Download as pdf or txt
Download as pdf or txt
You are on page 1of 22

Eur. J. Biocheni.

164,485 - 506 (1987)


0FEBS 1987

Review
Iron transport and storage
Robert R. CRICHTON and Mireille CHARLOTEAUX-WAUTERS
Unite de Biochimie, Universite Catholique de Louvain, Louvdin-la-Neuve

(Received October 22/December 12, 1986) - EJB 86 1157

In the development of human society, the utilisation of molecules are powerful complexants of Fe(II1) and are
metals both in decorative art forms and in objects of practical secreted into the extracellular medium by a large number
application passed through several phases: gold and copper, of unicellular organisms. The ferri-siderophores [the Fe(II1)-
malleable and easily workable, were successively replaced by chelate complexes] are then assimilated by the cells via a
bronze (an alliance of copper and tin) and thereafter by iron. receptor specific for the complex and once inside the cell the
It is not the object of the present review to consider the iron iron is released either by reduction of the iron to Fe(l1) (for
age nor to insist on the important contribution of the Celts in which the siderophore has little affinity) or by hydrolysis of
its development, but the importance of iron in our civilisation the ferri-siderophore accompanied by release of its iron.
is perhaps best summarized by the quotation from Rudyard For multicellular organisms, the problem of the transport
Kipling cited by Philip Aisen in his article on physico-chemical of iron to the different cell types of the organism and its
aspects of iron metabolism [l]: resorption from the dietary sources of iron available to it
(in general such organisms do not possess a system of
“Gold is,for the mistress - silver,for the maid -
siderophores with their appropriate receptors, and must
Copper.for the cruftsman cunning at his trade!
therefore find the iron that they require in their alimentation)
‘Good!’ said the Baron, sitting in his hall,
also demands a solution. As we shall see, this role is assured
‘But iron’ - Cold Iron - is master qf them all”.
by a serum globulin, transferrin, which complexes iron, trans-
If the Celts were among the first to recognize the practical ports it in the circulation and, via specific receptors, is taken
value of this element, it is certain that this was directly up by the cells of the organism. The release of iron is pH-
connected with its biological abundance in the earth’s crust, dependent and the availability of proton pumps within specific
particularly compared with elements such as gold, silver and intracellular compartments assures the intracellular release of
copper [2]. Iron is the second most abundant metal, after iron; the iron-free protein is recycled from the cell and released
aluminium, and the fourth most abundant element in the into the circulation for reutilisation.
earth’s crust. But it is also interesting to remark that in Once the iron is released within the cell, the same problems
contrast to the three elements mentioned above, iron is subject that we have evoked above again present their necessary
to an extremely important corrosion, such that there remain fidelity to the rules of the solution chemistry of iron: either
relatively few objects from prehistoric times made with this the iron which has been liberated from its soluble siderophore
metal in comparison with those made from gold, silver or complex will be complexed by an appropriate intracellular
copper! chelator, or else it will follow its inexorable hydrolysis
In fact, iron, element 26 in the periodic table, has two and polymerisation towards biological inaccessibility. We
properties at physiological pH and in aqueous medium which encounter here the second development in the evolution of
make it at the same time interesting for biological systems in the capacity of living organisms to assimilate and utilize iron:
terms of catalysis and transport, and also biologically in- the appearance of intracellular iron storage compounds,
accessible. Iron can exist in aqueous solution in two oxidation which allow the cell to dispose of a pool of bioavailable iron
states: Fe2+ and Fe3+. Secondly, the solution chemistry of in a form which is soluble in physiological conditions, and
iron is essentially dominated by the hydrolysis and poly- non-toxic. The archetype of these compounds is the iron
merisation of aqueous Fe(1II) to insoluble and potentially storage protein, ferritin.
biologically inaccessible ferric hydroxides and oxyhydroxides. A brief parenthesis imposes itself here: the insistence that
We do not consider further this problem, which has been dealt such iron storage compounds should be non-toxic. In fact the
with in detail in numerous review articles [l, 3, 41, but direct potential toxicity of iron, free in solution through complexa-
attention to the consequences which result from this situation. tion to whatever ligand it may be, manifests itself through the
For living organisms to be able to obtain iron from their reaction described in 1935 by Haber and Weiss [6]: this reac-
environment they have firstly evolved a series of molecules tion ( 3 ) is the sum of reactions (1) and ( 2 ) , respectively reduc-
(essentially derived from catechols or from a series of different tion of Fe(II1) by superoxide ion and the well-known Fenton
hydroxamic acids) designated as siderophores [5]. These reaction :
Correspondence to R. R. Crichton, Unit6 de Biochimie, Universitk
Catholique de Louvain, Place Louis Pasteur 1, B-1348 Louvain-la-
Fe(II1) + 0, 4 Fe(I1) + 0 2 (11
Neuve, Belgium
Dedicated to Professoi- Dr Gerhard Braunitzer on the occasion
Fe(I1) + Hz02 4 Fe(1II) + OH- + OH’ (2)
of his 65th birthday. (3)
486

Since the oxidation of ferrous iron by molecular oxygen are used as chelators in the treatment of iron overload in
can be represented by the sequence of reactions (4) and (5), it man.
is immediately apparent that if 'free' iron exists within a cell,
it could provoke the production of hydroxyl radicals, with
Siderophores
disastrous consequences for the cell. The system is in fact used
by macrophages in the killing of bacteria: The first siderophore to be isolated was ferrichrome [ll]
from the culture filtrate of the smut fungus Ustilago
Fe(I1) + O2 + Fe(II1) + 0 , (4) sphaerogena [Ill. It was shown to stimulate the growth of
microorganisms requiring chelated iron. Subsequently the
group of Keller-Scherlein et al. concluded that most actino-
Thus the complexation of iron within a cell necessitates myces produced iron complexing agents, which they called
that the iron storage protein must guard its iron in a soluble, ferrioxamines [12]. The term siderophore proposed by
bio-available and non-toxic form. These conditions are Lankford [13] was subsequently adopted and can be defined
fulfilled by ferritin. [ 5 ] as 'a low-molecular-mass, virtually ferric-specific ligand,
The importance of iron for living organisms is underlined the biosynthesis of which is carefully regulated by iron and
by its role in a large number of proteins which require its the function of which is to supply iron to the cell'. At present
presence for their activity [7]. Firstly, there are proteins about 200 different natural siderophores are known [5].
involved in the reversible binding of oxygen in animals
(haemoglobins and myoglobins), plants (leghaemoglobin) Classification and structure of siderophores
and in invertebrates (haemerythrin, this latter containing non-
haem iron). There are many enzymes catalysing reactions with Although siderophores display a considerable structural
oxygen : mono-oxygenases such as the pteridine-dependent variation, they may in general be classified as either
hydroxylases for the aromatic amino acids and haemoproteins hy droxamates
such as cytochrome P450; dioxygenases, both haem and non-
haem, for example, tryptophan pyrrolase and prolyl hydroxy-
lase; oxidases, and the terminal enzyme of the mitochondria1
respiratory chain, cytochrome c oxidase; reactions with
peroxides (hydroperoxidases, catalase, ferroxidase, lacto-
ferroxidase) with superoxide (microbial superoxide dis-
mutases) and, associated with other metals, with nitrogen
(Mo) and hydrogen (Ni). In the respiratory, photosynthetic or phenolates/catecholates (derivatives of 2,3-dihydroxyben-
and microsomal electron transport chains we find zoic acid).
cytochromes (of the types a, b, c and d)as well as iron-sulphur
proteins. Other iron-sulphur proteins catalyse oxidation reac-
tions (xanthine oxidase, xanthine dehydrogenase, aldehyde
13
oxidase and sulphite oxidase) and the Kreb's cycle enzyme
aconitase is also an iron-sulphur protein (of the 4Fe-4S class).
Finally, last but by no means least, there is the iron-containing
ribonucleotide reductase necessary for the transformation of
ribonucleotide diphosphates to their corresponding deoxy Siderophores present an extremely high affinity for ferric
derivatives. iron with formation constants ranging from for entero-
bactin to loz3 for aerobactin [13- 191. Ferric siderophores
are octahedral complexes in which the coordinated metal ion
IRON TRANSPORT IN PROKARYOTES is d5 high spin and rapidly exchangeable. Since the ligands
AND LOWER EUKARYOTES are hard bases they have relatively low affinity for soft acid
cations such as Fe(I1) [5]. Thus we could anticipate that iron
Introduction release from ferri-siderophores would involve a reduction
As was pointed out in the opening introduction, micro- step.
organisms have developed a high-affinity system for iron up- The structures of five siderophores, which can all be taken
take from their extracellular medium. This consists of three up as the corresponding ferric complex by Escherichia coli
components: (a) low-molecular-mass soluble, high-affinity strain K12, are given in Figs 1- 5 [5].
ferric-iron ligands released by the cells and called siderophores Ferrichrome, the prototype of hydroxamate-type
(from the Greek 'iron bearers'); (b) a membrane receptor for siderophores (Fig. 1A) is a cyclic hexapeptide consisting of
the iron-loaded form of the siderophore (ferri-siderophore) three residues of glycine and three residues of N-hydroxy-L-
that transports the ferric chelate across the microbial mem- ornithine in which hydroxamic acid groups are produced by
brane; (c) an enzymatic system capable of releasing the iron the acetylation of the three hydroxamine functions (Fig. 1B).
from the ferri-siderophore within the cell. The natural ferrichrome which is produced by many types of
Siderophores are probably found in all aerobic and fungi, has a preferred A geometry at the metal centre. Both
facultative anaerobic micro-organisms but they have so far Neurospora crassa and E. coli can take up this siderophore (in
never been reported in strict anaerobes, in lactic acid bacteria the case of the latter organism this despite the fact that it
or in higher organisms. cannot synthesise it). Moreover, the mirror image A cis-
The functions of siderophores are multiple: they act not enantio ferrichrome is ineffective in supplying iron to the fungi
only as receptor-dependent, high-affinity iron transport N. crassa [20] and Penicillium parvum [21] and less effective
systems but also as growth or germination factors, as anti- than the natural A cis ferrichrome in supplying iron to E. coli
biotics or as virulence factors [8 - 101. The iron-free molecules P11.
487

Fig. 1 . Structures of ( A ) ferrichrome, the prototype of the hydroxamate type of siderophores and ( B ) dejerrichrome. (A) Reproduced with
permission from Neilands [5]. (B) * = iron chelation sites. Reproduced with permission from Byers and Arceneaux [336]

A
B

HO -‘C-CO,H
/
B H j C H , O H
H

R n
- -
schizokinen H 2
C
aerobactin COOH 4
arthrobartin H 4
Fig. 3. Structure of siderophores based on citric acid. Reproduced with
permission from Neilands [5]

and chromic ions. A recent study has established the


stereospecificity of rhodotorulate-mediated iron uptake in R .
pilimanea [22].
Aerobactin is a derivative of citric acid (Fig. 3) in which
the distal carboxyl groups have been substituted with
hydroxamate functions. In E. coli only strains with the Col V
plasmid, which carries genes for the synthesis of the
siderophore and one gene for the uptake (receptor) system,
can use ferric aerobactin as a service of iron. A number of
Fig. 2. Structure of rhodotorulic acid and its derivatives. (A) Rhodo- other bacteria such as Aerobricter aerogenes can also produce
torulic acid; (B) dimerumic acid; (C) coprogen. Reproduced with this siderophore.
permission from Neilands [5] The ferrioxamine family of siderophores is represented
(Fig. 4) by desferrioxamine B, the mesylate salt of which is
marketed by Ciba-Geigy as Desferal, the major drug used in
the chelation therapy of secondary iron overload in man.
Rhodotorulic acid (Fig. 2) is a dipeptide of acetylated These siderophores, whch include the antibiotic ferriomycin,
N-hydroxyornithine which mediates iron uptake in the yeast are produced by most actinomycetes [12].
Rhodotorula pilimanea and is produced in vast quantities by The phenol/catechol type of siderophore enterochelin, or
the yeast when grown in iron-deficient culture media. This enterobactin as it is often called, (Fig. 5) is common to all
dihydroxamate siderophore forms a 3 :2 complex with ferric enteri bacteria. It forms a hexadentate complex with a forma-
488

Fig. 4. Structure of ferrioxumine fumily qf siderophores. Desferrioxamine 9, R = H. Mesylate salt = Desferal. Reproduced with permission
from Neilands [337]

9 A
B

Fig. 5. Structure of ( A ) ferric enterobactin (jerric complex of cyclo-tri-2,3-dihydroxyhenzoylserine) and ( B ) enterohactin (deferri) or
enterochelin. (A) Reproduced with permission from Neilands [5]. (B) * = iron chelation sites. Reproduced with permission from Bycrs and
Arceneaux [336]

tion constant of lo5’ between a ferric iron atom and six Outside Peripi. Cytoplasm
OM CM
deprotonated phenolic hydroxy groups derived from a triester
of 2,3-dihydroxybenzoylserine.The natural iron complex of
enterochelin has the A configuration; the synthetic mirror
image ferric enantioenterochelin does not support the growth
of E. coli [23]. Fez+
We could also add to this collection ferric citrate, for which
a well characterized outer membrane receptor exists [24] in
E. coli.
Fe”

RECEPTORS FOR SIDEROPHORES


Ferric siderophore transport has been extensively studied Fe3‘-Enterocheiin
in a number of gram-negative bacteria (E. coli, Salmonella FE”
typhimurium), gram-positive bacteria (Bacillus sp. and Myco-
bacterium sp) and in fungi ( N . crassa and U . sphaerogena) [25,
261. However, on account of the well established genetics of
E. coli strain K12, we know most about iron-siderophore Fig. 6. Ferri-siderophores uptuke systems of E. coli K12 pColv. Re-
transport in this organism. produced with permission from Hantke [34]
At this point it is important to recall that gram-negative
bacteria have both an outer membrane and a plasma (or
cytosolic) membrane separated by a rigid murein (peptido-
glycan) layer. In the outer membrane are located porins, pro- follows as a logical consequence of this observation that they
teins which form pores through which small molecules can require a receptor protein.
pass into the periplasm and gain access to the plasma mem- When E. coli is placed in low-iron growth conditions,
brane, where they may be taken up by specific transport the derepression of seven new outer membrane proteins of
systems. The molecular mass of siderophores, and more molecular mass 74- 83 kDa is observed [28]. For five of these
importantly of their iron complexes, varies over 500- proteins (Fig. 6) a receptor function for one, or for several
1000 Da and is thus too large to allow free diffusion through siderophores has been established: it is interesting to note that
the small water-filled pores of the outer membrane [27]. It some of these ferri-siderophore receptors also recognise a
489

number of colicines and bacteriophages. We shall consider In the first mechanism, a siderophore may be compared
each of these ferri-siderophore receptor systems in turn. with an ‘iron taxi’: the ferri-siderophore brings the iron to the
The receptor for ferrichrome, designated in Fig. 6 as fhuA cell membrane but does not penetrate into the cell. Dissocia-
(ferric hydroxamate uptake) was originally called tonA, the tion of the iron-chelate complex occurs in the membrane: iron
attachment site for bacteriophages T1, T5 and 6 80 as well as enters the cell while the ligand remains extracellular [17, 35-
for colicine and albomycin 1251. The discovery of tonA as 371. This mechanism is described in plants and yeast.
the receptor for ferrichrome in E. coli was preceded by the The second mechanism proposes the transport of the in-
discovery of a series of mutants in S . typhimurium resistant to tact ferri-siderophore into the cell where cellular dissociation
albomycin, an antibiotic analogue of ferrichrome [28]. The of the metal involves chemical breakdown of the ligand itself.
mutants were found to be defective in ferrichrome transport An esterase specific for the ferric complex hydrolyses the
and their mapping on the chromosome of S . typhimurium was ligand during iron release in the case of ferric enterobactin
found to coincide with the tonA locus in E. coli. As Nielands and fusarinine-type siderophores [38 - 421.
has pointed out 1251, this was the origin of the idea that the This mechanism of iron release from ferri-enterochelin is
biochemical function of the ronA gene product must be that typified in E. coli where an esterase, encoded by the gene,f;s,
of a receptor for ferrichrome [29]. While E. coli does not degrades the enterochelin by hydrolysis of the ester bonds in
synthesise ferrichrome, it one the less has an outer membrane the siderophore, and permits iron release with concomitant
receptor for this ferri-siderophore produced by Penicillium destruction of the siderophore [43, 441.
and many other fungi. In a third mechanism, the ferri-siderophore is also trans-
Subsequent to the discovery that the ferrichrome receptor ported intact into the cell but the iron is released via a reduc-
was the gene product of tonA, it became apparent that the tion step. This reaction probably requires a reductase
protective effect of ferric enterochelin against colicine B [30] [NAD(P)H oxidoreductase] which reduces the ferric iron to
could be simply explained by the fact that the colicine B the ferrous state; since the siderophore has little affinity for
outer membrane receptor was in reality the ferric enterochelin Fez+ the consequence is that the iron is released. Such re-
receptor. This outer membrane protein of molecular mass ductase activity was reported elsewhere [36, 45 -471. During
81 kDa, designated in Fig. 6 as fepA, is the receptor for this reduction, the ligand may or may not undergo chemical
enterochelin, the siderophore produced by E. coli under low modification. Enterochelin and ferrichrome were acetylated
iron stress. during iron release (during or after reduction of iron) and
Strains of E. coli which possess the Col V plasmid can also thus secreted into the external medium [48]. This modified
synthesise a second siderophore, aerobactin. The receptor product had a decreased affinity for ferric iron and so did not
protein for ferric aerobactin, iutA (Fig. 6), present in the outer serve as an effective iron siderophore.
membrane, has recently been sequenced [311. In contrast, aerobactin is not destroyed after delivery of
Iron can also be assimilated by E. coli from other iron: the iron-free siderophore is once again excreted into the
hydroxamate siderophores such as coprogen (from N . crasso), medium and reused in subsequent iron transport [49]. This
rhodotorulic acid (from R . pilimanea) and ferrioxamine B mechanism is known as the ‘iron shuttle mechanism’ because
(from various species of Streptomyces), via the fhuE protein one siderophore molecule compared, as a true ferric iono-
[321. phore, can shuttle more than one iron ion into the organism.
Finally, E. coli can take up iron from ferric citrate through As we will see in the next section of our review, this re-
the acceptor protein fecA, inducible under low iron stress in sembles in many respects the mode of iron transport by trans-
the presence of citrate. ferrin.
Iron uptake from ferrichromes requires, in addition to
fhuA, at least five other genes: tonB,,fhuCDBand exbB. Three
of them,JhuCDB, map close tofhuA [33], and the gene prod- PROTEINS OF IRON TRANSPORT
ucts offiuC and fhuD are localised in the plasma membrane. The purification of iron binding proteins in extracellular
The gene product of tonB is essential for all the ferri- biological fluids has a long history. Conalbumin (which we
siderophore uptake systems: mutations in tonB not only now refer to as ovotransferrin) was purified in 1900 [50] and
abolish ferrichrome uptake, but all the other ferri-siderophore identified as the antimicrobial agent of raw egg white in 1946
uptake systems and that for vitamin B12 are defective. The [51]. Two years earlier it had been established that the anti-
same is true for mutations in exbB [34]. Thus, in the microbial properties of egg white were abolished by the addi-
hydroxamate siderophore pathway the specificity resides in tion of iron [52]. The iron binding protein of human serum,
the outer membrane receptor, while the componentsFuCDB, serotransferrin was purified by Schade and Caroline in 1946
tonB and exbB are common to all three classes of [53] and its anti-bacterial properties, as well as their abolition
hydroxamates (Fig. 6). Ferric enterobactin and ferric citrate by iron, established. Working independently, Laurell and In-
uptake also requires tonB and exbB together with the receptor gelman [54] also purified the ‘red’ protein from pig plasma
protein genes fepA and,fecA and the gene products of,fepB and in the same year Holmberg and Laurell [55] proposed
andfecB respectively (Fig. 6). the name ‘transferrin’, which has since replaced the name
siderophilin originally utilised by the American workers. In
1949 Schade et al. [56] established that human serotransferrin
IRON RELEASE FROM FERRISIDEROPHORES
binds two atoms of iron per molecule and that iron binding
Once the ferri-siderophore has been taken up by the outer is accompanied by the concomitant binding of a bicarbonate
membrane receptor protein, iron release can take place by ion for each iron atom bound. In the same year it was estab-
three mechanisms which are briefly summarised below. It is lished that transferrin iron can be released [57] by acidification
somewhat surprising that, whereas iron siderophore uptake of the medium. Subsequent studies have clearly established
pathways are well established in E. coli, much of our informa- the importance of this mechanism within the cell. The iron
tion concerning iron release comes from other microorgan- binding protein of milk, lactotransferrin, is a relative new-
isms, notably fungi. comer to the scene [58,59] but its iinportance both as an iron
490
I S , 10 15

LTF
STF
OTF
LTF
STF
OTF

9*
Q P R T H
D P Q T F
G S T T S
bs w w

C S K T D E R P A S

LTF L L F ?o T c s ? K F Dy.-mQ sIA[G sr-p R s


STF LL Y N K I N H C R F D El FFSEGCAPGSK-KDS
OTF L I H NOR T G T c N F D EI Y F S E G C A P G S P- N S

0 6 a

LTF P R I D ? G L!P' S G:fF?A I Q N%!K S E?E V?

Fig. 7. Amino acid sequences of human serotransferrin ( S T F ) , hen ovotransferrin ( O T F ) and human lactotransferrin (LTF). Reproduced with
permission from Metz-Boutigue et al. [73]
49 1

NeuAc(aZ-b)-Gal(R1-4)-GlcNAc(~1-2)-Wan(al-3)

A Man( B1-4)-GlcNAc(B1-4)-G1 cNAc t l - A s n

NeuAc(crZ-6)-Gal(B1-4)-Cl cNAc(Rl-Z)-Man(a1-6) /

NeuAc(x2-6)-Gal(B1-4)-GlcNAc(B1-2)-Man(nl-3)

B \Man (?1-4)-G1 cAC( Bl-4)-G1 cNAc P 1- Asn

NeuAc(a2-6)-Gal(~l-4)-GlcNAc(B1-2)-Man(~l-6) / (31-6)
I
Fuc

GlcNAc( P I - 4 )
\
GlcNAc(~1-2)-Man(a1-3)

C
G1 cNAc( B1-4) 1 Man( p l - 4 ) - G 1 cNAc( 41-4) -G1 cNAc l11-Asn

GlcNAc( p l - E ) - Y a n ( B 1 - 6 ) /
Fig. 8. Structure of the principal glycans of ( A ) human serotransferrin, ( B ) human lactotransferrin and ( C ) hen ovotransferrin. Reproduced
with permission from Spik et al. [338]

transport protein and as a protective agent against bacterial and Waterman [77] strongly suggest the presence of two hom-
infections (it is found also in tears and is secreted by ologous domains (44% identical residues): ND 2 (108 -232)
leukocytes) is now well established. As we shall see, the ex- and CD 2 (463 - 589), which could be taken as an indication
tensive sequence homologies between the three types of trans- that each half of the molecule is composed of three distinct
ferrin, not only confirm the presence of two distinct domains domains. For the moment, the two-domain nature of trans-
but allow us to draw some conclusions concerning the nature ferrins seems to be well established and, as we will see, each
and localisation of their iron binding sites. of these domains contains an iron binding site.

Structure of trandferrins
The transferrins of all of the three classes described above T h e carbohydrate component of transferrins
are glycoproteins of molecular mass around 80 kDa which The transferrins are glycoproteins. Human serotransferrin
have the capacity to bind two atoms of ferric iron in associ- contains two branched oligosaccharide chains attached to the
ation with the binding of an anion, usually bicarbonate (for asparagine residues 415 and 608. The structure of the glycan,
reviews see [60, 611). The form of the transferrin molecule, as determined by chemical and enzymatic techniques and con-
deduced from preliminary X-ray crystallographic studies of
firmed by NMR and mass spectrometry [78 - 801 is given in
human and rabbit serotransferrin [62 - 641 and human
Fig. 8. The glycosylation sites conform to the rule that the
lactoferrin [65] and from hydrodynamic data [66], is that of
sequence which codes for glycosylation is the tripeptide Asn-
an ellipsoid of revolution with a ratio of 2 : l between the
major and minor axes. The rabbit serotransferrin molecule
Xaa-(Ser/Thr), namely Asn-Lys-Ser (residues 428 -430) and '
Asn-Val-Thr (635 - 637)'. Both are in the C-terminal half of
presents a clearly dilobal structure [64] but it is at present
the molecule, and no other potential glycosylation sites are
premature to suggest that the two lobes represent the two
apparent in the amino acid sequence [67-701. In human
distinct N- and C-terminal iron binding domains to which we
lactotransferrin [73] there are four potential sites for glycosyla-
will refer subsequently.
tion: Asn-Trp-Thr (137- 139), Asn-Ala-Ser (389-393), Asn-
The amino acid sequences of human serotransferrin [67 -
Gln-Thr (490 - 492) and Asn-Gly-Ser (635 - 637). Only two
701, of hen ovotransferrin [71, 721 and of human lacto- of them are glycosylated [73], namely the asparagine residues
transferrin [73] have been determined and are presented in at positions 137 and 490 of the lactoferrin sequence. Hen
Fig. 7. The sequence homology between human sero-
ovotransferrin contains three sequences in the C-terminal half
transferrin and human lactotransferrin or hen ovotransferrin of the molecule respectively Asn-Arg-Thr (490 -492) ', Asn-
is 59% and 51 7'0 and between human lactotransferrin and hen Gly-Ser (635 -637)' and Asn-Pro-Ser (690-692)', which
ovotransferrin is 49%. The homology is more pronounced in could serve as sites for glycosylation. Only one of them
the C-terminal half of the molecule in all three cases. When appears to be glycosylated, namely asparagine 490. We might
the N-terminal and C-terminal halves of the three transferrin conclude from this that the presence of a residue Xaa which
molecules are compared there is clear evidence of internal has a sufficiently small side chain, and thus permits flexibility
homology: 41 '/O for human serotransferrin, 37% for human of the polypeptide chain (Ala or Gly) or an amino acid which
lactotransferrin and 33% for hen ovotransferrin. For human imposes rigidity to the main chain (Pro) blocks the action of
lactoferrin [73] clear evidence was found for a sequence hom- the glycosylation in Asn.
ology of the N- and C-terminal halves (1 - 338; 339 - 703) by
the method of Staden [74], but no evidence was found for the ' The numbering of rcsidues in human serotransferrin and hen
fourfold or sixfold internal homology previously reported [71, ovotransferrin are based on the sequence of human lactotransferrin
75, 761. However, results obtained using the method of Smith (Fig. 7).
492

1
I 4&1

Fig. 9. Localisation of the disulfide bridges of human lactotransferrin ( L F T ) , human serotransferrin (STF) , and hen ovotransferrin (OTFj.
Reproduced with permission from Metz-Boutigue et al. [73]

The biantennary glycan given in Fig. 8 is found in both The iron and anion binding sites of transferrins
glycosylation sites A and B of human serotransferrin. Howev- The binding of Fe3+ to each of the two binding sites in
er, in addition, other structures, notably triantennary glycans, the N- and C-terminal domains of the transferrin molecule
are also found in normal human serum transferrin [81, 821 is accompanied by the binding of an anion (carbonate or
albeit in much smaller amounts. The structures of the human bicarbonate) and by the release of three protons. Whether
lactoferrin glycans [83] and of the ovotransferrin glycan [84] these latter are derived from hydrolysis of water coordinated
are also given in Fig. 8. While the core glycan remains the to the iron, from ionisable groups of the protein, or from both
same as for serotransferrin, there are quite substantial sources remains unclear [60]. The consequence of iron binding
differences in the degree of substitution, notably fucosylation is that the net negative charge of the transferrin molecule is
of GlcNAc residues in lactotransferrin, and introduction of increased by one unit for each iron bound [87]. These early
GlcNAc on mannose residues in ovotransferrin. studies of iron binding [87] suggested cooperativity between
the sites of ovotransferrin such that the affinity of the second
site for iron was increased by two orders of magnitude
Disulphide localisation in transferrins
following binding of iron to the first site. However, subsequent
The distribution of half-cystine residues and, for that studies on human serotransferrin indicated that iron binding
matter, of disulphide bridges in serotransferrin, lactotrans- was equivalent for the two sites [88-901. A number of
ferrin and ovotransferrin is remarkably well conserved spectroscopic studies (reviewed in 1601) however revealed
(Fig. 9). Thus, in the N-terminal half of the molecule, six differences between the two metal binding sites of ovo-
disulphide bridges (numbered 1 - 6 according to Williams transferrin. This view was substantiated by results which
[SS]) are found in essentially similar positions in all three showed that the one of the two iron binding sites of human
molecules. The same is also true of nine disulphide bridges in serotransferrin was much more susceptible to release by pro-
the C-terminal half of the molecule (for the clarity of the tons than the other [91, 921, and by the demonstration [93]
discussion, we ignore a number of additional disulphides pres- that the N- and C-terminal domains of ovotransferrin are
ent in one or other of the transferrins). Of these nine di- occupied by iron in a clearly non-equivalent manner. A
sulphides, six are in more or less homologous positions with detailed analysis was undertaken by Aisen, taking advantage
those of the N-terminal half of the molecule. On the basis of of the method developed by Makey and Seal [94] which, for
these data, Williams has proposed [85]that the precursor of the first time, enabled the four distinct species of transferrin,
transferrin as we know it today was a molecule which re- namely apotransferrin, monoferric transferrin A, monoferric
sembled the N-terminal half of the molecule with a molecular transferrin B and diferric transferrin, to be separated one from
mass of 40 kDa, one iron binding site and six disulphide the other on polyacrylamide gels in the presence of urea.
bridges. To explain the appearance of the product of gene Whereas at pH 7.4 the affinity of the C-terminal site is 5 -6
duplication that we find in the present day transferrins, he times greater than that of the N-terminal site, the difference
suggests that the 40-kDa protein was too small to avoid elimi- at pH 6.7 is 20 times [95]. The affinity of the two sites in
nation from the circulation by the kidneys, and cites results circulating serotransferrin under physiological conditions
with N- and C-terminal domains of ovotransferrin 1861which ranges over 1-6 x 10” M-’ [961.
confirm this hypothesis. The identification of the amino acid residues involved in
In conclusion, the conservative dispositon of the di- the iron and anion binding sites must ultimately await the
sulphide bridges in transferrins strongly supports the idea determination of the three-dimensional structure from X-ray
that the transferrins are the product of gene duplication and crystallographic studies. However, in view of the fact that the
reinforces the two-domain nature of these proteins. EPR and optical spectra of different transferrins are very
493

similar [88, 971 and that the amino acid residues which for the preparation of 59Fe-labelled transferrins [110, 1111,
constitute these sites would be expected to be conserved in the iron binding appears to involve a two-step process. First a
course of evolution, it is not unreasonable to attempt at pres- spectroscopically distinct and identifiable ternary complex of
ent to identify them. On the basis of results obtained by a ferric nitrilotriacetate and the protein is formed, in which
number of techniques, it seems likely that the iron ligands in the anion binding site is occupied by nitrilotriacetate [112].
transferrins involve two histidine residues [98, 991, two or Thereafter, in a much slower reaction, the bicarbonate ion
three tyrosine residues [loo, 1011, a water molecule or a replaces nitrilotriacetate at the anion binding site to give the
hydroxide ion [loo, 1021, and a carbonate or bicarbonate physiological form of transferrin. While this model of iron
anion [loo, 1031. This latter is bound not only to the metal binding may be true for ferric nitrilotriacetate, it is by no
but also to the protein: chemical modification studies [lo41 means sure that it applies to ferric citrate or for that matter
imply that an arginine is involved, whereas NMR to any other as yet unidentified physiological ferric chelate.
spectroscopy suggests that histidine is the anion binding An alternative model for iron binding based on the obser-
ligand of the protein [loll. vation that bicarbonate and oxalate bind, albeit weakly, to
A tentative analysis of the iron and anion binding sites of apotransferrin [113], assumes that anion binding precedes iron
transferrins was made by Chasteen [lo51 and in the present binding [114]. Anion binding neutralises positively charged
analysis we have included the sequence data of the human groups on the protein and thus facilitates the approach of
lactotransferrin molecule [65]. Fe(II1) or Fe(1I) (the latter is subsequently autoxidised) to the
The conclusions of Chasteen are in part supported and binding sites. The protonated groups in the vicinity of the
clarified by the inclusion of the human lactoferrin sequence metal binding site may be the source of some of the protons
(Fig. 7). For the two histidine ligands it seems likely that they which are released when apotransferrin binds iron.
are 117 and 252 for the N-domain and 472 and 609 for the C- The release of iron from transferrin is most readily
domain (all three proteins have histidines at these positions) achieved in vitro by lowering the pH to values of between
and that the histidine residues at positions 210 and 558 are 5.0 - 5.5 in the presence of appropriate Fe(II1) chelators,
not ligands since in human lactoferrin they are replaced by although for lactotransferrin much more extreme pH values
glutamate and aspartate respectively. The assigment of of 2 - 3 are required for iron release [115]. It is possible that
tyrosine ligands proposed by Chasteen (188 and 191 for the protonation of the anion in the metal binding site destabilises
N-domain, 538 and 540 for the C-domain) is compromised the iron-anion-protein ternary complex and that this leads to
by the presence of a lysine residue at position 188 of the release of the anion and of the metal. As we shall see in the
lactoferrin sequence, and also by the presence of tyrosine next part of this review, the physiological consequences of the
residues in all three transferrin sequences for both N- and C- inclusion of transferrin within an acidic intracellular vesicle
terminal domains at positions 83 and 427, and at positions 93 results de facto in the release of its iron within the cell.
and 447. Since the tyrosine residues at positions 93, 188 and A number of other models have been proposed for trans-
191 in ovotransferrin are protected by iron from reaction with ferrin iron release. One involves direct chelation of the trans-
tetranitromethane [loll, we might speculate that perhaps the ferrin iron by stronger iron chelators such as bacterial
two tyrosine ligands are those at positions 93 and 191 for the siderophores. However, whereas catechol-based siderophores
N-domain, and those at positions 447 and 540 for the C- and synthetic analogues are both thermodynamically and
domain. kinetically capable of removing transferrin iron [116, 1171,
For the arginine residues involved in the binding of the desferrioxamine B although thermodynamically capable, is
anion, Chasteen opts for the residues at positions 121 and 477 kinetically slow both in vivo and in vitro [118,119]. The kinetic
in the N- and C-domains, which are effectively conserved in all barrier to iron release by desferrioxamine B can be overcome
three transferrin sequences. The involvement of the arginine by a number of iron-chelating anions of which pyrophosphate
residues at positions 235 and 257 in the N-domain, and at and organic pyrophosphates are most effective [120, 1211. The
positions 592 and 614 in the C-domain, which Chasteen re- introduction of a catechol group on the terminal amino group
cognises cannot be excluded and which are conserved in all of desferrioxamine B gave a derivative which was kinetically
three transferrin molecules, might be eliminated by the prox- able to remove transferrin iron [122]. The cationic groups near
imity of conserved histidine residues at positions 117 and 472, the metal binding site may serve to direct anionic chelating
thereby accounting for the perturbation of the proton NMR agents to the iron and participate in recently observed
spectra of histidine residues concomitant on iron and anion quaternary complexes of the type chelate-iron-transferrin-
binding [99]. carbonate [123].
Mechanisms involving reducing agents have also been pro-
posed [124, 1251 although their physiological relevance re-
Transferrin iron uptake and release
mains unclear (in view of the nature of the reducing agents
The form in which iron is presented to the apotransferrin employed).
molecule in vivo is unknown. From in vitro studies it appears
that Fe(I1) and Fe(II1) can serve as a source of transferrin
Transferrin iron uptake by cells
iron. The former is autoxidised by the protein through a
chelate effect [106]; Fe(I1) is poorly bound by the protein, if In a paper published in 1963, Jandl and Katz [126] pro-
at all [107], whereas Fe(II1) is very tightly bound. When simple vided convincing evidence that doubly labelled transferrin
ferric salts are used they hydrolyse, polymerise to complex delivered its iron to reticulocytes by a process in which the
polynuclears and result in non-specific binding [log]. This can protein remained to a large extent in the stroma whereas the
be avoided by the use of ferric chelates or freshly prepared iron was internalised within the cells. This plasma to cell
solutions of ferrous salts [109]. An excess of HCO; in solution cycle of transferrin could best be explained by a receptor for
ensures the occupation of the anion binding site by the physio- transferrin on the plasma membrane of the cel!s which allowed
logical anion, and furthermore ensures specific binding [109]. the iron-loaded transferrin molecule to bind to the cell surface,
In the case of ferric nitrilotriacetate, which is often employed release its iron and thereafter the iron-depleted protein mole-
494

cule would be released to renew its search for iron and to


initiate a new cycle of transferrin-mediated iron uptake.
As we will see in what follows, the fundamental principle
of the transferrin cycle has proved to be valid for almost all
cell types which have been examined, and has been clarified
in a great many of its details for an already impressive number
of different cells. However, before discussing in detail the
mechanism by which cells take up transferrin, release its iron
90
within the cell, and recycle the apotransferrin back to the
circulation, we might well ask the question: how do cells
regulate their requirement for iron? The answer comes from
an elegant study by Morgan and his colleagues [127]. An
analysis of the number of transferrin receptors and the rate of
transferrin-bound iron uptake by various immature erythroid
cell precursors showed that during erythroid cell development
I
the number of transferrin receptors increased from 300000
per cell at the early normoblast stage to a maximum of 800000
per cell on intermediate normoblasts. The further maturation
of intermediate normoblasts was accompanied by a decline in
the number of transferrin receptors, which attained a level of Fig. 10. The human serotransferrin receptor. Reproduced with
105000 per cell in the circulating reticulocyte (and which permission from Newinan et al. [132]. Numbers on the right indicate
declines to almost imperceptible levels in mature red blood molecular mass in kDa
cells). A close correlation was observed between the number
of transferrin receptors and the iron uptake from transferrin
during erythroid cell development, and it was found that in
all of the immature erythroid cell populations studied the rate The gene for the human transferrin receptor has been mapped
of iron uptake was about 36 iron atoms per receptor per hour. on chromosome 3 [138, 1391 and two cDNA clones for the
The conclusion of the authors, and their circumspection in receptor have recently been isolated [140]. The gene for trans-
expressing it, merits citation: 'These results indicate that the ferrin is also on chromosome 3 [138] as is the gene for the
level of transferrin receptors may be the major factor which melanoma cell-surface antigen, p97, which has sequence ho-
determines the rate of iron uptake during erythroid cell mology with transferrin and binds iron [141]. The avian bursa1
development'. We can only add that this conclusion seems lymphoma oncogene (B-lym) codes for a polypeptide that is
likely to be true for other cell types than those of the erythron, structurally related to transferrin, lactoferrin and p97 [142].
since we know that many tumour cells express much greater
numbers of transferrin receptors than do the corresponding
normal cells, and that their reaction to a culture medium The transferrin cell cycle
which seeks to deprive them of iron is to increase the number As we have seen earlier, the idea of a plasma-to-cell cycle
of transferrin receptors at their cell surface severalfold [128]. of transferrin was first evoked by Jandl and Katz [126]. The
However, we do not yet know the mechanisms by which a half-life of human serotransferrin is 7-9 days [143]. When we
mammalian cell regulates its expression of transferrin re- take into account that 30-40 mg iron is exchanged between
ceptors as a function of its needs for iron. different cellular compartments in man each day [144], it is
immediately apparent that the transferrin molecule must be
recycled intact many times in the course of delivery of iron to
The transferrin receptor
cells.
The discovery of the transferrin receptor resulted from Our ideas concerning the serotransferrin cell cycle have
the preparation of monoclonal antibodies against a human evolved gradually over the past decade since the idea that iron
erythroid leukemia cell line. The monoclonal B3/25 was in release from transferrin might take place in a vesicle with a
fact found to be directed against the transferrin receptor [129], localised low pH was first mooted [145]. It was assumed from
as was a monoclonal anti-T cell reagent OKT 9 [130]. Indeed, early studies that transferrin uptake was only dependent on
it seems likely that many monoclonal antibodies directed the interaction with receptors at the cell surface [126], and this
against tumour cell lines are directed against the transferrin view point continues to receive support as we will see later.
receptor [131]. The structural features of the transferrin re- However, the fact that a number of features of the process of
ceptor are depicted in Fig. 10 (from [132]).The model is based transferrin and iron uptake were similar to those observed in
on data published in [129, 130, 132-1371. The receptor is a the endocytosis of other proteins by cells led to the suggestion
disulphide-linked dimer of molecular mass 180 kDa which that transferrin might enter immature red cells prior to the
presents two transferrin binding sites. Trypsin digestion release of its iron for haem synthesis [146]. Evidence was
liberates a 70-kDa fragment whch retains the capacity to bind accumulated which showed that subsequent to binding of the
transferrin and which is also able to bind the monoclonal iron-loaded transferrin to its plasma membrane receptor, the
antibodies B3/25 and OKT 9. The receptor has N-asparagine- transferrin-receptor complex entered erythroid cells by endo-
linked glycans probably composed of two high-mannose cytosis [145- 1501.
chains and one complex chain. The antigenic determinants What might be the intracellular pathway of the transferrin-
which react with OKT 9 and B3/25 are part of the polypeptide receptor complex which results in release of its iron within the
and are not the glycans. In the internal part of the receptor cell and in the recycling of the apoprotein back to the plasma
the transmembrane tail of molecular mass 5 kDa possesses membrane? Using antibodies directed against the plasma
phosphoryl serine residues and a covalently bound fatty acid. membrane of cultured rat fibroblasts it was concluded that
49 5
cytoplasm. In contrast to many other ligand-receptor com-
plexes, the apotransferrin-receptor complex retains a high
stability at acidicpH such that after its transition in the CURL
(compartment of uncoupling of ligand and receptor) it is
recycled back to the plasma membrane. At the neutral or
slightly alkaline pH of the extracellular fluid, apotransferrin
has a lowered affinity for its receptor, is released, and can
return to the circulation in search of iron.
That this mechanism occurs in other normal cell types is
supported by studies on erythroid, placental and hepatic cells
[170, 176- 1841. In rat liver most of the transferrin was found
in the hepatocytes with little in other cell types (Kupffer cells,
endothelial cells, fat cells) and the iron was found to be rapidly
incorporated into mitochondria1 and cytosolic ferritin. Evi-
dence was also found for the presence of transferrin within
a low-density membrane fraction [176, 1771. The uptake of
< Receptor transferrin by reticulocytes occurs by endocytosis through
It, Oiferric tronsferrin
coated pits [178,179] and the rate of endocytosis of transferrin
0 Apotronsferrin
+ Fe
is equivalent to that of iron uptake [160]. Further, the inhibi-
tion of iron uptake by inhibitors of endocytosis is pro-
Fig. 11. Transferrin uptake and recycling in human tumor cell: the portional to the inhibition of transferrin endocytosis [l SO] and
transferrin cell cycle. Reproduced with permission from Dautry- both transferrin and its receptor are recycled to the cell surface
Varsat et al. [I711 by exocytosis [181] with a recycling time of 3-4 min at 37°C
[160]. That receptor binding [182] and endocytosis [183] is also
involved in transferrin and iron uptake by placenta is also
well established.
plasma membrane fragments, after internalisation, gain access However in placenta and in hepatocytes there seem to be
to lysosomes and are thereafter recycled back to the cell two intracellular pathways of endocytosed transferrin, one
surface [151]. By analogy, we proposed that the transferrin- recycling rapidly (and responsable for the bulk of the iron
receptor complex passed into the lysosomal compartment uptake) and a second recycling more slowly [184].
where its iron was released, and that the apotransferrin mole- As has been noted above, if recent results seem to point
cule was returned to the cell surface and discharged into to the role of this transferrin cycle in many cell types, both
the extracellular medium [152]. Further, it was shown that normal and tumoral, there are still reports in the literature
a number of drugs which affect endocytosis and lysosomal which imply that at least part of iron release from transferrin
function greatly reduce iron uptake from transferrin by takes place at the plasma membrane, based on studies with
fibroblasts without significantly affecting the uptake of trans- rat and rabbit reticulocytes [185 - 1921.
ferrin protein [152 - 1541. Similar results were obtained from
studies on developmentally totipotent teratcarcinoma stem
cells [155] and it was again concluded that the lysosome was PROTEINS OF IRON STORAGF
an important intermediate in the iron uptake pathway. In Introduction
parallel investigations on reticulocytes, the group of Morgan
found support for the hypothesis that iron release from trans- In animals, the major pool of non-haem iron (ap-
ferrin occurred as the result of protonation of transferrin proximately 25% of total body iron) is stored in two
within acidic intracellular vesicles [156- 1611. Similar effects compounds: ferritin and haeinosiderin [143]. Ferritin was first
of lysosomotropic agents were observed in human leukemic crystallised from horse spleen by Laufberger in 1935 and in a
and hepatoma cell lines [162,163]. That the intracellular acidic detailed publication [193] he described its purification, its high
compartment was nonlysosomal in nature was established by iron content (over 20% by weight) and suggested that it was
Percoll gradient fractionation of postnuclear supernatants in probably a substance that served as an iron depot. It has a
K562 human erythroleukemic cells [164]. The intracellular widespread distribution in all kinds of living organisms; its
route of transferrin and its receptor via a 'short circuit' re- presence has been reported in mammals, birds, fish, in-
cycling ligand and receptor to the cell surface was found in a vertebrates, plants, fungi and in prokaryotes such as E. coli
number of tumour cells [165-1691. and Azotobucter sp (reviewed in [194-1981). Ferritin is also
The effect of pH on the binding of transferrin and present in serum, where it was first discovered as the active
apotransferrin to its receptor on rabbit reticulocytes and on component of a vasopressor substance [199]; its determination
the human tumour cell lines HepG2 and K562 was analysed in serum is useful in the assessment of iron storage status
and led to the formulation of the transferrin-cell cycle which [200]. Haemosiderin was first isolated from horse spleen in
is illustrated in Fig. 11 [170- 1721. Subsequent to the binding 1929 as insoluble red-brown granules [201]. Both ferritin and
of the iron-loaded transferrin to its receptor, the complex is haemosiderin have a high and variable content of iron: the
internalised, with formation of a coated pit and a coated former is found in the cytoplasm in a soluble form whereas
vesicle [173-1751 which sheds its clathrin to become an the latter is present as insoluble granules within secondary
endosome. An energy-dependent proton pump acidifies the lysosomes. Both iron storage compounds consist of an in-
pH of the endocytic vesicle to pH 5.5-5.0. Under these organic core, the iron mineral of which has a rather similar
conditions, but probably also with the help of an as yet un- composition and structure, and a protein component, which
identified intraendosomal chelator, the iron, bound to the is well characterised in the case of ferritin, but remains ill-
transferrin-receptor complex, is released and passes into the defined for haemosiderin.
496
copy [220]which are both compatible with a direct interaction
between the core and the protein shell. As we will see this
would also account for the presence of a number of lysine and
arginine residues in the interior of the apoferritin molecule
[215] which could interact with phosphate residues on the
surface of the iron core and help to bind the hydrated iron
oxide to the inner surface of the protein.
Haemosiderin has considerable spectroscopic and mor-
phological similarities to the ferritin iron core [221-2241 de-
spite its much higher ironlprotein ratio.
One hypothesis is that soluble cytosol ferritin is pro-
gressively transferred to and accumulated within scondary
. . . . . . . . . . . . . . . . . . . . .
lysosomes: the following sequence of events has been pro-
posed for the formation of haemosiderin [221]:
Fig. 12. Structure of ferrihydride. Reproduced with permission from
Ford et al. [264] Cytosol ferritin + lysosome-siderosome ferritin
1
denaturation of ferritin protein
(loss of solubility, aggregation)
Ferritin structure: the mineral core 5-
proteases
As we have noted above, the ferritin molecule consists of
two components: a mineral core of hydrated ferric oxide and
5-
decomposition of ferritin protein
a multi-subunit protein shell which englobes the former and
thereby assures its solubility in an aqueous environment. The 1
divested (FeO . OH) cores
ferritin molecule can maintain a concentration of 1 M ferric
iron in solution, in contrast with the extremely poor solubility 5-
core disintegration
of the variety of sols, gels, amorphous or crystalline precipates
that emerge from the hydrolysis of iron(II1) in aqueous solu- 1
amorphous haemosiderin (FeO ' OH).
tion [4].
The mineral core of ferritin is an inorganic hydrated This would be consistent with data which indicate the
iron(II1) oxide of variable iron content containing some presence in human and horse ferritins of high-molecular-mass
phosphate: for the ferritin of horse spleen an atomic ratio, polymers which are less soluble and more susceptible to pre-
Fe/P = 9, was reported in early studies [202] whereas a more cipitation than ferritin monomers and which have an iron/
recent investigation [203] found ratios of 21 for human spleen protein ratio consistent with loss of protein from this ferritin
ferritin and of only 1.7 for bacterial ferritin from Pseudomonas [225].
aeruginosa. The iron content of the horse spleen ferritin mole-
cule varies from 0 to a maximum of 4500 atoms [204]. Re-
Apoferritin: the protein subunit
constitution of ferritin in the absence of phosphate has no
effect on the X-ray or electron diffraction pattern [205], and Treatment of ferritin with reducing agents such as dithio-
treatment of ferritin with 1 M NaOH, which provokes the nite, together with an appropriate chelator of Fe(I1) [226]
dissociation of the protein shell, results in the loss of more or thioglycollate [ 1941, followed dialysis or gel permeation
than 80% of the inorganic phosphate originally associated chromatography to remove the iron-chelate complex and the
with the iron core [206]. Analysis of the X-ray and the electron reductant yields the intact protein shell, apoferritin. Native
diffraction patterns of ferritin iron cores 1207-2101 indicate apoferritin can be prepared by differential or density gradient
that the ferritin iron core has a structure which resembles that centrifugation of ferritin solutions. Both types of apoferritin
of the mineral ferrihydrite [211, 2121; it is illustrated in Fig. 12. are closely similar as judged by X-ray diffraction [204, 2271,
This mineral, (5Fe203 . 9H20), which is formed by the slow circular dichroism and hydrodynamic properties [228]. Most
heating of Fe(N03)3 . 9 H 2 0 at 80 - 85 "C [207], has been structural as well as functional studies have been carried out
found in the walls of old mine workings as a relatively recent with chemically prepared apoferritin. Horse spleen apoferritin
deposit [212]. The unit cell contains four oxygen layers (in- has a molecular mass of 476 kDa and a subunit molecular
stead of six in the closely related haematite structure) and the mass, based on the amino acid sequence [229], of 19824 Da.
Fe(II1) sites are underpopulated with a stoichiometry of 5Fe : All ferritins that have been characterised have the same 24-
120 (instead of 2: 3 in haematite). Alternative structures based subunit structure, although bacterial ferritins have a rather
on radial X-ray scattering [213] or on extended X-ray absorp- smaller subunit molecular mass, 15 kDa for the E. coli protein
tion fine structure (EXAFS) measurements [214, 2151 are ex- [230] and 17 kDa for that isolated from Azotobacter species
cluded by the absence of octahedral iron and incompatibility [231, 2321. Bacterioferritins are also unusual in that they
with the X-ray and electron diffraction patterns respectively. contain haem, and appear to be identical with cytochrome bl
The localization of the phosphate ions in the ferritin cores of E. coli [230, 2331 and with a h-type cytochrome of Azoto-
remains unknown. In vitro it can be replaced by dissolved bacter vinelandii [231, 2341 and of A . chroococcum [232].
phosphate ions [216, 2171. However, inclusion of phosphate The apoferritin shell can aggregate to give dimers, trimers
in the reaction mixture yields a more monodisperse core size etc. which are often observed on polyacrylamide gel electro-
[216] and a smaller apparent size when measured by Moss- phoresis. It is one of the more elementary criteria of the purity
bauer spectroscopy [218]. The phosphate ions might replace of ferritin preparations that all the protein bands on non-
surface hydroxyl functions. This could explain the evidence denaturing gels should stain not only for protein but should
from neutron low-angle scattering [219] and electron micros- also give a positive Prussian blue reaction. On SDS/polyacryl-
497

amide gels, purified ferritin preparations frequently present I = horse jpieen A p o l e r r i t i n Trorfi (229)
bands of lower molecular mass than the apoferritin subunit I1 = Hurnari H chairi from (254)
(6 - 8, 11 - 13 and 14- 15 kDa), which appear to result from
proteolytic cleavage of the 19-kDa subunit [235- 2391. Fur- I ~ I = H u r l x i L chain frorii (254)
ther heterogeneity is apparent on SDS gels of mammalian IV = Txlpoio Rerl c e l l A p o f e r r i t i n from (257)
ferritins since in addition to the 19-kDa subunit a second V = Rat Liver L chain f r o m (252)
subunit of 21 kDa is frequently observed; these two subunits
are designated H (heavy, 21 kDa, predominant in heart) and
L (light, 19 kDa, predominant in liver and spleen); tissue
ferritins are assumed to be a mixture of the two
homopolymers, HZ4 and LZ4, and the 23 heteropolymers
HZ3L1-H1LZ3 [240-2461. The multiple bands observed on
isoelectric focussing of tissue ferritins can be explained by this
model if we assume that H and L differ not only in size but
also in their isoelectric points; the ratio of H/L subunits is
both species- and tissue-specific and varies from 1 to 9 in
horse spleen and in human liver [247, 2481 to 8.5 to 1 in horse
heart ferritin [248]. Human H chains are more acidic than L
chains [240], whereas the inverse is the case for horse ferritins
[240, 2481.

Apoferritin: primary structure and comparative studies


The amino acid sequence of horse spleen apoferritin was
first established in 1981 [229] after some 12 years of effort.
Thereafter direct protein sequencing methods established the
primary structure of the human spleen L subunit and of some
70 residues of a minor sequence, assumed to be the H subunit
[249], and of the human liver L chain [250] as well as partial
sequences of human placenta ferritin and of the ferritin of
HeLa cells [251]. cDNA methods have subsequently estab-
lished the sequence of a rat liver L subunit [252], of human
liver H subunit [253,254], human liver L subunit [254], human
lymphocyte H subunit [255] (partial sequence), human mono-
cyte-like cell line U937 L subunit [256] and tadpole red cell
ferritin [257]. The sequences are presented in Fig. 13. The
cDNA studies indicated that both H and L subunits are
encoded by a family of genes in human [253] and rat [258].
However, more recent studies [259,260], including the cloning
and sequencing of the gene coding for human apoferritin H
subunit [260], has established the presence of a number of
pseudogenes. The apoferritin H gene has three introns and its I
exon sequence is identical to that of cDNAs isolated from I!
human liver, lymphocytes, HeLa cells and endothelial cells. I;!
There are at least 15 intronless pseudogenes whose features
1V
suggest that they originated by reverse transcription and inser-
tion; the authors conclude that there is probably only one
gene coding for the human apoferrin H subunit [260]. The
gene for ferritin has been assigned to chromosome 19 [261]
and for the H chain to chromosome 11 [262]. However more
recently ferritin genes have been found on 12 different 151 170 174 178
chromosomes [263]. It is normal, since ferritin is a housekeep-
ing gene, that it would be expressed in the germline of the LGEYLFERL~LKH~
species and so be available for reverse transcription, incorpor- 4----0KH--GDSONES
ation into the genome and transmission to the progeny. The
presence of so many pseudogene copies of the ferritin subunit
tl-----DKH-flGESS
makes the identification of active ferritin genes on chro-
mosomes difficult [259].
Comparison of the amino acid sequences reveals that
horse, human and rat L subunit sequences are considerably
more homologous than are the human L and H sequences:
there are only 14% substitutions between human and horse
L sequences, compared with 45% substitutions for the two
human subunits. It is difficult to assign the tadpole sequence
as H- or L-like since it is almost as similar to the human H Fig. 13. Amino acids sequences of ferritins
498

1
I Table 1. Structural regions of the apoferritin subunit
Reproduced with permission from Rice et al. [265]

Region Residues Region Residues

N-terminal loop 1-9 CD turn 121 - 123


A helix 10-39 D helix 124-155
AB turn 40 - 44 DE turn 156-159
B helix 45 - 72 E helix 160-169
L loop 73-91 T C-terminal tail 170- 174
C helix 92-120

and D (124- 155), with a shorter fifth helix E (160- 169). The
Fig. 14. Packing of the 24 subunits of the apojerritin molecule. Re- N-terminus, N, iies at the opposite end of the subunit to E,
produced with permission from Ford et al. [264] which is disposed at an acute angle to the main bundle of
helices. The loop L joins helices B and C; together with its
related counterpart in a neighbouring subunit it forms a sec-
tion of antiparallel /3-sheet within the dimer. The pairs of
helices A, B and C, D are connected by short turns such
that these helices are antiparallel. In the view of the subunit
presented in Fig. 15, the helices B and D have one face towards
the inside of the protein shell, and A and C have one face
towards the outside of the molecule. The loop L is also at the
outer surface.
Within the subunit the interior helices and their side chains
interact to form a tightly packed hydrophobic core over a
distance of some 3.5 nm. In the middle of the helical bundle
the character of the interior changes and we find several buried
polar or hydrophilic residues, Tyr-23, Lys-58, Glu-103 and
Glu-I 37, which form a network of hydrogen bonds with a salt
bridge between Lys-58 and Glu-103. The longest helix, D,
extends beyond the main helical bundle and the chain folds
back so that the helix E is disposed at an acute angle to the
Fig. 15. The three-dimensional structure of the subunit of the helical bundle. It interacts with the subunit through hydro-
apoferritin. Reproduced with permission from Ford et al. [264] phobic interactions involving Phe-166 and Leu-171 and
hydrogen bonds involving Glu-163. The structural regions of
the apoferritin subunit are summarised in Table 1. In the
intact apoferritin molecule there are two types of channels
sequence (67% homology) as to the human L chain (59% through which small molecules or ions could gain access to
homology). The human H subunit has only four amino acid the central cavity of the molecule. These channels are located
residues more than its L equivalent and yet has an apparent along the threefold and fourfold axes of symmetry and have
molecular mass of 2 kDa more as judged by SDS-PAGE. very different characters. The six fourfold channels are very
The rat L subunit has an eight-residue insertion near the C- hydrophobic (Fig. 16A). The symmetrical arrangement of
terminus compared to the other two L chains (with which four E helices buries a number of hydrophobic residues,
it has homology of 88% and 82% with horse and human notably Leu-161, Leu-165 and Leu-169. The channel is 0.3-
respectively). Surprisingly direct sequence analysis of peptides 0.4 nm in diameter and some 1.2 nm long. In contrast, the
derived from rat liver H subunits revealed that the H-subunit side-chain residues which project into the eight threefold
sequence did not contain this octapeptide. When we compare channels (Fig. 16B) are polar or hydrophilic, notably Asp-
all of the available sequences the residues 2 - 8,26 - 39, 106- 127 and Glu-130. In the crystal these channels are occupied
114, 122- 131 and 160- 169 show considerable homology by metal ions (probably Cd2+ of crystallisation). One is
with mostly conservative changes. coordinated to the three Asp residues in the interior of the
channel and the other by the three Glu residues at the outer
surface.
Apojerritin: the three-dimensional structure There are two hydrophobic patches on the surface of the
Horse spleen apoferritin crystallises from aqueous subunit which are buried within the subunit-subunit contacts
solutions of CdS04 yielding large octahedral crystals with of the intact apoferritin shell. The most important involves
unit cell dimension a = 18.48 nm belonging to the cubic space residues of helix A (Val-20, Leu-24, Tyr-28, Leu-31) and of
group F432 [227]. The 24 subunits pack in 4: 3 :2 symmetry, the loop L (Phe-78, Leu-81, Pro-84). This hydrophobic patch,
as illustrated in Fig. 14. The three-dimensional structure has some 2.2 nm in length, is buried from the solvent by interac-
been solved at a resolution of 0.28 nm, the amino acid se- tion with the equivalent region in a second subunit to form a
quence [229] incorporated into the atomic model and the twofold symmetry axis. This no doubt explains the observa-
structure has been refined to an R value of 0.20 [264, 2651. tion that dimers are intermediates in the dissociation of
The apoferritin subunit (Fig. 15) consists of a bundle of four apoferritin [266].The other hydrophobic region on the surface
long a-helices: A (residues 10-39), B (45-72), C (92- 120) of the subunit contains one face of the E helix (Leu-161, Tyr-
499

Fig. 16. Stereoviews of the apoferritin shell around the threefold ( A ) and,fourfold ( B ) axes. Reproduced with permission from Ford et al. [264]

164, Leu-165, Leu-169 and Leu-154 close to the C-terminus residue insertion at residues 158 of the rat protein [270]. This
of the D helix). These residues are buried in the fourfold substitution is in the DE corner, where the electron density is
channels. weak, perhaps on account of its flexibility. There is however
It is clear from X-ray diffraction that the three-dimen- a region of negative density near residue 48, which is consistent
sional structures of ferritins from horse, human, rat, mouse, with the substitution of Cys in the horse protein by Gly in rat
dog and ferret liver and spleen are isomorphous [267]. liver and with the loss of a bound Cd2+ ion.
Separated H and L chains from human, rat and horse ferritins Mouse ferritin exhibits structural homology with horse
can form hybrid molecules [268,269] and the physicochemical spleen ferritin as judged by X-ray diffraction patterns [270]
properties of horse spleen, liver and heart ferritins, which although it is reported to contain almost exclusively heavy
contain respectively lo%, 40% and 85% of H subunits are chains [271]. It is suggested [270] that insertions in the se-
consistent with the hypothesis that they are hybrids composed quence may occur without disrupting the quaternary structure
of variable proportions of the two subunit types [248]. Thus, of the apoferritin: known sites of insertion are at the N-
it would seem that the tertiary and quaternary structure is terminus and the C-terminus of the human H chain and in
conserved to a large extent in both H and L chains. The the DE turn in the rat liver L chain (see Fig. 13).
difference electron density map (rat liver minus horse spleen) When the different apoferritin sequences presented in
shows very few features which can be attributed to the eight- Fig. 13 are compared with the horse spleen subunit structure,
500

it is clear that residues which are buried within the subunit gen) show that iron incorporation is a complex process,
are much more conserved than are residues exposed on the characterised by two distinct phases [280,282- 2841. The first
outer or inner surfaces. Thus, over half of the 60 buried slow stage corresponds to initial iron binding and oxidation,
residues of the subunit are invariant and of the remainder implying specific sites on the apoferritin molecule. This is
two-thirds of the replacements are conservative. About a third often referred to as the nucleation phase. The subsequent
of the external residues are invariant, and less than a third growth of the iron micelle is accompanied by further oxida-
of the residues on the internal surface are conserved. In all tion, hydrolysis and polymerisation. This second phase may
apoferritin subunits the sequence of residues 122- 131 is occur on the surface of the micelle, and hence no longer
conserved (with the exception of the substitution of Leu-129 require catalysis by the protein [280], or may still necessitate
by Ile); this sequence includes Asp-127 and Glu-130, potential an active role for the protein [281]. The initial phase is at
metal-binding sites at the exterior and interior of the threefold least second order in iron [283-2851, and this together with
channels. inhibition studies [284- 2861 suggests that the first steps in
In the horse spleen apoferritin molecule there are a great iron binding and oxidation involve a bimetallic site [287,288].
number of hydrogen bonds and salt bridges both within the Carboxylate functions are clearly implied in the catalysis of
subunit and at the interfaces between subunits, which most iron oxidation as indicated by chemical modification studies
probably contribute to the considerable thermostability of the [289] and by EXAFS [290]. An EPR study of iron accumula-
molecule. Almost half of these interactions are conserved in tion in apoferritin has identified what appears to be a spin-
all of the sequences and most of the others are replaced by coupled Fe(I1)-Fe(II1) dimer with a net electron spin of 1/2
alternative interactions. However, in the centre of the subunit, [291]. The binding stoichiometries of Mn(II), VO(1V) and
in a region rich in polar residues and flanked on either side by Cd(I1) to apoferritin are close to 0.3 and 0.7 metal ion per
two densely packed hydrophobic regions, there is considerable subunit [292], close to the values of 0.33 and 0.67 predicted
variability, such as the change of Lys-58 (which forms a salt for the binding of one and two metal ions respectively per
bridge with Glu-103 that certainly contributes in a important three subunits. This would be consistent with the binding of
way to the stability of the subunit) to Glu in the human H one or two metal ions in each of the eight hydrophilic channels
sequence. Conservation of residues in potential metal binding along the threefold axes: the X-ray crystallographic study
sites is discussed below. Although ferritin from E. coli has shows [293] that cadmium binding sites are found on the
been crystallised in the same space group as horse spleen outside and inside of these channels (constituted respectively
ferritin [230], a detailed structure is not yet available. However by three Glu-130 residues and three Asp-127 residues from
partial amino acid sequence data on this protein suggests that symmetry-related subunits).
it has no structural homology with mammalian ferritins [272]. We should however note here that if Fe(I1) appears to be
more readily assimilated by apoferritin in vivo than Fe(II1)
[294, 2951, the iron which is released from transferrin within
Iron deposition in ferritin the cell, as we have seen above, is ferric and must thus be
We can imagine at least two totally different mechanisms reduced prior to its incorporation into the storage protein.
for iron incorporation into ferritin, namely the assembly of An alternative might be that in vivo iron is present as a low-
apoferritin subunits around preformed micellar iron cores or molecular-mass polynuclear form, as has been proposed for
else the penetration of Fe(I1) into the apoferritin shell and its the iron utilised for haem synthesis [296,297] and that it is in
subsequent oxidation, hydrolysis and polymerisation to give this form that it is incorporated into apoferritin.
the ferric oxyhydroxide polymer.
The former was suggested by Saltman and his colla-
borators [273] on the basis of studies on the hydrolysis and Storage iron mobilisation
polymerisation of Fe(II1) chelates. They synthesized a poly- In vitro iron can be mobilised from ferritin by direct chela-
mer of approximate composition Fe403(OH)4 . (NO,), tion of Fe(II1) or by reduction in the presence of an ap-
. 1.5 H 2 0 which not only had the same diameter, 7.0 nm, as
propriate chelator of Fe(I1). Iron release is rather slow both
the ferritin iron core, but which also had similar spectroscopic for a number of biological chelators [298-3011 and with
properties [213, 2741. They showed that when the cor- synthetic chelators of Fe(II1) [300, 3021 or with microbial
responding ferric citrate micelle was placed in a solution of siderophores [303- 3051. However much more rapid rates
noncrystalline apoferritin the synthetic core was surrounded were observed for the chelation of ferritin iron under argon by
by the protein subunits to give a product similar to ferritin in dihydrolipoate and dihydrolipoamide [306]. In general much
its gross morphology [273]. However, since the biosynthesis more rapid rates of iron release are observed with reducing
of apoferritin precedes that of ferritin it is unlikely that such agents such as dithionite, dihydroflavins or thioglycollate
a mechanism is involved. [307- 3111in the presence of a$-bipyridyl. Mobilisation rates
Ferritin-like products are formed when apoferrin is in- are consistently more rapid at acidic than at neutral pH values
cubated with ferrous salts in the presence of appropriate [309], and it has been proposed that specific interactions occur
oxidants [276-2811. Neiderer [279] advanced the hypothesis between products or educt and the interfacial iron(II1)
that Fe(I1) ions could penetrate the protein shell of the hydroxide of the ferritin core [311]. Although it has been
apoferritin molecule where their oxidation would be catalysed suggested that dihydroflavins must penetrate into the interior
by the protein: the nascent ferric ions would form an in- of the protein shell in order to reduce iron [309], the recent
tramolecular precipitate of FeO . OH which would soon be study found no evidence of hindered shell penetration [311].
too big to escape. This view has received considerable support This is consistent with the observation that a number of quite
over the years, and a certain number of facts have been estab- large molecules and ions can gain access to the interior of the
lished, even if we have not yet identified in the three-dimen- ferritin molecule [204, 219, 312, 3131. Reductive mobilisation
sional structure of the protein the precise sites involved.
Kinetic studies using ferrous salts (usually ferrous ammonium Educt: a substance separated unchanged from another sub-
sulphate) with an appropriate oxidant (usually molecular oxy- stance; distinguished from product.
501
of ferritin iron by superoxide [314-3171 and by ascorbate in form haem. The source of iron for ferrochelatase might be
the presence of oxygen [318] have also been reported: the transferrin, ferritin or the transit iron pool. While it has been
former is extremely slow, while the latter is attributed to the shown in vitro that transferrin can readily supply iron for
monodehydroascorbate radical. haem synthesis by isolated mitochondria [324- 3261, in view
We might now pose the question as to what is the chemical of the transferrin-to-cell cycle presented above, it is difficult
nature of the ferritin iron mobilising agent in vivo. Of the to see how a transferrin molecule bearing iron could come
biological reductants, dihydroflavins would seem to be into physical contact with a mitochondrion. It has been
unlikely on account of the extremely low intracellular concen- suggested that an FMN-dependent ferriductase associated
tration of free flavins and the absence of evidence in animal with the mitochondria could enable electrons to be siphoned
cells of a flavin reductase. Likewise the concentrations of from the respiratory chain to reductively mobilise ferritin iron
lipoate and its derivatives within cells are many orders of for haem synthesis [327- 3301.
magnitude below those used in the iron mobilisation experi- This does not seem to be likely [297] since the proposed
ments. Superoxide, whether produced by xanthine oxidase in specificity of interaction between ferritin and the mito-
its NAD-dependent dehydrogenase form or by haemoglobin chondrion is not observed when homologous and
or myoglobin in the presence of oxygen seems unlikely not hererologous systems are compared, no specific binding of
only on account of the high levels of superoxide dismutase ferritin to the mitochondrion is observed and the rate of haem
within cells, but also on account of the potential danger to synthesis is essentially identical for mitoplasts and
the cell of Fenton chemistry, as discussed previously. mitochondria. This leaves the possibility that the presumed
Not only is the nature of the iron mobilising agent un- low-molecular-mass polynuclears constituting the transit iron
known, we do not know with any certainty where storage is pool [4,296] are the principal source of iron for ferrochelatase
mobilised within the cell. However, since we find that ferritin together with cytosol ferritin. There are also reports in the
and haemosiderin iron is more readily mobilised at acidic pH literature of a pool of non-haem, non-iron-sulphur mito-
values than at neutrality both by reductants and by chelators chondrial iron which represents about 25% of total
[311] (and R. R. Crichton and collaborators, unpublished mitochondrial iron and which could function as a proximate
observations), lysosomal haemosiderin iron might be a good iron donor to ferrochelatase in vivo [331- 3331.
target for therapeutic chelators in the treatment of secondary At present we know very little about the pathways of
iron overload. storage iron mobilisation from cells. In normal human sub-
jects some 40 mg of tissue iron is mobilised per day, 80% due
Intracellular iron metabolism to the recycling of iron derived from the catabolism of effete
red blood cells by the macrophages of the reticuloendothelial
Within the cell, iron is present in several forms and is system to the erythroid bone marrow for haemoglobin syn-
distributed within different intracellular compartments. We thesis [143]. Although the presence of specific receptors for
have already mentioned the storage forms ferritin (in large apotransferrin has been reported on activated peritoneal
part in the cytoplasm) and haemosiderin (in secondary macrophages [334], the presence of apotransferrin in the
lysosomes). There are haem proteins, iron-sulphur proteins culture medium does not appear to be essential for the excre-
and proteins such as ribonucleotide reductase and the aromat- tion of iron by the cells [335]. It seems that iron is released
ic amino acid hydroxylases which contain non-haem iron from macrophages as newly synthesized macrophage ferritin
which is not in an iron-sulphur cluster [319]. There is however which, in the culture medium, releases its iron to
an intermediate pool of chelatable or transit iron the apotransferrin [335]. This transfer from ferritin to apo-
appearances of which are ‘somewhat like the Loch Ness transferrin is stimulated by low-molecular-mass serum factors
monster, only to disappear from view before its presence, or (R. R. Crichton and Y. Jin, unpublished results).
indeed its nature, can be confirmed’ [320]. While it has been In conclusion, we may compare intracellular iron metab-
claimed that this might represent up to 20% of intrahepatic olism with a symphony orchestra: we can, in the absence of
iron [321], this seems rather unlikely and it is more probable the conductor, identify the different instruments but not the
that this pool represents only a very small amount of iron. It role that they will play. From the view point of the brass
is presumably composed of iron (ferrous of ferric) complexed section, it seems clear that the horns and trumpets of cytosol
by low-molecular-mass chelators and may correspond to the ferritin have the lion’s share of the symphony to be performed,
low-molecular-mass polynuclear forms of ferric chelates with a supporting role for the lysosomal trombones and tubas
found in the course of controlled hydrolysis and polymerisa- of haemosiderin. In the string section it seems obvious that
tion of these latter [4, 2961. This cytosol iron pool is often the violins of mitochondrial ferrochelatase reinforced by the
referred to as the transit iron pool based on the assumption counterpoint of the altos, cellos and double bases of haem
that it represents iron in transit between transferrin and degradation have the principal role to play. The sharply in-
ferritin [322]. flected tones of the flutes and clarinets of the transit iron pool
Intracellular iron metabolism. not only involves iron are supported by the bassoons and oboes of the Schnei-
uptake by the cell from transferrin, but also the supply of iron der formation who have surreptitiously introduced low-
within the cell for synthesis of iron-containing proteins and molecular-mass polynuclears. And in the face of all this
the mechanisms which allow iron to be excreted from the cell. confusion, the transferrin percussion section resonates all of
We have already considered the first point and we address the its forces against the cell membrane, uncertain whether they
others in the paragraphs which follow. want to get out or to get in . . .
The incorporation of iron into non-haem iron proteins is Once the conductor has taken his place in the middle of
very poorly studied and we will not consider it here. How- this chaos, we begin to hear clearly, and we realise that the
ever the terminal step in haem biosynthesis, catalysed by principal role will be given to each section of the orchestra as
ferrochelatase, is somewhat better established. This enzyme, a function of the music to be played. And that is probably
located in the matrix face of the inner mitochondrial mem- how intracellular iron metabolism is regulated: not by a pre-
brane [323], incorporates Fe(I1) into protoporphyrin IX to ponderant role of any one of the protagonists but by the
harmonious repartition of their contributions in order to 24. Frost, G. E. & Rosenberg, H. (1973) Biochim. Biophys. Acta
assure the harmony, homogeneity and homeostasis of the 330,90-101.
ensemble. 25. Neilands, J. B. (1982) Annu. Rev. Microbiol. 36, 285-309.
26. Brdun, V. (1985) in The enzymes of biological membranes vol. 3,
(Martonosi, A. N., ed.) pp. 617-652, Plenum Press, New
Epigram York.
27. Nikaido, H. (1979) in Bacterial outer membranes (Inouye, M.,
While we have learned much in the last few years about ed.) pp. 361 -407, Wiley, New York.
iron transport and storage, there remains an important 28. Luckey, M., Pollack, J . R., Wayne, R., Ames, B. N. & Neilands,
number of fundamental questions as yet unanswered. What J. B. (1972) J. Bacteriol. 111, 731 -738.
mechanism regulates the iron requirements of a given cell, 29. Wayne, R. & Neilands, J. B. (1975) J. Bacteriol. 121, 497-503.
whether by the secretion of siderophores or by the number of 30. Guterman, S. K. (1973) J . Bacteriol. 114, 1217-1224.
transferrin receptors expressed at the cell surface? How 31. Krone, W. J. A., Stegehuis, F., Konigstein, G., de Graaf, F.
is storage iron assimilated and released? What are the K. & Oudega, B. (1985) FEMS Microbiol. Lett. 26, 153-
161.
mechanisms involved in intracellular iron metabolism, and
32. Matzanke, B. F., Muller, G. I. & Raymond, K. N. (1984) Bio-
how is iron released from cells; the transferrin-cell cycle is chem. Biophys. Res. Comrnun. 121,922-930.
well established, but how does iron get out of the cellular 33. Fecker, L. & Braun, V. (1984) J . Bacteriol. 156, 1301-1314.
prison in which it is incarcerated? Since we wish to end with
34. Hantke, K. (1985) in Proteins of iron storage and transport (Spik,
an epigram, we can conclude that, the more we know about G., Montreuil, J., Crichton, R. R. & Mazurier, J., eds)
iron storage and transport - the more we need to know! pp. 231 -243, Elsevier, Amsterdam.
35. Ratledge, C., Patel, P. V. & Mundy, J. (1982) J . Gen. Microbiol.
128, 1559-1565.
36. Emery, T. (1971) Biochemistry 10, 1483- 1488.
REFERENCES 31. Carrano, C. J. &Raymond, K . N. (1978) J . Bacteriol. 136,69-
74.
1. Aisen, P. (1977) Ciba Found. Symp. 51, 1 - 14. 38. O’Brien, I. G., Cox, G. B. &Gibson, F. (1971) Biochim. Biophys.
2. Cotton. F. A. & Wilkinson. G. (1980) in Advanced inorganic
~I
Acta 237, 531-549.
chemistry 4th edn, pp. 750-751, Wiley-Interscience, “New 39 Langman, L., Young, 1. G., Frost, G. E., Rosenberg, H. &
York. Gibson, F. (1972) J . Bacteriol. 112, 1142-1149.
3. Hill, H. A. 0.(1982) in The biologicalchemistry of iron (Dunford, 40 Porra, R. J., Langman, L., Young, I. G. & Gibson, F. (1972)
H. B., Dolphin, D., Raymond, K. N . & Seiker, L., eds) pp. I - Arch. Biochem. Biophys. 153, 74-78.
12, Reidel, Dordrecht. 41 Emery, T. (1976) Biochemistry 15,2723-2728.
4. Schneider, W. & Schwyn, B. (1987) in Aquatic surface chemistry 42 Lodge, J. S., Gaines, C. G., Arceneaux, J. E. L. & Byers, B. R.
(Stumm, W., ed.) Wiley-Intersciences, New York, in the press. (1980) Biochem. Biophys. Res. Commun. 97,1291 - 1295.
5. Neilands, J. B. (1981) Annu. Rev. Biochem. 50, 715-731. 43 Coderre, P. E. & Earhart, C. F. (1984) FEMS Microbiol. Lett.
6. Haber, F. & Weiss, J. (1934) Proc. R. Soc. Lond. A 147, 332- 25, 111 -116.
337. 44 Fleming, T. P., Nahlik, M. S., Neilands, J. B. & McIntosh, M.
I. Wrigglesworth, J. M. & Baum, H . (1980) in Iron in biochemistry A. (1985) Gene 34,47-54.
and medicine, 2nd edn (Jacobs, A. & Worwood, M., eds) 45. Straka, J. G. & Emery, T. (1979) Biochim. Biophys. Acta 569,
pp. 29-86, Academic Press, London. 277 - 286.
8. Neilands, J. B. (1981) Annu. Rev. Nutr. 1, 27-46. 46. Brown, K. A. & Ratledge, C. (1975) FEBS Lett. 53,262-266.
9. Crosa, J. H. & Hodges, L. L. (1981) Infect. Immunol. 31, 223- 47. Ernst, J. F. & Winkelmann, G. (1977) Biochim. Biophys. Acta
227. 500, 27 -41.
10. Walter, M. A., Potter, S. A. & Crosa, J. H. (1983) J . Bacteriol. 48. Hartmann, A. & Braun, V. (1980) J. Bacteriol. 143, 246-255.
156,880-887. 49. Braun, V., Brazel-Faisst, C. & Schneider, R. (1984) FEMS
11. Neilands, J. B. (1952) 1. Am. Chem. Soc. 74,4846-4847. Microbiol. Lett. 21, 99- 103.
12. Keller-Scherlein, W., Prelog, V. & Zahner, H. (1964) Fortschr. 50. Osborne, T. B. & Cambell, G. F. (1900) J . Am. Chem. Soc. 22,
Chem. Org. Naturst. 22, 279-333. 422 -426.
13. Lankford, C. E. (1973) Crit. Rev. Microbiol. 2, 273-331. 51. Alderton, G., Ward, W. H. & Ferold (1946) Arch. Biochem.
14. Snow, G. A. (1970) Bacteriol. Rev. 34, 99- 125. Biophys. 11, 9-13.
15. Wayne, R. & Neilands, J. B. (1975) J . Bacteriol. 121, 497-503. 52. Schade, A. L. & Caroline, L. (1944) Science (Wash. DC) 100,
16. Jacobs, A., White, G. P. & Tait, G. H. (1977) Biochern. Biophys. 14-15.
Res. Commun. 74, 1626- 1630. 53. Schade, A. L. & Caroline, L. (1946) Science (Wash. DC) 104,
17. Raymond, K. N. & Carrano, C. J. (1979) Acc. Chem. Res. 12, 340 - 341.
183- 196. 54. Laurell, C. B. &Ingelman, B. (1947) Acta Chem. Scand. 1,770-
18. Neilands, J. B., Peterson, T. & Leong, S. A. (1980) in Inorganic 776.
chemistry in biology andmedicine (Martell, A. E., ed.) pp. 263 - 55. Holmberg, C. G. & Laurell, C. B. (1947) Acfa Chem. Scand. 1,
278, American Chemical Society, Washington DC. 944-950.
19. Raymond, K. N., Muller, G. & Matzanke, B. F. (1984) Top. 56. Schade, A. L., Reinhart, R. W. & Levy, H. (1949) Arch. Biochem.
Curr. Chem. 123, 50-102. Biophys. 20, 170- 172.
20. Winkelman, G. (1979) FEBS Lett. 97, 43-46. 57. Surgenor, D. M., Koechlin, B. A. & Strong, L. E. (1949) J. Clin.
21. Winkelman, G. & Braun, V. (1981) FEMS Microbiol. Lett. 11: Invest. 28, 73 -96.
237-241. 58. Montreuil, J., Tornelat, J. & Mullet, S. (1960) Biochim. Biophys.
22. Muller, G., Isowa, Y. & Raymond, K . N. (1985) J . Biol. Chem. Acta 45,413-421.
260, 13921 - 13926. 59. Masson, P. L. & Heremans, J. F. (1968) Eur. J . Biochem. 6,
23. Neilands, J. B., Erickson, T. J. & Rastetter, W. H. (1981) J . Biol. 579 - 584.
Chem. 256,3831 - 3832. 60. Aisen, P. & Listowsky, I. (1980) Annu. Rev. Biochem. 49, 357-
393.
Definition: ‘Any terse, witty, pointed statement often anti- 61. Chasteen, N. D. (1977) Coord. Chem. Rev. 22,l-36.
thetical’. Example: ‘Experience is the name everyone gives to his 62. de Lucas, L. J., Suddath, F. L., Gams, R. A. & Bugg, C . E.
mistakes’. (1978) J. Mol. Bid. 123, 285-2286,
503
63. Al-Hilal, D., Baker, E., Carlisle, C. H., Gorinsky, B., Horsburgh, 95. Aisen, P., Leibman, A. & Zweier, J. (1978) J . Biol. Chem. 253,
R. C., Linley, P. F., Moss, D. S., Schneider, H. & Stimpson, 1930-1937.
R. (1976) J . Mol. Bid. 108, 255-257. 96. Aisen, P. & Leibman, A. (1978) in Trunsport byproteins(Blauer,
64. Gorinsky, B., Horsburgh, C., Lindley, P. F., Moss, D., Parkcr, G. & Sund, H., eds) pp. 277- 294, W. de Gruyter & Co., Berlin,
M. &Watson, J. L. (1979) Nature (Lond.) 281, 157-158.. New York.
65. Baker, E. N. & Rumball, S. V. (1977) J . Mol. Biol. 111, 207- 97. Aasa, R. (1972) Biochem. Biophys. Rex Commun. 49, 806-
210. 812.
66. Rossencu-Motreff, M. Y., Soctewey, F., Lamote, R. & Peeters, 98. Rogers, T. B., Gold, B. A. & Feeney, R. E. (1977) Biochemistry
H. (1971) Biopolymers 10, 1039-1048. 16,2299-2305.
67. Mac Gillevray, R. T. A,, Mendez, E., Sinha, S. K., Sutton, M. 99. Alsaadi, B. M., Williams, R. J. P. & Woodworth, R. C. (1981)
R., Linebach-Zins, J. & Brew, K. (1982) Proc. Nut1 Acad. Sci. J . Inorg. Biochem. 15, 1-6.
U S A 79, 2504-2508. 100. Pecorara, V. L., Harris, W. R., Carrano, C. J . & Raymond, K.
68. Mac Gillivray, R. T. A., Mendez, E., Shewale, J. G., Sinha, C. N. (1981) Biochemistry 20, 7033-7039.
K., Linebach-Zins, J. & Brcw, K. (1983) J . Biol. Chem. 258, 101. Williams, J. (1982) Biochem. J . 201, 647-651.
3543 - 3553. 102. Koenig, S. H. & Schillinger, W. E. (1969) J . Bid. Chem. 244,
69. Uxan, G., Frain, M., Park, I., Besmond, C., Haessen, G., Trepat, 6520-6526.
J. S., Zakin, M. M. & Kahn, A. (1984) Biochem. Biophys. Res. 103. Harris, D. C. & Gelb, M. H. (1980) Biochim. Biophys. Acta 623,
Commun. l l Y , 273-281. 1-9.
70. Yang, F., Lum, J. B., McGill, J. R., Moore, C. M., Naylor, S. 104. Rogers, T. B., Buerresen, T. & Feeney, J. (1978) Biochemistry
L., Van Bragt, P. H., Baldwin, W. D. & Bowman, B. H. (1984) 17,1105-1109.
Proc. Natl Acad. Sci. USA 81,2752-2756. 105. Chasteen, N. D. (1983) Trends Biochem. Sci. 8, 272-275.
71. Jeltsch, J. M. & Chambon, P. (1982) Ezcr. J . Biochem. 122,29 1 - 106. Harris, D. C. & Aisen, P. (1973) Biochim. Biophys. Actu 329,
296. 156- 158.
72. Williams, J., Elleman, T. C., Kingston, J. B., Wilkins, A. G. & 107. Gaber, B. P. & Aisen, P. (1970) Biochim. Biophys. Aciu 221,
Kuhn, K. A. (1982) Eur. J. Biochem. 122,297-303. 228 - 233.
73. Metz-Boutigue, M.-H., Jolles, J., Mazurier, J., Schoentgen, F., 108. Bates, G. W. & Schlabach, M. R. (1973) J . Biol. Chem. 248,
Legrand, D. S., Spik, G., Montreuil, J. & JollZs, P. (1984) Eur. 3228 - 3232.
J . Biochem. 145, 659-676. 109. Workman, E. F. Jr, Graham, G. & Bates, G. W. (1975) Biochim.
74. Staden, R. A. (1982) Nucleic Acids Res. 10, 2951 -2961. Biophys. Acta 399, 254 - 264.
75. Mac Gillivray, R. T. A,, Mendez, E. & Brew, K. (1977) in 310. Batcs, G. W. & Wernicke, J. (1917) J . Biol. Chem. 246, 3679-
Proteins ufiron metabolism (Brown, E. B., Aisen, P., Fielding, 3685.
J. & Crichton, R. R., eds) pp. 131-142, Grune & Stratton, 111. Bates, G. W., Billups, C. & Saltman, P. (1967) J . Biol. Chem.
New York. 242, 2810-2815.
76. Mazurier, J., Metz-Boutigue, M. H., Jollks, J., Spik, G., 112. Aisen, P., Aasa, R., Malmstrom, B. G. & Vanngard, T. (1967)
Montreuil, J. & Jollks, P. (1983) Experientia (Basel) 39, 135- J . Bid. Chem. 242, 2484 - 2490.
141. 113. Woodworth, R. C., Virkaitis, L. M., Woodbury, R. G. & Fava,
77. Smith, T. F. & Waterman, M. S. (1981) Adv. Appl. Math. 2, R. A. (1975) in Proteins of iron storage and transport in hiochem-
482 - 489. istry and medicine (Crichton, R. R., ed.) pp. 39 - 50, North-
78. Spik, G., Bayard, B., Fournet, B., Strccker, G., Bouquelet, S. & Holland, Amsterdam.
Montreuil, J. (1975) FEBS Lett. 50, 296-2299, 114. Bates, G . W. & Schlabach, M. R. (1975) in Proteins of iron
79. Dorland, L., Haverkamp, J., Schut, B. L., Vliegenthart, J. F. storage and transport in biochemistry m d medicine (Crichton,
G., Spik, G., Strecker, G., Fournet, B. & Montreuil, J. (1977) R. R., cd.) pp. 51 -58, North-Holland, Amsterdam.
FEBS Lett. 77, 15 - 20. 1 15. Masson, P. L. & Heremans, J. (1968) Eur. J . Biochem. 6 , 579 -
80. Karlsson, K. A., Pascher, I., Samuelsson, B. E., Finne, J., 584.
Krusius, T. & Rauvala, H. (1978) FEBS Lett. 94,413-417. 116. Carrano, C. J. & Raymond, K . N. (1979) J . Am. Chem. Soc.
81. Spik, G., Debruyne, V. & Montreuil, J. (1982) Falk Symp. 34, 101, 5401 - 5404.
477-483. 117. Pecoraro, V. L., Weitl, F. L. & Raymond, K. N. (1981) .I. Am.
82. Marz, L., Hatton, M. W. C., Berry, L. R. & Regoeczi, E. (1982) Chem. Soc. 103, 5133-5140.
Can. .I. Biochem. 60, 624- 630. 11 8. Hallberg, L. & Hedenberg, L. (1 965) Scand. J . Haemutol. 2,67 -
83. Spik, G . , Strecker, G., Fournet, B., Bouquelet, S., Montreuil, 75.
J., Dorland, L., Van Halbeek, H. &Vliegenthart, J. F. G. (1982) 119. Morgan, E. H. (1971) Biochim. Biophys. Acfu244, 103-116.
Eur. J . Biochem. 121,413-419. 120. Pollack, S., Vanderhoff, G. & Lasky, F. (1971) Biochinz. Biophys.
84. Dorland, L., Haverkamp, J., Vliegenthart, J. F. G., Spik, G., Act0 497, 481 -487.
Fournet, B. & Montreuil, J. (1979) Eur. J . Biochem. 100,569- 121. Morgan, E. H. (1977) Biochim. Biophys. Acta 449, 169-177.
574. 122. Rodgers, S. J. & Raymond, K . N. (1983) J . Med. Chem. 26,
85. Williams, J. (1982) Trends Biochem. Sci. 7, 394-397. 439 -443.
86. Williams, J., Grace, S. A. & Williams, J. M. (1982) Biochem. J . 123. Cowart, R. E., Kojima, N. & Bates, G. W. (1982) J . Biol. (Miem.
201, 417-419. 257,7560-7565.
87. Warner, R. C . & Weber, I. (1953) J . Am. Chem. Soc. 75, 5094- 124. Kojima, N. & Bates, G. W. (1979) J . Bid. Chem. 254, 8847-
5101. 8854.
88. Aasa, R., Malmstrom, B. G., Saltman, P. & Vanngird, T. (1963) 125. Sommer, J. H., O'Hara, P. B., Jonah, C. D. & Bersohn, R. (1982)
Biochim. Biophys. Acta 75, 203-222. Biochim. Biophys. Acta 703, 62 -68.
89. Aisen, P., Leibman, A. & Reich, H. A. (1966) J . Biol. Chem. 126. Jandl, J. M. & Katz, J. H. (1963) J . Clin. Invest. 42, 314-326.
241, 1666-1671. 127. Iacopetta, B. J., Morgan, E. H. & Yeoh, G. C. T. (1982) Biochim.
90. Wenn, R. V. & Williams, J. (1968) Biochem. J . 198, 69-74. Biophys. Acta 687, 204-210.
91. Princiotto, J . V. & Zapolski, E. J. (1968) Nature (Lond.) 255, 128. Mattia, E., Rao, K., Shapiro, D. S., Sussman, H. & Klausner,
87-88. R. D. (1984) J . Biol. Chem. 259, 2689-2692.
92. Lestas, A. N. (1976) Br. J . Huematol. 32, 341 -350. 129. Trowbridge, I. S. & Omary, M. B. (1981) Proc. Natl Acud. Sci.
93. Butterworth, R. M., Gibson, J . F. & Williams, J. (1975) Biochem. USA 78, 3039 - 3043.
J . 149, 559-563. 130. Sutherland, D. R., Delia, D., Schneider, C., Newman, R. A.,
94. Makey, D. G. & Seal, U. S. (1976) Biochim. Biophys. Acta 453, Fcmshcad, J. & Greaves, M. F. (1981) Proc. Nail Acad. Sci.
250 - 256. USA 78, 4535-4519.
131. Haynes, B. F., Hemler, M., Cotner, T., Mann, D. L., Eisenberth, 167. Lamb, J. E., Ray, F., Ward, J. H., Kushner, J. P. & Kaplan, J.
G. S., Strominger, J. L. & Fauci, A. S. (1981) J . Immunol. 127, (1983) J. Biol. Chem. 258,8751 -8758.
347 - 351. 168. Rao, K., van Renswoude, J., Kempf, C. & Klausner, R. D.
132. Newman, R., Schneider, C., Sutherland, R., Vodinelich, L. & (1983) FEBS Lett. 160, 213-216.
Greaves, M. F. (1982) Trends Biochem. Sci. 7, 397-400. 169. Hopkins, C. R. (1983) Cell35, 321-330.
133. Seligman, P. A,, Schleicher, R. B. & Allen, R. H. (1979) J . Biol. 170. Morgan, E. H. (1983) Biochim. Biophys. Acta 762,498-502.
Chem. 254,9943 - 9946. 171. Dautry-Varsat, A., Ciechanover, A. & Lodish, H. F. (1983) Proc.
134. Hu, H. Y. Y. & Aisen, P. (1978) J . Supramol. Struct. 8,349-360. Natl Acad. Sci. U SA 80,2258 -2262.
172. Klausner, R. D., Ashwell, G., van Renswoude, J., Harford, J.
135. Fernandez-Pol, J. A. & Klos, D. J. (1980) Biochemistry 19,
B. & Bridges, K. R. (1983) Proc. Nut1 Acad. Sci. USA 80,
3904-3912. 2263 - 2266.
136. Schneider, C., Sutherland, R., Newman, R. A. & Greaves, M. 173. Booth, A. G. &Wilson, M. J. (1981) Biochem. J . 196,355-362.
F. (1982) J . Biol. Chem. 257,8516-8522. 174. Bleil, J. D. & Bretscher, M. S. (1982) EMBO J . 1, 351 -355.
137. Omary, M. B. & Trowbridge, I. S. (1981) J . Biol. Chem. 256, 175. Pearse, B. M. F. (1982) Proc. Nut1 Acad. Sci. USA 79, 451-
471 5 -471 8. 455.
138. Goodfellow, P. N., Banting, G., Sutherland, D. R., Greaves, M. 176. Morgan, E. H., Smith, G. D. & Peters, T. J. (1986) Biochem. J.
F., Solomon, E. &Povey, S. (1982) Somat. CellGenet. 8,197- 237, 163-173.
206. 177. Sibille, J. C., Octave, J. N., Schneider, Y. J., Trouet, A. &
139. Enns, C. A,, Suomalainen, H. A., Gebhardt, J. E., Schroder, Crichton, R. R. (1986) Eur. J . Biochem. 155,47-55.
J. & Sussman, H. H. (1982) Proc. Natl Acad. Sci. USA 79, 178. Appleton, T. C., Morgan, E. H. & Baker, E. (1971) in The
3241 - 3245. regulation of erythropoiesis and hemoglobin synthesis
140. Schneider, C., Kurkinen, M. & Greaves, M. F. (1983) EMBO J . (Travnicek, T. & Neuwirt, J., eds) pp. 310-315, University
2,2259 - 2265. Karlova, Praha.
141. Plowman, G. D., Brown, J. P.,'Enns, C. A,, Schroder, J., 179. Morgan, E. H. (1981) Mol. Aspects Med. 4, 1 - 123.
Nikinma, B., Sussman, H. H., Hellstrom, K. E. & Hellstrom, 180. Iacopetta, B. J. & Morgan, E. H. (1984) Biochim. Biophys. Acta
I. (1983) Nature (Lond.) 303, 70-72. 805,211-216.
142. Goubin, G., Goldman, D. S., Luce, J., Neiman, P. E. & Cooper, 181. Harding, C., Heuser, J. & Stahl, P. (1983) J . Biol. Chem. 97,
G. M. (1983) Nature (Lond.) 302, 114-117. 129- 139.
143. Awai, M. & Brown, E. B. (1963) J. Lab. Clin. Med. 61, 363- 182. McArdle, H. J., Douglas, A. J. & Morgan, E. H. (1985) J. Cell
368. Physiol122,405 -409.
144. Bothwell, T. H., Charlton, R. W., Cook, J. D. & Finch, C. A. 183. Baker, E., van Bockxmeer, F. M. & Morgan, E. H. (1983) Q. J.
(1979) in Iron metabolism in man, p. 334, Blackwell, Oxford. Exp. Physiol. 69, 359 - 372.
145. Baker, E. (1977) in Ciba Found. Symp. 51, 365-373. 184. Baker, E., McArdle, H. J. & Morgan, E. H. (1985) in Proteins
146. Morgan, E. H. & Baker, E. (1969) Biochim. Biophys. Acta 184, of iron storage and transport (Spik, G., Montreuil, J., Crichton,
442 -454. R. R. & Mazurier, J., eds) pp. 131 - 142, Elsevier, Amsterdam.
147. Baker, E. &Morgan, E. H. (1968) Biochemistry 8, 1133-1141. 185. Verhoef, N. J. & Noordeloos, P. J. (1977) Clin. Sci. Mol. Med.
148. Morgan, E. H. & Appleton, T. C. (1969) Nature (Lond.) 223, 52, 87-96.
1371- 1372. 186. Glass, J., Nunez, M. T. & Robinson, S. H. (1977) Biochem.
149. Sullivan, A. L., Grasso, J. A. & Weintraub, L. R. (1976) Blood Biophys. Res. Commun. 75,226-232.
47,133-143. 187. Loh, T. T., Yeung, Y. G. & Yeung, D. (1977) Biochim. Biophys.
150. Hemmaplardh, D. & Morgan, E. H. (1977) Br. J. Haematol. 36, Acta471, 118-124.
85 - 96. 188. Zaman, Z., Heynen, M. J. & Venvilghen, R. L. (1980) Biochim.
151. Schneider, Y.-J., Tulkens, P., de Duve, C. & Trouet, A. (1979) Biophys. Acta 632, 553 - 561.
J . Cell Biol. 82, 466 -474. 189. Woodworth, R. C., Brown-Mason, A,, Christensen, T. G., Witt,
152. Octave, J. N., Schneider, Y. J., Hoffmann, P., Trouet, A. & D. P. & Comeau, R. D. (1982) Biochemistry 21,4220-4225.
Crichton, R. R. (1979) FEBS Lett. 108, 127-130.. 190. Egyed, A. (1984) Br. J . Haematol. 56, 563-570.
153. Octave, J. N., Schneider, Y. J., Crichton, R. R. & Trouet, A. 191. van der Heul, C., Veldlman, A,, Kroos, M. J. & van Eijk, H. G.
(1983) Eur. 1.Biochem. 115,611 -618. (1984) Int. J . Biochenz. 16, 383-389.
154. Octave, J. N., Schneider, Y. J., Hoffmann, P., Trouet, A. & 192. Nunez, M. T. & Glass, J. (1985) J . Biol. Chem. 260, 14707-
Crichton, R. R. (1982) Eur. J. Biochem. 123,235-240. 14711.
155. Karin, M. & Mintz, B. (1981) J. Biol. Chem. 256, 3245-3252. 193. Laufberger, V. (1973) Bull. SOC.Chim. Biol. 19, 1575-1582.
156. Paterson, S. & Morgan, E. H. (1980) J . Cell Physiol. 105,489- 194. Crichton, R. R. (1973) Struct. Bonding 17,67- 134.
502. 195. Munro, H. N. & Linder, M. C. (1978) Physiol. Rev. 58, 317-396.
157. Morgan, E. H. (1981) Biochim. Biophys. Acta 642, 119-134. 196. Harrison, P. M., Hoare, R. J., Hoy, T. G. &Macara, I. G. (1980)
158. Light, A. & Morgan, E. H. (1982) Scand. J . Haemutol. 28,205- in Iron in biochemistry and medicine, vol. I1 (Jacobs, A. &
214. Worwood, M., eds) pp. 73 - 114, Academic Press, London.
159. Armstrong, N. J. & Morgan, E. H. (1983) Biochim. Biophys. 197. Clegg, G. A., Fitton, J. E., Harrison, P. M. & Treffry, A. (1980)
Acta 762, 175- 186. Prog. Biophys. Mol. Biol. 36, 56 - 86.
160. Iacopetta, B. J. & Morgan, E. H. (1983) J. Biol. Chem. 258, 198. Theil, E. C. (1983) in Advances in inorganic biochemistry, vol. 5
9108-9115. (Theil, E. C., Eichorn, G. L. & Marzilli, L. G., eds) pp. 1-38,
161. Iacopetta, B. J. & Morgan, E. H. (1983) Eur. J . Cell Biol. 32, Elsevier, New York.
17-23. 199. Mazur, A. & Shorr, E. (1948) J . Biol. Chem. 176,771 -787.
162. Klausner, R. D., Van Renswoude, J., Ashwell, G., Kempf, C., 200. Jacobs, A. & Worwood, M. (1975) New Engl. J . Med. 292,951 -
Schechter, A. N., Dean, A. & Bridges, K. R. (1983) J. Biol. 956.
Chem. 258,4715 -4724. 203. Cook, S. F. (1929) J . Biol. Chem. 82, 595-609.
163. Ciechanover, A,, Schwartz, A. L., Dautry-Varsat, A. & Lodish, 202. Michaelis, L., Coryell, C. D. & Granick, S. (1943) J . Biol. Chem.
H. F. (1983) J . Biol. Chem. 258,9681 -9689. 148,463 -480.
164. Van Renswoude, J., Bridges, K. R., Harford, J. B. & Klausner, 203. Mann, S., Bannister, J. V. & Williams, R. J. P. (1986) J . Mol.
R. D. (1982) Proc. Natl Acad. Sci. USA 79, 6186-6190. Biol. 188, 225 - 232.
165. Enns, C. A,, Larrick, J. W., Suomalainen, H., Schroder, J. & 204. Fischbach, F. A. & Anderegg, J . W. (1965) J . Mol. Biol. 14,
Sussman, H. H. (1983) J . Cell Biol. 97, 579-585. 458-473.
166. Hopkins, C. R. & Trowbridge, I. S. (1983) J . CellBiol. 97, 508- 205. Harrison, P. M., Fischbach, F. A., Hoy, T. G. & Haggis, G. H.
521. (1967) Nature (Lond.) 216, 1188-1190.
505

206. Granick, S. (1942) J . Biol. Chem. 146,451 -461. 248. Stefanini, S., Chiancone, E., Arosio, P., Finazzi-Agro, A. &
207. Towe, K. M. & Bradley, W. F. (1967) J. Colloid Interface Sci. Antonini, E. (1982) Biochemistry 21, 2293 -2299.
24,384- 392. 249. Wustefeld, C. & Crichton, R. R. (1982) FEBS Lett. 150, 43-
208. Harrison, P. M., Fischbach, F. A., Hoy, T. G. & Haggis, G. H. 48.
(1967) Nature (Lond.) 216, 1188-1190. 250. Addison, T. M., Fitton, J. E., Lewis, W. G., May, K. &Harrison,
209. Massover, W. H. & Cowley, J. M. (1973) Proc. Nut1 Acad. Sci. P. M. (1983) FEBS Lett. 164, 139-144.
USA 70, 3847-3851. 251. Crichton, R. R. (1983) in Structure and transport of iron storage
210. Isaacson, M. & Ohtsuki, M. (1980) Scanning Electron Microsc. and transportproteins (Uroshizaki, I., Aisen, P., Listowsky, I. &
1, 72-80. Drysdale, J. W., eds) pp. 1 - 10, Elsevier, Amsterdam.
211. Towe, K. M. (1981) J. Biol. Chem. 256,9377-9378. 252. Liebold, E. A., Aziz, N., Brown, A. J. P. & Munro, H. N. (1984)
212. Chukrov, F. V., Zvyagin, B. B., Gorshkov, A. I., Yermilova, L. J . Biol. Chem. 259,4327-4334.
P. & Balashova, V. V. (1973) Znt. Geol. Rev. 16, 1131-1143. 253. Constanzo, F., Santoro, C., Colantuoni, V., Bensi, G., Raugei,
213. Brady, G. W., Kurkjian, C. R., Lyden, E. F. X., Robin, M . B., G., Romano, V. & Cortese, R. (1984) EMBO J. 3, 23 -27.
Saltman, P., Spiro, T. & Terzis, A. (1968) Biochemistry 7, 254. Boyd, D., Vecoli, C., Belcher, D. M., Jain, S. W. & Drysdale, J.
2185 -2192. W. (1985)J. Bid. Chem.240,11755-11761.
214. Heald, S. M., Stearn, E. A., Bunker, B., Holt, E. M. & Holt, S. 255. Boyd, D., Jain, S. K., Crampton, J., Barrett, K. J. & Drysdale,
L. (1979) J . Am. Chem. Soc. 101,67-73. J. (1984) Proc. Nut1 Acad. Sci. U S A 81,4751 -4755.
215. Theil, E. C., Sayers, D. E. & Brown, M. A. (1979) J . Biol. Chem. 256. Darner, M. H., Salfeld, J., Will, H., Leibold, E. A,, Vass, J. K. &
254, 8132-8134. Munro, H. N. (1985) Proc. Natl Acad. Sci. USA 82, 3139-
216. Treffry, A. & Harrison, P. M. (1978) Biochem. J. 171,313-320. 3143.
217. Imai, N., Terada, H., Arata, Y . & Fujiwara, S. (1978) Bull. 257. Didsbury, J. R., Theil, E. C., Kaufman, R. E. & Dickey, L. N.
Chem. Soc. Jpn. 51,2538-2542. (1986) J . Biol. Chem. 261, 949 -955.
218. Williams, J. M., Danson, D. P. & Janot, C. (1978) Phys. Med.' 258. Brown, A. J. P., Leibold, E. A. & Munro, H. N . (1983) Proc.
Biol. 23, 835-851. Natl Acad. Sci. USA 80, 1265- 1269.
219. Stuhrmann, H. B., Haas, J., Ibel, K., Koch, M. H. J. & Crichton, 259. Munro, H. N., Leibold, E. A., Vass, J. K., Aziz, N., Rogers, J.,
R. R. (1976) J . Mol. Biol. 100, 399-413. Murray, M. & White, K. (1985) in Proteins of iron storage and
220. Massover, W. M. (1978) J . Mol. Biol. 123, 721 -726. transport (Spik, G., Montreuil, J., Crichton, R. R. & Mazurier,
221. Richter, G. W. (1984) Lab. Invest. 50, 26-35. J., eds) pp. 331 -341, Elsevier, Amsterdam.
222. Iancu, T. C. (1983) Mol. Aspects Med. 6, 1-100. 260. Constanzo, F., Colombo, M., Staempfli, S., Santoro, C.,
223. Fischbach, F. A., Gregory, D. W., Harrison, P. M., Hoy, T. Marone, M., Frank, R., Delius, H. &Cortese, R. (1986) Nucleic
G. &Williams, J. M. (1971) J. Ultrastruct. Res. 37, 495-503. Acids Res. 14, 721 -736.
224. Bell, S. H., Weir, M. P., Dickson, D. P. E., Gibson, J. F., Sharp, 261. Caskey, J. H., Jones, C., Miller, Y . E. & Seligman, P. A. (19113)
G. A. & Peters, T. J. (1984) Biochim. Biophys. Acta 787,227- Proc. Nut1 Acad. Sci. U S A 80, 482 - 486.
236. 262. Worwood, M., Brook, J. D., Cragg, S. J., Helkuhl, B., Jones, B.
225. Hoy, T. G. & Jacobs, A. (1981) Br. J . Haematol. 49, 593-597. M., Perera, P., Roberts, S. H. & Shaw, D. J. (1985) Hum. Genet.
226. Granick, S. & Michaelis, L. (1943) J. Biol. Chem. 147, 91 -97. 69, 371 -374.
227. Harrison, P. M. (1959) J . Mol. Biol. I , 69-80. 263. Drysdale, J., Jain, S. K., Boyd, D., Barrett, C., Vecoli, C.,
228. Leach, B. S., May, M. E. & Fish, W. W. (1976) J . Biol. Chem. Belcher, D. M., Beaumont, C., Worwood, M., Lebo, R.,
251,3856-3861. McGill, J. & Crampton, J. (1985) in Proteins of iron transport
229. Heusterspreute, M. & Crichton, R. R. (1981) FEBS Lett. 129, and storage (Spik, G., Montreuil, J., Crichton, R. R. &
322 - 327. Mazurier, J., eds) pp. 343 -347, Elsevier, Amsterdam.
230. Yariv, J., Kalb, A. J., Sperling, R., Bauminger, E. R., Cohen, S. 264. Ford, G. C., Harrison, P. M., Rice, D. W., Smith, J . M. A.,
G. & Ofer, S. (1981) Biochem. J. 197, 171-175. Treffry, A., White, J. L. & Yariv, J. (1984) Phil. Trans. R. Soc.
231. Steifel, E. I. & Watt, G. D. (1979) Nature (Lond.) 279, 81 -83. Lond. B 304,551 -565.
232. Chen, M. & Crichton, R. R. (1982) Biochim. Biophys. Acta 707, 265. Rice, D. W., Ford, G. C., White, J. L., Smith, J. M. A. &
1-6. Harrison, P. M. (1983) in Advances in inorganic biochemistry,
233. Yariv, J. (1983) Biochem. J . 211, 527-535. vol. 5 (Theil, E. C., Eichorn, G. L. & Marzilli, L. G., eds)
234. Jiudi, L., Jiwen, W., Zepu, Z., Ya, T. & Bei, D. (1980) Sci. Sin. pp. 39-50, Elsevier, New York.
Engl. Ed. 23, 897 - 904. 266. Crichton, R. R. (1972) Biochem. J. 130, 35-36P.
235. Niitsu, Y . ,Ishitani, K. & Listowsky, I. (1973) Biochem. Biophyx. 267. Rice, D. W., Ford, G. C., White, J. L., Smith, J. M. A. &
Res. Commun. 55, 1134-1140. Harrison, P. M. (1983) in Structure andfunction ofiron storage
236. Linder, M. C., Moor, J . R. & Munro, H. N. (1974) J . Biol. Chem. and transportproteins (Uroshizaki, I., Aisen, P., Listowsky, I. &
249,7707-7710. Drysdale, J. W., eds) pp. 11 - 16, Elsevier, Amsterdam.
237. Ishitani, K., Niitsu, Y. & Listowsky, I. (1975) J . BioI. Chem. 268. Otsuka, S., Maruyama, H. & Listowsky, I. (1981) Biochemistry
250,3142-3148. 20, 5226 - 5232.
238. Linder, M. C., Moor, J. R., Munro, H. N. & Morris, H. P. 269. Otsuka, S., Listowsky, I., Niitsu, Y. & Uroshizahki, 1. (1980)
(1975) Biochim. Biophys. Acta 384,409-421. J. Biol. Chem. 255,6234 - 6237.
239. Bryce, C. F. A., Magnusson, C. G. M. & Crichton, R. R. (1978) 270. Rice, D. W., Dean, B., Smith, J. M. A,, White, J. L., Ford, G.
FEBS Lett. 94, 257-262. C. &Harrison, P. M. (1985) FEBS Lett. 181, 165-168.
240. Drysdale, J. W. (1977) Ciba Found. Symp. 51,41-57. 271. Massover, W. H. (1979) Biochim. Biophys. Acta 579,169-180.
241. Arosio, P., Adelman, T. G. & Drysdale, J. W. (1978) J. Biol. 272. Tsugita, A. & Yariv, J. (1985) Biochem. J . 231, 209-212.
Chem. 253,4451 -4458. 273. Pape, L., Multani, J. S., Saltman, P. & Stitt, C. (1968) Biochemis-
242. Arosio, P. (1979) FEBS Lett. 104, 51 - 54. rry 7, 606-612.
243. Lavoie, D. J., Ishikawa, K. & Listowsky, I. (1978) Biochemistry 274. Spiro, T. G., Allerton, S. E., Renner, J., Terzis, A,, Bils, R. &
17,5448 - 5454. Saltman, P. (1966) J . Am. Chem. Soc. 88,2721 - 2726.
244. Watanabe, N. & Drysdale, J. W. (1983) Biochim. Biophys. Acta 275. Reference deleted.
743, 98 - 105. 276. Bielig, H. J. & Bayer, E. (1955) Naturwissenschaften 42, 125-
245. Uroshizaki, I., Niitsu, Y., Ishitani, K., Matsuda, M. & Fukuda, 126.
M. (1971) Biochim. Biophys. Acta 243,187-192. 277. Loewus, M. W. & Fineberg. R. A. (1957) Biochim. Biophys. Acta
246. Drysdale, J. W. (1970) Biochim. Biophys. Acta 207, 256-258. 26,441 -443.
247. Bomford, A., Berger, M., Lis, Y .& Williams, R. (1978) Biochem. 278. Harrison, P. M., Fischbach, F. A,, Hoy, T. G. & Haggis, G. H.
Biophys. Res. Commun. 83, 334-341. (1967) Nature (Lond.) 216, 1188-1190.
279. Neiderer, W. (1970) Experientia (Basel) 26, 218 -220. 309. Jones, T., Spencer, R. & Walsh, C. (1978) Biochemistry 17,
280. Macara, I. G., Hoy, T. G. & Harrison, P. M. (1972) Biochem. 4011 -4017.
J . 126, 151-162. 310. Harrison, P. M., Banyard, S. H., Clegg, C. A,, Stammers, D.
281. Bryce, C. F. A. & Crichton, R. R. (1973) Biochern. J . 133, 301 - K. & Treffry, A. (1978) in Transporf by proteins (Blauer, G. &
309. Sund, H., eds) pp. 259-275, Walter de Gruyter, Berlin, New
282. Macara, I. G., Hoy, T. G. & Harrison, P. M. (1973) Biochem. York.
J . 135, 343-348. 31 1. Funk, F., Lenders, J. P., Crichton, R. R. & Schneider, W. (1985)
283. Pbques, E. P., Pbques, A. & Crichton, R. R. (1979) J. Mol. Eur. J . Biochem. 152,167-172.
Catal. 5, 363 - 375. 312. Banyard, S. H., Stammers, D. K . & Harrison, P. M. (1978)
284. Pbques, E. P., Piques, A. & Crichton, R. R. (1980) Eur. J . Nature (Lond.) 271,282 - 284.
Biochem. 107,447-453. 313. Hoare, R. J., Harrison, P. M. & Hoy, T. G. (1975) Nature
285. Wauters, M., Michelson, A. M. & Crichton, R. R. (1978) FEBS (Lond.) 255,653 - 654.
Lett. 91, 276-280. 314. Mazur, A., Green, S., Saha, A. & Carleton, A. (1958) J . Clin.
286. Chasteen, N. D. &The& E. C. (1982) J. Biol. Chem. 253,7672- Invest. 53, 1809-1817.
7677. 315. Williams, D. M., Lee, G. R. & Cartwright, G. E. (1974) J . Clin.
287. Crichton, R. R. & Roman, F. (1978) J . Mol. Catal. 4, 75-82. Invest. 53, 665 - 667.
288. Crichton, R. R. (1981) in The biological chemistry of iron 316. Topham. R. W., Walker, M. C. & Calisch, M. P. (1982) Biochem.
(Dunford, H. B., Dolphin, D., Raymond, K. N. & Sieker, L., Biophys. Res. Commun. 109,1240- 1246.
eds) pp. 45-61, Reidel, Dordrecht. 317. Thomas, C. E., Morehouse, L. A. & Aust, S. D. (1985) J . Biol.
289. Wetz, K. & Crichton, R. R. (1976) Eur. J . Biochem. 61, 545- Chem. 260,3275 -3280.
550. 318. Bienfait, H. F. &Van den Briel, M. L. (1980) Biochim. Biophys.
290. Sayers, D. E., Theil, E. C. & Rennick, F. J. (1983) J . Biol. Chem. Acta 631, 507-5510,
258, 14076- 14079. 319. Wrigglesworth, J. M. & Baum, H. (1980) in Iron in biochemistry
291. Chasteen, N. D., Antanaitis, B. C. & Aisen, P. (1985) J . B i d . andmedicine, vol. I1 (Jacobs, A. & Worwood, M., eds) pp. 29 -
Chem. 260,2926 - 2929. 86, Academic Press, London, New York.
292. Wardeska, J. G., Viglione, B. & Chasteen, N. D. (1986) J . Biol. 320. Crichton, R. R. (1984) Trends Biochem. Sci. 9, 283-286.
Chem. 261,6677-6683. 321. Bacon, B. R. & Tavill, A. S. (1984) Sem. Liver Diseases4,lSl-
293. Harrison, P. M., White, J. L., Smith, J. M. A., Farrant, G. W., 192.
Ford, G. C., Rice, D. W., Addison, J. M. & Treffry, A. (1985) 322. Jacobs, A. (1977) Ciba Found. Symp. 51,91- 100.
in Proteins of iron storage and transport (Spik, G., Montreuil, 323. Jones, M. S. & Jones, 0. T. G. (1969) Biochem. J. 223, 31 - 38.
J., Crichton, R. R. & Mazurier, J., eds) pp. 67-72, Elsevier, 324. Konopka, K. & Romslo, I. (1980) Eur. J . Biochem. 107, 433-
Amsterdam. 439.
294. Hoy, T. G. & Harrison, P. M. (1976) Br. J . Haematol. 33,497- 325. Konopka, K. & Romslo, I. (1981) Eur. J . Biochem. 117, 239-
504. 244.
295. Treffry, A. &Harrison, P. M. (1979) Biochem. J . 181,709-716. 326. Nilsen, T. & Romslo, I. (1985) Biochim. Biophys. Acta 842,
296. Schneider, W. & Erni, I. (1982) in The biochemistry andphysi- 162-169.
ology of iron (Saltman, P. & Hegenauer, J., eds) pp. 121 - 126, 327. Ulvik, R. J. & Romslo, I. (1978) Biochim. Biophys. Acta 541,
Elsevier, Amsterdam. 251 -262.
297. Funk, F., Lecrenier, C., Lesuisse, E., Crichton, R. R. & 328. Ulvik, R. J. & Romslo, I. (1979) Biochim. Biophys. Acta 588,
Schneider, W. (1986) Eur. J . Biochem. 157, 303-309. 256-271.
298. Mazur, A., Baez, S. & Shorr, E. (1955) J. Bid. Chem. 213, 147- 329. Ulvik, R. J., Romslo, I., Roland, F. & Crichton, R. R. (1981)
160. Biochim. Biophys. Acta 677, 50 - 56.
299. Bielig, H. J. & Bayer, E. (1955) Naturwissenschqften 42, 466- 330. Ulvik, R. J. (1982) Biochim. Biophys. Acta 715,42-51.
468. 331. Flatmark, T. & Tangeras, A. (1977) in Proteins of iron metab-
300. Pape, L., Multani, J. S., Stitt, C. & Saltman, P. (1968) Biochemis- olism (Brown, E. B., Aisen, P., Fielding, J. & Crichton, R. R.,
try 7, 613-616. eds) pp. 349-358, Grune & Stratton, New York.
301. Dognin, J. & Crichton, R. R. (1975) FEBS Left. 54, 234-236. 332. Tangeras, A,, Flatmark, T., Bickstrom, D. & Ehrenberg, A.
302. Tuffano, T. P., Pecoraro, V. L. & Raymond, K. N. (1981) (1980) Biochim. Biophys. Actu 589, 162-175.
Biochim. Biophys. Acta 668,420-428. 333. Tangeras, A. (1985) Biochim. Biophys. Acta. 843, 199-207.
303. Dognin, J., Girardet, J. L. & Chapron, Y. (1973) Biochim. Bio- 334. Nishisato, T. & Aisen, P. (1982) Br. J . Haematol. 52, 631 -640.
phys. Acta 297,276-284. 335. Saito, K., Nishisato, T., Grasso, J. A. & Aisen, P. (1986) Br. J .
304. Crichton, R. R., Roman, F. & Roland, F. (1980) J . Inorg. Haematol. 62, 275 -286.
Biochem. 13, 305-316. 336. Byers, B. R. & Arceneaux, E. L. (1977) in Microorganisms and
305. Tidmarsh, G. F., Klebba, P. E. & Rosenberg, L. N. (1983) minerals (Weinberg, E. D., ed.) pp. 21 5 - 249, Marcel Decker,
J. Inorg. Biochem. 18, 161-268. New York.
306. Bonomi, F. & Pagani, S. (1986) Eur. J . Biochem. 155,295-300. 337. Neilands, J. B. (1979) in Trace metals in health and disease
307. Sirivech, S., Frieden, E. & Osaki, S. (1974) Biochem. J . 143, (Kharasch, N., ed.) pp. 27-41, Raven Press, New York.
311 -315. 338. Spik, G., Fournet, B., Cheron, A., Strecker, G., Montreuil, J.,
308. Crichton, R. R., Roman, F. & Wauters, M. (1975) Biochem. Dorland, L. & Vliegenthart, J. F. G. (1979) 5th Int. Symp.
Soc. Trans. 3, 946-948. Glycoconjugates, Kiel, pp. 21 - 22.

You might also like