You are on page 1of 17

Structure

Review

Biology of Amyloid: Structure, Function,


and Regulation
Jason Greenwald1 and Roland Riek1,*
1ETH Zurich, Physical Chemistry, ETH Honggerberg, 8093 Zurich, Switzerland

*Correspondence: roland.riek@phys.chem.ethz.ch
DOI 10.1016/j.str.2010.08.009

Amyloids are highly ordered cross-b sheet protein aggregates associated with many diseases including Alz-
heimer’s disease, but also with biological functions such as hormone storage. The cross-b sheet entity
comprising an indefinitely repeating intermolecular b sheet motif is unique among protein folds. It grows
by recruitment of the corresponding amyloid protein, while its repetitiveness can translate what would be
a nonspecific activity as monomer into a potent one through cooperativity. Furthermore, the one-dimensional
crystal-like repeat in the amyloid provides a structural framework for polymorphisms. This review summa-
rizes the recent high-resolution structural studies of amyloid fibrils in light of their biological activities. We
discuss how the unique properties of amyloids gives rise to many activities and further speculate about
currently undocumented biological roles for the amyloid entity. In particular, we propose that amyloids could
have existed in a prebiotic world, and may have been the first functional protein fold in living cells.

Introduction one-dimensional nature of the order in the fibrils makes them


Amyloid fibrils have long been associated with dozens of poor candidates for three-dimensional crystallization, and to
diseases including Alzheimer’s disease (AD), Parkinson’s date, the only representative crystal structures are of amyloido-
disease, and prion diseases (Chiti and Dobson, 2006). Recently, genic peptides that are short enough to pack in a three-dimen-
however, it has become evident that there exist many amyloids, sional lattice. These structures indicate that the most basic
termed ‘‘functional amyloids,’’ that have normal biological activ- cross-b structure is in fact a one-dimensional crystal with single
ities (Fowler et al., 2007). Amyloid is a term whose definition has translational and rotational symmetry elements. Alternatively,
evolved along with our understanding of its underlying structure solid-state nuclear magnetic resonance (NMR) is a well-suited
(Sipe and Cohen, 2000). In this review, we adopt the modern high-resolution structure method for the study of amyloids, yet
biophysical definition: an unbranched protein fiber whose to date, only a single high-resolution NMR structure of an
repeating substructure consists of b strands that run perpendic- amyloid is available.
ular to the fiber axis, forming a cross-b sheet of indefinite length Amyloid Crystal Structures of Short Amyloidogenic
(Figure 1). Thus, amyloids are composed of an ordered arrange- Peptides: The Cross-b Spine at Atomic Resolution
ment of many (usually thousands) copies of a peptide or protein. Researchers in the Eisenberg lab have taken a reductionist
They are easily identified using electron microscopy (EM) as approach to determine atomic resolution structures of amyloids.
long, nonbranched filaments with diameters of 6–12 nm (Sunde By using short fibril-forming peptide segments of amyloid
and Blake, 1997) (Figure 1). The repeating cross-b sheet motif proteins (e.g., Sup35, insulin, Ab, tau, and amylin), they were
gives rise to characteristic X-ray fiber diffraction patterns with able to grow three-dimensional microcrystals that likely repre-
a meridional reflection at 4.7 Å corresponding to the inter-b sent the structure in the fibril form (i.e., in the one-dimensional
strand spacing and an equatorial reflection at 6–11 Å corre- crystal-like form) (Ivanova et al., 2009; Nelson et al., 2005;
sponding to the distance between stacked b sheets. (Astbury Sawaya et al., 2007; Wiltzius et al., 2009; Wiltzius et al., 2008).
et al., 1935; Sunde et al., 1997) (Figure 1). Among protein folds, Because all of the peptides formed both fibrils and microcrystals,
this structural entity is unique and it gives rise to a plethora of the researchers were able to demonstrate that the peptides
functions (both good and bad for the cell). It is the aim of this share a common structure in the two ordered states The
review to summarize the recently established high-resolution evidence for this includes that the microcrystals and fibrils
structural data on amyloids and to set this information in grow in the same conditions with some fibrils developing off of
perspective with their biological activities. the tips of crystals, that the long axis of the microcrystals is the
same as the fibril axis, that the microcrystals can seed the
High Resolution Structures and Their Implications growth of amyloid fibrils and that the calculated fiber diffraction
By definition, all amyloid fibrils have a translational symmetry of the microcrystals and the diffraction from their fibril counter-
element that lies parallel to the fibril axis, whereas electron parts are very similar.
micrographs indicate that most have in addition a rotational Using this approach, they have solved the structures of
element (combined rotational/translational) yielding a helical or 25 microcrystals representing 22 unique peptides. The struc-
screw symmetry. Thus, well-aligned fibrils can sometimes give tures represent a survey of the types of interactions that can
diffraction patterns that can be used to model (at low resolution) support a cross-b core including the b strand and b sheet
the fibril structure. Despite their highly ordered nature, amyloids symmetries that can exist in a cross-b fibril. Considering the
are difficult to study by high-resolution structural methods. The facts that several peptides form more than one distinct cross-b

1244 Structure 18, October 13, 2010 ª2010 Elsevier Ltd All rights reserved
Structure

Review

Figure 1. Underlying Structure of Amyloids


(A) Amyloid fibrils are composed of long filaments that are visible in negatively stained transmission electron micrographs.
(B) The schematic diagram of the cross-b sheets in a fibril, with the backbone hydrogen bonds represented by dashed lines, indicates the repetitive spacings that
give rise to (C) the typical fiber diffraction pattern with a meridional reflection at 4.7 Å (black dashed box) and an equatorial reflection at 6–11 Å (white dashed
box).

spine with distinct intersheet packing, and peptide fragments The Bigger Picture: Amyloid Fibrils of HET-s (218–289)
with overlapping sequences from the same amyloid proteins The only high-resolution model of a complete amyloid fibril
form distinct structures, the microcrystal-derived structures is the solid-state NMR structure of the prion-forming domain of
are not likely to represent the conformations of the peptides HET-s (Wasmer et al., 2008, 2009). HET-s is a functional prion
in the context of their biological amyloids. Furthermore, solid- from the filamentous fungi Podospora anserina that is involved
state NMR studies of both fibrils and microcrystals of the in a self-nonself discrimination process (Coustou et al., 1997).
peptide GNNQQNY show that the peptide conformation in the Like most prion proteins, HET-s is a multidomain protein. It has
crystals and fibrils differ from one another in subtle but distinct a globular N-terminal domain and a flexible and disordered
ways (van der Wel et al., 2007). Nonetheless, the wealth of infor- C-terminal prion-forming domain (PFD) (Balguerie et al.,
mation contained within these atomic resolution models is 2003). The PFD (comprising residues 218–289) can undergo
invaluable for understanding the fundamentals of amyloid struc- a large structural rearrangement to form a stable amyloid that
ture. For example, the collection of structures reveals two is itself the infectious entity, able to induce other HET-s mole-
classes of b sheet stacking interfaces, termed the ‘‘dry’’ and cules to form the amyloid structure. The three-dimensional
‘‘wet’’ interfaces. The dry interface is devoid of water mole- structure of HET-s(218–289) fibrils was determined using 134
cules, consisting of complementary side chain interdigitation experimental inter- and intramolecular distance restraints
that results in a high peptide packing density. Such a side chain collected in PDSD (proton-driven 13C spin diffusion) experi-
interdigitation is termed a ‘‘steric zipper’’ motif. The wet inter- ments (Wasmer et al., 2008). The fibril is a left-handed b-sole-
face is composed of hydrogen bonds between side chains, noid with each protein molecule forming two helical turns.
some via water molecules, in a manner similar to intermolecular Because there are two helical windings per molecule, the
contacts in protein crystals. This, along with the fact that some b strands alternate between inter- and intramolecular hydrogen
peptides were found in multiple crystal forms with the same dry bonding along the fiber axis. In addition, there is an offset in the
but different wet interfaces, suggests that the stable structural winding of the solenoid so that the side chain packing
unit of the microcrystals, and hence of the fibrils, is a pair of between the sheets is both inter- and intramolecular (Figure 3),
b sheets. The complete exclusion of water between the b sheets increasing the size and complexity of the intermolecular
maximizes the entropy gain for the formation of the interface interface (note: a similar intermolecular side chain arrangement
whereas the complementarity required to form such an inter- has been proposed for the Ab(1–42) fibrils associated with AD
face explains the sequence specificity of amyloid fibrils. [Luhrs et al., 2005]). In contrast to all of the small peptide struc-
Although the amyloids of full-length proteins should have tures in which none of the interstrand contacts are intramolec-
more complex structures, the types of interactions that stabilize ular, the HET-s fibril b strands pack with 50% intramolecular
them are likely to be the same as those observed in the short contacts (interstrand and intersheet). Furthermore, the pseudo-
peptides. As exemplified in Figure 2, these interactions (both repeat in the PFD amino acid sequence generates a pattern of
intra- and intersheet) can be classified as nonpolar (Van der alternating charges along the fibril axis that supports the
Waals and aromatic stacking) or polar (alternating charges correct in-register alignment of the b sheets (Figure 4). Thus,
and hydrogen-bond ladders). it seems that the HET-s monomer contains structural elements

Structure 18, October 13, 2010 ª2010 Elsevier Ltd All rights reserved 1245
Structure

Review

Figure 2. Steric Zippers, Hydrogen Bonding, and van der Waals Packing in Atomic Resolution Amyloid Crystal Structures
The interactions found in cross-b fibrils are exemplified by the structures of two peptides (A) NNQNTF (Protein Data Bank code: 3FVA) and (B) NVGSNTY (Protein
Data Bank code: 3FTL). The left panels are the views looking down the axes of the microcrystal ‘‘fibrils’’ with the neighboring dry interfaces depicted as a solvent-
accessible surface (gray) to show the tight interdigitation of the side chains. The intersheet hydrogen bonds are shown as yellow dashed lines (only a unique set
are shown, not symmetry equivalent bonds). The coloring scheme is white for main-chain carbon, yellow for side chain carbon, blue for nitrogen, and red for
oxygen. The position of the two-fold screw axes that relate the individual molecules in the fibril are indicated with black hurricane symbols. The right panels
are the view from the side of the fibrils showing only the b sheet corresponding to the colored strand from the left panel. The intrasheet hydrogen-bonds are shown
(black for main-chain only bonds and yellow for any side chain interactions) and the coloring of the main-chain carbons of individual strands alternates blue and
white. The front face of the sheet in (A) has two Asn ladders and a Phe ladder. The peptide in (B) is actually comprised of two short b strands with a kink between
them. The kink allows the Asn in the ladder to make a hydrogen bond to the main-chain oxygen of residue three (Gly) within the same molecule. In (A) the spacing
between the repetitive structures is indicated (the left panel has only the average spacing between main-chain atoms because the intersheet distances are not
related by crystallographic translations along unit cells).

that facilitate amyloid fibril nucleation (pseudo-repeats), growth microcrystals: a hydrophobic core, steric zipper-like interactions
(winding offset) (Ritter et al., 2005), and ‘‘in register’’ intermo- for the intersheet packing, and Asn ladders and p-p stacking
lecular alignment (alternate charged side chains). Within this running along the length of the fiber (Figures 3 and 4). However,
context, it is worth mentioning that, as a functional amyloid, solid-state NMR and distance restraint-based refinements in
HET-s has evolved to fold into a cross-b motif and therefore general cannot generate models with the accuracy required to
may be more complex than the disease-related amyloids for assign the conformation of every side chain. For example, as
which the cross-b structure is an unfortunate energy minimum shown in Figure 3, the side chains of the Asn residues within
on the folding landscape. The b-solenoid is in fact a large the double Asn ladder of HET-s are less well defined than those
structural family of soluble proteins (Kajava and Steven, in the peptide structures (Figure 2). This ‘‘disorder’’ in the HET-s
2006), and the complexity of the HET-s PFD amyloid structure model is unlikely to be a true representation of the structure
is on par with other protein folds. It is ironic though, that with its because the conformation of the Asn is dictated by the large
mere two helical turns per molecule, the HET-s PFD is the entropy gain achieved in satisfying a hydrogen-bond for every
shortest b-solenoid in sequence but a structure that can form Asn in the ladder without immobilizing any water molecules.
fibrils longer than 100 mm. Unlike in the peptide microcrystal In a long fibril with hundreds or thousands of repeats, this
structures, there is a pattern of order/disorder visible on all entropic effect can lead to a very large restraint on side chain
sides of the fibril (Figure 4) that arises from the flexible non- dynamics. Thus, much of the conformational variability in the
b stranded loops of the molecule and that is distributed around HET-s PFD structure is a result of the model refinement method.
the fibril by a twist in the HET-s fiber model of about 14 per Additionally, the twist rate of 14 per monomer is poorly
monomer. described by the NMR restraints and thus has an inherent
Despite its complexity, close inspection reveals that the HET-s uncertainty. It is therefore very useful to have the atomic
fibril consists of the same structural features that are found in the resolution peptide crystal structures as a complement to the

1246 Structure 18, October 13, 2010 ª2010 Elsevier Ltd All rights reserved
Structure

Review
Figure 3. The b-Solenoid Motif in HET-s
The left view is a ribbon representation of the
HET-s PFD fibril with the peptide chains alter-
nating blue and white and a ball on the N-termini
(residue 225). The four b strands are numbered
and with ‘‘a’’ and ‘‘b’’ designations for the codirec-
tional strands that are broken by a short kink and
‘‘–’’ and ‘‘+’’ designations for the strands from
the molecule below and above the central white
one. In both views, it is clear the side chain interac-
tions between the two sheets occur between
strands 1 and 4 of the same molecule and between
strands 2 and 3 of neighboring subunits. The right
view is 90 rotated from the left and is showing
only the Ca trace of the cross-b core and the
four Asn side chains in the interior of the core
that form a double Asn ladder. The absence of
the visible hydrogen-bond network in the Asn
ladder reflects the limitations of the technique
and refinement method rather than the actual
conformation of the Asn side chains (see main
text).

solid-state NMR-derived structure of the HET-s PFD, and 1999; Guijarro et al., 1998; Marcon et al., 2005; Polverino de Lau-
a synthesis of both NMR and crystallographic data can be reto et al., 2003) can alter the influence of the side chains in the
used to generate much more information about amyloids. aggregation process, perhaps driving any protein into an
The Amyloid as a Universal Structure of Polypeptides amyloid conformation.
The reductionist approach of the Eisenberg team has shown Structural Polymorphism of Amyloids
that the cross-b fold can be as simple as a single four-residue Although the current atomic resolution structural data on func-
b strand (the asymmetric unit from some of the microcrystals) tional amyloids is limited, it appears that under physiological
that assembles in a repetitive manner (Nelson et al., 2005). On conditions, the amyloid fibrils of highly evolved structures like
the other hand, the HET-s amyloid fold is as complex as that of HET-s are isomorphic structural entities, whereas ‘‘accidental’’
globular proteins. The predictive power of several algorithms for amyloids like the disease-associated amyloids are often found
the cross-b aggregation propensity of polypeptide sequences in multiple structural states, or polymorphs. One can argue that
(Fernandez-Escamilla et al., 2004; Tartaglia et al., 2008; Trovato because the disease-related amyloids result from a misfolding
et al., 2006) suggests that the cross-b state is less complex than of the polypeptide chain, their structures do not always represent
most tertiary structures, for which the only predictive capabilities the deepest or only accessible local minimum in the folding
are based on sequence similarity to known structures. Thus the energy landscape (or in this case, the aggregation energy land-
complexity of the cross-b fold can fall somewhere between scape). Furthermore, this landscape may also be sensitive to
a secondary and a tertiary structural feature. The simplicity of the environment such that changes in the conditions of aggrega-
the most basic repeating subunit of the amyloid (e.g., a b strand) tion will lead to different local minima, each representing
might suggest that any polypeptide that can form a b strand a different polymorph (Wang et al., 2010b). This energetic
could eventually form an amyloid. In fact, many proteins aggre- description explains why polymorphism exists, but currently
gate into amyloids or amyloid-like states when the delicate much more can be said about what are the structural origins of
balance between folding and aggregation is disturbed: overex- polymorphism.
pression in Escherichia coli can cause many proteins to form EM images can differentiate polymorphic fibrils based on the
inclusion bodies that have an amyloid-like substructure (Ventura degree of twisting, the number of filaments per fibril, and the
and Villaverde, 2006; Wang et al., 2008); some proteins can diameter or mass per length of the fibrils (Fandrich et al.,
aggregate into amyloid-like structures when heat denatured; 2009). Such structural heterogeneity can also be observed in
and some proteins that have stable soluble folds can be pushed solid-state NMR spectra, either as multiple NMR signals per
into an amyloid state in nonbiological conditions (Chiti et al., atom or as very broad resonance lines, indicating that the origin
1999; Guijarro et al., 1998). However, amyloid aggregation is of the macroscopically observed polymorphisms in distinct
highly amino acid sequence specific as demonstrated by the structures is at the atomic level. Indeed, the various steric zipper
‘‘steric zipper’’ side chain interactions observed in the micro- packings observed in the amyloid peptide crystal structures
crystals discussed above (Nelson and Eisenberg, 2006a; Nelson (Figure 4) represent two types of polymorphisms, packing and
et al., 2005; Sawaya et al., 2007). The essential involvement of segmental. Packing polymorphs (Figure 4A) have the same resi-
side chain interactions in the aggregation process is evident dues involved in the cross-b core but are packed differently (i.e.,
from the observed sequence-specific nature of amyloid aggre- parallel versus anti-parallel strands) whereas segmental poly-
gation (Margittai and Langen, 2006; Tjernberg et al., 2002; Zanuy morphs differ in which stretches of residues are involved in the
and Nussinov, 2003) and from the predictive power of the algo- cross-b core.
rithms for aggregation propensities. Nonetheless, an exhaustive In addition to packing and segmental polymorphs, another
screening of nonphysiological conditions such as high protein source of multiple energetically similar minima could be the
concentration, extreme pH, nonaqueous solvents (Chiti et al., repetitive nature of the aggregate state. In a soluble protein,

Structure 18, October 13, 2010 ª2010 Elsevier Ltd All rights reserved 1247
Structure

Review
Figure 4. Polymorphism in Amyloids
(A) The effect of packing polymorphism (left panel)
and side chain polymorphism (right panel) on the
surface features of amyloids. The left panel
compares the packing polymorphic structures of
the peptide NVGSNTY (PDB 3FTK top and 3FTL
bottom) with three orthogonal views. The right
panel shows a single view of the peptide LVEALYL
(PDB 3HYD) in the true crystal structure conforma-
tion on the left and with the Glu side chain rotated
to its most favored conformation while maintaining
its hydrogen bond network on the right. The
solvent-accessible surfaces are colored by atom
with yellow for carbon, blue for nitrogen and red
for oxygen.
(B) The HET-s PFD surface showing a complete
360 twist of the fibril (broken into two panels).
The amino acid residues are colored yellow for
nonpolar (F,W,Y,P,V,A,I,L,C,M,G) green for polar
(S,T,N,Q,H) and blue (R,K) and red (D,E) for
charged. The alternating surface charges on the
amyloid core are more visible in the top view
whereas the long loop connecting strands 2 and
3 is the largely yellow and green stripe of less
ordered features visible in the bottom view.

protofilaments. It has been postulated


that the tight binding between protofila-
ments of a fibril is based on weak
and nonspecific interactions that are
translated into potent interaction by the
repeat-induced cooperativity. Such
different supramolecular structures may
be the result of underlying packing,
segmental, or side chain polymorphisms
in the protofilments (Figure 4A) or may
themselves be an independent form of
polymorphism, termed ‘‘assembly poly-
small structural differences such as loop or side chain confor- morphism’’ (Paravastu et al., 2008). Regardless of their molec-
mations usually interchange on a fast time scale and comprise ular origins, all four classes of polymorphism (i.e., segmental,
a part of the large ensemble of structures that exists for one packing, side chain, and assembly polymorphism) can be prop-
particular protein. However, in highly ordered states such as agated by the self-templating nature of amyloids that ensures
protein crystals, many side chains that are known to be dynamic the persistence of distinct amyloid fibrils, each consisting of
in solution are found in only a single well-ordered state. Further- a unique conformation or polymorph. However, due to the
more, it is common to find the side chain conformations to be stochastic nature of nucleation and the fact that nucleation is
distinct between two crystals of the same protein. Thus, the often a rate-limiting step in the aggregation process, the Boltz-
one-dimensional crystal-like nature of the amyloid entity will mann distribution of peptide conformations is not necessarily
also place restrictions on the ensemble of conformations that reflected in mixtures of polymorphic amyloid aggregates. For
can exist within one fibril/crystal. An example of such side chain example, any polymorph, once it is established, may replicate
conformers is presented in Figure 4A, in which the observed itself more quickly than a more stable polymorph that nucleates
crystal structure conformation is shown on the left and the Glu only slowly.
side chain rotated to its most favored conformation while main- The Amyloid Compared to Other Protein Aggregates
taining its hydrogen bond network on the right. Although the two Although the amyloid-like conformation is a common feature of
amyloids differ only by the side chain orientation of Glu, their many protein aggregates, there are other mechanisms by which
surface features are markedly different. It is not possible to proteins can assemble into ordered aggregates without invoking
have a mixture of Glu side chain conformations within one fiber the cross-b motif. This includes domain-swapping, end-to-end
because the two are sterically mutually exclusive. This type of stacking (Bennett et al., 2006; Eisenberg et al., 2006; Nelson
polymorphism is subtler than the other two and we refer to it and Eisenberg, 2006a, 2006b), and silk-type b sheet polymeriza-
as side chain polymorphism. tion. The functional aggregation of proteins in an end-to-end or
Because amyloid fibers are often bundles of protofilaments lateral manner is most well studied in the cytoskeletal complexes
(the most basic unit of the cross-b entity), polymorphism is of the actins and tubulins. Their correct function requires that
often observed as different supramolecular assemblies of the there be a tight control over the assembly and disassembly of

1248 Structure 18, October 13, 2010 ª2010 Elsevier Ltd All rights reserved
Structure

Review

their ordered aggregates (i.e., actin filaments and microtubules). a mechanical force are required to achieve the ordered assembly
In the case of actin, the filaments retain the ability to make of fibroins into a dragline silk fiber.
branch points that are critical for the rigidity of the cytoskeleton Yet another type of protein aggregation occurs in the
by incorporating specialized branch-inducing complexes into assembly of the structural proteins like collagen and the keratins.
the filament. Because it does not involve a gross rearrangement These structural proteins comprise a variety of folds that we will
of the protein fold, the end-to-end type of aggregation is revers- not discuss here except to say that what separates them from
ible and does not limit much the speed at which the assembly other ordered aggregates is the fact that they do not spontane-
and disassembly occur. The regulation of the assembly process ously form ordered aggregates, rather they require a large
is similar for both actin and the tubulins in which nucleating degree of specialized cellular machinery and processing to
factors stimulate the initiation of aggregation whereas NTP achieve their final state. Finally, a group of aggregates that are
(nucleoside triphosphate) binding and hydrolysis regulates the classified solely on their macroscopic appearance are the amor-
assembly/disassembly. Another example of end-to-end aggre- phous aggregates, a type that is often obtained by heat precip-
gation is the assembly of hemoglobin S (HbS), the glutamate to itation or concentration precipitation of proteins. Although the
valine point mutant of hemoglobin A, into end-to-end and lateral classical macroscopic amorphous aggregate is on the meso-
fibers. The fibers of HbS distort the red blood cells into the scopic scale different from the fibrous aggregate, a recent study
abnormal rigid shape that gives the name to the resulting from our lab indicates that amorphous aggregates may also
disease: sickle cell anemia. The polymerization of HbS is contain a defined cross-b-structure, however without the long
affected by several factors including oxygen saturation, hemo- range (>mm) order found in fibrils (Wang et al., 2010b). This
globin concentration, and hemoglobin composition. This latter finding suggests that the manner in which a polypeptide back-
effect spares HbS/HbA individuals from severe anemia, and bone achieves a kinetically accessible local energy minimum
because there is no biological control over the assembly/disas- might require that there is some local order or repeated structure
sembly, HbS/HbS individuals are afflicted to various degrees. throughout the aggregate. Thus, there seems to be a caveat
In contrast to the end-to-end/lateral aggregates are those of about using the terms ‘‘ordered’’ versus ‘‘amorphous’’ protein
the arthropod silks whose ordered assembly is rapid and aggregates and it is perhaps more correct to describe aggre-
obtained on demand from specialized glands. The enormous gates based on an order/disorder continuum or an extent of
variety of silks represents many uncharacterized mechanisms long-range ordering (i.e., coherence length).
for polypeptide assembly, yielding structures that range from In summary, a comparison of the various types of aggregates
primarily a-helical content to parallel b sheets and even to described above reveals two significant features that set the
cross-b amyloid-like structures. However, the more well-studied amyloid apart from other ordered aggregates. First, the cross-b
spider fibroins present an interesting mechanism that involves spine of the amyloid can be formed from peptides as short as
dynamic control like the end-to-end aggregates, a large and irre- four residues. Second, and partly due to the first, the repeat
versible structural rearrangement like the amyloid aggregates distance is short; in the most basic amyloid it is the interstrand
and a specialized production machinery like the keratin/collagen distance of 4.7 Å. The close spacing of identical side chains
aggregates. The fibroins have multiple repeating domains of can generate specificity where none would exist without
several types with highly conserved terminal nonrepeating (NR) a repeating structure. For example, a single amphipathic b strand
globular domains. Protein constructs that contain just two types would have no affinity for a membrane, but a fibril made up of this
of repeats (Gly/Ala rich and Gly/Pro/Gln rich) can form a b-rich peptide could bind tightly to lipids. The same is true of a basic
aggregate whereas the addition of the C-terminal NR domain peptide interacting with DNA (see below). In addition to support-
to the construct produces a protein that in vitro displays many ing cooperative binding, the amyloid structure allows very short
of the aggregation properties of silk. The different types of peptides to assemble into complexes with regular secondary
repeating domains give rise to the many properties of silk, structure, thus mimicking some features of larger folded proteins.
most importantly its tensile strength and flexibility. On silk fiber Hence, amyloids have unique structural properties that can result
assembly, the Gly/Ala rich regions (thought to be helical in solu- in unique activities, as we shall see in the following section.
tion) form a b sheet rich structure in which, unlike amyloids, the
strands run parallel to the fiber axis, whereas the Gly/Pro/Gln Structure-Activity Relationship
segments adopt an uncharacterized structure that gives the The two-state (soluble/insoluble) nature of amyloids and the
elasticity to the silk. A recent study has determined the structure structural repetitiveness of the aggregates add a level of
of the C-terminal nonrepeating domain from the dragline silk of complexity that most soluble proteins cannot access. Hence,
Araneus diadematus, and showed that it is essential for amyloids can possess a variety of biophysical and biological
controlled aggregation and ordered fiber assembly (Hagn properties, supporting a diversity of functions that rivals that of
et al., 2010). The current data support the hypothesis that it is soluble proteins. The growing list of known amyloid related activ-
the terminal NR domains that allow for the storage of the aggre- ities (Table 1) shows that the formation of amyloids may result
gation prone repeats at a very high concentration in the spinning both in a loss of function of the polypeptide, as described for
gland (%50% w/v). The conversion from the soluble proteins the yeast prions (Osherovich and Weissman, 2002; True and
(thought to be in either a liquid crystal or micellar suspension) Lindquist, 2000), or in a gain of function, as in the case of the
into the ordered insoluble fibers is stimulated by a change in HET-s prion (Maddelein et al., 2002). Pmel17 amyloids serve
the salt type (from Cl to PO43 ), a decrease in pH, dehydration, as a template for ligand binding (Fowler et al., 2006), whereas
and by the shear forces as the proteins pass through the narrow other amyloids induce a specific toxic response as shown for
spinning duct. Thus it appears that both a prior ordering and the HET-s prion system (Maddelein et al., 2002). The structural

Structure 18, October 13, 2010 ª2010 Elsevier Ltd All rights reserved 1249
Structure

Review

Table 1. Functional Amyloids


Amyloid Function Reference
Curli Component of extracellular matrix involved in adhesion, aggregation, Chapman et al., 2002
invasion and biofilm formation
Microcin E492 Bacteriocin, membrane pore-forming peptide, amyloid form is Bieler et al., 2005
inactive
Chaplins Assisting aerial hyphae formation in Streptomycetes Claessen et al., 2003
Harpins Secreted by plant pathogenic bacteria, destabilize plant membranes, Oh et al., 2007
induce cell death
Sup35a Translation termination, prion form is inactive True and Lindquist, 2000
Ure2pa Regulatory function Baxa et al., 2005
in the nitrogen
catabolite repression pathway, prion form inactive
Rnq1pa Enhances the inducibility of other prions Derkatch et al., 2000
Swi1pa Chromatin remodeling factor, prion form inactive Du et al., 2008
a
Mot3 Transcriptional regulator of cell wall remodeling genes, prion form is Alberti et al., 2009
inactive
Hydrophobins Surface attachment and aerial hyphae formation Wösten and de Vocht, 2000
HET-s Heterokaryon incompatibility (see text) Coustou et al., 1997
Pmel17 Templates the synthesis of melanin Fowler et al., 2006
Neuronal CPEB Cytoplasmic polyadenylation element-binding protein regulates Si et al., 2003a
mRNA translation (see text)
Peptide hormones Sorting, storage, and release of diverse hormones (see text) Maji et al., 2009
a
Although the functional nature of these prions is still debated (Nakayashiki et al., 2005), they are included as functional because under certain stresses
they may confer some selective advantage, and there are dozens of other potential prions in yeast that have been conserved throughout fungal evolu-
tion (Alberti et al., 2009).

repetitiveness of the amyloid fiber provides an ideal template for tory peptides in highly concentrated membrane-enclosed secre-
making it into a transmissible or infectious fold. In fact, most tory granules (Arvan and Castle, 1998; Dannies, 2001; Kelly,
prions (infectious proteins) have been found in their infectious 1985) the cells can store a large quantity of the hormone until
form to be amyloids whereas the amyloid structure itself appears a signal triggers its release, at which point they can secrete
to be responsible for prion strain diversity (see below) (Toyama hormones much faster than their synthesis rates would permit.
et al., 2007). Although the in vitro formation of most amyloids During the maturation process, the prohormones are proteo-
can be nucleated or ‘‘seeded’’ by the amyloid itself from a soluble lytically processed into their active hormone entity followed by
pool of the amyloidogenic peptide or protein, not all amyloids hormone aggregation into hormone-specific granules that are
have been shown to be transmissible and thus not all amyloids membrane-enclosed. The granules are generally composed of
can be considered prions (Riek, 2006). Thus the simplicity of a single peptide species or, when coaggregated, the distinct
a cross-b-structure with its one-dimensional crystal-like nature peptides exist exclusively in a specific integer ratio. Until
can encompass a wide range of activities. As we will highlight recently, there was no explanation for how peptide hormones
in the following specific examples, their activity can be depen- self-segregated, but the amyloid nature of peptide hormones in
dent on the repetitive nature of the amyloid but also on its two- secretory granules (Maji et al., 2009) explains much of what
state nature and the compactness of the fold. were mysteries in the processes of granule formation, selection,
Peptide Hormone Amyloids in the Formation storage, and hormone release. The nature of amyloids provides
of Secretory Granules a series of controls over secretory granules that are semi-auton-
Our recent finding that the amyloid structure is involved in the omous, requiring minimal peripheral cell machinery.
formation of secretory granules in mammalian cells (Maji et al.,
2009) hints at the biological ubiquity of functional amyloids. 1. The timing and location of amyloid aggregation in the Golgi
Eukaryotic cells have specialized pathways for both the constitu- can be controlled by the processing of the prohormone.
tive and regulated transport of secretory proteins/peptides to Because the amyloid aggregation of a small peptide is
the extracellular space (Kelly, 1985, 1987). During constitutive sensitive to the sequence and chemical modifications of
secretion, a newly synthesized peptide hormone is transported the peptide, there are multiple ways in which processing
to the cell surface whereupon it is immediately released and could initiate aggregation. In addition to controlling aggre-
the rate of peptide secretion is limited by the rate of peptide gation through modification of the hormone, there is the
synthesis (Arvan and Castle, 1998; Lacy, 1975; Palade, 1975; possibility of controlling aggregation through pH or the
Tooze, 1998). In contrast, many secretory cells have an addi- presence of helper molecules such as glycosaminogly-
tional secretion pathway through which they can store peptide cans (GAGs) that can stabilize the amyloid conformation
hormones for extended periods of time. By packaging the secre- (Maji et al., 2009).

1250 Structure 18, October 13, 2010 ª2010 Elsevier Ltd All rights reserved
Structure

Review

2. The formation of amyloid fibrils is highly sequence-specific 2005). Therefore, the high-resolution structure was able to
so that once initiated, the amyloid aggregation of the confirm that the amyloid core is the infectious entity. However,
hormone is self-selective, yielding granule cores of its many functions, only the infectivity of the HET-s prion is
composed of specific hormones only. Specific coaggre- completely contained within its PFD. The heterokaryon incom-
gation of some hormones is possible as has been demon- patibility function requires a full-length protein that includes the
strated for ACTH and b-endorphin, two hormones that are N-terminal globular domain (termed the HeLo domain based
processed from a single prohormone. Because some on its conservation with other fungal proteins) and the PFD
amyloid proteins are able to cross-seed (Giasson et al., (Balguerie et al., 2004). Furthermore, the HeLo domain has
2003; Han et al., 1995), it should be expected that some a prion regulatory function that also requires a full-length protein
hormones will form mixed fibrils, although still in a specific with the proper spacing between the domains (Balguerie et al.,
manner. 2003; Greenwald et al., 2010) (see also below). In the latter
3. After initiation of aggregation and sorting of the peptides report, we conclude that the formation of the amyloid structure
into specific secretory granule cores, the amyloid struc- in the PFD causes a structural rearrangement of the HeLo
ture provides the most dense packing possible (Nelson domain that leads to the other activities of the HET-s prion. In
et al., 2005), both excluding nonaggregation-prone consti- this manner, the combination of a globular fold and an amyloid
tutively secreted proteins and providing an extremely could create an immeasurable functional landscape and one
stable storage system. that has certainly been explored by nature and that is awaiting
4. As part of the granule maturation process, the hormone discovery in the laboratory. The evolutionarily optimized fibril
amyloids are surrounded by membrane as they separate structure of HET-s has many features that are likely to be found
from the Golgi. There are many examples to suggest that in other highly evolved functional amyloids (Barnhart and
membrane binding is an inherent property of amyloid Chapman, 2006; Fowler et al., 2006, 2007). In contrast, the
aggregates (Gellermann et al., 2005; Sparr et al., 2004) disease-associated amyloids discussed in the following exam-
and so formation of a membrane around the hormone ples have functions that are detrimental to the organism and
aggregate may be spontaneous. structures that are not as easily characterized due to their high
5. Finally, on receiving the appropriate signal, the granules degree of polymorphism arising from their metastable nature.
are secreted and the amyloid structure of the hormone Distinct Aggregates, Distinct Activities
aggregates enables a controlled release of monomeric, Our recent structural studies on HypF-N aggregates shows that
functional hormone (Maji et al., 2008). Each peptide even the rapid, forced aggregation of a protein can lead to
hormone will have its own dissociation rate and this can several distinct amyloid-like states whose conformations
again be controlled by factors such as pH, ion concentra- depend on the conditions in which they were formed (Wang
tion, and extracellular chaperones. et al., 2010b). Furthermore, the activities of the aggregates in
several biochemical and biological assays varied with the
The Structure-Activity Relationship in the HET-s conformation of the aggregates. HypF-N is the N-terminal
Amyloid Prion domain of the prokaryotic hydrogenase maturation factor, which
The structure of the prion-forming domain (PFD) from HET-s has been shown to form toxic oligomers on way to forming
revealed that the infectious state of this functional amyloid is amyloid fibrils. In addition to these amyloid fibrils, we studied
a b-solenoid and thus a member of a large structural class of the bacterial inclusion bodies, heat-precipitated aggregates,
soluble proteins (Maddelein et al., 2002; Ritter et al., 2005; concentration-induced aggregates, and trichloroacetic acid-
Wasmer et al., 2008). The complexity of its structure reflects its precipitated aggregates of HypF-N. Despite the wide range of
evolutionarily optimized role as an amyloid. The b-solenoid conditions used to induce protein aggregation, all five types of
structure is not a chance misfolding of the PFD, but rather HypF-N aggregates contain the cross-b sheet motif. However,
a very stable conformation that can spontaneously occur at each of the aggregates is structurally distinct, having different
moderate protein concentration. The presence of a pseudo- segments of the protein involved in protected secondary struc-
repeat in the sequence allows a single PFD to adopt two turns tures. We compared the affinity of each aggregate to adenosine
of the b-solenoid, forming the nucleus of the amyloid fold. As triphosphate (ATP), thioflavin T, oligonucleotides, and micelles,
well, it gives rise to alternating charges along the fibril axis that as well as the ability of each to interfere with cell viability.
support the correct in-register alignment of the b sheets Although the highest activities were found in the fibrils, the other
(Figure 4). The pairing of these repeats guarantees that there is aggregates had significant activities that were not intercorre-
a unique low-energy fold without detectable polymorphism in lated (e.g., strong ATP binding was not correlated with cell
physiological HET-s fibrils (Wasmer et al., 2008). With its hydro- viability). We believe that the key to their varying properties is
phobic core and hydrophilic solvent-exposed side chains, the that each aggregate is built around a cross-b sheet motif
HET-s PFD fibril has many of the hallmarks of soluble proteins. composed of different amino acid segments. The repetitive
In fact, unlike many amyloidogenic proteins, there is no toxicity nature of the cross-b sheet motif supports cooperative interac-
associated with its overexpression in bacterial or fungal hosts tions that can generate unique and potent activities. This one
or even in rats (unpublished data). example of the landscape of aggregate structure-activity rela-
It had been previously shown that the introduction of the tionships highlights the existence of a multifactorial structure-
b sheet-breaking residue proline within the predicted b strands activity function for amyloids and begins to shed light on the
resulted in a loss of infectivity in the fungus, whereas proline complexities of polymorphs (see above) and prion strains (see
mutations in the loop positions had no effect (Ritter et al., below).

Structure 18, October 13, 2010 ª2010 Elsevier Ltd All rights reserved 1251
Structure

Review

The Infectious Nature of Amyloids toxic protein aggregates in biology, it is not surprising that
An amyloid can in principle grow by recruiting the corresponding complex regulatory systems have evolved to maintain the
soluble amyloid protein. Hence, under certain circumstances, balance of functional proteins necessary for life. These systems,
amyloid replication can occur in an organism once infected by collectively referred to as protein homeostasis, or ‘‘proteostasis’’
the corresponding amyloid (seeds). The question of what makes (Powers et al., 2009) comprise a vast network that ranges from
an amyloid infectious has been addressed in a recent mathemat- synthesis and folding (i.e., ribosome, chaperones, aggregases,
ical model that was experimentally validated in vivo (Tanaka et al., disaggregases) to degradation (i.e., proteases, autophagy, lyso-
2004). The infectivity of an amyloid depends on the concentration somal targeting). Outside of this proteostasis network, functional
of soluble and aggregated amyloid protein, the growth rate of the aggregates have their own controls that can involve external
amyloid, and the rate of breakage of the amyloid fiber into seeds. regulatory elements (i.e., enzymatic activation) or self-contained,
According to this theoretical framework, manipulation of these autoregulatory elements.
variables could transform any amyloid into a prion (Riek, 2006). Proteostasis and the Role of Chaperones in Aggregation
For example, an increase of soluble amyloid protein in the host Transgenic Caenorhabiditis elegans that express the 42-residue
might be sufficient to transform an amyloid into a prion (Meyer- peptide of Ab in their body wall muscle cells suffer an age-related
Luehmann et al., 2006). In contrast, the formation of long fibrils paralysis that is accompanied by Ab amyloid deposits. In this
decreases the seed to total amyloid protein ratio, thus reducing C. elegans model of Ab toxicity, two opposing protective path-
the prion titer (Silveira et al., 2005). Based on the nucleated poly- ways have been identified in the proteostasis network (Cohen
merization model of amyloids it is theoretically possible that all et al., 2006). Both pathways are activated by inhibition of the
amyloids (under the correct circumstances) can be infectious. IGF1-R (insulin/insulin growth factor 1-like receptor) signaling
The normally noninfections Ab amyloid-associated AD becomes pathway, which negatively regulates the heat shock transcription
infectious in a mouse that overexpresses the precursor to Ab factor HSF1. The first pathway, enabled by upregulation of HSF1
(Kane et al., 2000; Meyer-Luehmann et al., 2006) as well as in non- activity, leads to the induction of a disaggregase activity that
transgenic primates (Ridley et al., 2006). Other amyloids that decreases the amount of Ab amyloid aggregates. This protective
appear to have an infectious component to them are the amyloid disaggregase activity is detectable in an in vitro assay in which
entity of foie gras (Westermark and Westermark, 2009) and the homogenized worms are mixed with Ab fibrils with continuous
tau amyloid associated with AD. For further details, we refer the monitoring of fibril content by Thioflavin T fluorescence.
reader to an excellent review by Aguzzi and Calella (2009) that A second pathway, also downstream of IGF1-R signaling and
concentrates on this topic. dependent on the FOXO transcription factor activity, acts by
The Toxic Nature of an Amyloid enhancing the formation of large aggregates and thereby
The long list of diseases associated with amyloid formation protecting the cell from smaller, putatively more toxic oligomers.
suggests that something is inherently toxic about amyloids. The study of the yeast prion Sup35, whose amyloid state is
However, this observation is countered by the growing list of propagated in [PSI+] variants, has greatly expanded our under-
functional amyloids (Table 1). Thus, toxicity, like other protein standing of the role of chaperones in the aggregation and disag-
activities, is rather something to be associated with a particular gregation of proteins (Serio and Lindquist, 2001). Many chaper-
conformation of a peptide, such that one polymorph will have ones bind to aggregation-prone segments of proteins and
different activities from another polymorph. But what are the protect them from unfavorable interactions, thereby assisting
structural features of an amyloid that can lead to toxicity? The in protein folding and preventing unwanted aggregation. This
lack of toxicity of the functional HET-s fibrils might be due to their description can lead to a simplistic view that predicts that chap-
hydrophilic surface that would suppress nonspecific hydro- erones would negatively regulate the amyloid based prions.
phobic interactions with other proteins or membranes. Alterna- However, several examples from the yeast literature paint
tively, the folding/aggregation pathway of functional amyloids a more nuanced picture of chaperone activity. The chaperone
in general may be more cooperative, lacking oligomeric or proto- Hsp104 is a protein-remodeling factor whose disaggregase
fibril intermediates of the kind that have been suggested to be activity, which can reduce the size of Sup35 fibrils, ironically is
the toxic species in AD (Hardy and Selkoe, 2002; Klein et al., required for [PSI+] propagation. Whereas the deletion of
2004; Lazo et al., 2005). Although b sheet-rich (Chimon et al., Hsp104 leads to [PSI+] curing, so does its overexpression
2007), these conformational intermediates have a structural (Chernoff et al., 1995). Thus, the proper balance of disaggregase
motif that is distinct from fibrils as evidenced by the establish- activity is required to maintain the correct size and numbers of
ment of both oligomer-specific and fibril-specific antibodies fibril seeds for prion propagation as the larger Sup35 aggregates
(Kayed et al., 2003). Consequently, it has been suggested that formed in the absence of Hsp104 have lost their infectivity.
the disease-associated amyloid fibrils actually have a beneficial Hsp104 is not acting alone and members of the Hsp70 family
function in that they sequester the toxic oligomers into nontoxic of chaperones are also involved in [PSI+] maintenance and
mature amyloid fibrils (Hardy and Selkoe, 2002). Finally, as curing. Despite their highly conserved sequence and domain
discussed next, the fibrillogenesis of functional amyloids may organization, the Hsp70 proteins can also have opposing effects
be highly regulated, which represses any potentially toxic effects on the Sup35 aggregate load and on the [PSI+] prion state. Two
such as mislocalization or hyperaggregation. subclasses of cytosolic Hsp70 proteins in yeast are Ssa (Ssa1)
and Ssb (Ssb1 and Ssb2). Ssb behaves as expected for a chap-
Control Of Aggregation erone by inhibiting the spontaneous formation of [PSI+] in [psi ]
Considering the often delicate balance between properly folded cells and by increasing the curing efficiency of overexpressed
and misfolded proteins as well as the widespread occurrence of Hsp104. Ssa has the exact opposite effect: inhibiting [PSI+]

1252 Structure 18, October 13, 2010 ª2010 Elsevier Ltd All rights reserved
Structure

Review

curing by overexpressed Hsp104 and increasing the sponta- Self-Regulating Aggregates


neous formation of [PSI+]. Studies with Ssa/Ssb chimeras In contrast to the aggregation controls described above, there
show that the peptide-binding domain determines the [PSI+] exist both functional and nonfunctional aggregates whose
activity (Allen et al., 2005). It is proposed that Ssa stabilizes an aggregation control is encoded within their protein sequence
unfolded/prion-competent form of Sup35 whereas Ssb either or structure. Nearly all globular proteins contain these sorts of
stimulates refolding into nonprion conformation or targets mis- controls because their stable three-dimensional structure itself
folded proteins for degradation. It is also shown that in a [PSI+] prevents the aggregation of their aggregation-prone regions.
culture that overproduces Sup35, the proportion of [psi ] cells Furthermore, a multiproteome analysis has found that many
increases with the overexpression of Ssa, demonstrating that highly aggregation prone stretches of amino acids in proteins
Ssa can also have antagonistic effects on [PSI+] propagation. are flanked by ‘‘gatekeeper’’ residues of lysine, arginine, or
This latter effect, like the Hsp104 deletion, is likely to be due to proline (Rousseau et al., 2006). These gatekeeper residues
an increase in the size of the Sup35 aggregates and decrease inhibit the formation of aggregates in these aggregation-prone
in the number of seeds. Thus, the proteostasis network of chap- stretches. An analogous use of gatekeeper residues is also
erones in yeast demonstrates the importance of the balance of found in the functional amyloid, curli, where they act as a regu-
protein expression and folding for the fate of amyloidogenic lator of aggregation and toxicity (Wang et al., 2010c). The curli
proteins. Furthermore, these results highlight the fact that the gatekeepers are aspartate and glycine, but they probably act
infectivity of an amyloid is dependent on the number and size similarly to the arginine, lysine, and proline gatekeepers. Curli
of the aggregates in the cell. fibers are composed of two subunits, CsgA and CsgB. In vivo,
Enzymatic Control of Aggregation the polymerization of the major curli subunit protein, CsgA, is
Perhaps the most intricate enzymatic control of aggregation is in dependent on CsgB-mediated nucleation. The amyloid core of
the coagulation cascade. Here the control of a nonamyloid CsgA features five imperfect repeats (R1–R5) and the internal
aggregation is achieved by a series of proteolytic activations three repeats were shown to have nonconsensus (gatekeeper)
leading to the cleavage of fibrinogen by thrombin to yield fibrin, residues. When the gatekeeper residues were mutated to the
the protein that forms the fibrous aggregate that strengthens consensus residues, there was a CsgB-independent growth of
a clot. Plasminogen binds to the fibrin molecules in a clot and curli fibers in vivo. These fibers were mislocalized and were
is activated by tissue plasminogen activator, which also binds associated with cytotoxicity. Thus the curli functional amyloid
to fibrin and cleaves plasminogen to form plasmin. Plasmin, aggregation is spatially controlled through a multicomponent
proceeds to digest fibrin, thus dissolving the clot. Although fibrin nucleation with gatekeeper residue control. In addition to gate-
is not an amyloid, its regulation is analogous to the enzymatic keeper residues, other controls can be encoded in the amyloid
control of peptide hormone aggregation. The proteolytic pro- itself. The aggregation propensity of a peptide can be altered
cessing of the nonamyloidogenic prohormone by the corre- by pH or salt (e.g., spider silks) and as mentioned for the peptide
sponding prohormone convertase, which like thrombin is synthe- hormones, can require a cofactor such as a charged polymer
sized as an inactive zymogen, is required to form the mature (heparin). An example of pH control has recently been reported
hormone. Because only the mature hormones are found in the for the amyloid core of Pmel17 (McGlinchey et al., 2009) that
secretory granules, it follows that the prohormone convertases in vitro, only forms amyloids under mildly acidic conditions like
are required to initiate the amyloid aggregation of peptide those found in the melanosome lumen.
hormones. The subsequent disaggregation is not enzymatically Another type of self-regulation is found in the functional aggre-
controlled, but rather by a change in the environment of the gates of spider silk (see above) and the HET-s prion. These multi-
amyloid aggregate, through the dilution of the monomeric domain proteins have, in addition to their natively disordered
peptides as they are released from the aggregate, and possibly aggregation domain, a globular domain (i.e., HeLo in HET-s)
by extracellular chaperones. A disease-related example of enzy- whose functions include an aggregation regulatory role. In the
matic control of aggregation is the processing of the amyloid HET-s system, aggregation is used as an identity marker
precursor protein (APP) by sequential cleavage reactions to yield that is detected through amyloid-based intermolecular interac-
Ab. Because the ‘‘control’’ of Ab formation does not have a known tions: soluble HET-S recognizes amyloid HET-s. Thus the aggre-
biological function, it may be better classified as an aberrant gation control in the HET-s/S system is a little more complex in
degradation. However, there are striking similarities between that the two ‘‘isoforms’’ (95% identical) have very different
the processing of APP (the precursor to Ab) and the functional aggregation propensities. HET-S does not aggregate in vivo
amyloid Pmel17 (Hurbain et al., 2008). Pmel17 aggregation has whereas HET-s spontaneously forms aggregates in vivo and
many levels of control in that the protein must first pass through their in vitro behavior is consistent with the one in vivo. The prion
multivesicular endosomal compartments before it can be forming domains (PFDs) of HET-s and HET-S, when expressed in
proteolytically processed by a proprotein convertase. The pro- isolation, both aggregate 100 times faster than HET-s indi-
cessing is required for amyloidogenesis and the vesicular traf- cating that the globular HeLo domains of both proteins have
ficking is required for the processing (Berson et al., 2003; Hurbain some inhibitory effect on aggregation. However, in HET-S the
et al., 2008). Other similarities include the fact that like APP, effect is much stronger and Het-S can actually inhibit the aggre-
Pmel17 is a membrane glycoprotein and that their amyloidogenic gation of HET-s in trans (Balguerie et al., 2003). Our structural
fragments are without glycosylation. Although Ab still has no study of the HeLo domains of HET-S and HET-s led us to the
known function, its presence in the body and the above similar- conclusion that the thermodynamic stability of the HeLo domain
ities with Pmel17 suggest that it may have a function (Kimura and is what regulates aggregation of the PFD (Greenwald et al.,
Schubert, 1993), which even might involve the amyloid entity. 2010). The cross-b folding of the PFD puts a structural strain

Structure 18, October 13, 2010 ª2010 Elsevier Ltd All rights reserved 1253
Structure

Review

on the HeLo domain that for HET-S, but not HET-s, leads to nantly in E. coli can be polymerized into amyloid fibrils that are
a structural rearrangement that prevents further amyloid growth. infectious. However, the infectivity titer is very low and only
In this manner, HET-S can inhibit its own aggregation as well as observed on inoculation intracerebrally into transgenic mice
that of the nearly identical HET-s. It is this mechanism that allows that have a many-fold overexpression of the murine PrP frag-
the PFD of HET-S to act as an ‘‘amyloid-sensor’’ that sets off the ment PrP(89-231). Brain extracts from these infected mice con-
cell-death reaction when it encounters the prion form of HET-s. tained the protease resistant PrPres, and neuropathological
Although the downstream processes that lead to cell-death are findings suggest that a novel prion strain was created. Very
currently not known, we hypothesize that the structurally altered recently, much more compelling evidence has been presented
HeLo domain of HET-S is directly involved in a toxic species. in which cofactors were used to convert recombinant PrP into
highly infectious PrPres via PMCA in the presence of RNA and
Hypothesis membrane-mimetics (Wang et al., 2010a). These experiments
There are many hypotheses that concern the amyloid entity. The together with similar results from Castilla et al. (2005) suggest
two most often reviewed ones—summarized only briefly here that the protein-only hypothesis is now also proven for mamma-
while referring to published reviews—are the amyloid cascade lian prion disease. There is, however, one caveat concerning all
hypothesis of Ab and a-synuclein associated with AD and of the above experiments that support the protein-only hypoth-
Parkinson’s disease (Hardy and Selkoe, 2002), and the protein- esis: although they demonstrate that the presence of PrPC
only hypothesis first proposed by Alper et al. (1967) concerning (Bueler et al., 1993), PrPres, and PrPres growth are necessary
the origin of prions and prion strains (Prusiner, 1998). Other for infectivity, it is still possible that there is an endogenous
hypotheses of interest are the involvement of amyloids in the viral-like DNA/RNA that gets expressed through the presence
origin of life (Carny and Gazit, 2005) and in the evolution of of PrPres and uses PrPres as a container for infectivity.
protein folding. Prion Strains
Prions: The Protein-Only Hypothesis In mammals, more than a dozen prion strains are known (Weiss-
The ‘‘protein-only hypothesis’’ (Alper et al., 1967; Griffith, 1967; mann, 2005). Prion strains are distinguished by several factors
Prusiner, 1982) states that prion diseases such as scrapie in including the incubation time, the age of disease onset, the ability
sheep, bovine spongiform encephalopathy (BSE), and Creutz- to cross a species barrier, and the brain area of deposited prions.
feldt-Jakob disease in human are distinct from infectious Because according to the protein-only hypothesis (see above)
diseases caused by bacteria, viruses, or viroids, in that the origin the prion protein in its aggregated conformation is both the toxic
of the disease is related to conformational alterations of a ubiqui- and the infectious culprit of prion diseases, the strain-specific
tous protein and that nucleic acids are not essential for the prop- properties must be attributed to altered conformations of prion
agation of the infectious agent. Thus, prions are infectious protein amyloids. Although it is perhaps difficult to imagine
proteins (Prusiner, 1982). In the prion diseases, the prions prop- how all of the prion strain-specific properties are due to altered
agate by converting the normal form of the prion protein (PrPC) conformations of PrPres, there is experimental evidence to
into an altered infectious b sheet-rich, protease resistant confor- support this hypothesis: the mammalian prion strains can be
mation (PrPres) (Prusiner, 1998), which is proposed to be an differentiated by PrP glycosylation patterns (Asante et al.,
amyloid entity. The amyloid serves as an infectious seed that 2002), conformation-sensitive antibodies to PrP (Cali et al.,
recruits soluble host PrPC for growth. The growing amyloid 2009), amyloid conformation-specific fluorescent dyes (Sigurd-
may break into pieces, producing more infectious seeds. son et al., 2007), and PrP protease-resistance. Furthermore,
Prions have also been identified in lower eukaryotes, namely conformationally distinct amyloid aggregates of the yeast prion
yeast and the filamentous fungus Podospora anserina (Coustou Sup35 can be propagated in vitro that on infection, lead to
et al., 1997; Serio and Lindquist, 2001; Wickner et al., 2001). The several distinct prion strains (King and Diaz-Avalos, 2004;
[URE3] and [PSI] non-Mendelian genetic elements of yeast were Tanaka et al., 2004). Specifically, two polymorphs of Sup35
shown to correspond to the prion state of the Ure2p and Sup35p fibrils that lead to different strains have been shown to have
proteins, respectively. Ure2p and Sup35p are soluble proteins in distinct cross-b cores (Toyama et al., 2007).
the normal conformation but aggregate in vivo on conversion to The fact that there are more than a dozen documented
the prion state (Edskes et al., 1999; Patino et al., 1996). In vitro, prion strains can be explained by the multitude of possible poly-
purified Sup35p and Ure2p proteins undergo self-seeded poly- morphisms. The combination of packing and segmental
merization into amyloid-like fibrils (Glover et al., 1997; King polymorphisms with side chain polymorphisms, assembly poly-
et al., 1997). It has been shown that the HET-s prion and the morphisms (Figure 4), and possible chemical modifications (such
yeast prions that are expressed in E. coli are infectious once as distinct glycosylation patterns) may generate many amyloid
they have been converted into amyloid fibrils (Maddelein et al., states with distinct activities. As exemplified in Figure 4, just
2002; Sparrer et al., 2000). Hence, for these prions the protein- a single rotation of a side chain can change the surface charge
only hypothesis has been many times proven. However, the de distribution of the amyloid and hence its potential to bind
novo generation of the infectious prion-agent starting from charged molecules such as membranes, RNA, and DNA.
noninfectious recombinant PrPC has only recently been Polymorphs may differ in their stability of the amyloid fiber, which
achieved. The landmark study from the lab of Deleault et al. can affect the shearing rate, thus leading to more or fewer infec-
(2007) showed that PrPC purified from cell lines can be infectious tious seeds and thus a difference in infectivity or disease onset
on the formation of protease-resistant PrPres using protein-mis- rate. The strain specific species barrier may be explained by
folding cyclic amplification (PMCA). Legname et al. (2004) the conformation of the amyloid that either is able to incorporate
showed for the first time that mouse PrP produced recombi- the new host PrP that differs in the amino acid sequence (low

1254 Structure 18, October 13, 2010 ª2010 Elsevier Ltd All rights reserved
Structure

Review

species barrier) or is only poorly able to seed the new host PrP what is the cause of toxicity is under debate (Finder and Glock-
(high species barrier). The often-observed adaptation of the shuber, 2007; Selkoe, 2004). However, for an aggregation
prion strain to the new host on serial transmission can be disease there may be many different amyloid structures present
explained by a new side chain or packing polymorphism that is in a disease sufferer and each of these structural entities can
established and that enables the host PrP to replicate more have distinct activities. Hence, Ab aggregates may be toxic to
efficiently. It is more difficult to directly connect PrP polymor- cells and the host on many levels. The plurality of potential
phism to the strain-specific brain lesion profiles, but because aggregates, each having several toxic mechanisms, may
cell-specific properties such as pH, membrane composition, thereby account in addition to genetic factors for the docu-
glycosylation, and available chaperone systems will likely vary mented complex multi facet nature of amyloid diseases. It is
in different brain regions, they may influence the amyloid replica- evident that many important scientific challenges lie ahead in
tion. The structure-dependent activities of the prion amyloid may the amyloid disease field.
build the basis for the brain region-dependent toxicity. In Prebiotic Biology and the Origin of Protein Folds
summary, with the multiple types of polymorphisms that can The many activities of protein aggregates support the hypothesis
form in an environment-dependent manner (Wang et al., that under the harsh conditions of the prebiotic earth some
2010b), the presence of hundreds of subtly different amyloid 3.9 billion years ago, the first self-propagating biomolecules
entities that have distinct activities yet that replicate themselves were peptide aggregates composed of the cross-b sheet entity
with great accuracy seems to be possible. (Maury, 2009). The synthesis of amino acids under primitive earth
The Amyloid Cascade Hypothesis in AD conditions has been demonstrated in the lab (Miller, 1953) and
AD is the most common fatal neurodegenerative disorder, and the subsequent formation of peptides can be achieved by one
the devastating neuropathology of AD is tightly linked to the of several prebiotic syntheses: (1) by carbonyl sulfide, a volcanic
cortical deposition of fibrillar Ab plaques (Glenner and Wong, gas (Leman et al., 2004); or (2) by copper in high salt and high
1984). The major component of these deposits is Ab(1-42), which temperature, conditions that mimic a shallow sun-baked lagoon
is generated from the amyloid precursor protein (APP) by the (Schwendinger and Rode, 1992). In a prebiotic world, those
proteolytic activities of b- and g-secretase (Kang et al., 1987; peptides that were able to form amyloids would have been
Masters et al., 1985). Elevated physiological levels of Ab(1-42) protected from hydrolysis (Toledano et al., 2006) or other chem-
lead to early onset AD in Down Syndrome patients (Goldgaber ical modification while exposed to intense ultraviolet irradiation,
et al., 1987; Tanzi et al., 1987), and in several hereditary forms large temperature fluctuations, and high salt concentration. The
of AD that have either been linked to mutations in the APP preferential assembly of certain sequences over others could
gene, or in the presenilin genes, which code for the g-secretase have been an early evolutionary pressure that selected for the
(Suzuki et al., 1994). Mouse models that are transgenic for fittest amino acid sequences (Williams et al., 2009), whereas
human hereditary APP variants and that overexpress human some of these peptide amyloids may have replicated themselves
g-secretase rapidly develop AD-like neurological disorders in a template-assisted peptide synthesis (Takahashi and Mihara,
(Games et al., 1995). These in vivo findings and the dramatically 2004). Next, the amyloid entities may have recruited membranes
increased in vitro aggregation propensity of Ab(1-42) compared by their intrinsic activity to bind other repetitive structures (Wang
to Ab(1-40) (Riek et al., 2001) suggest fibril formation by Ab(1-42) et al., 2010b). There are numerous selective advantages of
as the process primarily responsible for the fatal neuropatholog- a membrane enclosure for such a self-replicating system, as it
ical manifestation of AD. would control the concentration of both the peptides and amino
However, because of the weak correlation between plaque acids that are critical for peptide aggregation and template-as-
load and AD pathology the amyloid hypothesis has been chal- sisted peptide synthesis, as well as protect the system further
lenged. More recent findings indicate that Ab dimers (McDonald from the harsh environmental conditions. At some time in their
et al., 2010; Shankar et al., 2008), small diffusible Ab oligomers, evolutionary history, amyloids may also have recruited DNA or
and a membrane-bound Ab oligomer are highly toxic and are RNA molecules, again via their activity to bind other quasi-repet-
associated with memory dysfunction in the early stage of AD itive structures, thereby stabilizing the nucleic acids while
(Cohen et al., 2006; Hardy and Selkoe, 2002; Lambert et al., optimizing its own replication (Deleault et al., 2003). This may
1998; Lesne et al., 2006). Although the above toxic oligomers have set the stage for an RNA-protein world with the required
are enriched in b sheet content, they are in a distinct conforma- complexity to evolve into life as we know it (Carny and Gazit,
tion from amyloid fibrils (Kayed et al., 2003). In vitro, Ab has been 2005).
described in many states including a membrane-bound mono- Even if life’s origins are not tied to a self-replicating amyloid, it
mer, a dimer, small oligomers, worm-like fibrils, ring-like oligo- is still likely that the first protein folds in a living cell would have
mers, protofibrils, and dozens of polymorphic mature fibrils, been amyloids. There are many arguments to support the idea
the latter observable even within the same preparation (Mein- that protein aggregation has played a key role in the early evolu-
hardt et al., 2009). It has also been shown that different solid- tion of proteins (Chernoff, 2001; Maury, 2009; True and Lind-
state NMR spectra can be obtained depending on how the Ab quist, 2000) the simplest being the improbability that folded
fibrils were prepared (Balbach et al., 2002; Petkova et al., soluble proteins existed early in the evolution of polypeptide
2005). These different NMR spectra were correlated with the sequences. It is probable that primitive polypeptides were
EM-observable macroscopic fibril morphologies, indicating comprised of only a subset of today’s amino acids, and a model
that the polymorphisms have their roots at the atomic level of the codon usage in the Last Universal Ancestor predicts that in
(see above). Each of these various structural entities appear to this subset (Ala, Val, Ile, Asn, Ser, Thr, His, Asp) amyloidogenic
be toxic, but which of them is the most toxic culprit in AD and amino acids are well represented. The observation that protein

Structure 18, October 13, 2010 ª2010 Elsevier Ltd All rights reserved 1255
Structure

Review

sequences have evolved away from aggregation-prone amino lins. Because the protein within microtubules switch between
acid sequence stretches (Monsellier and Chiti, 2007) and that two conformational states, it is speculated that this conforma-
organism complexity anticorrelates with proteomic b-aggrega- tional switch generates a quantum coherent state in neurons
tion propensity (Tartaglia et al., 2005) are further clues that point that somehow (e.g., through quantum gravity) could affect and
to an amyloid as the mother of protein folds. Once peptides were influence the classical physics of neurons (Penrose, 1994). One
aggregating reproducibly into an amyloid entity with functionality of the strongest counter arguments to this idea is one of feasi-
in the biologically useful range (Wang et al., 2010b), soluble bility due to the short coherence times that have been calculated
molten globule-like states (Pervushin et al., 2007) and later (Tegmark, 2000), although coherence times of electron spin pairs
well-folded soluble protein folds may have evolved from these of several ms in proteins at biological relevant temperatures are
protein aggregates. documented (Ritz et al., 2009). We propose amyloids as an
The Amyloid Entity for Long-Term Memory alternative to microtubules as a supramolecular structure that
Although the mechanism of long-term memory is not known, it is could support quantum coherence-based biology. Several
believed that long-term memory must at some level require reports have shown that the cross-b sheet motif of amyloids
stable changes in the molecular components of the neuronal has properties such as electron spin diffusion, electric conduc-
synapse, perhaps mediated by local regulation of protein tance, and autofluorescence (Del Mercato et al., 2007; Margittai
synthesis (Darnell, 2003). The cytoplasmic polyadenylation and Langen, 2008) that may support quantum coherence
element binding protein (CPEB) is thereby considered to be phenomena.
a leading candidate for synaptic translational regulation that
acts by activating dormant mRNAs. In three very exciting
reports, Si et al. (2003a) showed that a neuronal isoform of Conclusion
CPEB in Aplysia (ApCPEB) is necessary for long-term synaptic In 1935, the pioneering biophysicist Astbury published an X-ray
changes, and that ApCPEB undergoes a conformational switch diffraction pattern from poached, stretched egg white that dis-
into an amyloid state that appears to be the active form of the played the reflections typical for the cross b sheet entity (Astbury
protein (Si et al., 2003b). This aggregated state is stably trans- et al., 1935). The authors concluded that when proteins/peptides
missible in yeast and is necessary for long-term memory in aggregate, they go into their energetically most favorable confor-
Aplysia (Si et al., 2010), and thus may provide a basis for mational state, i.e., the cross-b sheet motif. In other words, when
a long-lasting and permissive mark for long-term changes in a protein aggregates it generally forms an amyloid-like entity
the synapse. The N-terminal region of ApCPEB, unique to the comprising a specific structure. Protein aggregation can thus
neuronal isoform, is responsible for the amyloid-like properties be viewed as a primitive folding process that results in a defined
of the molecule. Like yeast prions that propagate as an amyloid, set of aggregated conformations. This makes the amyloid entity
it has a Gln and Asn content of 48% and lacks predictable a prime candidate both for the origin of life and for the first folds
secondary structure. Because the mouse homolog has only of the known protein universe. Furthermore, it is consistent with
18% Gln and Asn content in the N-terminal segment, it is not the fact that there are many functional amyloids that are involved
clear yet whether the corresponding process may also be in diverse mechanisms such as protection, storage, infectivity,
present in mammals. However, the concept of an amyloid entity and perhaps long-term memory. Although 75 years have passed
as the key to long-term memory, although perhaps surprising, is since the amyloid was proposed as a common structural motif,
interesting. The requirement of a lasting change to the synapse the structural and functional characterization of the amyloid
after a single signaling event could be fulfilled by an amyloid, world is just in the starting phase of discovery with much yet to
a structural entity whose soluble-to-aggregate transition can be revealed.
be tightly regulated (see above). Because the formation of the
ApCPEB amyloid results in a gain of function that leads to new REFERENCES
protein expression, the temporal regulation of the amyloid (i.e.,
increase or decrease in levels) may also allow for ‘‘strength- Aguzzi, A., and Calella, A.M. (2009). Prions: protein aggregation and infectious
diseases. Physiol. Rev. 89, 1105–1152.
ening’’ the memory during repetitive stimulations of the synapse.
While on the topic of functional amyloids in the brain, we would Alberti, S., Halfmann, R., King, O., Kapila, A., and Lindquist, S. (2009).
like to go one step further to propose an intriguing hypothesis, A systematic survey identifies prions and illuminates sequence features of
prionogenic proteins. Cell 137, 146–158.
although one that lies outside the range of the current scientific
testability. An important unanswered question is whether Allen, K.D., Wegrzyn, R.D., Chernova, T.A., Muller, S., Newnam, G.P., Winslett,
a computational machine (a Turing machine) that is governed P.A., Wittich, K.B., Wilkinson, K.D., and Chernoff, Y.O. (2005). Hsp70 chaper-
ones as modulators of prion life cycle: novel effects of Ssa and Ssb on the
by classical physics can also be a source of intelligence or Saccharomyces cerevisiae prion. Genetics 169, 1227–1242.
even consciousness. Penrose (1994) argues that the brain can
go beyond what can be achieved by algorithms, and unlike Alper, T., Cramp, W.A., Haig, D.A., and Clarke, M.C. (1967). Does the agent of
scrapie replicate without nucleic acid? Nature 214, 764–766.
a computer, can carry out a noncomputable type of functioning.
He proposes that consciousness and free will can only be Arvan, P., and Castle, D. (1998). Sorting and storage during secretory granule
biogenesis: looking backward and looking forward. Biochem. J. 332, 593–610.
explained by a new kind of physics that, for example, may
involve the translation of quantum mechanical states into Asante, E.A., Linehan, J.M., Desbruslais, M., Joiner, S., Gowland, I., Wood,
classical physics states (i.e., collapse of the wave function). A.L., Welch, J., Hill, A.F., Lloyd, S.E., Wadsworth, J.D., and Collinge, J.
(2002). BSE prions propagate as either variant CJD-like or sporadic CJD-like
Penrose (1994) suggests that such a situation could be taking prion strains in transgenic mice expressing human prion protein. EMBO J.
place in microtubules, the rigid repetitive polymers of the tubu- 21, 6358–6366.

1256 Structure 18, October 13, 2010 ª2010 Elsevier Ltd All rights reserved
Structure

Review
Astbury, W.T., Dickinson, S., and Bailey, K. (1935). CCLXXIX. The X-ray inter- Cohen, E., Bieschke, J., Perciavalle, R.M., Kelly, J.W., and Dillin, A. (2006).
pretation of denaturation and the structure of the seed globulins. Biochem. J. Opposing activities protect against age-onset proteotoxicity. Science 313,
29, 2351–2360. 1604–1610.

Balbach, J.J., Petkova, A.T., Oyler, N.A., Antzutkin, O.N., Gordon, D.J., Mere- Coustou, V., Deleu, C., Saupe, S., and Begueret, J. (1997). The protein product
dith, S.C., and Tycko, R. (2002). Supramolecular structure in full-length Alz- of the Het-s heterokaryon incompatibility gene of the fungus Podospora anser-
heimer’s b-amyloid fibrils: evidence for a parallel b-sheet organization from ina behaves as a prion analog. Proc. Natl. Acad. Sci. USA 94, 9773–9778.
solid-state nuclear magnetic resonance. Biophys. J. 83, 1205–1216.
Dannies, P.S. (2001). Concentrating hormones into secretory granules: layers
Balguerie, A., Dos Reis, S., Coulary-Salin, B., Chaignepain, S., Sabourin, M., of control. Mol. Cell. Endocrinol. 177, 87–93.
Schmitter, J.M., and Saupe, S.J. (2004). The sequences appended to the
amyloid core region of the HET-s prion protein determine higher-order aggre- Darnell, R.B. (2003). Memory, synaptic translation, and.prions? Cell 115,
gate organization in vivo. J. Cell Sci. 117, 2599–2610. 767–768.

Balguerie, A., Dos Reis, S., Ritter, C., Chaignepain, S., Coulary-Salin, B., Del Mercato, L.L., Pompa, P.P., Maruccio, G., Della Torre, A., Sabella, S., Tam-
Forge, V., Bathany, K., Lascu, I., Schmitter, J.M., Riek, R., and Saupe, S.J. burro, A.M., Cingolani, R., and Rinaldi, R. (2007). Charge transport and intrinsic
(2003). Domain organization and structure-function relationship of the HET-s fluorescence in amyloid-like fibrils. Proc. Natl. Acad. Sci. USA 104, 18019–
prion protein of Podospora anserina. EMBO J. 22, 2071–2081. 18024.

Barnhart, M.M., and Chapman, M.R. (2006). Curli biogenesis and function. Deleault, N.R., Harris, B.T., Rees, J.R., and Supattapone, S. (2007). Formation
Annu. Rev. Microbiol. 60, 131–147. of native prions from minimal components in vitro. Proc. Natl. Acad. Sci. USA
104, 9741–9746.
Baxa, U., Cheng, N., Winkler, D.C., Chiu, T.K., Davies, D.R., Sharma, D.,
Inouye, H., Kirschner, D.A., Wickner, R.B., and Steven, A.C. (2005). Filaments Deleault, N.R., Lucassen, R.W., and Supattapone, S. (2003). RNA molecules
of the Ure2p prion protein have a cross-b core structure. J. Struct. Biol. 150, stimulate prion protein conversion. Nature 425, 717–720.
170–179.
Derkatch, I.L., Bradley, M.E., Masse, S.V., Zadorsky, S.P., Polozkov, G.V.,
Bennett, M.J., Sawaya, M.R., and Eisenberg, D. (2006). Deposition diseases Inge-Vechtomov, S.G., and Liebman, S.W. (2000). Dependence and indepen-
and 3D domain swapping. Structure 14, 811–824. dence of [PSI(+)] and [PIN(+)]: a two-prion system in yeast? EMBO J. 19, 1942–
1952.
Berson, J.F., Theos, A.C., Harper, D.C., Tenza, D., Raposo, G., and Marks,
M.S. (2003). Proprotein convertase cleavage liberates a fibrillogenic fragment Du, Z., Park, K.W., Yu, H., Fan, Q., and Li, L. (2008). Newly identified prion
of a resident glycoprotein to initiate melanosome biogenesis. J. Cell Biol. 161, linked to the chromatin-remodeling factor Swi1 in Saccharomyces cerevisiae.
521–533. Nat. Genet. 40, 460–465.

Bieler, S., Estrada, L., Lagos, R., Baeza, M., Castilla, J., and Soto, C. (2005). Edskes, H.K., Gray, V.T., and Wickner, R.B. (1999). The [URE3] prion is an
Amyloid formation modulates the biological activity of a bacterial protein. aggregated form of Ure2p that can be cured by overexpression of Ure2p frag-
J. Biol. Chem. 280, 26880–26885. ments. Proc. Natl. Acad. Sci. USA 96, 1498–1503.

Bueler, H., Aguzzi, A., Sailer, A., Greiner, R.A., Autenried, P., Aguet, M., and Eisenberg, D., Nelson, R., Sawaya, M.R., Balbirnie, M., Sambashivan, S.,
Weissmann, C. (1993). Mice devoid of PrP are resistant to scrapie. Cell 73, Ivanova, M.I., Madsen, A.O., and Riekel, C. (2006). The structural biology of
1339–1347. protein aggregation diseases: fundamental questions and some answers.
Acc. Chem. Res. 39, 568–575.
Cali, I., Castellani, R., Alshekhlee, A., Cohen, Y., Blevins, J., Yuan, J.,
Langeveld, J.P., Parchi, P., Safar, J.G., Zou, W.Q., and Gambetti, P. (2009). Fandrich, M., Meinhardt, J., and Grigorieff, N. (2009). Structural polymorphism
Co-existence of scrapie prion protein types 1 and 2 in sporadic Creutzfeldt- of Alzheimer Ab and other amyloid fibrils. Prion 3, 89–93.
Jakob disease: its effect on the phenotype and prion-type characteristics.
Brain 132, 2643–2658. Fernandez-Escamilla, A.M., Rousseau, F., Schymkowitz, J., and Serrano, L.
(2004). Prediction of sequence-dependent and mutational effects on the
Carny, O., and Gazit, E. (2005). A model for the role of short self-assembled aggregation of peptides and proteins. Nat. Biotechnol. 22, 1302–1306.
peptides in the very early stages of the origin of life. FASEB J. 19, 1051–1055.
Finder, V.H., and Glockshuber, R. (2007). Amyloid-b aggregation. Neurodege-
Castilla, J., Saa, P., Hetz, C., and Soto, C. (2005). In vitro generation of infec- ner. Dis. 4, 13–27.
tious scrapie prions. Cell 121, 195–206.
Fowler, D.M., Koulov, A.V., Alory-Jost, C., Marks, M.S., Balch, W.E., and Kelly,
Chapman, M.R., Robinson, L.S., Pinkner, J.S., Roth, R., Heuser, J., Hammar, J.W. (2006). Functional amyloid formation within mammalian tissue. PLoS Biol.
M., Normark, S., and Hultgren, S.J. (2002). Role of Escherichia coli curli 4, e6.
operons in directing amyloid fiber formation. Science 295, 851–855.
Fowler, D.M., Koulov, A.V., Balch, W.E., and Kelly, J.W. (2007). Functional
Chernoff, Y.O. (2001). Mutation processes at the protein level: is Lamarck amyloid–from bacteria to humans. Trends Biochem. Sci. 32, 217–224.
back? Mutat. Res. 488, 39–64.
Games, D., Adams, D., Alessandrini, R., Barbour, R., Berthelette, P.,
Chernoff, Y.O., Lindquist, S.L., Ono, B., Inge-Vechtomov, S.G., and Liebman, Blackwell, C., Carr, T., Clemens, J., Donaldson, T., Gillespie, F., et al. (1995).
S.W. (1995). Role of the chaperone protein Hsp104 in propagation of the yeast Alzheimer-type neuropathology in transgenic mice overexpressing V717F
prion-like factor. Science 268, 880–884. b-amyloid precursor protein. Nature 373, 523–527.

Chimon, S., Shaibat, M.A., Jones, C.R., Calero, D.C., Aizezi, B., and Ishii, Y. Gellermann, G.P., Appel, T.R., Tannert, A., Radestock, A., Hortschansky, P.,
(2007). Evidence of fibril-like beta-sheet structures in a neurotoxic amyloid Schroeckh, V., Leisner, C., Lutkepohl, T., Shtrasburg, S., Rocken, C., et al.
intermediate of Alzheimer’s beta-amyloid. Nat. Struct. Mol. Biol. 14, 1157– (2005). Raft lipids as common components of human extracellular amyloid
1164. fibrils. Proc. Natl. Acad. Sci. USA 102, 6297–6302.

Chiti, F., and Dobson, C.M. (2006). Protein misfolding, functional amyloid, and Giasson, B.I., Forman, M.S., Higuchi, M., Golbe, L.I., Graves, C.L., Kotzbauer,
human disease. Annu. Rev. Biochem. 75, 333–366. P.T., Trojanowski, J.Q., and Lee, V.M.Y. (2003). Initiation and synergistic fibril-
lization of tau and a-synuclein. Science 300, 636–640.
Chiti, F., Webster, P., Taddei, N., Clark, A., Stefani, M., Ramponi, G., and Dob-
son, C.M. (1999). Designing conditions for in vitro formation of amyloid proto- Glenner, G.G., and Wong, C.W. (1984). Alzheimer’s disease and Down’s
filaments and fibrils. Proc. Natl. Acad. Sci. USA 96, 3590–3594. syndrome: sharing of a unique cerebrovascular amyloid fibril protein. Bio-
chem. Biophys. Res. Commun. 122, 1131–1135.
Claessen, D., Rink, R., de Jong, W., Siebring, J., de Vreugd, P., Boersma, F.G.,
Dijkhuizen, L., and Wosten, H.A. (2003). A novel class of secreted hydrophobic Glover, J.R., Kowal, A.S., Schirmer, E.C., Patino, M.M., Liu, J.J., and Lindquist,
proteins is involved in aerial hyphae formation in Streptomyces coelicolor by S. (1997). Self-seeded fibers formed by Sup35, the protein determinant of
forming amyloid-like fibrils. Genes Dev. 17, 1714–1726. [PSI+], a heritable prion-like factor of S. cerevisiae. Cell 89, 811–819.

Structure 18, October 13, 2010 ª2010 Elsevier Ltd All rights reserved 1257
Structure

Review
Goldgaber, D., Lerman, M.I., McBride, O.W., Saffiotti, U., and Gajdusek, D.C. Lazo, N.D., Maji, S.K., Fradinger, E.A., Bitan, G., and Teplow, D.B. (2005). The
(1987). Characterization and chromosomal localization of a cDNA encoding amyloid b protein. In Amyloid Protein-The b Sheet Conformation and Disease,
brain amyloid of Alzheimer’s disease. Science 235, 877–880. J.D. Sipe, ed. (Weinheim, Germany: Wiley-VCH Publishers), pp. 385–491.

Greenwald, J., Buhtz, C., Ritter, C., Kwiatkowski, W., Choe, S., Maddelein, Legname, G., Baskakov, I.V., Nguyen, H.O., Riesner, D., Cohen, F.E., DeAr-
M.L., Ness, F., Cescau, S., Soragni, A., Leitz, D., et al. (2010). The mechanism mond, S.J., and Prusiner, S.B. (2004). Synthetic mammalian prions. Science
of prion inhibition by HET-S. Mol. Cell, in press. 305, 673–676.

Griffith, J.S. (1967). Self-replication and scrapie. Nature 215, 1043–1044. Leman, L., Orgel, L., and Ghadiri, M.R. (2004). Carbonyl sulfide-mediated
prebiotic formation of peptides. Science 306, 283–286.
Guijarro, J.I., Sunde, M., Jones, J.A., Campbell, I.D., and Dobson, C.M. (1998).
Amyloid fibril formation by an SH3 domain. Proc. Natl. Acad. Sci. USA 95, Lesne, S., Koh, M.T., Kotilinek, L., Kayed, R., Glabe, C.G., Yang, A., Gallagher,
4224–4228. M., and Ashe, K.H. (2006). A specific amyloid-b protein assembly in the brain
impairs memory. Nature 440, 352–357.
Hagn, F., Eisoldt, L., Hardy, J., Vendrely, C., Coles, M., Scheibel, T., and
Kessler, H. (2010). A conserved spider silk domain acts as a molecular switch Luhrs, T., Ritter, C., Adrian, M., Riek-Loher, D., Bohrmann, B., Dobeli, H.,
that controls fibre assembly. Nature 465, 239–242. Schubert, D., and Riek, R. (2005). 3D structure of Alzheimer’s amyloid-b
(1-42) fibrils. Proc. Natl. Acad. Sci. USA 102, 17342–17347.
Han, H., Weinreb, P.H., and Lansbury, P.T., Jr. (1995). The core Alzheimer’s
peptide NAC forms amyloid fibrils which seed and are seeded by beta- Maddelein, M.L., Dos Reis, S., Duvezin-Caubet, S., Coulary-Salin, B., and
amyloid: is NAC a common trigger or target in neurodegenerative disease? Saupe, S.J. (2002). Amyloid aggregates of the HET-s prion protein are infec-
Chem. Biol. 2, 163–169. tious. Proc. Natl. Acad. Sci. USA 99, 7402–7407.

Hardy, J., and Selkoe, D.J. (2002). The amyloid hypothesis of Alzheimer’s Maji, S.K., Perrin, M.H., Sawaya, M.R., Jessberger, S., Vadodaria, K., Riss-
disease: progress and problems on the road to therapeutics. Science 297, man, R.A., Singru, P.S., Nilsson, K.P., Simon, R., Schubert, D., et al. (2009).
353–356. Functional amyloids as natural storage of peptide hormones in pituitary secre-
tory granules. Science 325, 328–332.
Hurbain, I., Geerts, W.J., Boudier, T., Marco, S., Verkleij, A.J., Marks, M.S., and
Raposo, G. (2008). Electron tomography of early melanosomes: implications Maji, S.K., Schubert, D., Rivier, C., Lee, S., Rivier, J.E., and Riek, R. (2008).
for melanogenesis and the generation of fibrillar amyloid sheets. Proc. Natl. Amyloid as a depot for the formulation of long-acting drugs. PLoS Biol. 6, e17.
Acad. Sci. USA 105, 19726–19731.
Marcon, G., Plakoutsi, G., Canale, C., Relini, A., Taddei, N., Dobson, C.M.,
Ivanova, M.I., Sievers, S.A., Sawaya, M.R., Wall, J.S., and Eisenberg, D. Ramponi, G., and Chiti, F. (2005). Amyloid formation from HypF-N under
(2009). Molecular basis for insulin fibril assembly. Proc. Natl. Acad. Sci. USA conditions in which the protein is initially in its native state. J. Mol. Biol. 347,
106, 18990–18995. 323–335.

Margittai, M., and Langen, R. (2008). Fibrils with parallel in-register structure
Kajava, A.V., and Steven, A.C. (2006). b-rolls, b-helices, and other b-solenoid
constitute a major class of amyloid fibrils: molecular insights from electron
proteins. Adv. Protein Chem. 73, 55–96.
paramagnetic resonance spectroscopy. Q. Rev. Biophys. 41, 265–297.
Kane, M.D., Lipinski, W.J., Callahan, M.J., Bian, F., Durham, R.A., Schwarz,
Margittai, M., and Langen, R. (2006). Side chain-dependent stacking modu-
R.D., Roher, A.E., and Walker, L.C. (2000). Evidence for seeding of b-amyloid
lates tau filament structure. J. Biol. Chem. 281, 37820–37827.
by intracerebral infusion of Alzheimer brain extracts in b-amyloid precursor
protein-transgenic mice. J. Neurosci. 20, 3606–3611.
Masters, C.L., Simms, G., Weinman, N.A., Multhaup, G., McDonald, B.L., and
Beyreuther, K. (1985). Amyloid plaque core protein in Alzheimer disease and
Kang, J., Lemaire, H.G., Unterbeck, A., Salbaum, J.M., Masters, C.L., Grzes-
Down syndrome. Proc. Natl. Acad. Sci. USA 82, 4245–4249.
chik, K.H., Multhaup, G., Beyreuther, K., and Muller-Hill, B. (1987). The
precursor of Alzheimer’s disease amyloid A4 protein resembles a cell-surface Maury, C.P. (2009). Self-propagating b-sheet polypeptide structures as prebi-
receptor. Nature 325, 733–736. otic informational molecular entities: the amyloid world. Orig. Life Evol. Biosph.
39, 141–150.
Kayed, R., Head, E., Thompson, J.L., McIntire, T.M., Milton, S.C., Cotman,
C.W., and Glabe, C.G. (2003). Common structure of soluble amyloid oligomers McDonald, J.M., Savva, G.M., Brayne, C., Welzel, A.T., Forster, G., Shankar,
implies common mechanism of pathogenesis. Science 300, 486–489. G.M., Selkoe, D.J., Ince, P.G., and Walsh, D.M. (2010). The presence of
sodium dodecyl sulphate-stable Ab dimers is strongly associated with
Kelly, R.B. (1985). Pathways of protein secretion in eukaryotes. Science 230, Alzheimer-type dementia. Brain 133, 1328–1341.
25–32.
McGlinchey, R.P., Shewmaker, F., McPhie, P., Monterroso, B., Thurber, K.,
Kelly, R.B. (1987). Protein transport. From organelle to organelle. Nature 326, and Wickner, R.B. (2009). The repeat domain of the melanosome fibril protein
14–15. Pmel17 forms the amyloid core promoting melanin synthesis. Proc. Natl. Acad.
Sci. USA 106, 13731–13736.
Kimura, H., and Schubert, D. (1993). Amyloid b-protein activates tachykinin
receptors and inositol trisphosphate accumulation by synergy with glutamate. Meinhardt, J., Sachse, C., Hortschansky, P., Grigorieff, N., and Fandrich, M.
Proc. Natl. Acad. Sci. USA 90, 7508–7512. (2009). Ab(1-40) fibril polymorphism implies diverse interaction patterns in
amyloid fibrils. J. Mol. Biol. 386, 869–877.
King, C.Y., and Diaz-Avalos, R. (2004). Protein-only transmission of three yeast
prion strains. Nature 428, 319–323. Meyer-Luehmann, M., Coomaraswamy, J., Bolmont, T., Kaeser, S., Schaefer,
C., Kilger, E., Neuenschwander, A., Abramowski, D., Frey, P., Jaton, A.L., et al.
King, C.Y., Tittmann, P., Gross, H., Gebert, R., Aebi, M., and Wuthrich, K. (2006). Exogenous induction of cerebral b-amyloidogenesis is governed by
(1997). Prion-inducing domain 2-114 of yeast Sup35 protein transforms agent and host. Science 313, 1781–1784.
in vitro into amyloid-like filaments. Proc. Natl. Acad. Sci. USA 94, 6618–6622.
Miller, S.L. (1953). A production of amino acids under possible primitive earth
Klein, W.L., Stine, W.B., Jr., and Teplow, D.B. (2004). Small assemblies of conditions. Science 117, 528–529.
unmodified amyloid b-protein are the proximate neurotoxin in Alzheimer’s
disease. Neurobiol. Aging 25, 569–580. Monsellier, E., and Chiti, F. (2007). Prevention of amyloid-like aggregation as
a driving force of protein evolution. EMBO Rep. 8, 737–742.
Lacy, P.E. (1975). Endocrine secretory mechanisms. A review. Am. J. Pathol.
79, 170–188. Nakayashiki, T., Kurtzman, C.P., Edskes, H.K., and Wickner, R.B. (2005).
Yeast prions [URE3] and [PSI+] are diseases. Proc. Natl. Acad. Sci. USA
Lambert, M.P., Barlow, A.K., Chromy, B.A., Edwards, C., Freed, R., Liosatos, 102, 10575–10580.
M., Morgan, T.E., Rozovsky, I., Trommer, B., Viola, K.L., et al. (1998). Diffusible,
nonfibrillar ligands derived from Ab1-42 are potent central nervous system Nelson, R., and Eisenberg, D. (2006a). Recent atomic models of amyloid fibril
neurotoxins. Proc. Natl. Acad. Sci. USA 95, 6448–6453. structure. Curr. Opin. Struct. Biol. 16, 260–265.

1258 Structure 18, October 13, 2010 ª2010 Elsevier Ltd All rights reserved
Structure

Review
Nelson, R., and Eisenberg, D. (2006b). Structural models of amyloid-like fibrils. Selkoe, D.J. (2004). Cell biology of protein misfolding: the examples of
Adv. Protein Chem. 73, 235–282. Alzheimer’s and Parkinson’s diseases. Nat. Cell Biol. 6, 1054–1061.

Nelson, R., Sawaya, M.R., Balbirnie, M., Madsen, A.O., Riekel, C., Grothe, R., Serio, T.R., and Lindquist, S.L. (2001). [PSI+], SUP35, and chaperones. Adv.
and Eisenberg, D. (2005). Structure of the cross-b spine of amyloid-like fibrils. Protein Chem. 57, 335–366.
Nature 435, 773–778.
Shankar, G.M., Li, S., Mehta, T.H., Garcia-Munoz, A., Shepardson, N.E.,
Oh, J., Kim, J.G., Jeon, E., Yoo, C.H., Moon, J.S., Rhee, S., and Hwang, I. Smith, I., Brett, F.M., Farrell, M.A., Rowan, M.J., Lemere, C.A., et al. (2008).
(2007). Amyloidogenesis of type III-dependent harpins from plant pathogenic Amyloid-b protein dimers isolated directly from Alzheimer’s brains impair
bacteria. J. Biol. Chem. 282, 13601–13609. synaptic plasticity and memory. Nat. Med. 14, 837–842.

Osherovich, L.Z., and Weissman, J.S. (2002). The utility of prions. Dev. Cell 2, Si, K., Choi, Y.B., White-Grindley, E., Majumdar, A., and Kandel, E.R. (2010).
143–151. Aplysia CPEB can form prion-like multimers in sensory neurons that contribute
to long-term facilitation. Cell 140, 421–435.
Palade, G. (1975). Intracellular aspects of the process of protein synthesis.
Science 189, 347–358. Si, K., Giustetto, M., Etkin, A., Hsu, R., Janisiewicz, A.M., Miniaci, M.C., Kim,
J.H., Zhu, H., and Kandel, E.R. (2003a). A neuronal isoform of CPEB regulates
Paravastu, A.K., Leapman, R.D., Yau, W.M., and Tycko, R. (2008). Molecular local protein synthesis and stabilizes synapse-specific long-term facilitation in
structural basis for polymorphism in Alzheimer’s b-amyloid fibrils. Proc. Natl. aplysia. Cell 115, 893–904.
Acad. Sci. USA 105, 18349–18354.
Si, K., Lindquist, S., and Kandel, E.R. (2003b). A neuronal isoform of the aplysia
Patino, M.M., Liu, J.J., Glover, J.R., and Lindquist, S. (1996). Support for the CPEB has prion-like properties. Cell 115, 879–891.
prion hypothesis for inheritance of a phenotypic trait in yeast. Science 273,
622–626. Sigurdson, C.J., Nilsson, K.P., Hornemann, S., Manco, G., Polymenidou, M.,
Schwarz, P., Leclerc, M., Hammarstrom, P., Wuthrich, K., and Aguzzi, A.
Penrose, R. (1994). Shadows of the Mind, First Edition (Oxford, England: (2007). Prion strain discrimination using luminescent conjugated polymers.
Oxford University Press). Nat. Methods 4, 1023–1030.
Pervushin, K., Vamvaca, K., Vogeli, B., and Hilvert, D. (2007). Structure and Silveira, J.R., Raymond, G.J., Hughson, A.G., Race, R.E., Sim, V.L., Hayes,
dynamics of a molten globular enzyme. Nat. Struct. Mol. Biol. 14, 1202–1206. S.F., and Caughey, B. (2005). The most infectious prion protein particles.
Nature 437, 257–261.
Petkova, A.T., Leapman, R.D., Guo, Z., Yau, W.M., Mattson, M.P., and Tycko,
R. (2005). Self-propagating, molecular-level polymorphism in Alzheimer’s Sipe, J.D., and Cohen, A.S. (2000). Review: history of the amyloid fibril.
b-amyloid fibrils. Science 307, 262–265. J. Struct. Biol. 130, 88–98.
Polverino de Laureto, P., Taddei, N., Frare, E., Capanni, C., Costantini, S.,
Sparr, E., Engel, M.F., Sakharov, D.V., Sprong, M., Jacobs, J., de Kruijff, B.,
Zurdo, J., Chiti, F., Dobson, C.M., and Fontana, A. (2003). Protein aggregation
Hoppener, J.W., and Killian, J.A. (2004). Islet amyloid polypeptide-induced
and amyloid fibril formation by an SH3 domain probed by limited proteolysis.
membrane leakage involves uptake of lipids by forming amyloid fibers.
J. Mol. Biol. 334, 129–141.
FEBS Lett. 577, 117–120.
Powers, E.T., Morimoto, R.I., Dillin, A., Kelly, J.W., and Balch, W.E. (2009). Bio-
Sparrer, H.E., Santoso, A., Szoka, F.C., Jr., and Weissman, J.S. (2000).
logical and chemical approaches to diseases of proteostasis deficiency. Annu.
Evidence for the prion hypothesis: induction of the yeast [PSI+] factor by
Rev. Biochem. 78, 959–991.
in vitro- converted Sup35 protein. Science 289, 595–599.
Prusiner, S.B. (1982). Novel proteinaceous infectious particles cause scrapie.
Science 216, 136–144. Sunde, M., and Blake, C. (1997). The structure of amyloid fibrils by electron
microscopy and X-ray diffraction. Adv. Protein Chem. 50, 123–159.
Prusiner, S.B. (1998). Prions. Proc. Natl. Acad. Sci. USA 95, 13363–13383.
Sunde, M., Serpell, L.C., Bartlam, M., Fraser, P.E., Pepys, M.B., and Blake,
Ridley, R.M., Baker, H.F., Windle, C.P., and Cummings, R.M. (2006). Very long C.C. (1997). Common core structure of amyloid fibrils by synchrotron X-ray
term studies of the seeding of b-amyloidosis in primates. J. Neural Transm. diffraction. J. Mol. Biol. 273, 729–739.
113, 1243–1251.
Suzuki, N., Cheung, T.T., Cai, X.D., Odaka, A., Otvos, L., Jr., Eckman, C.,
Riek, R. (2006). Cell biology: infectious Alzheimer’s disease? Nature 444, Golde, T.E., and Younkin, S.G. (1994). An increased percentage of long
429–431. amyloid b protein secreted by familial amyloid b protein precursor
(b APP717) mutants. Science 264, 1336–1340.
Riek, R., Guntert, P., Dobeli, H., Wipf, B., and Wuthrich, K. (2001). NMR studies
in aqueous solution fail to identify significant conformational differences Takahashi, Y., and Mihara, H. (2004). Construction of a chemically and confor-
between the monomeric forms of two Alzheimer peptides with widely different mationally self-replicating system of amyloid-like fibrils. Bioorg. Med. Chem.
plaque-competence, Ab(1-40)(ox) and Ab(1-42)(ox). Eur. J. Biochem. 268, 12, 693–699.
5930–5936.
Tanaka, M., Chien, P., Naber, N., Cooke, R., and Weissman, J.S. (2004).
Ritter, C., Maddelein, M.L., Siemer, A.B., Luhrs, T., Ernst, M., Meier, B.H., Conformational variations in an infectious protein determine prion strain differ-
Saupe, S.J., and Riek, R. (2005). Correlation of structural elements and infec- ences. Nature 428, 323–328.
tivity of the HET-s prion. Nature 435, 844–848.
Tanzi, R.E., Gusella, J.F., Watkins, P.C., Bruns, G.A., St George-Hyslop, P.,
Ritz, T., Wiltschko, R., Hore, P.J., Rodgers, C.T., Stapput, K., Thalau, P., Van Keuren, M.L., Patterson, D., Pagan, S., Kurnit, D.M., and Neve, R.L.
Timmel, C.R., and Wiltschko, W. (2009). Magnetic compass of birds is based (1987). Amyloid b protein gene: cDNA, mRNA distribution, and genetic linkage
on a molecule with optimal directional sensitivity. Biophys. J. 96, 3451–3457. near the Alzheimer locus. Science 235, 880–884.

Rousseau, F., Serrano, L., and Schymkowitz, J.W. (2006). How evolutionary Tartaglia, G.G., Pawar, A.P., Campioni, S., Dobson, C.M., Chiti, F., and Ven-
pressure against protein aggregation shaped chaperone specificity. J. Mol. druscolo, M. (2008). Prediction of aggregation-prone regions in structured
Biol. 355, 1037–1047. proteins. J. Mol. Biol. 380, 425–436.

Sawaya, M.R., Sambashivan, S., Nelson, R., Ivanova, M.I., Sievers, S.A., Tartaglia, G.G., Pellarin, R., Cavalli, A., and Caflisch, A. (2005). Organism
Apostol, M.I., Thompson, M.J., Balbirnie, M., Wiltzius, J.J., McFarlane, H.T., complexity anti-correlates with proteomic b-aggregation propensity. Protein
et al. (2007). Atomic structures of amyloid cross-b spines reveal varied steric Sci. 14, 2735–2740.
zippers. Nature 447, 453–457.
Tegmark, M. (2000). Importance of quantum decoherence in brain processes.
Schwendinger, M.G., and Rode, B.M. (1992). Investigations on the mechanism Phys. Rev. E Stat. Phys. Plasmas Fluids Relat. Interdiscip. Topics 61, 4194–
of the salt-induced peptide formation. Orig. Life Evol. Biosph. 22, 349–359. 4206.

Structure 18, October 13, 2010 ª2010 Elsevier Ltd All rights reserved 1259
Structure

Review
Tjernberg, L., Hosia, W., Bark, N., Thyberg, J., and Johansson, J. (2002). Wasmer, C., Lange, A., Van Melckebeke, H., Siemer, A.B., Riek, R., and Meier,
Charge attraction and b propensity are necessary for amyloid fibril formation B.H. (2008). Amyloid fibrils of the HET-s(218-289) prion form a b solenoid with
from tetrapeptides. J. Biol. Chem. 277, 43243–43246. a triangular hydrophobic core. Science 319, 1523–1526.

Toledano, S., Williams, R.J., Jayawarna, V., and Ulijn, R.V. (2006). Enzyme-
Wasmer, C., Schutz, A., Loquet, A., Buhtz, C., Greenwald, J., Riek, R.,
triggered self-assembly of peptide hydrogels via reversed hydrolysis. J. Am.
Bockmann, A., and Meier, B.H. (2009). The molecular organization of the
Chem. Soc. 128, 1070–1071.
fungal prion HET-s in its amyloid form. J. Mol. Biol. 394, 119–127.
Tooze, S.A. (1998). Biogenesis of secretory granules in the trans-Golgi network
of neuroendocrine and endocrine cells. Biochim. Biophys. Acta 1404, Weissmann, C. (2005). Birth of a prion: spontaneous generation revisited. Cell
231–244. 122, 165–168.

Toyama, B.H., Kelly, M.J., Gross, J.D., and Weissman, J.S. (2007). The struc-
tural basis of yeast prion strain variants. Nature 449, 233–237. Westermark, G.T., and Westermark, P. (2009). Serum amyloid A and protein
AA: molecular mechanisms of a transmissible amyloidosis. FEBS Lett. 583,
Trovato, A., Chiti, F., Maritan, A., and Seno, F. (2006). Insight into the structure 2685–2690.
of amyloid fibrils from the analysis of globular proteins. PLoS Comput. Biol. 2,
e170. Wickner, R.B., Taylor, K.L., Edskes, H.K., Maddelein, M.L., Moriyama, H., and
Roberts, B.T. (2001). Yeast prions act as genes composed of self-propagating
True, H.L., and Lindquist, S.L. (2000). A yeast prion provides a mechanism for protein amyloids. Adv. Protein Chem. 57, 313–334.
genetic variation and phenotypic diversity. Nature 407, 477–483.

van der Wel, P.C., Lewandowski, J.R., and Griffin, R.G. (2007). Solid-state Williams, R.J., Smith, A.M., Collins, R., Hodson, N., Das, A.K., and Ulijn, R.V.
NMR study of amyloid nanocrystals and fibrils formed by the peptide (2009). Enzyme-assisted self-assembly under thermodynamic control. Nat.
GNNQQNY from yeast prion protein Sup35p. J. Am. Chem. Soc. 129, 5117– Nanotechnol. 4, 19–24.
5130.
Wiltzius, J.J., Landau, M., Nelson, R., Sawaya, M.R., Apostol, M.I., Gold-
Ventura, S., and Villaverde, A. (2006). Protein quality in bacterial inclusion schmidt, L., Soriaga, A.B., Cascio, D., Rajashankar, K., and Eisenberg, D.
bodies. Trends Biotechnol. 24, 179–185. (2009). Molecular mechanisms for protein-encoded inheritance. Nat. Struct.
Mol. Biol. 16, 973–978.
Wang, F., Wang, X., Yuan, C.G., and Ma, J. (2010a). Generating a prion with
bacterially expressed recombinant prion protein. Science 327, 1132–1135.
Wiltzius, J.J., Sievers, S.A., Sawaya, M.R., Cascio, D., Popov, D., Riekel, C.,
Wang, L., Maji, S.K., Sawaya, M.R., Eisenberg, D., and Riek, R. (2008). Bacte- and Eisenberg, D. (2008). Atomic structure of the cross-b spine of islet amyloid
rial inclusion bodies contain amyloid-like structure. PLoS Biol. 6, e195. polypeptide (amylin). Protein Sci. 17, 1467–1474.

Wang, L., Schubert, D., Sawaya, M.R., Eisenberg, D., and Riek, R. (2010b).
Wösten, H.A., and de Vocht, M.L. (2000). Hydrophobins, the fungal coat unrav-
Multidimensional structure-activity relationship of a protein in its aggregated
elled. Biochim. Biophys. Acta 1469, 79–86.
states. Angew. Chem. Int. Ed. Engl. 49, 3904–3908.

Wang, X., Zhou, Y., Ren, J.J., Hammer, N.D., and Chapman, M.R. (2010c). Zanuy, D., and Nussinov, R. (2003). The sequence dependence of fiber orga-
Gatekeeper residues in the major curlin subunit modulate bacterial amyloid nization. A comparative molecular dynamics study of the islet amyloid poly-
fiber biogenesis. Proc. Natl. Acad. Sci. USA 107, 163–168. peptide segments 22-27 and 22-29. J. Mol. Biol. 329, 565–584.

1260 Structure 18, October 13, 2010 ª2010 Elsevier Ltd All rights reserved

You might also like