2
Editorial Wrestling with restless legs While most people take comfort in the knowledge that a peaceful rest awaits them at the end of a weary day, the many who suffer from restless legs syndrome (RLS) find no such reward in recumbency. The therapeutic dilemma of how to bring about resolution of symptoms for those patients suffering from RLS has thus disturbed the slumber of more than one neurologist, primary care physician, and yes, even the lowly nephrologist. In many disease states, serendipitous discoveries provide clues that help unlock key pathophysiologic processes, eventually providing satisfactory therapeutic solutions. So appears to be the case with RLS, where iron seems to be lynchpin to the disorder [1,2], serving as a probe that is allowing pieces of the mechanistic puzzle to gradually fall into place. Iron is a critical element in healthy neuronal cell function, acting as an essential co-factor for neurotransmit- ter synthesis [3]. Dopamine synthesis in the substantia nigra is dependent on iron acting as a co-factor for cytosolic tyrosine hydroxylase. In particular, iron is a co-factor for the rate-limiting step required to convert tyrosine to levodopa. The latter is then decarboxylated to form dopamine. As such, low levels of iron in the basal ganglia and other ‘movement centers’ of the brain can reduce levels of this neurotransmitter critical to movement and cognition. The presence of diminished iron stores in the substantia nigra of individuals afflicted with RLS adds support to this hypothesis [4]. Alternatively, studies in iron-deficient rats have shown decreased activity of dopamine transporter (DAT) in pre-synaptic neurons, perhaps mediated by decreased D 2 receptor expression [5]. The resultant decrease in reuptake of dopamine from the synaptic cleft may trigger ligand-induced down-regulation of D 1 receptors and aberrant neurotransmission. Some reversibility of DAT and dopamine receptor levels has been demonstrated with iron repletion in these animals [6]. Iron metabolism is tightly regulated within the neuron owing not only to its importance in normal cellular physiologic functioning, but the vulnerability of the cell to excessive accumulation. Ferrous (Fe C2 ) iron is lethal to neuronal cell membranes given its capability of forming intracellular hydroxyl radicals [7]. Transferrin receptors expressed on the neuronal cell surface serve to transport ferric iron (Fe C3 ) transferrin complex into the cell [8]. Of note, neuronal transferrin receptor expression is increased by iron deficiency. Once the iron-transferrin complex is endocytosed, iron is released from transferrin into endosomes, reduced again to its ferrous form, and transported into the cytosol by divalent metal transport 1 (DMT1). Excessive cytosolic iron build-up is further precluded by ferroportin1, a membrane-bound protein that mediates ferrous iron egress from the cell. In concert with the ferrous oxidase activity of hephastine, a ceruloplasmin- like protein, ferrous iron is oxidized and exported into the brain interstitium to be bound again by transferrin. Iron can then be transported back into the blood, carried to other tissues, or to serum ferritin for storage. Neural storage of iron by intracellular ferritin occurs only in a few selected areas of the brain, of which the substantia nigra, putamen, and other basal ganglia nuclei are excluded. Conversely, dopaminergic neurons in these areas contain neuromelanin, an iron-storage pigment with less iron-binding capacity than ferritin. The exact nature and function of neuromelanin is currently not fully understood [9]. As noted above, a considerable body of evidence now suggests that a defect in the metabolism of neurocellular iron plays a causative role in RLS. Recent studies have demonstrated salutary effects of high dose intravenous iron in the treatment of subjects with both primary and secondary RLS [10,11]. In this issue of Sleep Medicine, Earley et al. report the effects of repetitive intravenous iron adminis- tration to patients experiencing a relapse of RLS after achieving a beneficial response to a previous dose of infused iron. They observed an apparent attenuated rate of iron loss measured by decelerated ferritin decay after each sequential dose. Sustained serum ferritin levels and RLS response might well be explained here by iron supersaturation of neurocellular ferritin and neuromelanin, co-factor sites for a neurotransmitter synthesis and D 2 receptor expression, and decreased expression of transferrin receptors and/or increased ferroportin1 activity. This would be not all that unexpected given the abundance of iron provided by intravenous administration. Recent studies of leukocyte iron dysmetabolism in end-stage renal disease [12],a condition where secondary RLS reaches a prevalence of 20–57% [8], reveal up-regulated transferrin receptor and down-regulated ferroportin1 activity. If these two iron transporter defects also exist in neurons of ESRD patients, Sleep Medicine 6 (2005) 295–296 www.elsevier.com/locate/sleep 1389-9457/$ - see front matter q 2005 Elsevier B.V. All rights reserved. doi:10.1016/j.sleep.2005.02.006

Wrestling with restless legs

Embed Size (px)

Citation preview

Page 1: Wrestling with restless legs

Editorial

Wrestling with restless legs

While most people take comfort in the knowledge that a

peaceful rest awaits them at the end of a weary day, the

many who suffer from restless legs syndrome (RLS) find no

such reward in recumbency. The therapeutic dilemma of

how to bring about resolution of symptoms for those

patients suffering from RLS has thus disturbed the slumber

of more than one neurologist, primary care physician, and

yes, even the lowly nephrologist.

In many disease states, serendipitous discoveries provide

clues that help unlock key pathophysiologic processes,

eventually providing satisfactory therapeutic solutions. So

appears to be the case with RLS, where iron seems to be

lynchpin to the disorder [1,2], serving as a probe that is

allowing pieces of the mechanistic puzzle to gradually fall

into place.

Iron is a critical element in healthy neuronal cell

function, acting as an essential co-factor for neurotransmit-

ter synthesis [3]. Dopamine synthesis in the substantia nigra

is dependent on iron acting as a co-factor for cytosolic

tyrosine hydroxylase. In particular, iron is a co-factor for the

rate-limiting step required to convert tyrosine to levodopa.

The latter is then decarboxylated to form dopamine. As

such, low levels of iron in the basal ganglia and other

‘movement centers’ of the brain can reduce levels of this

neurotransmitter critical to movement and cognition. The

presence of diminished iron stores in the substantia nigra of

individuals afflicted with RLS adds support to this

hypothesis [4]. Alternatively, studies in iron-deficient rats

have shown decreased activity of dopamine transporter

(DAT) in pre-synaptic neurons, perhaps mediated by

decreased D2 receptor expression [5]. The resultant decrease

in reuptake of dopamine from the synaptic cleft may trigger

ligand-induced down-regulation of D1 receptors and

aberrant neurotransmission. Some reversibility of DAT

and dopamine receptor levels has been demonstrated with

iron repletion in these animals [6].

Iron metabolism is tightly regulated within the neuron

owing not only to its importance in normal cellular

physiologic functioning, but the vulnerability of the cell to

excessive accumulation. Ferrous (FeC2) iron is lethal to

neuronal cell membranes given its capability of forming

intracellular hydroxyl radicals [7]. Transferrin receptors

expressed on the neuronal cell surface serve to transport

1389-9457/$ - see front matter q 2005 Elsevier B.V. All rights reserved.

doi:10.1016/j.sleep.2005.02.006

ferric iron (FeC3) transferrin complex into the cell [8].

Of note, neuronal transferrin receptor expression is

increased by iron deficiency. Once the iron-transferrin

complex is endocytosed, iron is released from transferrin

into endosomes, reduced again to its ferrous form, and

transported into the cytosol by divalent metal transport 1

(DMT1). Excessive cytosolic iron build-up is further

precluded by ferroportin1, a membrane-bound protein that

mediates ferrous iron egress from the cell. In concert with

the ferrous oxidase activity of hephastine, a ceruloplasmin-

like protein, ferrous iron is oxidized and exported into the

brain interstitium to be bound again by transferrin. Iron can

then be transported back into the blood, carried to other

tissues, or to serum ferritin for storage. Neural storage of

iron by intracellular ferritin occurs only in a few selected

areas of the brain, of which the substantia nigra, putamen,

and other basal ganglia nuclei are excluded. Conversely,

dopaminergic neurons in these areas contain neuromelanin,

an iron-storage pigment with less iron-binding capacity than

ferritin. The exact nature and function of neuromelanin is

currently not fully understood [9].

As noted above, a considerable body of evidence now

suggests that a defect in the metabolism of neurocellular

iron plays a causative role in RLS. Recent studies have

demonstrated salutary effects of high dose intravenous iron

in the treatment of subjects with both primary and secondary

RLS [10,11]. In this issue of Sleep Medicine, Earley et al.

report the effects of repetitive intravenous iron adminis-

tration to patients experiencing a relapse of RLS after

achieving a beneficial response to a previous dose of infused

iron. They observed an apparent attenuated rate of iron loss

measured by decelerated ferritin decay after each sequential

dose. Sustained serum ferritin levels and RLS response

might well be explained here by iron supersaturation of

neurocellular ferritin and neuromelanin, co-factor sites for a

neurotransmitter synthesis and D2 receptor expression, and

decreased expression of transferrin receptors and/or

increased ferroportin1 activity. This would be not all that

unexpected given the abundance of iron provided by

intravenous administration. Recent studies of leukocyte

iron dysmetabolism in end-stage renal disease [12], a

condition where secondary RLS reaches a prevalence of

20–57% [8], reveal up-regulated transferrin receptor and

down-regulated ferroportin1 activity. If these two iron

transporter defects also exist in neurons of ESRD patients,

Sleep Medicine 6 (2005) 295–296

www.elsevier.com/locate/sleep

Page 2: Wrestling with restless legs

Editorial / Sleep Medicine 6 (2005) 295–296296

one would expect copious cytosolic iron available to

facilitate neurotransmitter synthesis. That ESRD patients

with RLS experience a beneficial response to administered

iron despite apparent normal or supranormal iron stores

implies the contrary, however. Assuming that defects are

similar in primary and secondary RLS, the above studies

taken together might imply that iron is not available in the

cytosol for neurotransmitter synthesis or D2 receptor

expression, perhaps due to abnormal offloading of iron

from DMT1 or subtle changes in microenvironmental pH

that preclude dissociation of iron from its carrier or storage

vehicle.

There is obviously more work to be done. However, the

sharing of experiences and information across subspecial-

ties should allow future collaborative efforts to pinpoint

single or multiple subcellular defects in this ubiquitous

disorder. One can only hope that this will translate

into successful, long lasting therapeutic interventions that

will guarantee both the patients with RLS and their care

providers a good night’s rest.

References

[1] Ekbom KA. Restless legs syndrome. Neurology 1960;10:868–73.

[2] Sun ER, Chen CA, Ho G, Earley CJ, Allen RP. Iron and restless legs

syndrome. Sleep 1998;21:371–7.

[3] Blake D, Williams A, Pall H, Fonseca A, Beswick T. Iron and

akathisia. Br Med J 1986;292:1393.

[4] Allen RP, Barker PB, Wehrl F, Song HK, Early CJ. MRI measurement

of brain iron in patients with restless legs syndrome. Neurology 2001;

56:263–5.

[5] Erikson KM, Byron BC, Beard JL. Iron deficiency alters dopamine

transporter functioning in rat striatum. J Nutr 2000;130:2831–7.

[6] Beard J, Erikson KM, Byron BC. Neonatal iron deficiency results in

irreversible changes in dopamine function in rats. J Nutr 2003;133:

1174–9.

[7] Nappi AJ, Vass E. Iron, metalloenzymes, and cytotoxic reactions. Cell

Mol Biol 2000;46:637–47.

[8] Moos T, Morgan EH. The metabolism of neuronal iron and its

pathogenic role in neurologic disease. Ann NY Acad Sci 2004;1012:

14–26.

[9] Zucca FA, Giaveri G, Gallorini M, et al. The neuromelanin of human

substantia nigra: physiological and pathogenic aspects. Pigment Cell

Res 2004;17:610–7.

[10] Earley CJ, Heckler D, Horska A, Barker PB, Allen RP. The treatment

of restless legs syndrome with intravenous iron dextran. Sleep Med

2004;5:231–5.

[11] Sloand JA, Shelly MA, Feigin A, Bernstein P, Monk MD. A double-

blind, placebo-controlled trial of intravenous iron dextran therapy in

patients with end stage renal disease and restless legs syndrome. Am

J Kidney Dis 2004;43:663–70.

[12] Otaki Y, Nakanishi T, Hasuike Y, et al. Defective regulation of iron

transporters leading to iron excess in polymorphonuclear leukocytes

of patients on maintenance hemodialysis. Am J Kidney Dis 2004;43:

1030–9.

James A. Sloand

Nephrology Division,

University of Rochester, Rochester, NY, USA

E-mail address: [email protected]

Received 17 February 2005

Accepted 17 February 2005