You are on page 1of 12

Toxicol Rev 2003; 22 (1): 53-64

REVIEW ARTICLE 1176-2551/03/0001-0053/$30.00/0

© Adis Data Information BV 2003. All rights reserved.

Ricin
Mechanisms of Cytotoxicity
Michael J. Lord, Nicholas A. Jolliffe, Catherine J. Marsden, Cassandra S.C. Pateman, Daniel C. Smith,
Robert A. Spooner, Peter D. Watson and Lynne M. Roberts
Department of Biological Sciences, University of Warwick, Coventry, UK

Contents
Abstract . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 53
1. Structure and Function of Ricin . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 54
1.1 Ricin Structure . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 54
1.2 The Substrate for Ricin . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 54
1.3 Mode of Action of Ricin A Chain . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 55
1.4 Novel Enzymatic Activities of Ricin . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 56
1.5 Ricin Biosynthesis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 56
2. Getting Into the Cytosol . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 56
2.1 Binding of Ricin to Mammalian Cells . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 56
2.2 Endocytosis of Ricin . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 57
2.3 Transport of Ricin from Endosomes to the Golgi . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 57
2.4 Transport of Ricin from Golgi to Endoplasmic Reticulum . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 58
2.5 Membrane Translocation of Ricin . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 59
2.6 Retro-Translocation of Ricin A Chain in Other Systems . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 60
3. Conclusions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 61

Abstract Ricin is a heterodimeric protein produced in the seeds of the castor oil plant (Ricinus communis). It is
exquisitely potent to mammalian cells, being able to fatally disrupt protein synthesis by attacking the Achilles
heel of the ribosome. For this enzyme to reach its substrate, it must not only negotiate the endomembrane system
but it must also cross an internal membrane and avoid complete degradation without compromising its activity in
any way. Cell entry by ricin involves a series of steps: (i) binding, via the ricin B chain (RTB), to a range of cell
surface glycolipids or glycoproteins having β-1,4-linked galactose residues; (ii) uptake into the cell by
endocytosis; (iii) entry of the toxin into early endosomes; (iv) transfer, by vesicular transport, of ricin from early
endosomes to the trans-Golgi network; (v) retrograde vesicular transport through the Golgi complex to reach the
endoplasmic reticulum; (vi) reduction of the disulphide bond connecting the ricin A chain (RTA) and the RTB;
(vii) partial unfolding of the RTA to render it translocationally-competent to cross the endoplasmic reticulum
(ER) membrane via the Sec61p translocon in a manner similar to that followed by misfolded ER proteins that,
once recognised, are targeted to the ER-associated protein degradation (ERAD) machinery; (viii) avoiding, at
least in part, ubiquitination that would lead to rapid degradation by cytosolic proteasomes immediately after
membrane translocation when it is still partially unfolded; (ix) refolding into its protease-resistant, biologically
active conformation; and (x) interaction with the ribosome to catalyse the depurination reaction.
It is clear that ricin can take advantage of many target cell molecules, pathways and processes. It has been
reported that a single molecule of ricin reaching the cytosol can kill that cell as a consequence of protein
synthesis inhibition. The ready availability of ricin, coupled to its extreme potency when administered
intravenously or if inhaled, has identified this protein toxin as a potential biological warfare agent. Therapeuti-
cally, its cytotoxicity has encouraged the use of ricin in ‘magic bullets’ to specifically target and destroy cancer
54 Lord et al.

cells, and the unusual intracellular trafficking properties of ricin potentially permit its development as a vaccine
vector.
Combining our understanding of the ricin structure with ways to cripple its unwanted properties (its
enzymatic activity and promotion of vascular leak whilst retaining protein stability and important immunodomi-
nant epitopes), will also be crucial in the development of a long awaited protective vaccine against this toxin.

Ricin is a heterodimeric protein produced in the seeds of the ence of a B chain that mediates the binding and entry of holotoxin
castor oil plant (Ricinus communis). It is a potent and opportunistic (the disulphide-linked A- and B-chain complex) into susceptible
ribotoxin that can bind to the surface of many kinds of cell. What cells.
is very striking is the way ricin can exploit intracellular transport Amongst ribosome-inactivating proteins, the tertiary structure
pathways to travel from the plasma membrane to its target ribo- of ricin was the first to be determined.[14-16] It was shown to be a
somes in the cytosol. This journey involves retrograde routing to globular, glycosylated heterodimer in which ricin A chain and
the endoplasmic reticulum lumen, and retro-translocation from ricin B chain are joined by a single disulphide bond (figure 1,
this site to the cytosol.[1] It has been reported that a single molecule panel a). The 262 amino acyl residues of ricin B chain form a
of ricin reaching the cytosol can kill that cell as a consequence of bilobal structure lacking α-helices or β-sheets that probably arose
protein synthesis inhibition.[2] Indeed, the lethal dose of ricin in by gene duplication.[17] Each of the structurally homologous lobes
humans has been estimated at between 5–10 μg/kg bodyweight.[3] contains three related subdomains and at least one of these in each
The ready availability of ricin, coupled to its extreme potency lobe possesses a sugar-binding pocket. In contrast, ricin A chain,
when administered intravenously[3] or if inhaled[4] has identified comprising 267 amino acyl residues, has three structural domains
this protein toxin as a potential biological warfare agent.[5] Thera- with approximately 50% of the polypeptide arranged into α-
peutically, its cytotoxicity has encouraged the use of ricin in helices or β-sheet [18-20] (figure 1, panel a).
‘magic bullets’ to specifically target and destroy cancer cells,[6,7]
and the unusual intracellular trafficking properties of ricin poten- 1.2 The Substrate for Ricin
tially permit its development as a vaccine vector.[8,9]
The way in which ricin inactivates ribosomes was first de-
This review will focus on the structure, function and intracellu- scribed by Endo and Tsurugi[22] who showed that ricin A chain
lar trafficking of ricin, highlighting the features that underlie its acted by cleaving a specific glycosidic bond within the large
cytotoxic potency. ribosomal RNA (rRNA) of the 60S subunit of eukaryotic ribo-
somes. Ricin A chain must be separated from ricin B chain to
1. Structure and Function of Ricin display this activity.[23,24] Subsequently, Endo and Tsurugi[25] dem-
onstrated that ricin A chain was an RNA-specific N-glycosidase
1.1 Ricin Structure that hydrolytically cleaved the N-glycosidic bond of the adenine
residue at position 4324 (A4324) within the 28S rRNA of rat liver
Ricin is a prototype ribosome-inactivating protein. Such pro- ribosomes leaving the phosphodiester backbone of the RNA at this
teins have been isolated from many sources including bacteria, site intact. It is very striking that this single, specific depurination
fungi and the leaves, seeds and roots of plants.[10-12] In general, completely and irreversibly inactivates the ribosome. Closer
most ribosome-inactivating proteins can be classified as either examination revealed that the target adenine lay in a universally
Type 1 or Type 2.[13] Type 1 ribosome-inactivating proteins are the conserved sequence of 12 nucleotides, 5′-AGUACGAGAGGA-3′
most common (~100 described to date), and consist of a single (target adenine shown in italics), known as the sarcin-ricin loop.[26]
enzymatic chain of ~30 kDa. Type 2 ribosome-inactivating pro- The sarcin-ricin loop is associated with the binding of elongation
teins, typified by ricin, are potent cytotoxins with an A chain that is factors.[27] Modification of this region by ricin A chain thereby
structurally and functionally equivalent to the Type 1 ribosome- prevents the binding of such factors and inhibits the elongation
inactivating proteins, disulphide-linked to a lectin referred to as cycle of protein synthesis.[28,29] However, the relevance of
the B chain. The ~30 kDa B chain is able to bind terminal ribosomal proteins in rendering ribosomes susceptible to attack by
galactose residues on cell surface components. It follows that ricin A chain is highlighted by the fact that the kcat (a measure of
although Type 1 and Type 2 ribosome-inactivating proteins are the catalytic rate of an enzyme reaction) for depurination of naked
active against ribosomes in vitro, only the Type 2 ribosome- 28S rRNA by ricin A chain is nearly 105-fold lower than the kcat
inactivating proteins, such as ricin, are cytotoxic due to the pres- for rRNA within a ribosome.[25] Moreover, although ricin A chain

© Adis Data Information BV 2003. All rights reserved. Toxicol Rev 2003; 22 (1)
Ricin Toxicity 55

a b

RTA

Glu177
Arg180

Tyr123

Tyr80

RTB
FMP

c
E177 NH2 E177 NH2 NH2

N N N E177 N
C C N N
− O −
O O O
N C
N N N − N N
O O O
H H
C O
O H H O H
C O H NH NH
H +
NH H OH
+ R180
+ R180 R180
H2N NH H2N NH
H2N NH

Fig. 1. Structure of ricin and catalytic mechanism of the A chain. (a) The α-carbon backbone of RTB is shown in green whilst that of RTA is coloured
according to its structure with α-helices in pink, β-strands in yellow and coil in white. The site of the single interchain disulphide bond is shown in blue. (b)
The active site of RTA bound with a substrate analogue, formycin-5′-monophospate (FMP), is shown with critical catalytic residues, Arg180, Glu177,
Tyr123 and Tyr80 coloured with carbon atoms in grey, oxygen atoms in red, nitrogen atoms in blue and FMP in green. (c) A schematic of the proposed
catalytic mechanism of RTA.[21] RTA = ricin A chain; RTB = ricin B chain.

is active against naked 23S prokaryotic rRNA, intact Escherichia residue at position 180 (R180), all of which are located within the
coli ribosomes are completely refractory to depurination by ricin active site (figure 1, panel b). Although the precise mechanism of
A chain,[30] and plant ribosomes are ~5000-fold less sensitive to N-glycosidase action for ricin A chain remains unclear, Monzingo
ricin A chain than mammalian ribosomes.[31] The ribosome is a and Robertus[21] have proposed a mechanistic model based on the
large, complex substrate that operates via structural transitions, crystal structures of ricin A chain in complex with small molecules
and it is likely that the ricin A chain polypeptide also interacts with and site-directed mutagenesis studies.[20,21,32-36] The kinetic ana-
sites outside the immediate vicinity of the sarcin-ricin loop. The lysis of ricin A chain variants with mutations at either Arg180 or
presence of particular ribosomal proteins may therefore allow or Glu177 showed little change in Km, the binding affinity of the
prevent access of ricin A chain to the sarcin-ricin loop and might enzyme for its substrate. However, the kcat (a measure of the
thereby contribute to substrate specificity. catalytic rate of an enzyme reaction) of such mutants was affected
suggesting that these two residues are involved in the catalytic
1.3 Mode of Action of Ricin A Chain mechanism of the enzyme rather than in substrate binding.
The Monzingo and Robertus[21] model (figure 1, panel c) took
The catalytic activity of ricin lies exclusively within the A these studies into account, and proposed that the rRNA substrate
chain. Ricin A chain, along with other ribosome-inactivating becomes bound in the active site cleft with the target adenine
proteins, contains several invariant amino acid residues. These making stacking interactions with tyrosine residues Y80 and
include tyrosine residues at positions 80 and 123 (Y80 and Y123), Y123. Residue R180 is positioned such that it can protonate the
a glutamic acid residue at position 177 (E177) and an arginine leaving group at N-3 of adenine, facilitating an electron flow that

© Adis Data Information BV 2003. All rights reserved. Toxicol Rev 2003; 22 (1)
56 Lord et al.

results in the breaking of the bond between N-9 of the adenine ring chain.[50] Five disulphide bonds are also formed, four within ricin
and the C-1′ of the ribose. This bond cleavage results in the B chain and one between the mature subunit domains.[51]
production of an oxycarbonium ion on the ribose, which is The resulting core-glycosylated, folded proricin is then trans-
stabilised by the presence of E177. The protonation of N-3 of ported to storage vacuoles via the Golgi complex.[50,52] This intra-
adenine by R180 increases the basic character of the R180 side cellular transport is accompanied by several poorly characterised
chain, allowing it to pull a proton from a nearby water molecule modifications, including trimming of oligosaccharides and addi-
thus activating it and allowing solvent attack on the ribose carboni- tion of fucose to the ricin toxin A glycan.[53] Upon deposition in
um creating a neutral ribose. Through this action, it has been vacuoles, the mature disulphide-linked ricin A chain/ricin B chain
reported that a single A chain molecule in the cytosol can heterodimer is finally generated by the endoproteolytic removal of
depurinate ~1500 ribosomes per minute, leading to a rapid inhibi- the N-terminal and internal propeptides.[54] The intramolecular
tion of protein synthesis. propeptide has been shown to be both necessary[55] and suffi-
cient[56,57] for vacuolar targeting.
1.4 Novel Enzymatic Activities of Ricin Recombinant proricin has been produced and its properties
examined.[58] It is an active prolectin containing functional sugar
It has become evident that intact ricin can modify non-
binding sites. In contrast, proricin lacks N-glycosidase activity,
ribosomal nucleic acid substrates in vitro, allowing it to be consid-
consistent with reports that in mature holotoxin, ricin A chain must
ered a polynucleotide:adenosine glycosidase.[37] Nuclear DNA
be reductively separated from ricin B chain in order to have
damage has also been reported in human endothelial cells,[38]
activity.[23,24] It appears that in both the dimeric ricin holotoxin and
possibly as a prelude to the induction of apoptosis.[39,40] While
in proricin, the ricin B chain moiety can sterically obstruct the
certain ribonuclease and deoxyribonuclease activities ascribed to
active site of ricin A chain. Manufacture as a precursor perhaps
some ribosome-inactivating proteins are likely to be due to con-
explains why Ricinus cells are able to synthesise large amounts of
tamination,[41,42] it is unclear whether additional enzyme activities
ricin even though endogenous ribosomes are susceptible (albeit
reported for ricin and other ribosome-inactivating proteins (e.g.
much less so than animal ribosomes[31,59,60]), to the action of ricin
chitinase, phosphatase, phospholipase and superoxide dismutase
A chain.[25] Since the endosperm cells of Ricinus show no signs of
activities) are in any way physiologically relevant.[13,43,44]
ribosome damage whilst storing large amounts of ricin, it can
Recently, it has been shown that ricin contains a neutral lipase
further be deduced that active ricin A chain does not escape from
activity in vitro, correlating to the presence of a catalytic triad
low pH vacuoles to reach the cytosol. The generation of an active
between the two subunits.[45] Again, however, it remains unclear
holotoxin only within the safe haven of vacuoles therefore enables
whether this activity is functionally relevant since abolition of the
Ricinus seeds to synthesise and store large quantities of a potent
lipase activity had only a marginal effect on cytotoxicity.
toxin that can effectively function in plant defence against
predators without compromising its own survival.
1.5 Ricin Biosynthesis

Ricin is made in the fatty endosperm cells of maturing castor oil 2. Getting Into the Cytosol
seeds (Ricinis communis) where it accumulates in protein storage
vacuoles to 5% of total organelle protein.[46] Although ricin is a 2.1 Binding of Ricin to Mammalian Cells
heterodimer, its individual subunits are initially synthesised to-
gether in the form of a precursor.[47] The first 26 residues constitute When ricin is applied to the exterior of most animal cells, it will
a signal peptide for translocation into the endoplasmic reticulum bind to complex carbohydrates containing terminal N-acetyl
lumen of the plant cell[48] followed by a nine-residue propeptide of galactosamine or β-1,4-linked galactose residues.[3] The abun-
no known function. This combined 35 residue presequence is dance of cell surface galactolipids and glycoproteins provides an
followed by the mature ricin A chain sequence, which in turn is explanation for the diversity of cell lines that are sensitive to ricin.
joined to ricin B chain by a 12-residue intramolecular propep- Indeed, HeLa cells have been shown to possess around 3 × 107
tide.[49] The signal peptide and the two propeptides are absent in potential ricin binding sites,[61] although not all of these sites may
mature ricin, with the former being removed during synthesis and be functionally involved in the productive internalisation of the
segregation into the endoplasmic reticulum lumen, and the latter toxin. The relative contribution of glycoproteins and/or
being removed in vacuoles. Within the endoplasmic reticulum glycolipids in the uptake of ricin by target cells remains uncertain,
lumen, the nascent protein becomes core-glycosylated at one site although a role for glycosphingolipids as receptors has recently
in ricin A chain (unpublished data) and at two sites within ricin B been ruled out.[62] Native ricin also possesses the ability to bind

© Adis Data Information BV 2003. All rights reserved. Toxicol Rev 2003; 22 (1)
Ricin Toxicity 57

ER
Recycling TGN Golgi
endosome
Caveolae
COP1-independent?

Clathrin-dependent
pH
Cytosol
Rab9
EE
Clathrin-independent
COP1-dependent?
LE

Lysosome

Fig. 2. Schematic representation of the pathways that may be used by ricin to enter eukaryotic cells. COP1 = coatomer protein 1; EE = early endosomes;
ER = endoplasmic reticulum; LE = late endosomes; TGN = trans-Golgi network.

and enter some cell types by virtue of its own carbohydrate side internalisation was reduced to half of that seen in control cells,
chains. A limited number of cell lines express mannose receptors suggesting that ~50% of surface membrane uptake occurs by a
on their surface, to which ricin, with its high mannose-type gly- clathrin-independent mechanism.[65,66,80] Although ricin uptake
cans typical of plant glycoproteins, can bind. Mannose receptor- has been used to probe different endocytic pathways, careful
mediated uptake of ricin has been described in both macro- interpretation is needed since the area of the plasma membrane
phages[63] and rat liver endothelial cells.[64] occupied by, for example, clathrin-coated pits and caveolae invag-
inations, can differ dramatically from one cell type to another.[81,82]
2.2 Endocytosis of Ricin Furthermore, the perturbation of one route of uptake might actual-
ly up-regulate entry by a different pathway.[71,77]
Due to its promiscuous binding, ricin becomes localised to all
types of membrane invaginations[65] and as such, is a valuable tool 2.3 Transport of Ricin from Endosomes to the Golgi
in the study of different endocytic mechanisms[66,67] (figure 2). The
binding and internalisation of ricin into Vero cells (African Green
The majority of ricin that initially enters the cell is delivered to
Monkey kidney cells) was first visualised in clathrin-coated pits
early endosomes from where most appears to be either recycled to
using a ricin-gold conjugate.[68] However, evidence emerged to
the cell surface or delivered via late endosomes to lysosomes for
suggest that clathrin-independent mechanisms were also involved
degradation (figure 2). Raising the pH of endosomes/lysosomes
in toxin uptake. For example, Hep-2 cells, in which coated pit
does not protect against ricin. This, taken together with an effect of
formation was blocked by hypotonic shock and potassium ion
the ionophore monensin that influences Golgi structure and func-
depletion, remained sensitive to ricin.[69] Furthermore, in HeLa
tion, and a protection against ricin when endosome-to-Golgi trans-
cells, over-expression of a trans-dominant negative mutant of the
port is blocked by low temperature,[65] suggested a requirement for
protein dynamin (a guanosine triphosphate [GTP]-binding mecha-
the Golgi apparatus in ricin intoxication. A proportion (~5%) of
noenzyme that constricts lipids to aid vesicle formation) failed to
internalised ricin was also visualised within the perinuclear Golgi
protect cells against ricin, although it did abolish clathrin-depen-
dent endocytosis.[70] In a contradictory study, ricin uptake was with 70–80% of this small fraction being located in the trans-
shown to depend on dynamin.[71] Golgi network.[83]
Clathrin-independent endocytosis includes uptake by caveolae To date, the most direct evidence for the transport of ricin to the
(specialised, invaginated lipid rafts) and macropinocytosis.[72] Golgi complex comes from biochemical data. A recombinant form
When both caveolae and clathrin-dependent endocytosis were of ricin A chain was engineered to contain an internal tyrosine
perturbed by the removal of membrane cholesterol, ricin uptake sulphation site.[84] Internalisation of the unlabelled holotoxin into
still occurred.[70,73] The molecular machinery controlling this cells metabolically labelled with [35S]-sulphate led to the radio-
clathrin-independent endocytosis has yet to be elucidated, but labelling of a proportion of toxin. This outcome demonstrated that
uptake may occur by more than one mechanism.[65,71,74-79] In cells transport to the trans-Golgi network, the location of tyrosyl
lacking functional clathrin-dependent uptake, the rate of ricin sulphotransferase, had occurred.

© Adis Data Information BV 2003. All rights reserved. Toxicol Rev 2003; 22 (1)
58 Lord et al.

The question arises as to how ricin is trafficked from en- using electron microscopy, it has not been possible to visually
dosomes to the Golgi apparatus? Studies have revealed the exis- detect this toxin within either the Golgi cisternae or the endoplas-
tence of more than one route,[67,85-88] one of which is utilised by the mic reticulum. Such studies do not allow us to determine whether
mannose-6-phosphate receptor[89,90] in a ‘classical’ pathway in- ricin can be translocated from this location or whether an even
volving transport from early to late endosomes to the trans-Golgi smaller fraction, too low to allow detection, is sorted and trans-
network. Early to late endosome transport requires the low pH- ported in a retrograde manner to the endoplasmic reticulum.
dependent formation of carrier vesicles.[91] However, ricin trans- The question as to whether ricin escapes to the cytosol from the
port to the Golgi is not affected by reagents that perturb the low pH Golgi or whether it needs to traffic elsewhere for the membrane
of endosomes.[65,92] Additionally, over-expression of a dominant- transport step has been more successfully addressed using bio-
negative mutant of Rab9, a small GTPase involved in regulating chemical assays. An early study demonstrated that ricin cytotoxic-
vesicular transport between late endosomes and the trans-Golgi ity required a brefeldin A-sensitive step.[98-100] In most cells,
network,[89,90] failed to protect cells from intoxication by ricin[70] brefeldin A interferes with a membrane bound GTP exchange
and did not abrogate sulphation of internalised toxin.[67] factor,[101] leading to a reorganisation of internal membranes and a
Collectively, these studies indicate that ricin may be transport- redistribution of Golgi contents to the endoplasmic reticu-
ed to the Golgi apparatus by a pathway that is independent of late lum.[102,103] Ilimaquinone, a drug that causes Golgi vesiculation in
endosomes (figure 2). Existence of a direct pathway from early/ some cell types, can also protect against ricin.[104] The simplest
recycling endosomes to the Golgi apparatus has been more thor- interpretation of these protective phenotypes is that ricin must
oughly investigated using a related bacterial ribotoxin, Shiga travel through the Golgi before it gains access to its ribosomal
toxin.[72,86,93] Recently, specific proteins, e.g. Rab6a′ and cognate substrates in the cytosol. Furthermore, transient expression of
SNAREs,1 have been implicated in the direct early endosome-to- mutant GTPases that regulate vesicle transport steps in the early
trans-Golgi network transport step utilised by Shiga toxin.[94] The secretory pathway partially protected cells from ricin,[105] Intro-
Rab9-independent transport of ricin suggests that this protein can duction of a C-terminal endoplasmic reticulum retrieval signal
also exploit the direct route from early endosomes to the Golgi, (KDEL2) into ricin A chain increased the cytotoxicity of reconsti-
although further work is required to correlate such transport with tuted holotoxin,[106,107] and of free ricin A chain,[108,109] suggesting
other markers of this pathway. It is indeed conceivable that ricin an encounter of incoming toxin with Golgi-to-endoplasmic reticu-
can utilise multiple transport pathways from the endosomal com- lum cycling KDEL receptors. Under these conditions, free ricin A
partments to the Golgi apparatus due to the multiplicity of its chain would enter the cells by fluid phase uptake.[110] Ricin
binding partners. holotoxin has been shown to interact in vivo with the highly
Other molecules have a lesser or uncertain relevance. Retro- abundant endoplasmic reticulum chaperone calreticulin.[111]
grade routing to the Golgi apparatus of ricin and Shiga toxin was The most direct evidence for Golgi-to-endoplasmic reticulum
only marginally affected by mutations in the recycling endosome- transport of ricin, however, has been shown by the brefeldin A-
localised GTPase, Rab11.[67,93] This would suggest that a sensitive uptake and subsequent core-glycosylation of a non-
Rab11-dependent route from the recycling endosome to the trans- glycosylated holotoxin.[84] Post-translational core-glycosylation of
Golgi network is of minor significance. The importance of lipids an endocytosed protein is atypical, but it provides compelling
such as cholesterol, and cholesterol-binding proteins, in regulating evidence for complete retrograde transport of ricin from the cell
the intracellular transport of ricin is unclear at present. Indeed, surface to the endoplasmic reticulum where oligosaccharyl trans-
only marginal effects on ricin sulphation have been reported when ferase is exclusively located. The presence of this glycosylated A-
cholesterol homeostasis was perturbed,[88,95] or when signalling chain in a cytosolic fraction further indicated that membrane
via protein kinase A was inhibited.[96] translocation takes place once transport of the toxin to endoplas-
mic reticulum is accomplished.[84]
2.4 Transport of Ricin from Golgi to
Endoplasmic Reticulum
How might ricin traffic through the Golgi apparatus? The
classic pathway of retrograde transport involves movement via
Ricin has been visualised in the Golgi.[97] However, as moni- coatomer protein 1-coated vesicles.[112,113] A second, less well
tored by immunogold labelling, only ~5% of toxin was accumulat- characterised pathway is coatomer protein 1-independent[114,115]
ed in this compartment after a 1-hour internalisation.[83] Indeed, (figure 2). This pathway mediates the transport of some cycling

1 SNARE is the SNAP receptor, SNAP is the soluble NSF attachment protein, and NSF is the N-ethyl-maleimide-sensitive fusion protein.
2 KDEL is tetrapeptide lysine-aspartic acid-glutamic acid-leucine.

© Adis Data Information BV 2003. All rights reserved. Toxicol Rev 2003; 22 (1)
Ricin Toxicity 59

Golgi glycosylation enzymes and possibly lipids, in a manner chaperones and associated proteins that assist in the folding of
controlled by Rab6[114] (a small GTPase that modulates intra-Golgi nascent proteins. Some proteins serve to ‘time-out’ interactions
transport). It should be noted, however, that the overall signifi- with chaperones, whilst others somehow couple recognition of a
cance of coatomer protein 1-independent transport and the nature defective protein with the retrotranslocation machinery for extrac-
of any carriers involved remain unclear. Both pathways are tion and destruction in the cytosol. Genetic and biochemical
utilised by a number of bacterial toxins. For example, cholera evidence support a role for the Sec61 protein translocons in the
toxin and Pseudomonas exotoxin A exploit KDEL receptors that retro-translocation of endoplasmic reticulum-associated protein
in turn are sequestered into coatomer protein 1 coated vesi- degradation substrates.[127] Tightly coupled to this process is the
cles.[116-119] In contrast, Shiga toxin utilises the coatomer protein ubiquitination of internal lysyl residues of endoplasmic reticulum
1-independent route.[114,115] Only one study has been made with degradation substrates. However, whether this modification is
ricin, where a block in coatomer protein 1 function did not protect required only for subsequent targeting to proteasomes or whether
cells from intoxication.[120] it is needed for the actual retro-translocation process remains
unclear. Although understanding of retro-translocation is incom-
2.5 Membrane Translocation of Ricin plete, there is a sufficient framework of implicated components to
test their involvement in ricin dislocation.
Although ricin translocation has been reported from fractionat- A pre-requisite for the activity of ricin A chain is that it is
ed endosomes,[121] this may represent a minor transport step or one reductively separated from ricin B chain. At present, it is unclear
that occurred from contaminating microsomes, since a large body whether this occurs in the endoplasmic reticulum as it does for
of evidence (reviewed in section 2.4) has since accumulated to cholera toxin[129-131] and Pseudomonas exotoxin A[132] or whether
support the idea that ricin is routed to the endoplasmic reticulum. it occurs in the reducing environment of the cytosol. An early
Why might ricin need to traffic to this organelle to exert a toxic study implicated translocation of a non-reducible ricin holotoxin
effect? It is likely that ricin’s complex intracellular journey is that was as cytotoxic as native ricin[133] although this has never
connected to the membrane translocation step. Many bacterial been further characterised and, as yet, the precise fate of ricin B
toxins form pores in biological membranes, either directly or upon chain is unknown.
exposure to low pH. Ricin, however, does not form pores in To date, no chaperone interactions other than holotoxin with
membranes. One appealing possibility is that ricin must be endo- calreticulin have been detected[111] and, as such, it remains unclear
cytosed to a compartment that contains in its membrane pre- how ricin/ricin A chain is selected and targeted to the retro-
existing protein-conducting channels through which the toxin can translocation apparatus. It remains possible that reduction of the
exit.[122] In the endomembrane system, the only organelle that is holotoxin in the endoplasmic reticulum exposes region(s) within
known to meet this criterion is the endoplasmic reticulum, whose ricin A chain normally masked in ricin that allow this subunit to
membrane contains abundant protein translocases (known as interact with the membrane. Significant structural changes have
Sec61 translocons)[123] and peptide transporters.[124] While peptide been shown to occur in ricin A chain, but not in ricin B chain or
transporter-independent cells have been shown to be as sensitive holotoxin, when added to artificial liposomes containing negative-
to ricin as parental peptide transporter-positive cells,[8] effort has ly charged lipids.[134] The physiological significance of this in
focused on showing that protein translocon complexes provided relation to the endoplasmic reticulum membrane requires clarifica-
the conduits for toxin export. However, the tightly gated Sec61 tion. This situation contrasts with knowledge of cholera toxin
channels were originally thought to be involved only in the import interactions where protein disulphide isomerase serves as an un-
of nascent proteins, making it difficult to explain how this machin- foldase to present toxin to the retro-translocation machinery.[135]
ery might be used in the reverse direction. The involvement of protein translocase complexes in toxin
The possible mechanism for this step remained elusive until retro-translocation has been shown by co-immunoprecipitation of
recognition of a quality control system in the endoplasmic reticu- the protein translocase alpha subunit with the post-translationally
lum whereby newly made proteins detected as aberrant or orphan core-glycosylated ricin A chain variant that was endocytosed to
become dislocated to the cytosol for degradation by proteasomes. the endoplasmic reticulum as part of a holotoxin.[107] Interestingly,
This quality control pathway is termed endoplasmic reticulum- Chinese hamster ovary cell lines with genetic defects in endoplas-
associated protein degradation (ERAD).[125,126] A schematic to mic reticulum-associated protein degradation also exhibited in-
outline the fate of a typical misfolded soluble protein in the yeast creased resistance to ricin intoxication.[136]
endoplasmic reticulum is shown in figure 3. In mammalian cells, One problem faced by toxins that hijack the retro-translocation
ERAD is more complex, with an elaborate system of molecular machinery is that to be toxic they must avoid the ultimate fate of

© Adis Data Information BV 2003. All rights reserved. Toxicol Rev 2003; 22 (1)
60 Lord et al.

Deglycosylation Proteasomal
degradation

PNGase

Import via
Cytosol Sec61 translocon
Hrd1p Ubiquitination

Sec61p
Sec61p
Ubc1p/7p

Hrd3 Cue1p
ER

Export via
Kar2p
Sec61 translocon
Glycosylation

Misfolded protein Protein folded


targeted for degradation correctly for activity
Fig. 3. Schematic representation of the endoplasmic reticulum (ER)-associated protein degradation (ERAD) pathway in the yeast Saccharomyces
cerevisiae. Newly made proteins are delivered through the translocon (comprising the Sec61p complex and accessory proteins) into the ER lumen. After
scrutiny by the quality control system, many persistently misfolded proteins are targeted via the translocon back to the cytosol for destruction (for a recent
review see Jarosch et al.[128]). The chaperone Kar2p, protein disulphide isomerase family members and mannose-binding lectins have been implicated in
the early events relating to the turnover of misfolded proteins. Membrane proteins (Hrd3p and Der1p) serve as potential receptors for these substrate
recognition components, whilst ubiquitin-conjugating enzymes (e.g. Ubc1p, Ubc6p and Ubc7p), and membrane bound ubiqutin ligases (e.g. Hrd1p) are
responsible for the polyubiquitination of internal lysyl residues of ERAD substrates. Note that the protein Cue1p recruits soluble Ubc7p to the surface of the
ER membrane. Ubiquitination of ERAD substrates on the cytosolic surface of the membrane appears to be crucial for the retrotranslocation process,
serving to provide a ratcheting mechanism or to recruit other complexes such as proteasomes or the Cdc48p complex that in turn leads to eventual
degradation by proteasomes. Peptide N-glycanase (PNGase) is a cytosolic peptide that removes glycans from glycoproteins at some point prior to their
degradation. For clarity, not all proteins are shown in the schematic.

endoplasmic reticulum-associated protein degradation substrates, suggests that ricin A chain can be targeted to proteasomes in a
namely degradation by proteasomes. It was noticed that the cata- ubiquitin-independent manner, but that uncoupling from the ubi-
lytic subunits of a number of toxins shown to reach the endoplas- quitination process provides an opportunity for refolding, and
mic reticulum contained very few, if any, lysine residues.[137] This hence degradation avoidance, of at least a fraction of the toxin.
intriguing observation led to a hypothesis that a paucity of lysyl Partially unfolded ricin A chain has been shown to refold in the
residues may serve to assist the uncoupling of toxin retro-translo- presence of ribosomes to regain full catalytic activity.[140] This
cation from downstream degradation by avoidance of the ubiqui- work has suggested that ribosomes may act as suicidal chaperones
since, by the very act of refolding ricin A chain, the ribosome
tination process.[137] This hypothesis has been tested by modulat-
becomes irreversibly modified.
ing the lysine content in ricin, abrin[138] and cholera toxin.[139] In all
cases, the lysine content altered cytotoxicity in the predicted way,
2.6 Retro-Translocation of Ricin A Chain in Other Systems
such that the greater the lysine content the lower the cytotoxicity
of the toxin unless proteasomes were inhibited. It would appear The genetically tractable budding yeast, Saccharomyces cerevi-
that ricin A chain is not a natural substrate for ubiquitination and siae, has routinely been utilised to investigate eukaryotic cellular
that its low lysine content attenuates what might otherwise be a processes that are more complex (and hence more difficult to
very efficient process for removing and thereby disarming this study) in mammalian systems. However, exogenously adminis-
toxin in the cytosol. Interestingly, when proteasomes are inhibited, tered ricin is not lethal to yeast due to the absence of an appropriate
cells are typically sensitised 2- to 3-fold to ricin.[8,111,138] This galactosyl transferase. Yeast ribosomes are sensitive to ricin A

© Adis Data Information BV 2003. All rights reserved. Toxicol Rev 2003; 22 (1)
Ricin Toxicity 61

chaintoxicity, however,[141] and ricin A chaincan be synthesised de unwanted properties (its enzymatic activity and promotion of
novo and delivered into the yeast endoplasmic reticulum, render- vascular leak[147] whilst retaining protein stability and important
ing this system appropriate for the study of retro-translocation. immunodominant epitopes[148]), will also be crucial in the develop-
Quality control is less stringent in this organism than it is in ment of a long awaited protective vaccine against this toxin.[149]
mammalian cells and there are some differences in the required
components, including the absence of a calreticulin homo- Acknowledgements
logue.[126] Nevertheless, many of the players involved in the pro-
Work cited from the Warwick laboratory was supported by the UK
cess of ERAD in yeast have been identified (figure 3), although the
BBSRC and The Wellcome Trust. The authors have no conflicts of interest
relevance and sequential involvement of most of these in the retro- that are directly relevant to the content of this manuscript.
translocation of ricin A chain is unknown. As in mammalian cells,
the Sec61 translocon is known to be the channel through which
References
most misfolded yeast ERAD substrates are exported from the 1. Lord JM, Roberts LM. Toxin entry: retrograde transport through the secretory
endoplasmic reticulum.[142,143] Simpson et al.[144] investigated the pathway. J Cell Biol 1998; 140: 733-6
degradation kinetics of ricin A chain in a number of yeast endo- 2. Eiklid K, Olsnes S, Pihl A. Entry of lethal doses of abrin, ricin and modeccin into
the cytosol of HeLa cells. Exp Cell Res 1980; 126: 321-6
plasmic reticulum-associated protein degradation mutants. Using 3. Olsnes S, Pihl A. Toxic lectins and related proteins. In: Cohen P, van Heyningen S,
protein translocation mutants that were competent for import into editors. Molecular action of toxins and viruses. Amsterdam: Elsevier, 1982:
the endoplasmic reticulum while being cold sensitive for endo- 51-105
4. Griffiths GD, Rice P, Allenby AC, et al. Inhalation toxicology and histopathology
plasmic reticulum to cytosol export,[142] it was shown that ricin A of ricin and abrin toxins. Inhal Toxicol 1995; 7: 269-88
chain mutants could be stabilised relative to their rapid degrada- 5. United Nations Convention on the prohibition of the development, production,
stockpiling and use of chemical weapons and on their destruction; 1993 Jan 13;
tion in wild-type strains.[144] As in mammalian cells, it was also
Paris
evident that the ricin A chain was degraded in a ubiquitin-indepen- 6. Kreitman RJ. Immunotoxins in cancer therapy. Curr Opin Immunol 1999; 11:
dent, proteasome-dependent manner. The need for a recycling of 570-8
7. Frankel AE, Kreitman RJ, Sausville EA. Targeted toxins. Clin Cancer Res 2000; 6:
ricin A chain between Golgi and endoplasmic reticulum, as re- 326-34
quired for some soluble yeast endoplasmic reticulum-associated 8. Smith DC, Gallimore A, Jones E, et al. Exogenous peptides delivered by ricin
protein degradation substrates,[145,146] is not known. require processing by signal peptidase for transporter associated with antigen
processing-independent MHC class 1-restricted presentation. J Immunol 2002;
169: 99-107
3. Conclusions 9. Smith DC, Lord JM, Roberts LM, et al. 1st class ticket to class 1: protein toxins as
pathfinders for antigen presentation. Traffic 2002; 3: 697-704
Ricin is exquisitely potent to mammalian cells, being able to 10. Stirpe F, Barbieri L, Battelli MG, et al. Ribosome inactivating proteins from
fatally disrupt protein synthesis by attacking the Achilles heel of plants: present studies and future prospects. Biotechnology (N Y) 1992; 10:
405-11
the ribosome. For this enzyme to reach its substrate, it must not 11. Barbieri L, Battelli MG, Stirpe F. Ribosome inactivating proteins from plants.
only negotiate the endomembrane system but it must also cross an Biochim Biophys Acta 1993; 1154: 237-82
internal membrane and avoid complete degradation without com- 12. Hartley MR, Chaddock JA, Bonness MS. The structure and function of ribosome
inactivating proteins. Trends Plant Sci 1996; 1: 254-9
promising its activity in any way. It is clear that ricin can take 13. Peumans WJ, Hao Q, van Damme EJ. Ribosome-inactivating proteins from plants:
advantage of many target cell molecules, pathways and processes. more that RNA N-glycosidases? FASEB J 2001; 15: 1493-506
Future studies will attempt to clarify more exactly the range and 14. Montford W, Villafranca JE, Monzingo AF, et al. The three-dimensional structure
of ricin at 2.8A resolution. J Biol Chem 1987; 262: 5398-403
regulation of endocytic pathways used by ricin, the nature of intra- 15. Rutenber E, Katzin BJ, Ernst S, et al. Crystallographic refinement of ricin to 2.5Å.
molecular carriers and transport intermediates, the interactions Proteins 1991; 10: 240-50
16. Rutenber E, Robertus JD. Sructure of ricin B-chain at 2.5Å resolution. Proteins
that lead to subversion of endoplasmic reticulum-associated pro-
1991; 10: 260-9
tein degradation and toxin refolding, and the precise downstream 17. Villafranca JE, Robertus JD. Ricin B chain is the product of gene duplication. J
steps leading to apoptosis. In parallel with such fundamental Biol Chem 1981; 256: 554-6
18. Katzin BJ, Collins EJ, Robertus JD. Structure of ricin A-chain at 2.5 Å. Proteins
studies, ricin will continue to be used therapeutically for the 1991; 10: 251-9
selective killing of cancer cells.[6,7] Where delivery of material to 19. Mlsna D, Monzingo AF, Katzin BJ, et al. Structure of recombinant ricin A-chain at
the endoplasmic reticulum is required, the unusual intracellular 2.3 Å resolution. Protein Sci 1993; 2: 429-35
20. Weston SA, Tucker AD, Thatcher DR, et al. X-ray structure of recombinant ricin
trafficking of this protein (as well as other bacterial toxins) can A-chain at 1.8 Å resolution. J Mol Biol 1994; 244: 410-22
also be exploited. In this way, disarmed (catalytically inactive) 21. Monzingo AF, Robertus JD. X-ray analysis of substrate analogs in the ricin A-
versions will be developed as molecular carriers to trigger an chain active site. J Mol Biol 1992; 227: 1136-45
22. Endo Y, Tsurugi K. RNA N-glycosidase activity of ricin A-chain: mechanism of
immune response against certain tumours and viruses.[8] Combin- action of the toxic lectin ricin on eukaryotic ribosomes. J Biol Chem 1987; 262:
ing our understanding of the ricin structure with ways to cripple its 8128-30

© Adis Data Information BV 2003. All rights reserved. Toxicol Rev 2003; 22 (1)
62 Lord et al.

23. Lewis MS, Youle RJ. Ricin subunit association: thermodynamics and the role of 48. Ferrini JB, Martin M, Taupiac MP, et al. Expression of functional ricin B chain
the disulfide bond. J Biol Chem 1986; 261: 11571-7 using the baculovirus system. Eur J Biochem 1995; 233: 772-7
24. Wright HT, Robertus JD. The intersubunit disulfide bridge of ricin is essential for 49. Lamb FI, Roberts LM, Lord JM. Nucleotide sequence of cloned cDNA coding for
cytotoxicity. Arch Biochem Biophys 1987; 256: 280-4 preproricin. Eur J Biochem 1985; 148: 265-70
25. Endo Y, Tsurugi K. The RNA N-glycosidase activity of ricin A-chain. J Biol Chem 50. Lord JM. Precursors of ricin and Ricinus communis agglutinin: glycosylation and
1988; 263: 8735-9 processing during synthesis and intracellular transport. Eur J Biochem 1985;
26. Wool IG, Correll CC, Yuen-Ling C. Structure and function of the sarcin-ricin 146: 411-6
domain. In: Garrett J, Douthwaite L, Lilias PJ, et al., editors. The ribosome: 51. Roberts LM, Lord JM. The synthesis of Ricinus communis agglutinin. Eur J
structure, function, antibiotics and cellular interactions. Washington, DC: ASM Biochem 1981; 119: 31-4
Press, 2000: 461-473 52. Lord JM. Synthesis and intracellular transport of lectin and storage protein precur-
27. Ban N, Nissen P, Hansen J, et al. Placement of protein and RNA structures into a sors in endosperm from castor bean. Eur J Biochem 1985; 146: 403-9
5Å-resolution map of the 50S ribosomal subunit. Nature 1999; 400: 841-7 53. Lord JM, Harley SM. Ricinus communis agglutinin B chain contains a fucosylated
28. Sperti S, Montanaro L, Mattioli A, et al. Inhibition by ricin of protein synthesis in oligosaccharide side chain not present on ricin B chain. FEBS Lett 1985: 189;
vitro: 60 S ribosomal subunit as the target of the toxin. Biochem J 1973; 136: 72-6
813-5 54. Harley SM, Lord JM. In vitro endoproteolytic cleavage of castor bean lectin
29. Nilsson L, Nygard O. The mechanism of the protein synthesis elongation cycle in precursors. Plant Sci 1985; 41: 111-6
eukaryotes: effect of ricin on the ribosome interaction with elongation factors. 55. Frigerio L, Vitale A, Lord JM, et al. Free ricin A chain, proricin and native toxin
Eur J Biochem 1986; 161: 111-7 have different cellular fates when expressed in tobacco protoplasts. J Biol Chem
30. Hartley MR, Legname G, Osborn R, et al. Single-chain ribosome-inactivating 1998; 273: 14194-9
proteins from plants depurinate Escherichia coli 23S ribosomal RNA. FEBS 56. Frigerio L, Jolliffe NA, Di Cola A, et al. The internal propeptide of the ricin
Lett 1991; 290: 65-8 precursor carries a sequence-specific determinant for vacuolar sorting. Plant
31. Cawley DB, Hedbolm ML, Hoffman EJ, et al. Differential sensitivity of rat liver Physiol 2001; 126: 167-75
and wheat germ ribosomes to inhibition of polyuridylic acid translation by ricin 57. Jolliffe NA, Ceriotti A, Frigerio L, et al. The position of the proricin vacuolar
A chain. Arch Biochem Biophys 1977; 182: 690-5 targeting signal is functionally important. Plant Mol Biol 2003; 41: 825-35
32. Schlossmann D, Withers D, Welsh P, et al. Role of glutamic acid 177 of the ricin 58. Richardson PT, Westby M, Roberts LM, et al. Recombinant proricin binds
toxin A chain in enzymatic inactivation of ribosomes. Mol Cell Biol 1989; 9: galactose but does not depurinate 28S ribosomal RNA. FEBS Lett 1989; 255:
5012-21 15-20
33. Frankel A, Welsh P, Richardson J, et al. Role of arginine 180 and glutamic acid 59. Harley SM, Beevers H. Ricin inhibition of in vivo protein synthesis by plant
177 of ricin toxin A chain in enzymatic inactivation of ribosomes. Mol Cell Biol ribosomes. Proc Natl Acad Sci U S A 1982; 79: 5935-8
1990 Dec; 10: 6257-63 60. Taylor S, Massiah A, Lomonossoff G, et al. Correlation between the activities of
34. Ready MP, Kim Y, Robertus JD. Site-directed mutagenesis of ricin A-chain and five ribosome-inactivating proteins in depurination of tobacco ribosomes and
implications for the mechanism of action. Proteins 1991; 10: 270-8 inhibition of tobacco mosaic virus infection. Plant J 1994; 5: 827-35
35. Kim Y, Robertus JD. Analysis of several active site resiues of ricin A chain by 61. Sandvig K, Olsnes S, Pihl A. Kinetics of binding of the toxic lectins abrin and ricin
mutagenesis and x-ray crystallography. Protein Eng 1992; 5: 775-9 to surface receptors of human cells. J Biol Chem 1976; 251: 3977-84
36. Chaddock JA, Roberts LM. Mutagenesis and kinetic analysis of the active site 62. Spilsberg B, van Meer G, Sandvig K. Role of lipids in the retrograde pathway of
Glu177 of ricin A-chain. Protein Eng 1993; 6: 425-31 ricin intoxication. Traffic 2003; 4: 544-52
37. Barbieri L, Valbonesi P, Bonora E, et al. Polynucleotide: adenosine glycosidase 63. Simmons BM, Stahl PD, Russell JH. Mannose receptor-mediated uptake of ricin
activity of ribosome-inactivating proteins: effect on DNA, RNA and poly A. toxin and ricin A chain by macrophages: multiple intracellular pathways for A
Nucleic Acids Res 1997; 25: 518-22 chain translocation. J Biol Chem 1986; 261: 7912-20
38. Brigotti M, Alfies R, Sestili P, et al. Damage to nuclear DNA induced by Shiga 64. Magnusson S, Kjeken R, Berg T. Characterization of two distinct pathways of
toxin 1 and ricin in human endothelial cells. FASEB J 2002; 16: 365-72 endocytosis of ricin by rat liver endothelial cells. Exp Cell Res 1993; 205:
39. Komatsu N, Oda T, Muramatsu T. Involvement of both caspase-like proteases and 118-25
serine proteases in apoptotic cell death induced by ricin, modeccin, diphtheria 65. Sandvig K, van Deurs B. Endocytosis, intracellular transport, and cytotoxic action
toxin and Pseudomonas toxin. J Biochem (Tokyo) 1998; 124: 1038-44 of Shiga toxin and ricin. Physiol Rev 1996; 76: 949-66
40. Hu R, Zhai Q, Liu W, et al. An insight into the mechanism of cytotoxicity of ricin 66. Sandvig K, van Deurs B. Entry of ricin and Shiga toxin into cells: molecular
to hepatoma cells: roles of Bcl-2 family proteins, caspases, Ca2+-dependent mechanisms and medical perspectives. EMBO J 2000; 19: 5943-50
proteases and protein kinase C. J Cell Biochem 2001; 81: 583-93 67. Iversen TG, Skretting G, Llorente A, et al. Endosome to Golgi transport of ricin is
41. Barbieri L, Valbonesi P, Righi F, et al. Polynucleotide: adenosine glycosidase is independent of clathrin and of the Rab9- and Rab11-GTPases. Mol Biol Cell
the sole activity of ribosome-inactivating proteins on DNA. J Biochem (Tokyo) 2001; 12: 2099-107
2000; 128: 883-9 68. van Deurs B, Petersen OW, Sundan A, et al. receptor-mediated endocytosis of a
42. Day PJ, Lord JM, Roberts LM. The deoxyribonuclease activity attributed to ricin-colloidal gold conjugate in Vero cells: intracellular routing to vacuolar
ribosome-inactivating proteins is due to contamination. Eur J Biochem 1999; and tubulo-vesicular portions of the endosomal system. Exp Cell Res 1985;
258: 540-5 159: 287-304
43. van Damme EJM, Hao Q, Chen Y, et al. Ribosome-inactivating proteins: a family 69. Moya M, Dautry-Varsat A, Goud B, et al. Inhibition of coated pit formation in
of plant proteins that do more than inactivate ribosomes. Crit Rev Plant Sci Hep2 cells blocks the cytotoxicity of diphtheria toxin but not that of ricin toxin.
2001; 20: 395-465 J Cell Biol 1985; 101: 548-59
44. Barbieri L, Valbonesi P, Bonora E. Polynucleotide:adenosine glycosidase activity 70. Simpson JC, Smith DC, Roberts LM, et al. Expression of mutant dynamin protects
of ribosome-inactivating proteins: effect on DNA, RNA and poly A. Nucleic cells against diphtheria toxin but not against ricin. Exp Cell Res 1998; 239:
Acids Res 1997; 25: 519-22 293-300
45. Morlon-Guyot J, Helmy M, Lombard-Frasca S, et al. Ricin lipase activity: identifi- 71. Llorente A, Rapak A, Schmid SL, et al. Expression of mutant dynamin inhibits
cation of the catalytic site and implication in cytotoxicity. J Biol Chem 2003; toxicity and transport of endocytosed ricin to the Golgi apparatus. J Cell Biol
278: 17006-11 1998; 140: 553-63
46. Lord JM, Roberts LM, Robertus JD. Ricin: structure, mode of action, and some 72. Nichols BJ, Lippincott-Schwartz J. Endocytosis without clathrin coats. Trends Cell
current applications. FASEB J 1994; 8: 201-8 Biol 2001; 11: 406-12
47. Butterworth AG, Lord JM. Ricin and Ricinus communis agglutinin subunits are all 73. Rodal SK, Skretting G, Garred O, et al. Extraction of cholesterol with methyl-beta-
derived from a single-size polypeptide precursor. Eur J Biochem 1983; 137: cyclodextrin perturbs formation of clathrin-coated endocytic vesicles. Mol Biol
57-65 Cell 1999; 10: 961-74

© Adis Data Information BV 2003. All rights reserved. Toxicol Rev 2003; 22 (1)
Ricin Toxicity 63

74. Herskovits JS, Burgess CC, Obar RA, et al. Effects of mutant rat dynamin on 99. Prydz K, Hansen SH, Sandvig K, et al. Effects of brefeldin A on endocytosis,
endocytosis. J Cell Biol 1993; 122: 565-78 transcytosis and transport to the Golgi complex in polarized MDCK cells. J Cell
75. van der Bliek AM, Redelmeier TE, Damke H, et al. Mutations in human dynamin Biol 1992; 119: 259-72
block an intermediate stage in coated vesicle formation. J Cell Biol 1993; 122: 100. Okimoto T, Seguchi T, Ono M, et al. Brefeldin A protects ricin-induced cytotoxic-
553-63 ity in human cancer KB cell line, but not in its resistant counterpart with altered
76. Artalejo CR, Henley JR, McNiven MA, et al. Rapid endocytosis coupled to Golgi structures. Cell Struct Funct 1993; 18: 241-51
exocytosis in adrenal chromaffin cells involves Ca2+, GTP, and dynamin but 101. Boman AL, Kahn RA. Arf proteins: the membrane traffic police? Trends Biochem
not clathrin. Proc Natl Acad Sci U S A 1995; 92: 8328-32 Sci 1995; 20: 147-50
77. Damke H, Baba T, van der Bliek AM, et al. Clathrin-independent pinocytosis is 102. Helms JB, Rothman JE. Inhibition by brefeldin A of a Golgi membrane enzyme
induced in cells overexpressing a temperature-sensitive mutant of dynamin. J that catalyses exchange of guanine nucleotide bound to ARF. Nature 1992; 360:
Cell Biol 1995; 131: 69-80 352-4
78. Werbonat Y, Kleutges N, Jakobs KH, et al. Essential role of dynamin in 103. Klausner RD, Donaldson JG, Lippincott-Schwartz J. Brefeldin A: insights into the
internalization of M2 muscarinic acetylcholine and angiotensin AT1A recep- control of membrane traffic and organelle structure. J Cell Biol 1992; 116:
tors. J Biol Chem 2000; 275: 21969-74 1071-80
79. Lamaze C, Dujeancourt A, Baba T, et al. Interleukin-2 receptors and detergent- 104. Namibar MP, Wu HC. Ilimaquinone inhibits the cytotoxicities of ricin, diphtheria
resistant membrane domains define a chlathrin-independent endocytic pathway. toxin, and other protein toxins in Vero cells. Exp Cell Res 1995; 219: 671-8
Mol Cell 2001; 7: 661-71 105. Simpson JC, Dascher C, Roberts LM, et al. Ricin cytotoxicity is sensitive to
80. Ray B, Wu HC. Internalization of ricin in chinese hamster ovary cells. Mol Cell recycling between the endoplasmic reticulum and the Golgi complex. J Biol
Biol 1981; 1: 544-51 Chem 1995; 270: 20078-83
81. van Deurs B, Petersen OW, Olsnes S, et al. The ways of endocytosis. Int Rev Cytol 106. Wales R, Chaddock JA, Roberts LM, et al. Addition of an ER retention signal to
1989; 117: 131-77 the ricin A chain increases the cytotoxicity of the holotoxin. Exp Cell Res 1992;
82. Scherer PE, Tang Z, Chun M, et al. Caveolin isoforms differ in their N-terminal 203: 1-4
protein sequence and subcellular distribution. J Biol Chem 1995; 270: 107. Wesche J, Rapak A, Olsnes S. Dependence of ricin toxicity on translocation of the
16395-401 toxin A-chain from the endoplasmic reticulum to the cytosol. J Biol Chem
1999; 274: 34443-9
83. van Deurs B, Sandvig K, Petersen OW, et al. Estimation of the amount of
internalized ricin that reaches the trans-Golgi network. J Cell Biol 1988; 106: 108. Wales R, Roberts LM, Lord JM. Addition of an endoplasmic reticulum retrieval
53-67 sequence to ricin A chain significantly increase its cytotoxicity to mammalian
cells. J Biol Chem 1993; 268: 23986-90
84. Rapak A, Falnes PO, Olsnes S. Retrograde transport of mutant ricin to the
endoplasmic reticulum with subsequent translocation to cytosol. Proc Natl 109. Zhan J, Stayton P, Press OW. Modification of ricin A chain, by addition of
Acad Sci U S A 1997; 94: 783-8 endoplasmic reticulum (KDEL) or Golgi (YQRL) retention sequences, en-
hances its cytotoxicity and translocation. Cancer Immunol Immunother 1998;
85. Ghosh RN, Mallet WG, Soe TT, et al. An endocytosed TGN38 chimeric protein is
46: 55-60
delivered to the TGN after trafficking through the endocytic recycling compart-
110. Sandvig K, van Deurs B. Selective modulation of the endocytic uptake of ricin and
ment in CHO cells. J Cell Biol 1998; 142: 923-36
fluid phase markers without alteration in transferring endocytosis. J Biol Chem
86. Mallard F, Antony C, Tenza D, et al. Direct pathway from early/recycling
1990; 265: 6382-8
endosomes to the Golgi apparatus revealed through the study of shiga toxin B-
111. Day PJ, Owens SR, Wesche J, et al. An interaction between ricin and calreticulin
fragment transport. J Cell Biol 1998; 143: 973-90
that may have implications for toxin trafficking. J Biol Chem 2001; 276: 7202-8
87. Mallet WG, Maxfield FR. Chimeric forms of furin and TGN38 are transported with
112. Cosson P, Letourneur F. Coatomer interaction with di-lysine endoplasmic reticu-
the plasma membrane in the trans-Golgi network via distinct endosomal path-
lum retention motifs. Science 1994; 263: 1629-31
ways. J Cell Biol 1999; 146: 345-59
113. Cosson P, Letourneur F. Coatomer (COP1)-coated vesicles: role in intracellular
88. Sandvig K, Grimmer S, Lauvrak SU, et al. Pathways followed by ricin and Shiga
transport and protein sorting. Curr Opin Cell Biol 1997; 7: 484-7
toxin into cells. Histochem Cell Biol 2002; 117: 131-41
114. Girod A, Storrie B, Simpson JC, et al. Evidence for a COP1-independent transport
89. Lombardi D, Soldati T, Riederer MA, et al. Rab9 functions in the transport route from the Golgi complex to the endoplasmic reticulum. Nat Cell Biol 1999;
between late endosomes and the trans-Golgi network. EMBO J 1993; 12: 1: 423-30
677-82
115. White J, Johannes L, Mallard F, et al. Rab6 coordinates a novel Golgi to ER
90. Riederer MA, Soldati T, Shapiro AD, et al. Lysosome biogenesis requires Rab9 retrograde transport pathway in live cells. J Cell Biol 1999; 147: 743-59
function and receptor recycling from late endosomes to the trans-Golgi net-
116. Chaudhary V, Jinno Y, FitzGerald D, et al. Pseudomonas exotoxin contains a
work. J Cell Biol 1994; 125: 573-82
specific sequence at the carboxyl terminus that is required for cytotoxicity. Proc
91. Clague MJ, Urbe S, Aniento F, et al. Vacuolar ATPase activity is required for Natl Acad Sci U S A 1990; 87: 308-12
endosomal carrier vesicle formation. J Biol Chem 1994; 269: 21-4 117. Lencer WI, Constable C, Moe S, et al. Targeting of cholera toxin and Escherichia
92. Melby EL, Prydz K, Olsnes S, et al. Effect of monensin on ricin and fluid phase coli heat labile toxin in polarized epithelia: role of COOH-terminal KDEL. J
transport in polarized MDCK cells. J Cell Biochem 1991; 47: 251-60 Cell Biol 1995; 131: 951-62
93. Mallard F, Tang BL, Galli T, et al. Early/recycling endosomes-to-TGN transport 118. Majoul I, Sohn K, Wieland FT, et al. KDEL receptor (Erd2p)-mediated retrograde
involves two SNARE complexes and a Rab6 isoform. J Cell Biol 2002; 156: transport of the cholera toxin A subunit from the Golgi involves COP1, p23, and
653-64 the COOH-terminus of Erd2p. J Cell Biol 1998; 143: 601-12
94. Johannes L. The epithelial cell cytoskeleton and intracellular trafficking: 1 Shiga 119. Jackson ME, Simpson JC, Girod A, et al. The KDEL retrieval system is exploited
toxin B-subunit system, retrograde transport, intracellular vectorization, and by Pseudomonas exotoxin A, but not by Shiga-like toxin-1, during retrograde
more. Am J Physiol Gastroint Liver Physiol 2002; 283: G1-7 transport from the Golgi complex to the endoplasmic reticulum. J Cell Sci 1999;
95. Grimmer S, Iversen TG, van Deurs B, et al. Endosome to Golgi transport of ricin is 112: 467-75
regulated by cholesterol. Mol Biol Cell 2000; 11: 4205-16 120. Chen A, Hu T, Mikoryak C, et al. Retrograde transport of protein toxins under
96. Birkeli KA, Llorente A, Torgersen ML, et al. Endosome-to-Golgi transport is conditions of COPI disfunction. Biochim Biophys Acta 2002; 1589: 124-39
regulated by protein kinase A type II alpha. J Biol Chem 2003; 278: 1991-7 121. Beaumelle B, Alami M, Hopkins CR. ATP-dependent translocation of ricin across
97. van Deurs B, Tonnessen TI, Peterson OW, et al. Routing of internalised ricin and the membrane of purified endosomes. J Biol Chem 1993; 268: 23661-9
ricin conjugates to the Golgi complex. J Cell Biol 1986; 102: 37-47 122. Pelham HRB, Roberts LM, Lord JM. Toxin entry: how reversible is the secretory
98. Yoshida T, Chen C, Zhang M, et al. Disruption of the Golgi apparatus by brefeldin pathway? Trends Cell Biol 1992; 2: 183-5
A inhibits the cytotoxicity of ricin, modeccin and Pseudomonas toxin. Exp Cell 123. Johnson AE, van Waes MA. The translocon: a dynamic gateway at the ER
Res 1991; 192: 389-95 membrane. Annu Rev Cell Dev Biol 1999; 15: 799-842

© Adis Data Information BV 2003. All rights reserved. Toxicol Rev 2003; 22 (1)
64 Lord et al.

124. Russ G, Esquivel F, Yewdell JW, et al. Assembly, intracellular localization and 139. Rodighiero C, Tsai B, Rapoport TA, et al. Role of ubiquitination in retro-
nucleotide binding properties of the human peptide transporters TAP1 and translocation of cholera toxin and escape of cytosolic degradation. EMBO Rep
TAP2 expressed by recombinant vaccinia viruses. J Biol Chem 1995; 270:
2002; 3: 1222-7
312-24
140. Argent RH, Parrott AW, Day PJ, et al. Ribosome-mediated folding of partially
125. Ivessa NE, Kitzmuller C, de Virgilio M. ER-associated protein degradation inside
and outside of the endoplasmic reticulum. Protoplasma 1999; 207: 16-23 unfolded ricin A chain. J Biol Chem 2000; 275: 9263-9

126. Brodsky JL, McCracken AA. ER protein quality control and proteasome-mediated 141. Frankel A, Schlossman D, Welsh P, et al. Selection and characterisation of ricin
protein degradation. Semin Cell Dev Biol 1999; 10: 507-13 toxin A-chain mutations in Saccharomyces cerevisiae. Mol Cell Biol 1989; 9:
127. Tsai B, Ye Y, Rapoport TA. Retro-translocation of proteins from the endoplasmic 415-20
reticulum to the cytosol. Nat Rev Mol Cell Biol 2002; 3: 246-55 142. Pilon M, Shekman R, Römisch K, et al. Sec61p mediates export of a misfolded
128. Jarosch E, Geiss-Friedlander R, Meusser B, et al. Protein dislocation from the secretory protein from the endoplasmic reticulum to the cytosol for degradation.
endoplasmic reticulum: pulling out the suspect. Traffic 2002; 3: 530-6 EMBO J 1997; 16: 4540-8
129. Orlandi PA. Protein disulfide isomerase reduction of the A subunit of cholera toxin 143. Plemper RK, Böhmler S, Bordallo J, et al. Mutant analysis links to the translocon
in a human intestinal cell line. J Biol Chem 1997; 272: 4591-9
and BiP to retrograde protein transport for ER degradation. Nature 1997; 388:
130. Majoul I, Ferrari D, Söling HD. Reduction of protein disulfide bonds in an 891-5
oxidizing environment: the disulfide bridge of cholera toxin A subunit is
reduced in thye endoplasmic reticulum. FEBS Lett 1997; 401: 104-8 144. Simpson JC, Roberts LM, Römisch K, et al. Ricin A chain utilises the endoplasmic
reticulum-associated protein degradation pathway to enter the cytosol of yeast.
131. Tsai B, Rapoport TA. Unfolded cholera toxin is transferred to the ER membrane
and released from protein disulfide isomerase upon oxidation by Ero1. J Cell FEBS Lett 1999; 459: 80-4
Biol 2002; 159: 207-15 145. Vashist S, Kim W, Belden WJ, et al. Distinct retrieval and retention mechanisms
132. McKee ML, FitzGerald DJ. Reduction of furin-nicked Pseudomonas exotoxin A: are required for the quality control of endoplasmic reticulum protein folding. J
an unfolding story. Biochemistry 1999; 38: 16507-5513 Cell Biol 2001; 155: 355-68
133. Mohanraj D, Ramakrishnan D. Cytotoxic effects of ricin without an interchain 146. Caldwell SR, Hill KJ, Cooper AA. Degradation of endoplasmic reticulum (ER)
disulfide bond: genetic modification and chemical crosslinking studies. Bi-
quality control substrates requires transport between the ER and Golgi. J Biol
ochim Biophys Acta 1995; 1243: 399-406
Chem 2001; 276: 23296-303
134. Day PJ, Pinheiro TJT, Roberts LM, et al. Binding of ricin A chain to negatively
charged phospholipid vesicles leads to protein structural changes and destabi- 147. Smallshaw JE, Ghetie V, Rizo J, et al. Genetic engineering of an immunotoxin to
lizes the lipid bilayer. Biochemistry 2002; 41: 2836-43 eliminate pulmonary vascular leak in mice. Nat Biotechnol 2003 Apr; 21 (4):
135. Tsai B, Rodighero C, Lencer WI, et al. Protein disulfide isomerase acts as a redox- 387-91
dependent chaperone to unfold cholera toxin. Cell 2001; 104: 937-48 148. Tommasi M, Castelletti D, Pasti M, et al. Identification of ricin A chain HLA class
136. Teter K, Holmes RK. Inhibition of endoplasmic reticulum-associated degradation II-restricted epitopes by human T cell clones. Clin Exp Immunol 2001; 125:
in CHO cells resistant to cholera toxin, Pseudomonas aeruginosa exotoxin A, 391-400
and ricin. Infect Immun 2002; 70: 6172-9
149. Smallshaw JE, Firan A, Fulmer TR, et al. A novel recombinant vaccine which
137. Hazes B, Read RJ. Accumulating evidence suggests that several A-B toxins
protects mice against ricin intoxication. Vaccine 2002; 20: 3422-7
subvert the endoplasmic reticulum-associated protein degradation pathway to
enter target cells. Biochemistry 1997; 36: 11051-4
138. Deeks ED, Cook JP, Day PJ, et al. The low lysine content of ricin A chain reduces
Correspondence and offprints: Michael J. Lord, Department of Biological
the risk of proteolytic degradation after translocation from the endoplasmic
reticulum to the cytosol. Biochemistry 2002; 41: 3405-13 Sciences, University of Warwick, Coventry, CV4 7AL, UK.

© Adis Data Information BV 2003. All rights reserved. Toxicol Rev 2003; 22 (1)

You might also like