touchNEUROLOGY touchNEUROLOGY
Movement Disorders
Read Time: 4 mins

Epidemiology and Pathophysiology of Restless Legs Syndrome

Copy Link
Published Online: Jun 4th 2011
Authors: David B Sommer, Mark Stacy
Quick Links:
Article
Article Information
Article:

In 1945 Karl Ekbom coined the term ‘restless legs syndrome’ (RLS), which is still sometimes referred to as Ekbom’s syndrome. RLS is a common sensorimotor condition characterized by predominantly nocturnal dysesthesias relieved by limb movement. RLS is not a new disorder, having been documented in the medical literature for over half a century.1 However, in recent years RLS has been receiving increased attention following approval, in May 2005, by the US Food and Drug Administration (FDA) of the first agent indicated for the treatment of the condition. Also in 2005 the epidemiology and pathophysiology of RLS were reviewed by the RLS Foundation in the RLS Medical Bulletin2 (available at www.rls.org).

This article summarizes the current understanding in these fields and highlights the advances made since 2005. The clinical diagnosis and management of RLS are beyond the scope of this article and have been reviewed elsewhere.2–4

Epidemiology

Definition

Epidemiological characterization necessitates a clear definition of disease. The first formal criteria for the diagnosis of RLS were published by the International RLS Study Group (IRLSSG) in 1995. These criteria were updated in 2002 (see Table 1) and represent the current ‘gold standard’ for diagnosis of RLS.5 The presence of RLS correlates with periodic leg movements of sleep (PLMS) on polysomnography and periodic leg movements of wakefulness (PLMW) on the suggested immobilization test,6 but the diagnosis remains purely clinical. Hence, epidemiological studies rely on subjective reports. The IRLSSG criteria do not specify a cutoff of symptom frequency or severity, although there are many people in whom RLS symptoms are mild and do not impair quality of life (QOL). There has not been a universally accepted definition of clinically significant RLS, although the International RLS Study Group Rating Scale (IRLS) is currently the best validated measure of severity, with a score greater than 10 points (of 40 possible total points) indicating moderate to severe disease.7 Future research will likely employ published validated instruments to define clinically significant RLS.

Prevalence

Ekbom estimated the prevalence of RLS to be 5% in the general population. Subsequent survey studies have estimated the population prevalence of the disorder to be 1–29%.8 The RLS Epidemiology, Symptoms, and Treatment (REST) trial is the largest survey study to date of RLS prevalence. Questionnaires were completed by 23,052 patients presenting to primary care practices in the US and four Western European countries during a two-week period. Any degree of RLS symptoms was experienced by 11.9% of respondents, 9.6% had at least weekly symptoms, and 551 (2.4% of total respondents, 3.4% of respondents with complete data) had at least twice-weekly symptoms and endorsed a negative impact on QOL.9 Recently, similar figures have been reported in the Irish10 and Spanish11 primary care populations.

Prevalence estimates derived from primary care patients may overestimate general population prevalence because older and sicker persons are over-represented. However, a 1996 random digit dialing survey of adult Kentucky residents estimated a 10% prevalence of RLS in the adult population,12 which is similar to that found in the REST study.

International variations in the prevalence of RLS and the genetic and sociocultural differences that may account for this variation remain poorly understood. Most prevalence studies of RLS have been conducted on Caucasian populations. Prevalence estimates in East Asian studies included 0.1% in a Singaporean survey13 and 12.1% in a large Korean survey.14 (The latter study used a single question—“Have you ever experienced symptoms of RLS?”—so the prevalence of clinically significant RLS is probably much lower.) A Japanese survey found RLS symptoms in 5% of respondents but clinically significant RLS in 1.5%.15 Recently, Rangarajan et al. reported the results of a door-to-door survey of 1,266 individuals in Bangalore, India. They found a prevalence of RLS of 2.1% using the IRLSSG criteria, and 1.2% reported RLS that disrupted sleep.16 The prevalence of RLS in African populations has not been studied.

In summary, symptoms of RLS are quite common, and the population prevalence of RLS severe enough to cause impairment in QOL and warrant medical treatment is probably about 2–3%. This makes clinically significant RLS two to six times as prevalent as epilepsy in developed countries (estimated prevalence of 0.5–1%).17

Demographic Distribution

Many epidemiological studies have found some degree of female predominance among RLS sufferers, but reported female–male ratios vary from one to three. The female–male ratio among subjects with RLS in REST was 2.1, but this ratio did not differ significantly from the female–male ratio of all survey respondents.9 In the Kentucky telephonebased survey, the odds ratio (OR) for RLS in females was 1.33.12 In contrast, Lee et al. found an OR of 2.64 (95% confidence interval (CI) 1.24–7.74) in an American community sample,18 and Berger et al. found an OR of 1.98 (95% CI 1.25–3.13) in a German population.19 The weight of evidence would suggest that about twice as many women as men suffer from RLS, but the physiological basis for this difference and the degree to which this figure may be exaggerated by reporting bias are not known.

Population-based studies have found an increasing prevalence of RLS with age. Phillips et al. found a 3% prevalence of RLS symptoms in persons <30 years of age, about 10% in persons 30–79 years of age, and 19% in persons 80 years of age and older.12 Berger et al. found a steady increase from 4% in the third decade to 15% in the eighth decade.19 The IRLSSG has defined criteria for the diagnosis of RLS in children,5 and Kotagal and Silber retrospectively diagnosed RLS in 32 of 538 patients presenting to a pediatric sleep disorders clinic.20 However, populationbased studies assessing the prevalence of RLS in the pediatric population are lacking. As many adult RLS patients report an onset of symptoms in childhood,21 there is reason to believe that substantial numbers of children and adolescents are affected.

Race

Racial differences in RLS have not been completely explored. Anecdotally, African-Americans are under-represented in RLS specialty clinics.8,18 However, an analysis of a community-based sample of 1,071 adults in Baltimore found the prevalence of clinically significant RLS to be 4.7% in African-Americans and 3.8% in Caucasians. There was no statistically significant difference in the prevalence between groups before or after adjustment for other demographic variables.18 Thus, the relative absence of African-Americans in specialty clinics likely has much more to due with access to and utilization of healthcare than with biological differences.

Associated Conditions and Exposures

RLS is encountered with greater than expected frequency in several general medical conditions, and the RLS occurring in these conditions is often referred to as secondary RLS. The most well-established secondary causes of RLS are renal failure, iron deficiency, and pregnancy. RLS is common, with 20% prevalence, in the hemodialysis population, tends to be more severe than primary RLS, and is a predictor of mortality in this population.22 Iron deficiency has long been thought to be associated with RLS.23 O’Keffe et al. demonstrated significantly lower levels of ferritin (a sensitive indicator of body iron stores) in RLS patients than in controls,24 and this finding has been reproduced in several subsequent studies.8 Ekbom first noted an increased prevalence of RLS in pregnancy, and Manconi et al. have applied IRLSSG criteria to 200 consecutively enrolled pregnant women and found prevalences of 27 and 12% for symptoms at least twice monthly and at least three times weekly, respectively.25

Less well-characterized associations have been found with myelopathy, diabetes mellitus, neuropathy, and Parkinson’s disease. RLS has anecdotally been seen in spinal lesions, and Hogl et al. prospectively demonstrated incidence of transient RLS following spinal anesthesia.26 Although their findings failed to reach statistical significance (p=0.1), Skomro et al. found double the prevalence of RLS among type II diabetics compared with controls.27

28 Although a higher than expected prevalence of neuropathy is found in RLS populations, a higher than expected prevalence of RLS has not been seen in populations presenting with neuropathy.8 Gomez-Esteban et al. recently found RLS in 25 of 114 (21.9%) Parkinson’s disease patients—a higher than expected rate—but there was no significant additional sleep disruption or reduction in QOL associated with RLS.29

Dopamine antagonists (antipsychotics and anti-emetics) have been shown to cause motor restlessness and increased PLMS, and may cause RLS.2 Although antidepressant medications have been anecdotally reported to cause or exacerbate RLS, a review of 200 patients presenting to a sleep clinic (45% with RLS and 38% on antidepressants) found no association between RLS and antidepressant use.30

There is a higher prevalence of periodic limb movement disorder and RLS among children with attention deficit hyperactivity disorder (ADHD) versus controls,31 and a small non-controlled trial of dopaminergic treatment in children with RLS and ADHD demonstrated significant improvement in both conditions, suggesting an etiological link.32 The association between RLS and ADHD is deserving of, and likely to receive, more study in the near future.

The association between physical inactivity and RLS is another area deserving further study. Phillips et al. reported an OR for RLS of 3.32 (95% CI 1.67–6.30) for persons exercising for less than three hours per month,12 and a single small randomized trial of prescribed exercise for RLS demonstrated significant improvement in RLS severity.33 While inconclusive, these findings suggest that physical inactivity may be a modifiable risk factor for RLS.

Medical Attention Seeking and Physician Awareness

In the REST population, 65% of respondents with clinically significant RLS (at least twice-weekly symptoms) reported seeking medical attention for their symptoms within the preceding 12 months. Of this 65%, only 25% received a diagnosis of RLS.9 In the more recent Irish primary care study, only 32.4% of patients with moderate to severe RLS reported previously seeking medical attention for the symptoms, and only four of 24 patients (16.7%) who sought medical attention had received a diagnosis of RLS despite the majority of these patients reporting disruption of sleep and impairment in QOL.10 Clearly, improved patient and physician awareness of RLS would increase the rates of diagnosis. Currently, the pharmaceutical industry is investing heavily in promoting both patient and physician awareness of RLS.34 There has been exponential growth in the number of scientific articles about RLS published in recent years (see Figure 1). Even with this growth, RLS is relatively infrequently discussed in the medical literature. In the first eight months of 2007, 94 articles were indexed in Medline with the major medical subject heading (MeSH) of RLS compared with 1,828 articles indexed during the same time period with the major MeSH of epilepsy.

Genetics

The proportion of RLS patients with an affected first-degree relative has consistently been found to be higher than would be expected by chance alone, suggesting an inherited susceptibility to disease. Among a nonrandom sample of RLS patients, Walters et al. found that 81% of patients with onset of symptoms before 20 years of age had a positive family history compared with 58% of patients with later-onset symptoms.32 The results of a segregation analysis of 238 RLS patients, 537 first-degree relatives, and 133 spouses revealed strong evidence for an autosomal dominant pattern of inheritance in patients with early-onset RLS (symptoms prior to 30 years of age), but no similar indirect evidence for a single gene responsible for later-onset disease.35

Before 2006, three susceptibility loci had been identified on chromosomes 12q (RLS1), 14q (RLS2), and 9p (RLS3).36 Three more susceptibility loci, on chromosomes 20p (RLS4), 2q (RLS5), and 6p (RLS6), have subsequently been identified.37 Despite the strong evidence for a genetic component to RLS, attempts to identify culpable genes have so far been disappointing.38 Given that a subset of RLS patients probably have disease associated with single gene mutations, the potential discovery of disease-causing genes holds promise for better elucidating the molecular mechanisms of RLS.

Pathophysiology

The basic pathophysiological mechanisms of RLS remain poorly understood. Pharmacological, physiological, pathological, and neuroimaging studies have implicated dopaminergic systems, brain iron metabolism, and the endogenous opioid system. However, deciphering which observed changes are causative and which are secondary phenomena remains elusive.39

Dopaminergic Systems

Since the serendipitous finding by Akpinar in 1982 that relatively small doses of levodopa provided relief in RLS, it has been shown conclusively that levodopa and the dopamine agonists improve the symptoms of RLS.2 This has led to the conclusion that dopaminergic systems must play some

role in the pathophysiology of RLS. The fact that co-administration of domperidone (a peripheral dopamine antagonist) does not decrease the efficacy of pergolide (a dopamine agonist) in treating RLS lends support to the hypothesis that central rather than peripheral dopaminergic systems are responsible for RLS.40

Multiple single positron emission tomography (SPECT) studies have failed to find a difference between RLS patients and controls in dopamine transporter (DAT) binding in the striatum.2 However, in a positron emission tomography (PET) study of 16 RLS patients and 16 matched controls, Cervenka et al. found significantly reduced D2-binding potential in the thalamus and anterior cingulate cortex of RLS patients. They hypothesized that since these regions are involved in central sensory processing, impaired central dopaminergic transmission may account for the sensory symptoms of RLS.41

The diencephalospinal dopaminergic tract originating from the A11-A14 nuclei has received much attention as the potential anatomical site of dopaminergic dysfunction in RLS. This system projects to the limbic system, sensory cortex, and spinal cord.8 The proximity to circadian control centers in the hypothalamus may offer some explanation for the circadian pattern of symptoms observed in RLS.4 A rat model for the study of this system has been developed.42

Paulus and Trenkwalder have published a detailed hypothesis of how downregulation of D2 receptors (and overstimulation of D1 receptors with increasing dopaminergic therapy) may lead to augmentation and the delayed clinical worsening of symptoms seen in 60% of patients treated with levodopa.43

Brain Iron Metabolism

Iron deficiency has long been associated with RLS, and iron therapy has been effective in some cases.23 Earley at al. demonstrated markedly reduced cerebrospinal fluid ferritin levels and elevated transferrin levels in RLS patients compared with controls, even though there was no difference in serum ferritin levels.44 Thus, brain iron metabolism may be implicated in RLS even in the absence of total body iron deficiency.

A magnetic resonance imaging (MRI) study in five RLS patients and five controls demonstrated reduced iron concentration in the substantia nigra and putamena of RLS patients,45 and a subsequent larger study found decreased regional brain iron concentration in early-onset RLS patients relative to controls (but not late-onset RLS compared with controls).46 A pathological examination of the substantia nigra of RLS patients provided further evidence for regional iron deficiency. Surprisingly, transferrin receptor quantity was decreased (the opposite of what is seen in an irondeficiency state) and iron-regulatory protein 1 (IRP1) was deficient, leading to the hypothesis that a primary protein dysfunction in the brain iron-regulation system may be responsible for RLS.47

Relationship Between Brain Iron and Dopamine

Multiple potential biochemical links exist between the dopaminergic system and brain iron metabolism. Iron is a co-factor for tyrosine hydroxylase, the rate-limiting enzyme in the synthesis of dopamine.48 The dopamine transporter (DAT) is also impaired during iron deficiency.39 Further research will shed light on a unifying theory of RLS pathogenesis.

Opioid Systems

Successful amelioration of RLS symptoms has been demonstrated with oral opioids.4 A PET study assessing opioid binding found no group differences between 15 RLS patients and 12 controls, but did find significant negative correlation with RLS severity and 11C-diprenorphine uptake in the caudate nuclei, medial thalami, and anterior cingulated gyri.49 The pathophysiological significance of these findings remains unclear.39

Conclusion

RLS is a common disorder and likely causes significant morbidity in about 2% of the population. Women are more commonly affected, and the prevalence increases with age. The etiology and pathophysiology of RLS remain poorly characterized, but there has been a surge of interest in the disorder in recent years, and continued research will likely refine our growing understanding of this disorder. Enhanced awareness among patients and clinicians will likely continue to make RLS a common topic in clinical practice.

Article Information:
Received

2007-01-01T00:00:00

References

1. Tatlow WFT, Restless legs, CMAJ, 1954;71:191–2.
2. Rye DB, Adler CH, Allen RP, et al., RLS Medical Bulletin 2005, Rochester, Minnesota: Restless Legs Syndrome Foundation.
3. Gamaldo CE, Earley CJ, Restless legs syndrome: a clinical update, Chest, 2006;130:1596–1604.
4. Trenkwalder C, Paulus W,Walters AS, The restless legs syndrome, Lancet Neurol, 2005;4:465–75.
5. Allen RP, Picchietti D, Hening WA, et al., Restless legs syndrome: diagnostic criteria, special considerations, and epidemiology. A report from the restless legs syndrome diagnosis and epidemiology workshop at the National Institutes of Health, Sleep Med, 2003;4:101–19.
6. Garcia-Borreguero D, Larrosa O, de la Llave Y, et al., Correlation between rating scales and sleep laboratory measurements in restless legs syndrome, Sleep Med, 2004;5:561–5.
7. Kohnen R, Allen RP, Benes H, et al., Assessment of restless legs syndrome. Methodological approaches for use in practice and clinical trials, Mov Disord, 2007;in press, electronically published May 29, 2007.
8. Ondo WG, Restless Legs Syndrome. In: Jankovic J, Tolosa E (eds), Parkinson’s Disease and Movement Disorders, 5th edition, Philidelphia: Lippincott, Williams, and Wilkins, 2007;409–20.
9. Hening W,Walters AS, Allen RP, et al., Impact, diagnosis and treatment of restless legs syndrome (RLS) in a primary care opulation: the REST (RLS epidemiology, symptoms, and treatment) primary care study, Sleep Med, 2004;5:237–46.
10. O’Keeffe ST, Egan D, Myers A, Redmond S, The frequency and impact of restless legs syndrome in primary care, Ir Med J, 2007;100:539–42.
11. Pérez-Romero T, Comas-Fuentes A, Debán-Fernández M, et al., Prevalence of restless legs syndrome at the office in primary health care, Rev Neurol, 2007;44:647–51.
12. Phillips B, Young T, Finn L, et al., Epidemiology of restless legs symptoms in adults, Arch Intern Med, 2000;160:2137–41.
13. Tan EK, Seah A, See SJ, et al., Restless legs syndrome in an Asian population: A study in Singapore, Mov Disord, 2001;16:577–9.
14. Kim J, Choi C, Shin K, et al., Prevalence of restless legs syndrome and associated factors in the Korean adult population: the Korean Health and Genome Study, Psychiatry Clin Neurosci, 2005;59:350–53.
15. Kageyama T, Kabuto M, Nitta H, et al., Prevalences of periodic limb movement-like and restless legs-like symptoms among Japanese adults, Psychiatry Clin Neurosci, 2000;54:296–8.
16. Rangarajan S, Rangarajan S, D’Souza GA, Restless legs syndrome in an Indian urban population, Sleep Med, 2007; in press, Epub September 5, 2007.
17. Trescher WH, Lesser RP, The Epilepsies. In: Bradley WG, Daroff RB, et al. (eds.), Neurology in Clinical Practice, Philidelphia: Butterworth Heinemann, 2004.
18. Lee HB, Hening WA, Allen RP, et al., Race and restless legs syndrome symptoms in an adult community sample in east Baltimore, Sleep Med, 2006;7:642–5.
19. Berger K, Luedemann J, Trenkwalder C, et al., Sex and the risk of restless legs syndrome in the general population, Arch Intern Med, 2004;164:196–202.
20. Kotagal S, Silber MH, Childhood-onset restless legs syndrome, Ann Neurol, 2004;56:803–7.
21. Walters AS, Hickey K, Maltzman J, et al., A questionnaire study of 138 patients with restless legs syndrome: the ‘Night-Walkers’ survey, Neurology, 1996;46:92–5.
22. Winkelman JW, Chertow GM, Lazarus JM, Restless legs syndrome in end-stage renal disease, Am J Kidney Dis, 1996;28:372–8.
23. Chaudhuri KR, Iron and restless legs syndrome: the story unfolds, Sleep Med, 2006;7:395–6.
24. O’Keeffe ST, Gavin K, Lavan JN, Iron status and restless legs syndrome in the elderly, Age Ageing, 1994;23:200–203.
25. Manconi M, Govoni V, De Vito A, et al., Pregnancy as a risk factor for restless legs syndrome, Sleep Med, 2004;5:305–8.
26. Högl B, Frauscher B, Seppi K, et al., Transient restless legs syndrome after spinal anesthesia: a prospective study, Neurology, 2002;59:1705–7.
27. Skomro RP, Ludwig S, Salamon E, Kryger MH, Sleep complaints and restless legs syndrome in adult type 2 diabetics, Sleep Med, 2001;2:417–22.
28. Merlino G, Fratticci L, Valente M, et al., Association of restless legs syndrome in type 2 diabetes: a case-control study, Sleep, 2007;30:866–71.
29. Gómez-Esteban JC, Zarranz JJ, Tijero B, et al., Restless legs syndrome in Parkinson’s disease, Mov Disord, 2007;in press, electronically published Jun 19, 2007.
30. Brown LK, Dedrick DL, Doggett JW, Guido PS, Antidepressant medication use and restless legs syndrome in patients presenting with insomnia, Sleep Med, 2005;6:443–50.
31. Picchietti DL, England SJ,Walters AS, et al., Periodic limb movement disorder and restless legs syndrome in children with attention-deficit hyperactivity disorder, J Child Neurol, 1998;13: 588–94.
32. Walters AS, Mandelbaum DE, Lewin DS, et al., Dopaminergic therapy in children with restless legs/periodic limb movements in sleep and ADHD. Dopaminergic Therapy Study Group, Pediatr Neurol, 2000;22:182–6.
33. Aukerman MM, Aukerman D, Bayard M, et al., Exercise and restless legs syndrome: a randomized controlled trial, J Am Board Fam Med, 2006;19:487–93.
34. Woloshin S, Schwartz LM, Giving legs to restless legs: a case study of how the media helps make people sick, PLoS Med, 2006;3: e170.
35. Winkelmann J, Muller-Myhsok B, Wittchen HU, et al., Complex segregation analysis of restless legs syndrome provides evidence for an autosomal dominant mode of inheritance in early age at onset families, Ann Neurol, 2002;52:297–302.
36. Dhawan V, Ali M, Chaudhuri KR, Genetic aspects of restless legs syndrome, Postgrad Med J, 2006:626–9.
37. Online Mendelian Inheritance in Man. http://www.ncbi.nlm.nih.gov/sites/entrez?db=omim (Accessed August 10, 2007)
38. Xiong L, Levchenko A, Montplaisir J, et al., Genetic association studies of neurotensin gene and restless legs syndrome in French Canadians, Sleep Med, 2007; in press, Epub July 16, 2007.
39. Paulus W, Dowling P, Rijsman R, et al., Pathophysiological concepts of restless legs syndrome, Mov Disord, 2007;22:1451–6.
40. Wetter TC, Stiasny K, Winkelmann J, et al., A randomized controlled study of pergolide in patients with restless legs syndrome, Neurology, 1999;52:944–50.
41. Cervenka S, Pålhagen SE, Comley RA, et al., Support for dopaminergic hypoactivity in restless legs syndrome: a PET study on D2-receptor binding, Brain, 2006;129:2017–28.
42. Ondo WG, He Y, Rajasekaran S, Le WD, Clinical correlates of 6-hydroxydopamine injections into A11 dopaminergic neurons in rats: a possible model for restless legs syndrome, Mov Disord, 2000;15:154–8.
43. Paulus W, Trenkwalder C, Less is more: pathophysiology of dopaminergic-therapy-related augmentation in restless legs syndrome, Lancet Neurol, 2006;5:878–86.
44. Earley CJ, Connor JR, Beard JL, et al., Abnormalities in CSF concentrations of ferritin and transferrin in restless legs syndrome, Neurology, 2000;54:1698–1700.
45. Allen RP, Barker PB,Wehrl F, et al., MRI measurement of brain iron in patients with restless legs syndrome, Neurology, 2001;56: 263–5.
46. Earley CJ, Barker PB, Horska A, Allen RP, MRI-determined regional brain iron concentrations in early- and late-onset restless legs syndrome, Sleep Med, 2006;7:458–61.
47. Connor JR,Wang XS, Patton SM, et al., Decreased transferrin receptor expression by neuromelanin cells in restless legs syndrome, Neurology, 2004;62:1563–7.
48. Earley CJ, Allen RP, Beard JL, Connor JR, Insight into the pathophysiology of restless legs syndrome, J Neurosci Res, 2000; 62:623–8.
49. von Spiczak S, Whone AL, Hammers A, et al., The role of opioids in restless legs syndrome: an [11C]diprenorphine PET study, Brain, 2005;128:906–17.

Further Resources

Share this Article
Related Content In Movement Disorders
  • Copied to clipboard!
    accredited arrow-down-editablearrow-downarrow_leftarrow-right-bluearrow-right-dark-bluearrow-right-greenarrow-right-greyarrow-right-orangearrow-right-whitearrow-right-bluearrow-up-orangeavatarcalendarchevron-down consultant-pathologist-nurseconsultant-pathologistcrosscrossdownloademailexclaimationfeedbackfiltergraph-arrowinterviewslinkmdt_iconmenumore_dots nurse-consultantpadlock patient-advocate-pathologistpatient-consultantpatientperson pharmacist-nurseplay_buttonplay-colour-tmcplay-colourAsset 1podcastprinter scenerysearch share single-doctor social_facebooksocial_googleplussocial_instagramsocial_linkedin_altsocial_linkedin_altsocial_pinterestlogo-twitter-glyph-32social_youtubeshape-star (1)tick-bluetick-orangetick-red tick-whiteticktimetranscriptup-arrowwebinar Sponsored Department Location NEW TMM Corporate Services Icons-07NEW TMM Corporate Services Icons-08NEW TMM Corporate Services Icons-09NEW TMM Corporate Services Icons-10NEW TMM Corporate Services Icons-11NEW TMM Corporate Services Icons-12Salary £ TMM-Corp-Site-Icons-01TMM-Corp-Site-Icons-02TMM-Corp-Site-Icons-03TMM-Corp-Site-Icons-04TMM-Corp-Site-Icons-05TMM-Corp-Site-Icons-06TMM-Corp-Site-Icons-07TMM-Corp-Site-Icons-08TMM-Corp-Site-Icons-09TMM-Corp-Site-Icons-10TMM-Corp-Site-Icons-11TMM-Corp-Site-Icons-12TMM-Corp-Site-Icons-13TMM-Corp-Site-Icons-14TMM-Corp-Site-Icons-15TMM-Corp-Site-Icons-16TMM-Corp-Site-Icons-17TMM-Corp-Site-Icons-18TMM-Corp-Site-Icons-19TMM-Corp-Site-Icons-20TMM-Corp-Site-Icons-21TMM-Corp-Site-Icons-22TMM-Corp-Site-Icons-23TMM-Corp-Site-Icons-24TMM-Corp-Site-Icons-25TMM-Corp-Site-Icons-26TMM-Corp-Site-Icons-27TMM-Corp-Site-Icons-28TMM-Corp-Site-Icons-29TMM-Corp-Site-Icons-30TMM-Corp-Site-Icons-31TMM-Corp-Site-Icons-32TMM-Corp-Site-Icons-33TMM-Corp-Site-Icons-34TMM-Corp-Site-Icons-35TMM-Corp-Site-Icons-36TMM-Corp-Site-Icons-37TMM-Corp-Site-Icons-38TMM-Corp-Site-Icons-39TMM-Corp-Site-Icons-40TMM-Corp-Site-Icons-41TMM-Corp-Site-Icons-42TMM-Corp-Site-Icons-43TMM-Corp-Site-Icons-44TMM-Corp-Site-Icons-45TMM-Corp-Site-Icons-46TMM-Corp-Site-Icons-47TMM-Corp-Site-Icons-48TMM-Corp-Site-Icons-49TMM-Corp-Site-Icons-50TMM-Corp-Site-Icons-51TMM-Corp-Site-Icons-52TMM-Corp-Site-Icons-53TMM-Corp-Site-Icons-54TMM-Corp-Site-Icons-55TMM-Corp-Site-Icons-56TMM-Corp-Site-Icons-57TMM-Corp-Site-Icons-58TMM-Corp-Site-Icons-59TMM-Corp-Site-Icons-60TMM-Corp-Site-Icons-61TMM-Corp-Site-Icons-62TMM-Corp-Site-Icons-63TMM-Corp-Site-Icons-64TMM-Corp-Site-Icons-65TMM-Corp-Site-Icons-66TMM-Corp-Site-Icons-67TMM-Corp-Site-Icons-68TMM-Corp-Site-Icons-69TMM-Corp-Site-Icons-70TMM-Corp-Site-Icons-71TMM-Corp-Site-Icons-72