You are on page 1of 5

General and Comparative Endocrinology 172 (2011) 39–43

Contents lists available at ScienceDirect

General and Comparative Endocrinology


journal homepage: www.elsevier.com/locate/ygcen

Review

The role of ubiquitin–proteasome system in ageing


Peter Löw ⇑
Department of Anatomy, Cell and Developmental Biology, Eötvös Loránd University, Budapest, Hungary

a r t i c l e i n f o a b s t r a c t

Article history: Maintenance of cellular homeostasis influences ageing and it is determined by several factors, including
Available online 13 February 2011 efficient proteolysis of damaged proteins. The ubiquitin–proteasome system is the major protein degra-
dation pathway in the cell. Specifically, the proteasome is responsible for clearance of abnormal, dena-
Keywords: tured or in general damaged proteins as well as for the regulated degradation of short-lived proteins.
Insulin In this review the involvement of the ubiquitin–proteasome pathway in protein degradation at different
Insulin-like growth factor levels of cellular life is discussed in relation with ageing. Though the exact underlying mechanism is
Ubiquitin
unclear, an age-related decrease in proteasome activity weakens cellular capacity to remove oxidatively
Proteasome
Ageing
modified proteins and favours the development of diseases. Up-regulation of proteasome activity is char-
acteristic of muscle wasting conditions, but may not be rate limiting. Meanwhile, enhanced presence of
immunoproteasomes in ageing brain and muscle tissue could reflect a persistent inflammatory defence
and anti-stress mechanism. Insulin/IGF-1 signalling regulates ageing in worms, flies and mammals.
The insulin/IGF-1 receptor inhibits the forkhead transcription factor, FoxO through activating a cascade
of conserved kinases. Longevity increases when FoxO becomes activated in response to reduced insu-
lin/IGF-1 signalling. The ubiquitin–proteasome system plays a major role in signal transduction associ-
ated with stress and ageing. The understanding of specific proteolytic targeting paves the way for a
new generation of active molecules that may control particular steps of normal and pathological ageing.
Ó 2011 Elsevier Inc. All rights reserved.

1. Introduction and ageing associated diseases. Since in most instances ubiquityla-


tion is the primary event in target selection, the system of ubiqui-
Protein degradation by the ubiquitin–proteasome system is an tylation and deubiquitylation might be of similar importance.
essential cellular mechanism that has come into focus of ageing re-
search recently. One of the most important machineries for degra-
dation of endogenous proteins is the proteasome, whose activity
decreases during the ageing process. Several major ageing related 2. The ubiquitin–proteasome system
diseases have been characterised as protein degradation failures.
Two major pathways accomplish regulated protein catabolism The majority of cellular proteins are degraded by the UPS, which
in eukaryotic cells: the ubiquitin–proteasome system (UPS) and consists of both substrate-recruiting and substrate-degrading
the autophagy–lysosomal system. The UPS serves as the primary machinery. The former is composed of three enzymes, the first of
route for degradation for thousands of short-lived proteins and which (E1) activates the polypeptide ubiquitin in an ATP depen-
many regulatory proteins. UPS-mediated catabolism is also essen- dent manner, enabling its transfer onto a ubiquitin carrier enzyme
tial to maintain amino acid pools in acute starvation and contrib- (E2). Activated ubiquitin is further transferred by a ubiquitin pro-
utes to the degradation of defective proteins. Autophagy, by tein ligase (E3) to a substrate protein [11]. The substrate-recruiting
contrast, is primarily responsible for degrading long-lived proteins machinery then catalyses the formation of an isopeptide bond be-
and maintaining amino acid pools in chronic starvation. tween the C-terminal glycine residue of ubiquitin and the e-amino
During the recent years evidence has increased that proteasome group of a substrate protein lysine residue. Repeated addition of
activity is decreased during the ageing process in various model ubiquitin moieties onto the first results in a polyubiquitylated sub-
systems and that these changes might be causally related to ageing strate protein that is recognised by the proteolytic machinery of
the UPS, the 26S proteasome. The requirement for the addition of
multiple ubiquitin molecules may be part of a proof-reading mech-
⇑ Address: Eötvös Loránd University, Department of Anatomy, Cell and Develop- anism, as a large number of deubiquitylating enzymes (DUBs) are
mental Biology, Pazmany Peter setany 1/C, H-1117 Budapest, Hungary. Fax: +36 1 also found within cells [29]. DUBs remove ubiquitin molecules be-
381 2184. fore a sufficiently large branch structure has been synthesised to
E-mail address: peterlow@elte.hu activate proteasomal destruction (Fig. 1).

0016-6480/$ - see front matter Ó 2011 Elsevier Inc. All rights reserved.
doi:10.1016/j.ygcen.2011.02.005
40 P. Löw / General and Comparative Endocrinology 172 (2011) 39–43

Fig. 2. Structure of 20S, 26S and immunoproteasome. 26S proteasome is a large


multisubunit protease complex that selectively degrades polyubiquitylated pro-
teins. It is composed of two subcomplexes: a barrel-shaped 20S core particle
containing the protease active sites and two 19S regulatory particles that cap the
barrel and control access of substrates to the core. The 20S particle consists of four
Fig. 1. The ubiquitylation cascade. Ubiquitin (Ub) is a small 8 kDa protein, which is
axially stacked heptameric rings, two inner b-rings and two outer a-rings, forming
first transferred to the ubiquitin-activating enzyme, E1, in an ATP dependent
three continuous chambers inside. This structure keeps the proteolytic active sites,
manner. This activated ubiquitin is then transferred to the ubiquitin conjugating
located at the N-termini of three of the seven b-subunits, sequestered in the central
enzyme, E2. Finally, the ubiquitin is covalently attached to the target protein by E3
chamber and prevents unregulated substrate degradation. The 19S regulatory
ubiquitin ligase, leading to the formation of a polyubiquitin chain. The polyubiq-
particles are composed of approximately 20 subunits which form a ‘‘lid’’ and a
uitylated protein is recognised by the 26S proteasome, and is destroyed in an ATP-
‘‘base’’. During host infection cellular stimulation with interferon-c results in the
dependent manner. Deubiquitinating enzymes (DUB) can rescue proteins from
new synthesis of immunoproteasomes which contain three inducible catalytic b
degradation and recycle ubiquitin or ubiquitin oligomers from ubiquitin–protein
subunits in place of the three standard catalytic subunits. The immunoproteasomes
conjugates. (Ub, ubiquitin; E1, ubiquitin-activating enzyme; E2, ubiquitin-conju-
can also be activated by association with an 11S regulator, which is composed of
gating enzyme; E3, ubiquitin ligase; DUB, deubiquitinating enzyme.)
two interferon-c-inducible subunits PA28a and PA28b.

The overall shape of the 20S proteasome is an elongated cylin- synthesis by b1i, b2i and b5i subunits, respectively. As a result,
der with large central cavities and narrow constrictions. The seven although these particles digest proteins at rates similar to those
different a-subunits are located at the ends, whereas the seven dif- for constitutive proteasomes, they generate a higher fraction of
ferent b-subunits form the two inner rings. a-ring is closed by the peptides with the appropriate C-termini and length to serve in
amino-terminal extensions of the a-subunits, whereas the b-ring antigen presentation. In immunoproteasomes, PA28 (11S) activa-
has an open space in its centre. There are two antichambers inside tor can replace the 19S complex [3] (Fig. 2).
the proteasome and a central catalytic chamber. Only three b sub-
units are active among the b subunits in both b rings. Subunits b1,
b2 and b5 of both adjacent b-rings expose their proteolytically 3. Proteasomal activity during ageing
active sites, exhibiting caspase-like, trypsin-like and chymotryp-
sin-like cleavage specificity, respectively. Other b-subunits form Homeostasis is a key feature of cellular lifespan. Maintenance of
the active pocket for them. In mammalian cells, the 20S core is cellular homeostasis influences the rate of ageing and its efficiency
found associated with activator complexes PA700 (19S) or PA28 is determined by the cooperation between protein stability and
(11S) [6]. PA700 contains at least 18 subunits, including six ATPas- resistance to stress, protein refolding, protein repair and proteoly-
es in its ‘‘base’’, that exert a chaperone-like activity and can unfold sis of damaged proteins. Protein degradation is predominately cat-
proteins prior to entry in the proteolytic chamber, and eight sub- alysed by the proteasome which is responsible for cell clearance of
units in its ‘‘lid’’, that recognise the poly-ubiquitin (poly-Ub) sig- abnormal or damaged proteins as well as for the regulated degra-
nals [28]. When capped with PA700, the 20S proteasome forms dation of short-lived proteins. Impaired proteasome function has
the 26S complex, which is the competent structure for Ub-medi- been tightly correlated to ageing both in vivo and in vitro. A number
ated proteolysis. Binding of the substrate protein to the protea- of studies have shown that the proteasome can be activated by ge-
some is followed by protein unfolding by the half-dozen ATPases netic manipulations as well as by factors that affect either its con-
that encircle the pore of the proteasome catalytic core, and trans- formation and stability or the expression of its subunits and the
location of the unfolded protein into the central proteolytic cham- rate of proteasome assembly. This ‘‘readjustment’’ has been shown
ber, where it is cleaved into short peptides. The 20S proteasome in to have a great impact on retention of cellular homeostasis since it
turn, works independently of ATP and ubiquitin and has been promotes lifespan extension.
implicated in the degradation of damaged or unfolded proteins. An age-related decrease in proteasome activity has been ob-
26S proteasome activity can be identified as the ATP-associated served in different tissues (including skin, muscle, heart, liver, kid-
activity. ATP stabilises the 26S complex and allows the opening ney, lung and eye lens). In all of these investigations, proteasome
of channels in the rings of the 20S core by ATPases located in the activity was measured using fluorigenic peptide substrates and
19S regulatory particle [17] (Fig. 2). caspase-like activity was found to be consistently depressed with
Proteasome has also been implicated in antigen presentation increasing age. Decreased activity could be due to reduced
process and immune response, mainly through immunoprotea- amounts of proteasome. When the specific activity of 20S protea-
somes. In this proteasome type, the constitutively expressed b1, somes purified from different tissues was calculated, a loss of cas-
b2 and b5 subunits are substituted during de novo proteasome bio- pase-like and chymotrypsin-like or trypsin-like activity was
P. Löw / General and Comparative Endocrinology 172 (2011) 39–43 41

detected, supporting the link between intrinsic changes in 20S pro- late in and/or outside of the cells. Damaged proteins are removed
teasomes and increasing age. The concentration of ubiquitylated by enzymatic degradation by cytosolic proteases, lysosomes and
proteins was also found to be increased in aged tissue and cells, the proteasome, and there is evidence that these three mecha-
suggesting that the activity of 26S proteasomes could be affected nisms are altered in neurons during ageing [14,21]. In rats, protea-
by ageing in addition to 20S proteasomes [8]. Earlier work has some activity decreases with advancing age in the cerebral cortex,
attributed the observed decrease of proteasome function to the hippocampus and spinal cord, but not in the cerebellum or brain-
accumulation of damaged proteins, such as lipofuscin, during age- stem [15]. Analyses of brain tissue samples suggest a much more
ing and senescence. Later it was revealed that the decreased func- severe malfunction of the proteasomal system in Alzheimer’s dis-
tion of proteasome in senescence is primarily due to the reduced ease [18] and Parkinson’s disease [20]. Several of the adverse con-
rates of proteasome biosynthesis and assembly. Moreover, it was sequences of ageing on neuronal function and survival can be
determined that replicative senescence is accompanied with mimicked by pharmacological inhibition of proteasomes [25] or
reduced levels of the b-type subunits, thus acting as the ‘‘rate-lim- lysosomes [2]. In addition, increasing evidence suggests that im-
iting’’ subunits for efficient proteasome assembly [5]. paired autophagy is important in neuronal dysfunction and death
In the mammalian muscle the proteasome-dependent proteo- in ageing and age-related disease [22].
lytic pathway is sequentially regulated by amino acids during mat- In several neurodegenerative diseases, such as Huntington’s,
uration and ageing. The study of the respective role of insulin and Alzheimer’s and Parkinson’s, autophagy tries to protect the cells
amino acids in the inhibition of proteolysis in the fed state sug- from the accumulation and aggregation of defective misfolded
gested a threshold of insulin induction below which proteasome- cytosolic proteins caused by mutations in their genes, e.g. the poly-
dependent proteolysis may not be regulated. The lack of regulation glutamine-tract at the N-terminus of the Huntingtin protein in the
of skeletal muscle proteasome-dependent proteolysis in old rats is case of Huntington’s, b-amyloid precursor protein and the microtu-
mainly mediated by a resistance to amino acids [4]. bule-associated protein tau in Alzheimer’s, and a-synuclein in Par-
As ageing is a progressive and irreversible (but not pathological) kinson’s disease. Under normal conditions, these proteins are
phenomenon, a decline in proteasome activity may be regarded as degraded by the ubiquitin–proteasome system, but in these neuro-
the natural answer to an age-related decrease in the rate of protein degenerative diseases this system becomes overwhelmed and
synthesis, a process counterbalanced by protein degradation. autophagy acts as a rescue system.
Therefore, it is no surprise that experimental inhibition of protea- In rats, proteasome activity decreases with advancing age in the
some activity in primary neuronal cells, for example, induces cerebral cortex, hippocampus and spinal cord, but not in the cere-
impairments in protein synthesis, especially since many transcrip- bellum or brainstem. Analyses of brain tissue samples suggest a
tion factors and some translation-initiation factors, as well as ribo- much more severe malfunction of the proteasomal system in Alz-
some biogenesis, are regulated by proteasomal processing. heimer’s and Parkinson’s disease. Several of the adverse conse-
However, proteasome activity may also be necessary to remove quences of ageing on neuronal function and survival can be
products of other processes that become imbalanced during age- mimicked by pharmacological inhibition of proteasomes or lyso-
ing, such as the antioxidant response system. For example, a low- somes. In addition, increasing evidence suggests that impaired
ered capacity to eliminate oxidatively damaged proteins due to a autophagy is important in neuronal dysfunction and death in age-
decrease in all three proteasome peptidase activities in eye lens ing and age-related disease. It has been shown that autophagy dis-
nuclei favours cataract formation in elderly individuals and, in rupted during normal ageing can be restored by dietary restriction,
heart muscle, reduces tolerance of the aged heart to ischaemia/ which is consistent with a possible role for impaired removal of
reperfusion. damaged organelles in neuropathologies of ageing. Proteins can
Structural alterations of proteasome subunits have been shown be altered by mutations, post-translational modifications, or by
to affect proteasome activity through changes of the 20S barrel intracellular or extracellular aggressors (oxidative stress, ultravio-
conformation. Initially, SDS and some fatty acids have been shown let radiation and toxic compounds) that often induce a change in
to stimulate proteasome activities in vitro [9], whereas potassium their conformation. The altered protein is usually recognised by
chloride has a negative effect [17] by favouring the open or the molecular chaperones, which are proteins that allow protein repair
closed conformation of the proteasome, respectively. Protea- or refolding. If the chaperones do not succeed in this repairing
some-activating hydrophobic peptides have been shown to be activity, they send the altered protein to degradation—usually to
bound as modifiers at noncatalytic sites, thus mimicking the effect the ubiquitin–proteasome system.
of 11S complex by opening the a-gated pore [16]. The increased However, if the activity of these systems is impaired, or the pro-
activities promote cellular resistance to oxidants and confer exten- tein has already been organised into insoluble complex structures
sion of human fibroblasts lifespan [13]. (fibres or oligomers), macroautophagy is the only system that is
able to remove those toxic complexes. In conditions in which not
even macroautophagy can remove the altered proteins, these
4. Proteasome and neurodegeneration aggregates usually trap other still-functional proteins inside them-
selves [19].
A life-long steady decrease in proteasome activity is proposed
to be responsible for accumulation of abnormally folded proteins,
formation of inclusion bodies and development of neurodegenera- 5. Insulin/IGF-1 signalling and the proteasome
tive diseases such as Alzheimer’s and Parkinson’s disease and Hun-
tington’s disease. Thus, age-related proteasomal dysfunction could The ubiquitin–proteasome pathway is a new partner for the
be regarded as a factor in these disease processes, which involve control of insulin signalling. The insulin-like growth factor type 1
the formation of plaques, filaments and aggregates. Once gener- (IGF-I) plays an important role in neuronal physiology. Reduced
ated, these protein inclusions have been found to further inhibit IGF-I levels are observed during ageing and this decrease may be
proteasome activity and thus amplify the formation of inclusion important to age-related changes in the brain. IGF-I stimulates pro-
bodies. teasome activity, in a process mediated by the IGF-I receptor, PI3-
A major problem encountered by neurons during ageing that is kinase/mTOR (mammalian target of rapamycin) signalling [7].
strongly linked to neurodegenerative disorders is the accumulation In frontal cortex the level of oxidised proteins is significantly
of damaged proteins that form insoluble aggregates that accumu- reduced in transgenic mice designed to overproduce IGF-I com-
42 P. Löw / General and Comparative Endocrinology 172 (2011) 39–43

pared with wild-type animals. The frontal cortex of IGF-I overpro- destruction by the proteasome [24]. Their work revealed two
ducing mice exhibited high chymotrypsin-like activity of the 20S important observations. First, SOCS-1 and SOCS-3 were shown to
and 26S proteasomes. The proteasome can also be activated in re- have distinct binding sites for IRS-1 or IRS-2 and for elongin BC
sponse to IGF-I in cell cultures. Kinetic studies revealed peak acti- ubiquitin ligase. Secondly, SOCS-mediated ubiquitylation of IRS-1
vation of the proteasome within 15 min following IGF-I or IRS-2 is a necessary step for inhibition of insulin action in vari-
stimulation. The effects of IGF-I on proteasome were not observed ous cell lines and mouse liver. This mechanism is consistent with
in R cells lacking the IGF-I receptor. Experiments using specific ki- the function of SOCS-1 and SOCS-3 as adapter molecules linking
nase inhibitors suggested that activation of proteasome by IGF-I in- tyrosine-phosphorylated proteins to an elongin BC-based E3 ubiq-
volves phosphatidyl inositol 3-kinase and mammalian target of uitin-ligase [12]. It appears that the ‘SOCS box’ acts as an adapter to
rapamycin signalling. IGF-I also attenuated the increase in protein facilitate the ubiquitylation of signalling proteins and their subse-
carbonyl content induced by proteasome inhibition. Thus, appro- quent targeting to the proteasome.
priate levels of IGF-I may be important for the elimination of oxi-
dised proteins in the brain in a process mediated by activation of
the proteasome. IGF-I can increase proteasome activity through 7. Conclusions
the mTOR pathway in vitro and that IGF-I levels in the brain corre-
late with proteasome activity. This suggests that the neuroprotec-  Proteasome deficiency is both a cause and a consequence of
tive effect of IGF-I may involve the clearance of deleterious ageing, and contributes to the general decrease in protein integ-
substances in the adult brain and favour the idea that maintaining rity and functionality with age.
IGF-I levels in brain may be beneficial during ageing [7,10].  Longevity increases when FoxO becomes activated in response
Insulin/IGF-1 signalling regulates ageing in worms, flies and to reduced insulin/IGF-1 signalling. IGF-I or insulin can reduce
mammals. The insulin/IGF-1 receptor, through activating a cascade protein degradation rapidly by suppressing FoxO transcription,
of conserved kinases, inhibits the forkhead transcription factor, which also inhibits proteasomal degradation.
FoxO. Longevity increases when FoxO becomes activated in re-  Insulin-induced degradation of IRS-1 is mediated by the protea-
sponse to reduced insulin/IGF-1 signalling. These findings indicate some degradation pathway.
that IGF-I or insulin can reduce protein degradation rapidly by sup-  Damaged proteins are removed by the proteasome, and this is
pressing autophagy via mTOR activation and independently Akt altered in neurons during ageing. Age-associated neurodegener-
suppressing FoxO transcription, which also inhibits proteasomal ative diseases such as Alzheimer’s disease and Parkinson’s dis-
degradation through the reduction of transcription of ubiquitin li- ease are accompanied by an accumulation of damaged,
gases atrogin-1 and MuRF1. Phosphorylation of FoxO in response misfolded and biologically inactive proteins.
to insulin not only leads to its nuclear exclusion, but also to its
ubiquitin-mediated degradation through the 26S proteasome [27]. The importance of the ubiquitin–proteasome system for molec-
ular and cellular biology, as well as for medical sciences is increas-
ing. In regard to ageing of cells and tissues, the examples presented
6. Ubiquitin–proteasomal degradation of IRS-1
in this review suggest that deregulation and changes of the protea-
some activity might have serious implications for ageing of organ-
While aberrations in the ubiquitin–proteasome pathway have
isms as well as for ageing associated diseases. In any case, further
been implicated in certain malignancies and neurodegenerative
understanding of the influence of the ubiquitin–proteasome sys-
disorders, recent studies indicate a role for this system in the path-
tem on the ageing process might help to identify targets for pre-
ogenesis of diabetes and its complications. Inappropriate degrada-
vention of deleterious loss of cell and tissue function and
tion of insulin signalling molecules such as insulin receptor
pathogenesis of ageing related diseases.
substrates (IRS-1 and IRS-2) has been demonstrated in experimen-
tal diabetes, mediated in part through the up-regulation of sup-
pressors of cytokine signalling (SOCS). It appears that altered References
ubiquitin–proteasome system might be one of the molecular
[1] M. Balasubramanyam, R. Sampathkumar, V. Mohan, Is insulin signaling
mechanisms of insulin resistance in many pathological situations.
molecules misguided in diabetes for ubiquitin–proteasome mediated
Drugs that modulate the SOCS action and/or proteasomal degrada- degradation? Mol. Cell. Biochem. 275 (2005) 117–125.
tion of proteins could become novel agents for the treatment of [2] E. Bednarski, C.E. Ribak, G. Lynch, Suppression of cathepsins B and L causes a
insulin resistance and Type 2 diabetes [1]. proliferation of lysosomes and the formation of meganeurites in hippocampus,
J. Neurosci. 17 (1997) 4006–4021.
Insulin receptor substrate (IRS) proteins are important intracel- [3] L. Borissenko, M. Groll, Diversity of proteasomal missions: fine tuning of the
lular molecules that mediate insulin receptor tyrosine kinase sig- immune response, Biol. Chem. 388 (2007) 947–955.
nalling. A decreased content of IRS proteins has been found in [4] F. Capel, M. Prod’homme, D. Bechet, D. Taillandier, M. Balage, D. Attaix, L.
Combaret, Lysosomal and proteasome-dependent proteolysis are differentially
insulin-resistant states in animals, humans and cultured cells un- regulated by insulin and/or amino acids following feeding in young mature
der various conditions. However, the molecular mechanism that and old rats, J. Nutr. Biochem. 20 (2009) 570–576.
controls cellular levels of IRS proteins is unknown. Chronic insulin [5] N. Chondrogianni, F.L. Stratford, I.P. Trougakos, B. Friguet, A.J. Rivett, E.S.
Gonos, Central role of the proteasome in senescence and survival of human
treatment induces the degradation of IRS-1, but not IRS-2, protein fibroblasts: induction of a senescence-like phenotype upon its inhibition and
in cultured cells. The insulin-induced degradation of IRS-1 can be resistance to stress upon its activation, J. Biol. Chem. 278 (2003) 28026–28037.
prevented by pretreatment with lactacystin, a specific inhibitor [6] O. Coux, K. Tanaka, A.L. Goldberg, Structure and functions of the 20S and 26S
proteasomes, Annu. Rev. Biochem. 65 (1996) 801–847.
for proteasome degradation. These data demonstrate that insu- [7] E. Crowe, C. Sell, J.D. Thomas, G.J. Johannes, C. Torres, Activation of proteasome
lin-induced degradation of IRS-1 is mediated by the proteasome by insulin-like growth factor-I may enhance clearance of oxidized proteins in
degradation pathway. the brain, Mech. Ageing Dev. 130 (2009) 793–800.
[8] B. Dahlmann, Role of proteasomes in disease, BMC Biochem. 8 (suppl. 1) (2007)
Recent studies have revealed that IRS proteins are ubiquitylated
S3.
and subsequently degraded by the 26S proteasome during insulin [9] B. Dahlmann, M. Rutschmann, L. Kuehn, H. Reinauer, Activation of the
stimulation or during cellular stress [23,26]. In an elegant study, multicatalytic proteinase from rat skeletal muscle by fatty acids or sodium
White and his colleagues indicated that SOCS-1 and SOCS-3 (a fam- dodecyl sulphate, Biochem. J. 228 (1985) 171–177.
[10] S. Fernandez, A.M. Fernandez, C. Lopez-Lopez, I. Torres-Aleman, Emerging
ily of proteins called the suppressor of cytokine signalling, SOCS) roles of insulin-like growth factor-I in the adult brain, Growth Horm. IGF Res.
modulate insulin signalling by targeting IRS-1 and IRS-2, for 17 (2007) 89–95.
P. Löw / General and Comparative Endocrinology 172 (2011) 39–43 43

[11] A. Hershko, A. Ciechanover, The ubiquitin system, Annu. Rev. Biochem. 67 [20] K.S. McNaught, C.W. Olanow, B. Halliwell, O. Isacson, P. Jenner, Failure of the
(1998) 425–479. ubiquitin–proteasome system in Parkinson’s disease, Nat. Rev. Neurosci. 2
[12] T. Kamura, S. Sato, D. Haque, L. Liu, W.G. Kaelin Jr., R.C. Conaway, J.W. (2001) 589–594.
Conaway, The Elongin BC complex interacts with the conserved SOCS-box [21] R.A. Nixon, The calpains in aging and aging-related diseases, Ageing Res. Rev. 2
motif present in members of the SOCS, ras, WD-40 repeat and ankyrin repeat (2003) 407–418.
families, Genes Dev. 12 (1998) 3872–3881. [22] R.A. Nixon, J. Wegiel, A. Kumar, W.H. Yu, C. Peterhoff, A. Cataldo, A.M. Cuervo,
[13] M. Katsiki, N. Chondrogianni, I. Chinou, A.J. Rivett, E.S. Gonos, The olive Extensive involvement of autophagy in Alzheimer disease: an immuno-
constituent oleuropein exhibits proteasome stimulatory properties in vitro electron microscopy study, J. Neuropathol. Exp. Neurol. 64 (2005) 113–122.
and confers life span extension of human embryonic fibroblasts, Rejuvenation [23] L. Rui, T.L. Fisher, J. Thomas, M.F. White, Regulation of insulin/insulin-like
Res. 10 (2007) 157–172. growth factor-1 signaling by proteasome-mediated degradation of insulin
[14] J.N. Keller, J. Gee, Q. Ding, The proteasome in brain aging, Ageing Res. Rev. 1 receptor substrate-2, J. Biol. Chem. 276 (2001) 40362–40367.
(2002) 279–293. [24] L. Rui, M. Yuan, D. Frantz, S. Shoelson, M.F. White, SOCS-1 and SOCS-3 block
[15] J.N. Keller, K.B. Hanni, W.R. Markesbery, Possible involvement of proteasome insulin signaling by ubiquitin-mediated degradation of IRS-1 and IRS-2, J. Biol.
inhibition in aging: implications for oxidative stress, Mech. Ageing Dev. 113 Chem. 277 (2002) 42394–42398.
(2000) 61–70. [25] P.G. Sullivan, N.B. Dragicevic, J.H. Deng, Y.D. Bai, E. Dimayuga, Q.X. Ding, Q.H.
[16] A.F. Kisselev, D. Kaganovich, A.L. Goldberg, Binding of hydrophobic peptides to Chen, A.J. Bruce-Keller, J.N. Keller, Proteasome inhibition alters neural
several non-catalytic sites promotes peptide hydrolysis by all active sites of 20 mitochondrial homeostasis and mitochondria turnover, J. Biol. Chem. 279
S proteasomes. Evidence for peptide-induced channel opening in the alpha- (2004) 20699–20707.
rings, J. Biol. Chem. 277 (2002) 22260–22270. [26] X.J. Sun, J.L. Goldberg, L.Y. Qiao, J.J. Mitchell, Insulin-induced insulin receptor
[17] A. Köhler, P. Cascio, D.S. Leggett, K.M. Woo, A.L. Goldberg, D. Finley, The axial substrate-1 degradation is mediated by the proteasome degradation pathway,
channel of the proteasome core particle is gated by the Rpt2 ATPase and Diabetes 48 (1999) 1359–1364.
controls both substrate entry and product release, Mol. Cell 7 (2001) 1143– [27] T. Vellai, K. Takacs-Vellai, M. Sass, D.J. Klionsky, The regulation of aging: does
1152. autophagy underlie longevity? Trends Cell Biol. 19 (2009) 487–494.
[18] Y.A. Lam, C.M. Pickart, A. Alban, M. Landon, C. Jamieson, R. Ramage, R.J. Mayer, [28] D. Voges, P. Zwickl, W. Baumeister, The 26S proteasome: A molecular machine
R. Layfield, Inhibition of the ubiquitin–proteasome system in Alzheimer’s designed for controlled proteolysis, Ann. Rev. Biochem. 68 (1999) 1015–1068.
disease, Proc. Natl. Acad. Sci. USA 97 (2000) 9902–9906. [29] K.D. Wilkinson, Ubiquitination and deubiquitination: targeting of proteins for
[19] M. Martinez-Vicente, A.M. Cuervo, Autophagy and neurodegeneration: when degradation by the proteasome, Semin. Cell Dev. Biol. 11 (2000) 141–148.
the cleaning crew goes on strike, Lancet Neurol. 6 (2007) 352–361.

You might also like