wide phase and a multinational replication sample of 3,935 cases and 5,754 controls of European descent revealed two new loci of genome-wide significance: an intergenic region on chromosome 2p14 approximately 1.3 Mb downstream of MEIS1 (rs6747972, p nominal = 9.03 × 10−11, OR = 1.23) as well as a locus on chromosome 16q12.1 encompassing an LD block of 140 kb containing both the 5′-end of TOX3 and the noncoding RNA BC034767 (rs3104767, p nominal = 9.4 × 10−19, OR = 1.35) [72].

While the two most recent loci still await replication in independent studies, the first four loci have been replicated in independent case/control samples [6567]. In individuals with secondary RLS due to end-stage renal insufficiency, the lead SNPs in BTBD9 were also associated with increased susceptibility to RLS in a combined German/Greek sample of 341 dialysis patients with RLS and 836 without RLS, while MEIS1 lead SNPs showed significant association only in the German sample [68]. A single association study in a non-European population corroborated the link between the intronic variants in BTBD9 (rs3923809 and rs9296249) and RLS in the Korean population [73]. To date, no GWAS in non-European populations or considering specific endophenotypes have been performed for RLS.

Single SNPs at the RLS-associated loci identified by the above studies bear effect sizes between 1.22 and 1.77 and risk allele frequencies between 0.19 and 0.82 (Table 15.3) [72]. Although the conferred risk is large when compared to common variants associated with other complex traits, when taken together, the most significant SNPs at these loci only explain about 6.8 % of the heritability of RLS [72]. Arguing for—most likely—both the existence of additional independent RLS-related variants within these loci as well as a number of additional loci. It is also important to realize that the lead SNPs may not be identical to the causal genetic variants at these loci, which makes functional follow-up studies indispensible in order to utilize genetic variants to inform the pathophysiology of RLS.


Following Up on GWAS


The link between the most likely candidate genes at the associated GWAS loci and RLS is not readily apparent. Functionally, most of the candidate genes highlighted by the GWAS are not well characterized. Transcriptional regulation especially in developmental processes in the nervous system seems to be the largest common denominator.


MEIS1


The transcription factor MEIS1 belongs to the family of highly conserved TALE homeobox genes and interacts with PBX and HOX proteins to increase the affinity and specificity of HOX proteins [74] as well as CREB1 [75] in DNA binding. In Xenopus laevis, meis1 is known to be involved in neural crest development [76]. Murine Meis1 is essential for proximo-distal limb patterning [77] and plays a role in the Hox transcriptional regulatory network that specifies spinal motor neuron pool identity and connectivity [78]. In the CNS of the adult mouse, it is known to be expressed in cerebellar granule cells, the forebrain, and the substantia nigra. While MEIS1 was initially identified in the context of acute myeloid leukemia [79, 80], in recent years, a role in murine heart development has also been recognized [81], and SNPs in intron 8 (but in weak LD with the known RLS SNPs) play a role in determining atrioventricular conduction velocity as reflected by the length of the PR interval of the electrocardiogram in both Europeans and African-Americans [82, 83]. Meis1 −/− mice develop ocular and vascular defects, fail to produce megakaryocytes, and display extensive hemorrhaging. They also die by embryonic day 14.5 [84]. Meis1 −/+ mice, however, survive into adulthood and exhibit hyperactivity reminiscent of the human RLS phenotype [85].

A second independent association signal is located in an intergenic region approximately 1.3 Mb downstream of MEIS1 and potentially possesses long-range regulatory function with MEIS1 and ETAA1 as potential target genes [72].

Several rare non-synonymous variants in MEIS1 have been identified in RLS patients. However, coding variants in MEIS1 are very rare in general (13 out of approximately 4,250 individuals with a non-synonymous variant in the NHLBI-ESP exomes [86]), possibly owing to the fact that MEIS1 represents one of the most highly conserved genes in the human genome, and, therefore, remain ambiguous with regard to possible causality of the RLS phenotype [8790]. On the whole, however, non-synonymous variants with MAF <0.1 % were found significantly more frequently in individuals with RLS compared to the general population [90]. Functional annotation using an in vivo complementation assay in zebra fish further revealed that variants harboring a loss of MEIS1 function were significantly enriched in individuals with RLS [90]. The same study also identified a low-frequency variant in the 3′ untranslated region (UTR) of MEIS1 (rs11693221; MAFcases = 13.55 % vs. MAFcontrols = 3.58 %; p = 1.27 × 10−89, OR = 4.42) associated with RLS [90]. Yet, at present, it cannot be determined whether this or any other variant tagged by rs11693221 represents the causal factor underlying this association signal. What does become clear, though, is the fact that, at the MEIS1 locus, an allelic series of genetic variants of different frequencies and different effect sizes contributes to the genetic framework of RLS.

Since the publication of the first GWAS, which identified common variants in MEIS1 as susceptibility factors for RLS, three studies have been reported which examine the functional differences brought about by the RLS-associated intronic variants. In the first, a significant decrease in MEIS1 mRNA and protein expression was found in lymphoblastoid cell lines and brain tissue (pons and thalamus) from homozygous carriers of the risk haplotype when compared to homozygous carriers of the non-risk haplotype [89]. In a second study, knockdown of the MEIS1 orthologue unc-62 by RNA interference in Caenorhabditis elegans was related to increased ferritin expression and an extended life span. In thalamus but not in pons samples of RLS patients homozygous for the MEIS1 risk haplotype (n = 9), ferritin light and heavy chains as well as divalent metal transporter 1 (DMT1) mRNA and protein expression were significantly increased when compared to RLS patients carrying the protective haplotype (n = 7). Several other key players in the iron metabolism such as transferrin, the transferrin receptors 1 and 2, aconitase 1, iron-responsive element binding protein 2, ceruloplasmin, hepcidin, and ferroportin were unchanged [91]. The authors argue that these data are in support of a disruption of physiological iron transport into the brain and—in conjunction with the also observed decrease of MEIS1 expression in in vitro cell models of iron deprivation—provide a functional link between the RLS gene MEIS1 and the iron metabolism, which is believed to play a role in RLS pathogenesis [91]. In the third study, it was shown that the risk allele of the best-associated SNP (rs12469063) in MEIS1 from the GWAS reduces enhancer activity in the Meis1 expression domain of the ganglionic eminences, which constitute the primordial basal ganglia, in mouse embryos at E12.5 [85]. In vitro studies suggest that CREB1 binds more strongly to this enhancer when the risk allele is present [85].


MAP2K5/SKOR1


Another locus encompasses both mitogen-activated protein kinase MAP2K5 and transcriptional corepressor, SKOR1. MAPK pathways are highly conserved among different species and are activated in response to signals that mediate the transduction of extracellular signals to the cytoplasmic nuclear effectors [92]. More specifically, MAP2K5 phosphorylates and activates ERK5 in response to oxidative stress, hyperosmolarity, and growth factors. It is expressed in the heart and skeletal muscle and critical in muscle cell differentiation [92]. Interestingly, the MAP2K5/ERK5 pathway has also been implicated in neuroprotection of dopaminergic neurons [93]. Not much is known about the physiologic function of SKOR1. It acts as a transcriptional corepressor of homeobox gene LBX1, which has been recognized as a factor in the development of pain and touch relay via sensory pathways in the dorsal horn of the spinal cord [94]. The genomic locus comprising MAP2K5 and SKOR1 was shown to harbor nine blood-based cis-eSNPs, that is, common variants that are located within ± 500 kb of the lead SNP, which alter blood-based gene expression [95]. None of these affected the expression of SKOR1, and only two affected the expression of MAP2K5, while seven altered expression of CALML4, thus highlighting the possibility that other genes in the vicinity of the current candidate genes at a given locus could also play a role in RLS pathophysiology [95].

Only gold members can continue reading. Log In or Register to continue

Stay updated, free articles. Join our Telegram channel

Jun 14, 2017 | Posted by in NEUROLOGY | Comments Off on Genetics of Restless Legs Syndrome (RLS)

Full access? Get Clinical Tree

Get Clinical Tree app for offline access