Você está na página 1de 16

Seminars in Cell & Developmental Biology 18 (2007) 716–731

Review

The endoplasmic reticulum and the unfolded protein response


Jyoti D. Malhotra a,b , Randal J. Kaufman a,b,∗
a Howard Hughes Medical Institute, Department of Biological Chemistry, University of Michigan Medical Center,
Ann Arbor, MI 48109, United States
b Howard Hughes Medical Institute, Department of Internal Medicine, University of Michigan Medical Center,

Ann Arbor, MI 48109, United States


Available online 8 September 2007

Abstract
The endoplasmic reticulum (ER) is the site where proteins enter the secretory pathway. Proteins are translocated into the ER lumen in an
unfolded state and require protein chaperones and catalysts of protein folding to attain their final appropriate conformation. A sensitive surveillance
mechanism exists to prevent misfolded proteins from transiting the secretory pathway and ensures that persistently misfolded proteins are directed
towards a degradative pathway. In addition, those processes that prevent accumulation of unfolded proteins in the ER lumen are highly regulated
by an intracellular signaling pathway known as the unfolded protein response (UPR). The UPR provides a mechanism by which cells can
rapidly adapt to alterations in client protein-folding load in the ER lumen by expanding the capacity for protein folding. In addition, a variety of
insults that disrupt protein folding in the ER lumen also activate the UPR. These include changes in intralumenal calcium, altered glycosylation,
nutrient deprivation, pathogen infection, expression of folding-defective proteins, and changes in redox status. Persistent protein misfolding initiates
apoptotic cascades that are now known to play fundamental roles in the pathogenesis of multiple human diseases including diabetes, atherosclerosis
and neurodegenerative diseases.
© 2007 Elsevier Ltd. All rights reserved.

Keywords: Endoplasmic reticulum; Unfolded protein response; ER; Secretory pathway; Apoptosis

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 717
2. Protein folding and quality control in the ER . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 717
3. UPR signaling . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 718
3.1. Perk phosphorylates eIF2␣ to attenuate mRNA translation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 718
3.2. IRE1␣ initiates non-conventional splicing of Xbp1 mRNA . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 719
3.3. ATF6-mediated transcriptional activation requires regulated intramembrane proteolysis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 719
3.4. BiP is a master regulator of UPR sensor activaton . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 721
4. ER stress-induced apoptosis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 721
4.1. CHOP is a UPR-induced transcription factor that mediates apoptosis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 722
4.2. BCL2-family members regulate ER stress-induced apoptosis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 722
4.3. IRE1␣-dependent activation of JNK mediates ER stress-induced apoptosis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 722
4.4. ER stress-induced caspase-dependent apoptosis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 723
5. ER stress and oxidative stress . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 723
5.1. Oxidative stress disrupts ER function . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 723
5.2. Protein folding in the ER causes oxidative stress . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 723
6. ER stress and disease pathogenesis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 725
6.1. ER stress contributes to metabolic disease . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 725

∗ Corresponding author at: University of Michigan Medical Center/HHMI, MSRB II, Room 4570, 1150 W. Medical Center Drive, Ann Arbor, MI 48109,
United States. Tel.: +1 734 763 9037; fax: +1 734 763 9323.
E-mail address: kaufmanr@umich.edu (R.J. Kaufman).

1084-9521/$ – see front matter © 2007 Elsevier Ltd. All rights reserved.
doi:10.1016/j.semcdb.2007.09.003
J.D. Malhotra, R.J. Kaufman / Seminars in Cell & Developmental Biology 18 (2007) 716–731 717

6.2. ER stress contributes to neurodegenerative disease . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 726


6.3. ER stress contributes to hyperhomocysteinemia and atherosclerosis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 727
7. Future perspectives . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 727
Acknowledgments . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 728
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 728

1. Introduction all contribute the accumulation of unfolded proteins in the ER


lumen.
Protein folding is an essential process for protein function Accumulation of unfolded protein initiates activation of
in all organisms. As a consequence, all cells have evolved an adaptive signaling cascade known as the unfolded protein
sophisticated mechanisms to ensure proper protein folding response (UPR). Appropriate adaptation to misfolded protein
occurs and to dispose of irreversibly misfolded proteins. All accumulation in the ER lumen requires regulation at all levels of
proteins that transit the secretory pathway in eukaryotic cells gene expression including transcription, translation, transloca-
first enter the endoplasmic reticulum (ER) where they fold tion into the ER lumen, and ERAD. Coordinate regulation of all
and assemble into multi-subunit complexes prior to transit these processes is required to restore proper protein folding and
to the Golgi compartment [1]. ‘Quality control’ is a surveil- ER homeostasis [1–6]. Conversely, if the protein folding defect
lance mechanism that permits only properly folded proteins is not resolved, chronic activation of UPR signaling occurs
to exit the ER en route to other intracellular organelles and which eventually induces an apoptotic (programmed cell death)
the cell surface. Misfolded proteins are either retained within response.
the ER lumen in complex with molecular chaperones or are In this review we summarize the signaling pathways that
directed toward degradation through the 26S proteasome in a mediate the UPR, mechanisms that signal cell death, the role of
process called ER-associated degradation (ERAD) or through the UPR in mammalian physiology, and the clinical implications
autophagy. of the UPR in health and disease.
The efficiency of protein-folding reactions depends on
appropriate environmental, genetic and metabolic conditions. 2. Protein folding and quality control in the ER
Conditions that disrupt protein folding present a threat to cell
viability. The ER provides a unique environment that chal- Protein folding and maturation in vivo is a highly assisted
lenges proper protein folding as nascent polypeptide chains process. The ER lumen contains molecular chaperones, fold-
enter the ER lumen. A high concentration of partially folded ing enzymes and quality control factors that assist in folding
and unfolded proteins predisposes protein-folding intermediates and trafficking of newly synthesized polypeptides. Nascent
to aggregation. Polypeptide binding proteins, such as BiP and polypeptide chains enter the ER lumen through a proteinaceous
GRP94, act to slow protein-folding reactions and prevent aber- channel, the Sec 61 translocon complex. The nascent chains
rant interactions and aggregation. The ER lumen is an oxidizing of most translocated polypeptides are subject to addition of
environment so disulfide bond formation occurs. As a conse- a preassembled oligosaccharide core (N-acetylglucosamine2 -
quence, cells have evolved sophisticated machinery composed mannose9 -glucose3 ), i.e., Glc3 Man9 GlcNac2 to selective
of multiple protein disulfide isomerases (PDIs) that are required asparagine (N) residues. N-Glycosylation is catalyzed by the
to ensure proper disulfide bond formation and prevent forma- oligosaccharyltransferase (OST), a multisubunit enzyme asso-
tion of illegitimate disulfide bonds. The ER is also the primary ciated with the translocon complex. Subsequently, sequential
Ca2+ storage organelle in the cell. Both protein-folding reac- action by the ER ␣-glucosidases I and II removes the two out-
tions and protein chaperone functions require high levels of ER ermost glucose residues to produce a mono-glucosylated core
intralumenal calcium. Protein folding in the ER requires exten- glycan. The mono-glucosylated glycoprotein can then interact
sive amounts of energy and depletion of energy stores prevents with two homologous ER lectins calnexin (CNX) and calreti-
proper protein folding. ATP is required for chaperone func- culin (CRT) that associate with Erp57, an oxidoreductase that
tion, to maintain Ca2+ stores and redox homeostasis, and for catalyzes disulfide bond formation. Upon release of folding sub-
ERAD. Finally, proteins that enter the ER lumen are subject to strates from CNX and/or CRT, the innermost glucose residue is
numerous post-translational modifications including N-linked rapidly removed by glucosidase II. Two potential mechanisms
glycosylation, amino acid modifications such as proline and by which chaperones may monitor protein folding are through
aspartic acid hydroxylation and ␥-carboxylation of glutamic acid exposed hydrophobic patches or through excessive surface
residues, and addition of glycosylphosphatidylinositol anchors. dynamics associated with the non-compact partially folded state.
All these processes are highly sensitive to alterations in the ER The protein chaperone BiP binds to the hydrophobic patches
luminal environment. As a consequence, innumerable environ- exposed on protein folding intermediates. Highly dynamic, non-
mental insults alter protein-folding reactions in the ER through native deglucosylated glycoproteins are recognized by the ER
mechanisms that include depletion of ER calcium, alteration folding sensor UDP-glucose:glycoprotein glucosyltransferase
in the redox status, and energy (sugar/glucose) deprivation. In (UGT1). UGT1 specifically re-glucosylates folding intermedi-
addition, gene mutations, elevated protein traffic through the ates released from CNX/CRT cycle. Reglucosylation mediates
ER compartment, and altered post-translational modification ER retention of immaturely folded glycoproteins so they enter
718 J.D. Malhotra, R.J. Kaufman / Seminars in Cell & Developmental Biology 18 (2007) 716–731

another round of CNX/CRT-assisted folding. Polypeptides that 3.1. Perk phosphorylates eIF2α to attenuate mRNA
fail to acquire their native transport-competent structure are translation
eventually removed by ER-associated degradation (ERAD).
During ERAD, folding-defective proteins are retranslocated to The most immediate response to ER stress in metazoan
the cytosol, in a process possibly mediated by EDEM and Derlin, cells is reversibly, transient attenuation of mRNA translation,
and are degraded by the 26S proteasome. Native, properly folded thereby preventing influx of newly synthesized polypeptides
polypeptides released from CNX/CRT are transported from the into the stressed ER lumen [12]. This translational attenua-
ER to the Golgi compartment, in an event possibly assisted by tion is signaled through PERK-mediated phosphorylation of the
mannose-binding lectins, such as ERGIC-53, VIPL, ERGL [7] eukaryotic translation initiation factor 2 on the alpha subunit
(Fig. 1). (eIF2␣) at Ser51. eIF2␣ phosphorylation inhibits the guanine
nucleotide exchange factor eIF2B that recycles the eIF2 com-
3. UPR signaling plex to its active GTP-bound form. The formation of the ternary
translation initiation complex eIF2-GTP-tRNAMet is required
In response to ER stress, three ER-localized transmembrane for AUG initiation codon recognition and joining of the 60S ribo-
signal transducers are activated to initiate adaptive responses. somal subunit that occurs during initiation phase of polypeptide
These transducers are two protein kinases inositol requiring chain synthesis. Lower levels of active ternary complex result in
kinase 1 (IRE1) [8,9], and double stranded RNA-activated pro- lower levels of translation initiation [10,13–15] (Fig. 1).
tein kinase-like ER kinase (PERK) [10] and the transcription PERK is an ER-associated transmembrane serine/threonine
factor activating transcription factor 6 (ATF6) [9,11]. These protein kinase. Upon accumulation of unfolded proteins in the
three UPR transducers are constitutively expressed in all known ER lumen, PERK dimerization and trans-autophosphorylation
metazoan cells (Fig. 1). IRE1 was the first component of the leads to activation of its eIF2␣ kinase function. [10,16]. In addi-
UPR that was identified, initially in yeast, and is conserved in tion to translational attenuation, activation of PERK also induces
all eukaryotic cells. The essential and unique properties of IRE1 transcription of approximately 1/3 of the UPR-dependent genes
signaling in the UPR have been conserved in all eukaryotic [13–15,17]. Although phosphorylation of eIF2␣ inhibits general
cells but higher eukaryotes also possess the additional sen- translation initiation, it is required for the selective translation
sors PERK and ATF6 that promote stress adaptation or cell of several mRNAs. One fundamental transcription factor for
death in a more complex and diverse, yet coordinated man- which translation is activated upon PERK-mediated phospho-
ner. rylation of eIF2␣, is the activating transcription factor 4 (ATF4)

Fig. 1. Protein trafficking from the ER: upon translocation of polypeptides through the Sec61 proteinaceous channel, asparagine residues are frequently modified by
covalent addition of a preassembled oligosaccharide core (N-acetylglucosamine2 -mannose9 -glucose3 ). This reaction is catalyzed by the oligosaccharyltransferase
(OST), a multisubunit complex associated with translocon. To facilitate unidirectional transport through the translocon, nascent polypeptide chains in the ER lumen
interact with BiP, a molecular chaperone that binds to exposed hydrophobic residues. Subsequently, rapid deglucosylation of the two outermost glucose residues
on the oligosaccharide core structures, mediated by glucosidase I and II (GlcI and GlcII), prepares glycoproteins for association with the ER lectins calnexin and
calreticulin. The calnexin/calreticulin-associated oxidoreductase ERp57 facilitates protein folding by catalyzing formation of intra- and inter-molecular disulfide
bonds, a rate-limiting step in the protein folding process. Release from calnexin/calreticulin followed by glucosidase II cleavage of the innermost glucose residue
prevents further interaction with calnexin and calreticulin. At this point, natively folded polypeptides transit the ER to the Golgi compartment, in a process possibly
assisted by mannose-binding lectins, such as ERGIC-53, VIPL, ERGL. As an essential component of protein-folding quality control, non-native polypeptides
are tagged for reassociation with calnexin/calreticulin by the UDP-glucose:glycoprotein glucosyltransferase (UGT1) to facilitate their ER retention and prevent
anterograde transport. Polypeptides that are folding incompetent are targeted for degradation by retrotranslocation, possibly mediated by EDEM and Derlins, into
the cytosol and delivery to the 26S proteosome. Triangles represent glucose residues, squares represent N-acetylglucosamine residues, and circles represent mannose
residues.
J.D. Malhotra, R.J. Kaufman / Seminars in Cell & Developmental Biology 18 (2007) 716–731 719

[13–15,17]. Expression profiling identified that genes encoding Two mammalian homologues of yeast IRE1 have been iden-
amino acid biosynthesis and transport functions, anti-oxidative tified; IRE1␣ [25] and IRE1␤ [26]. IRE1␣ is expressed in most
stress responses, and apoptosis, such as growth arrest and DNA cells and tissues, with highest levels of expression in the pan-
damage 34 (GADD34) and CAAT/Enhancer binding protein creas and placenta [25]. IRE1␤ expression is prominent only
(C/EBP) homolgous protein (CHOP/GADD153) [16,18] require in intestinal epithelial cells [26]. The cleavage specificities of
PERK, eIF2␣ phosphorylation, and ATF4 [13–15,17]. IRE1␣ and IRE1␤ are quite similar, thereby suggesting that they
Although the majority of PERK signaling is mediated do not recognize distinct substrates but rather confer temporal-
through phosphorylation of eIF2␣, studies suggest that the bZiP and tissue-specific expression [27].
Cap ‘n’ Collar transcription factor nuclear respiratory factor 2 Transcriptional analysis of UPR gene targets, such as BiP,
(NRF2) may also be a substrate for the PERK kinase activity GRP94 and calreticulin, identified a mammalian ER stress
[19]. NRF1 and NRF2 are transcription factors that integrate response element (ERSE, CCAAT(N9 )CCACG) that is neces-
a variety of responses to oxidative stress. NRF2 is distributed sary and sufficient for UPR gene activation [28]. Yoshida et al.
in the cytoplasm through its association with the microtubule- used a yeast one-hybrid screen to isolate factors that interact
associated protein Keap1 (Kelch-like Ech-associated protein with the ERSE. This screen identified one ERSE-binding pro-
1). Upon ER stress, PERK phosphorylates NRF2 to promote tein as the bZIP-containing transcription factor XBP1 (X-box
its dissociation from Keap1, leading to the nuclear accumula- binding protein) [28]. Subsequently, several groups using dif-
tion of NRF2. Nrf2−/− cells are sensitive to ER stress-induced ferent approaches demonstrated that XBP1 mRNA is a substrate
apoptosis. NRF2 is a direct PERK substrate and effector of for the endoribonuclease activity of metazoan IRE1 [9,29–31].
PERK-dependent cell survival [19]. NRF2 binds to the antioxi- Upon activation of the UPR, the IRE1 RNase activity initiates
dant response element (ARE) to activate transcription of genes removal of a 26 nucleotide intron from XBP1 mRNA. This splic-
encoding detoxifying enzymes like A1 and A2 subunits of ing reaction creates a translational frameshift to produce a larger
glutathione S-transferase, NAD(P)H:quinone oxidoreductase, form of XBP1 that contains a novel transcriptional activation
␥-glutamylcysteine synthetase, Heme oxygenase-1 (HO-1) and domain it its C-terminus. Spliced XBP1 is a transcriptional acti-
UDP-glucoronosyl transferase [20]. Possibly in a similar man- vator that plays a fundamental role activation of wide variety of
ner, NRF1 is localized to the ER membrane and translocates UPR target genes. Some of the genes identified that require the
to the nucleus upon ER stress [21]. These data support the IRE1/XBP1 pathway are those that encode functions involved
notion that PERK phosphorylates multiple substrates to protect in ERAD, such as the ER degradation-enhancing mannosidase-
cells from oxidative stress. Consistent with this idea, Perk−/− like protein EDEM. Consistent with this observation, cells that
cells accumulate ROS when they are exposed to ER stress are deficient in either IRE1 or XBP1 are defective in ERAD
[14]. [32](Fig. 2).
Analysis of gene-deleted mice has provided insight into the
3.2. IRE1α initiates non-conventional splicing of Xbp1 physiological roles of IRE1 and XBP1 in mammals. Dele-
mRNA tion of Ire1α or Xbp1 in mice creates an embryonic lethality
at E11.5–E14 [30,33]. Although deletion of Ire1β had no
The first component in the UPR pathway was isolated through developmental phenotype, Ire1β−/− mice were susceptible to
a genetic screen to identify mutants in UPR signaling in the bud- experimental-induced intestinal colitis [34]. Mice with heterozy-
ding yeast Saccharomyces cerevisiae. In this screen, Ire1p/Ern1p gous Xbp1 deletion appear normal but develop insulin resistance
was identified as an ER transmembrane protein kinase that is when fed a high-fat diet [35]. Thus, it was proposed that the UPR
required for the UPR [3,22]. Subsequently, it was discovered might be important in insulin signaling (see below). In addition,
that Ire1p is a bifunctional protein that also has a site-specific both IRE1 and XBP1 have critical roles in B cell differentiation.
endoribonuclease (RNase) activity [3,22]. Under non-stress Antigenic stimulation of mature B lymphocytes activates the
conditions, Ire1p protein kinase is maintained in an inactive UPR and signaling through IRE1-mediated XBP1 mRNA splic-
monomeric form through interactions with the protein chap- ing is required to drive B lymphocyte differentiation into plasma
erone Kar2p/BiP. Upon accumulation of unfolded proteins in cells [29,36–38]. These studies suggest that the IRE1/XBP1 sub-
the ER lumen, Ire1p is released from Kar2p/BiP and undergoes pathway of the UPR might be required for differentiation of cell
homodimerization and trans-autophosphorylation to activate its types that secrete high levels of protein. This is consistent with
RNase activity. The RNase activity of Ire1p cleaves a 252-base a requirement for XBP1 in pancreatic acinar cell development
intron from mRNA encoding the basic leucine zipper (bZIP)- [39].
containing transcription factor Hac1p. This splicing reaction
alters the carboxy-terminus of Hac1p to introduce a potent tran- 3.3. ATF6-mediated transcriptional activation requires
scriptional activation domain. The protein encoded by spliced regulated intramembrane proteolysis
HAC1 mRNA binds and activates transcription from the UPR
element [UPRE, minimal motif TGACGTG(C/A)] upstream The bZiP-containing activating transcription factor 6 (ATF6)
of many UPR target genes [2,23]. In S. cerevisiae, the UPR was identified as another regulatory protein that, like XBP1,
activates transcription of approximately 381 genes [24], more binds to the ERSE1 element in the promoters of UPR-responsive
than 50% of which provide functions in the secretory path- genes [28]. In mammals, there are two alleles of ATF6, ATF6␣
way. (90 kDa) and ATF6␤ (110 kDa), both synthesized in all cell types
720 J.D. Malhotra, R.J. Kaufman / Seminars in Cell & Developmental Biology 18 (2007) 716–731

Fig. 2. Signaling the unfolded protein response. Three proximal sensors IRE1, PERK and ATF6 regulate the UPR through their respective signaling cascades. Under
non-stressed conditions, BiP binds to the lumenal domains of IRE1, ATF6, and PERK. Upon accumulation of unfolded proteins in the ER lumen, IRE1 released
from BIP dimerizes to activate its kinase and RNase activities to initiate XBP1 mRNA splicing thereby creating a potent transcriptional activator. Primary targets that
require IRE1/XBP1 pathway for induction include genes encoding functions in ERAD. Similarly, ATF6 released from BiP transits to the Golgi compartment where
for cleavage by S1P and S2P proteases to yield a cytosolic fragment that migrates to the nucleus to further activate transcription of UPR-responsive genes. Finally,
PERK released from BiP dimerizes, autophosphorylates, and phosphorylates eIF2␣ on Ser 51 leading to general attenuation of translational initiation. Paradoxically,
eIF2␣ phosphorylation induces translation of ATF4 mRNA. The PERK/eIF2␣/ATF4 regulatory axis also induces expression of anti-oxidative stress response genes
and expression of genes encoding proteins with proapoptotic functions, such as CHOP.

as ER transmembrane proteins. In the unstressed cells, ATF6 is Pro-inflammatory cytokines IL6, 1L-1␤ and TNF␣ increase
localized at the ER membrane and bound to BiP. In response to transcription of CREBH to produce an inert protein that is local-
ER stress, BiP dissociation permits trafficking of ATF6 to the ized to the ER. Upon ER stress, CREBH transits to the Golgi
Golgi complex, where ATF6 is sequentially cleaved by two pro- compartment where it is cleaved by S1P and S2P processing
teases [40–42]. The serine protease site-1 protease S1P cleaves enzymes. However, cleaved CREBH does not activate transcrip-
ATF6 in the luminal domain. The N-terminal portion is sub- tion of UPR genes, but rather, induces transcription of a subset
sequently cleaved by the metalloprotease site-2 protease S2P of acute phase response genes, such as C-reactive protein and
[43]. The processed forms of ATF6␣ and ATF6␤ translocate murine Serum Amyloid P component (SAP) in hepatocytes.
to the nucleus and bind to the ATF/cAMP response element These studies identified CREBH as a novel ER-localized tran-
(CRE) and to the ER stress responsive element (ERSE-1) to scription factor that has an essential role in induction of innate
activate target genes [41]. ATF6␣ and ATF6␤ both require the immune response genes and links for the first time ER stress to
presence of the transcription factor CBF (also called NF-Y) to inflammatory responses [46].
bind ERSEI [40–42]. The proteases S1P and S2P were origi- In addition to ATF6 and CREBH, there are additional sim-
nally identified for their essential role in processing of the sterol ilarly related factors that are likely regulated through ER
response element binding protein (SREBP) transcription factor, stress-induced proteolytic processing, although their physiolog-
which is activated upon cholesterol deprivation [44] (Fig. 1). ical roles remain unknown. Old astrocyte specifically induced
Over-expression of the cleaved form of ATF6␣ identified a class substance (OASIS) and BBF2 human homolog on chromosome
of UPR genes that may be transcriptional targets of ATF6␣ [45]. 7 (BBF2H7) are cleaved by S1P and S2P in response to ER
Recently ATF6␣ and ATF6␤ have been deleted in the mouse. stress in astrocytes and neurons, respectively [47,48]. Tran-
Although deletion of either alone produce no significant phe- script induced in spermiogenesis 40 (Tisp40) is cleaved by S1p
notype, combined deletion is an early embryonic lethal. Where and S2P to activate transcription of EDEM [49]. These tissue-
ATF6␣ contributes significantly to UPR induced genes, no genes specific ATF6-like molecules may contribute to the ER stress
were identified that are regulated through ATF6␤. response. Finally, Luman/LZIP/CREB3 and CREB4 are also
Recently, additional bZIP-containing transcription factors two ATF6-like molecules that are cleaved by S1P and S2P to
that are localized to the ER and regulated by RIP have been activate UPR transcription, although their cleavage appears to
identified. CREBH was identified as a liver-specific bZiP not be activated by ER stress [50–52]. These transcription fac-
transcription factor of the CREB/ATF family with a trans- tors might be activated under conditions other than ER stress to
membrane domain that directs localization to the ER [46]. activate transcription of ER chaperones.
J.D. Malhotra, R.J. Kaufman / Seminars in Cell & Developmental Biology 18 (2007) 716–731 721

3.4. BiP is a master regulator of UPR sensor activaton tion, the luminal domain was shown to form dimers in vitro
in the absence of added polypeptide [59]. These observations
In non-stressed cells, the luminal domains of IRE1, PERK, bring into question the requirement for peptide binding to the
and ATF6 are bound to the protein chaperone BiP. In response MHC1-type cleft to promote dimerization. It is possible that
to stress, unfolded proteins accumulate and bind BiP, thereby a combination of BiP binding as well as peptide binding both
sequestering BiP and promoting BiP release from the UPR sen- regulate IRE1 dimerization. Future studies should resolve this
sors. When these sensors are bound to BiP, they are maintained issue.
in an inactive state [53]. This BiP-mediated negative-regulation
model for UPR activation is also supported by the observation
that BiP over-expression prevented activation of the UPR upon 4. ER stress-induced apoptosis
ER stress [54]. In addition, sufficiently high levels of expression
of any protein that binds BiP can activate the UPR. In con- If the UPR fails to resolve the protein-folding defect, apop-
trast, the accumulation of unfolded proteins that do not bind tosis is activated. In response to ER stress, apoptosis is signaled
BiP does not activate the UPR [55,56]. Analysis of the interac- through both mitochondrial-dependent and -independent path-
tion between BiP and ATF6 suggested that this dissociation is ways (Fig. 2). The ER might actually serve as a site where
not merely a consequence of competition between ATF6 and apoptotic signals are generated and integrated to elicit the death
unfolded protein for binding to BiP, but rather may involve response. Several mechanisms by which apoptotic signals are
an active ER stress-dependent release of BiP from ATF6 [57]. generated at the ER include: PERK/eIF2␣-dependent transcrip-
Recently, based on the X-ray crystal structure of the yeast Ire1p tional induction of the proapoptotic transcription factor CHOP;
luminal domain, Credle et al. identified a deep, long MHC1-type Bak/Bax-regulated Ca2+ release from the ER; IRE1-mediated
groove that exists in an Ire1p dimer and proposed that unfolded activation of apoptosis signal-regulating kinase 1 (ASK1)/c-Jun
polypeptides directly bind Ire1p to mediate its dimerization [58]. amino terminal kinase (JNK); and cleavage and activation of
However, although the X-ray crystal analysis of the human IRE1 procaspase 12 (Fig. 3). Recent evidence also suggests that oxida-
luminal domain indicated a similar structure as yeast Ire1p, the tive stress may provide a significant contributing factor to ER
MHC1-type groove was not solvent accessible [59]. In addi- stress-induced apoptosis.

Fig. 3. Pathways of ER stress-induced apoptosis. ER stress leads to several redundant pathways for caspase activation that involve mitochondrial-dependent and
independent pathways. Activated IRE1 recruits TRAF2 to elicit JNK phosphorylation and activation. Caspase 12 is a murine ER-associated proximal effector in
the caspase activation cascade that activates procaspase 9 to cleave procaspase 3, the primary executioner of cell death. A second cell death signaling pathway
activated by ER stress is mediated by transcriptional induction of genes encoding proapoptotic functions. Activation of PERK, ATF6 and possibly IRE1 lead to
transcriptional induction of CHOP that induces apoptosis possibly through up-regulating expression of the genes Gadd34, Dr5 and Trb3 or by inhibiting expression
of the anti-apoptotic gene Bcl2. Mitochondrial ROS can be generated as a result of ER stress-induced Ca2+ release and depolarization of the inner mitochondrial
membrane. Thus, oxidative stress in association of unresolved ER stress contributes to multiple pathways of cell death.
722 J.D. Malhotra, R.J. Kaufman / Seminars in Cell & Developmental Biology 18 (2007) 716–731

4.1. CHOP is a UPR-induced transacription factor that cascades [70,71]. In vitro experiments support the idea that
mediates apoptosis the increase in the cytosolic Ca2+ concentration (from micro-
molar to millimolar) activates the calcium-dependent protease
Probably the most significant ER stress-induced apoptotic m-calpain, which subsequently cleaves and activates the ER-
pathway is mediated through CHOP. CHOP/GADD153 (growth resident procaspase-12 to initiate caspase-dependent apoptosis
arrest and DNA damage 153) is a bZIP transcription factor that [72]. The Ca2+ released from the ER also enters mitochondria to
is induced through the ATF6 and PERK UPR pathways [18,60]. depolarize the inner membrane, and cause mitochondrial outer
Chop−/− cells are protected from ER stress-induced apopto- membrane pore transition to release cytochrome c and activate
sis [61], indicating the significance of this apoptotic pathway apoptosis protease-activating factor 1 (APAF-1)/procaspase-9-
during ER stress. Although the precise mechanism by which dependent apoptosis. Thus, BCL2 family members may regulate
CHOP mediates apoptosis in unknown, CHOP activates the tran- ER stress-induced apoptotic responses through Ca2+ signaling.
scription of several genes that may potentiate apoptosis. These The regulation of BH3-only members of the BCL2 family
include GADD34, ERO1, DR5 (death receptor 5), TRB3, and during ER stress is quite complex. BAX and BAK are required
carbonic anhydrase VI. GADD34 encodes a subunit of protein for most forms of apoptosis [73]. Additional BH3 domain-
phosphatase 2C that enhances dephosphorylation of eIF2␣ and only family members, p53 up-regulated modulator of apoptosis
promotes protein synthesis [62]. Persistent protein synthesis dur- (PUMA) and neutrophil NADPH oxidase factor (NOXA), are
ing periods of ER stress would chronically activate the UPR and up-regulated by p53 during ER stress. In addition, Puma−/−
initiate cell death pathways. ERO1 encodes an ER oxidase that cells and Noxa−/− cells are resistant to ER stress-induced apop-
increases the oxidizing potential of the ER [63]. DR5 encodes tosis [74]. BAX activation during ER stress is inhibited by
a cell surface death receptor that may activate caspase cascades the ER-localized anti-apoptotic factor Bax inhibitor-1 (BI-1).
[64]. Tribbles homolog 3 (TRB3) encodes a human ortholog BI-1−/− mice are sensitive to ER stress, whereas mice overex-
of Drosophila tribble, and TRB3-knock-down cells are resis- pressing BI-1 are resistant [75]. BCL2-interacting killer (BIK)
tant to ER stress-induced apoptosis [65]. Carbonic anhydrase is an ER-localized pro-apoptotic component that enhances the
VI may decrease the intracellular pH during ER stress [66]. recruitment of BAX and BAK to the ER membrane [76]. Finally,
CHOP has also been implicated in repressing transcription of BAX and BAK associate with IRE1␣ and potentiate its signaling
the anti-apoptotic BCL2 protein [67]. However the ability of during ER stress [77].
CHOP to induce ER stress-associated apoptosis has recently
been demonstrated to be dependent on the duration of the stress 4.3. IRE1α-dependent activation of JNK mediates ER
state. Chronic exposure to a mild stress can lead to adaptation by stress-induced apoptosis
selective attenuation of CHOP expression mediated by degrada-
tion of CHOP mRNA and CHOP protein, whereas expression In addition to initiating splicing of Xbp1 mRNA, activa-
of downstream targets encoding adaptive functions, such as ER tion of IRE1 signals into the MAP kinase cascade. The IRE1
chaperones like BiP, is persistent due to long-lived mRNAs and cytoplasmic domain interacts with the adaptor protein, tumor
proteins [68]. necrosis factor receptor-associated factor 2 (TRAF2). TRAF2
couples the activation of death receptors at the plasma mem-
4.2. BCL2-family members regulate ER stress-induced brane to activation of Jun kinase (JNK) and stress-activated
apoptosis protein kinase (SAPK) [78]. IRE1 and TRAF2 interact with
the mitogen-activated protein kinase kinase kinase, apopto-
During periods of ER stress, pro-apoptotic members of sis signal-regulating kinase 1 (ASK1), which subsequently
the Bcl2 family are recruited to the ER surface and activate phosphorylates and activates JNK [79]. Therefore, ER stress-
caspase-12. In contrast, the anti-apoptotic members prevent induced JNK activation and apoptosis are reduced in Ire1−/−
this recruitment, although the exact relationship between and Ask1−/− cells. However, this mechanism cannot account
these factors remains unclear. Over-expression of BCL2 fam- for the observation that Traf2−/− cells are more susceptible
ily members can prevent ER stress-induced apoptosis. BIM to ER stress-induced apoptosis [80]. TRAF2 also associates
(BCL2-interacting mediator of cell death) translocates from the with caspase-12 and regulates its activation [81]. IRE1–TRAF2
dynein-rich compartment to the ER membrane and activates activates the transcriptional repressor ATF3 as well, leading to
caspase-12 in response to ER stress, whereas an antiapoptotic apoptosis [82]. There is also evidence that UPR activation of
factor, BCL-xL (BCL2-like 1), binds to BIM and inhibits its IRE1 may initiate the extrinsic apoptotic pathway. IRE1 inter-
translocation [69]. Consistent with this notion, BIM is required acts with tumor necrosis factor receptor 1 (TNFR1) to form a
for ER stress-induced death [170]. complex with TRAF2 and ASK1 to mediate JNK activation.
BH3 domain (Bcl2-homology domain 3)-only containing The activation of JNK by ER stress is impaired in Tnfr1−/−
pro-apoptotic factors, such as Bax (Bcl2-associated X pro- cells. In addition, the expression of TNF␣ is up-regulated by
tein) and Bak (Bcl-2 homologous antagonist/killer), are present the IRE1 pathway during ER stress [83,84]. ROS can directly
at the mitochondrial and ER membranes [70,71]. During ER activate ASK1 by disrupting an ASK1 thioredoxin (TDX) com-
stress, BAX and BAK oligomerize to possibly permit Ca2+ plex through oxidation of TDX, and thereby lead to activation
efflux into the cytoplasm. Increased cytosolic Ca2+ can acti- of JNK, p38 MAP kinase, and cell death [85]. The Jun activa-
vate both mitochondrial- dependent and independent caspase tion domain-binding protein (JAB1) may be a feedback regulator
J.D. Malhotra, R.J. Kaufman / Seminars in Cell & Developmental Biology 18 (2007) 716–731 723

since it can interact with IRE1 and inhibit XBP1 mRNA splicing potential. In the presence of mitochondrial SOD, superoxide
and BiP transcription [86]. Thus, oxidative stress and ER stress is converted to hydrogen peroxide (H2 O2 ) that can diffuse out
may induce cell death by using the same molecular complex con- of mitochondria into the cytoplasm. In the presence of iron,
sisting of IRE1/TRAF2/ASK1/TDX/JNK. Finally, TNF␣ can hydrogen peroxide forms the highly reactive hydroxyl radical
activate the UPR in a ROS-dependent manner. These finding (OH• ) via the Fenton reaction. The superoxide anion radical
indicate an intricate relationship exists between death receptor (O2 •− ) also generates other toxic metabolites such as per-
signaling, oxidative stress, and activation of the UPR. oxynitrile (ONOO–), hypochlorous acid (HOCl) and singlet
oxygen (1 O2 ). Under physiological conditions ROS accumu-
4.4. ER stress-induced caspase-dependent apoptosis lation is guarded by numerous endogenous antioxidant defense
systems that include both enzymatic and non-enzymatic antiox-
Caspases 2, 3, 4, 7, 9 and 12 are reported to be involved in idant mechanisms that can either scavenge ROS or prevent their
ER stress-induced cell death [87–90]. Caspase-12 is associated formation. The enzymatic antioxidant defense mechanisms are
with the ER membrane, and activated by ER stress, possibly by mediated through superoxide dismutase (SOD), glutathione per-
calpain [91]. In addition, proapoptotic BCL2 family members oxidase (GPX), catalase, and thioredoxin reductase. Vitamins
BAX and BAK colocalize to the ER membrane and function provide a non-enzymatic antioxidant defense [100]. Finally,
to activate apoptosis through caspase 12 [87–90]. Caspase-12 redox homeostasis is contributed by several redox systems
activates caspase-9, which in turn activates caspase-3 [92] lead- including NAD+/NADH, NADP+/NADPH, and oxidized glu-
ing to cell death. Caspase-12−/− mice are partially resistant to tathione/reduced glutathione (GSSG/GSH).
ER stress-induced apoptosis but sensitive to other death stimuli, Both ER stress and oxidative stress, through ROS genera-
suggesting that caspase-12 is a regulator specific to ER stress- tion, can increase leak of Ca2+ from the ER lumen [101–103].
induced apoptosis [93]. However, the involvement of caspase-12 Increases in cytosolic Ca2+ can stimulate mitochondrial ROS
in apoptosis of human cells is still open to question, as the human production through multiple mechanisms. The mitochondrial
caspase-12 gene contains several inactivating mutations [94]. It electron transport chain generates ROS as a consequence of
is possible that caspase 4 mediates ER stress-induced apoptosis increased mitochondrial Ca2+ loading. The amount of mito-
in human cells [90,95]. chondrial ROS production primarily reflects the quantity of
the ubisemiquinone radical intermediate (QH• ), an intermedi-
5. ER stress and oxidative stress ate in the Q cycle at complex III [104,105]. The level of QH•
increases when either complex III is inhibited or with greater
ROS can be produced in all cellular compartments and flux through the respiratory chain. Ca2+ leak stimulates the TCA
ultimately results in protein damage [96]. Furthermore, the expo- cycle, thereby increasing O2 consumption and ROS generation.
sure of biological systems to various conditions of oxidative Ca2+ also stimulates nitric oxide synthase which generates NO•
stress leads to age-dependent increases in the cellular levels of that inhibits complex IV and can thereby enhance ROS produc-
oxidatively modified proteins, lipids and nucleic acids, and sub- tion. In addition, Ca2+ opens the permeability transition pore to
sequently predisposes to the development of well-recognized, release cytochrome c from the inner membrane thereby block-
age-related disorders that cause impaired cognitive function and ing the respiratory chain at complex III. Finally, Ca2+ -induced
metabolic integrity [97]. There is accumulating evidence to sug- permeability transition pore opening may cause leak of GSH
gest that protein folding, ER stress and production of ROS are from the matrix and as a consequence, deplete reducing equiv-
intimately intertwined, however, this area of ER stress is not alents.
well explored. Since oxidative protein folding occurs in the ER High levels of ROS generation within the mitochondria
and perturbations in protein folding can cause deleterious conse- further increases Ca2+ release from the ER. The very close prox-
quences, alterations in redox status or generation of ROS could imity of ER and mitochondria leads to accumulation of Ca2+
directly and/or indirectly affect ER homeostasis and protein near mitochondria, further increasing mitochondrial ROS pro-
folding. Elucidating the relationship between oxidative stress duction and leading to opening of the permeability transition
and oxidative protein folding represents a major area for future pore [106]. Furthermore, ROS can also feedback to sensitize the
research. Ca2+ release channels at the ER membrane [107,108]. For exam-
ple, this may occur through ROS or reactive nitrogen species that
5.1. Oxidative stress disrupts ER function can oxidize a critical thiol in the ryanodine receptor and cause
its inactivation, thereby enhancing Ca2+ release from the sar-
ROS can be produced both as a result of exposure to toxic coplasmic reticulum [109,110]. As the anti-oxidation potential
agents such as irradiation and environmental pollutants, and of the cell diminishes, the vicious cycle of Ca2+ release and ROS
also as byproducts of oxygen-utilizing enzymatic reactions, production becomes more threatening to cell survival.
such as the mitochondrial respiratory chain, the arachidonic
acid pathway, the cytochrome P450 family, glucose oxidase, 5.2. Protein folding in the ER causes oxidative stress
amino acid oxidases, xanthine oxidase, NADPH/NADPH oxi-
dases, or NO synthases [98,99] (Fig. 3A). The electron transport The ER is an organelle where proper protein folding and
chain produces membrane impermeable superoxide anion and disulfide formation of proteins is dependent on the redox sta-
the rate is dependent on the mitochondrial inner membrane tus within the lumen of the ER. In contrast to the cytosol
724 J.D. Malhotra, R.J. Kaufman / Seminars in Cell & Developmental Biology 18 (2007) 716–731

that has a reducing environment, the lumen of the ER is oxi- depletion of glutathione did not interfere with disulfide bond
dizing with a high ratio of oxidized to reduced glutathione formation [118,119].
(GSSG/GSH) [111]. Glutathione is a tripeptide (l-␥-glutamyl- Extensive genetic and biochemical studies using the yeast S.
l-cysteinyl-glycine) that is synthesized in the cytosol. The cell cerevisiae have provided detailed insights into the mechanisms
contains up to 10 mM GSH that is maintained in a reduced form underlying oxidative protein folding. A genetic screen in yeast
through a cytosolic NADPH-dependent reaction catalyzed by identified a conserved ER-membrane-associated protein Ero1p
glutathione reductase. Cellular redox homeostasis is maintained (ER oxidoreductin 1) [116,120] that plays a role similar to that
by a dynamic interaction between reduced glutathione (GSH) of the bacterial periplasmic protein DsbB in oxidative folding.
and protein thiols with ROS. Reduced glutathione GSH serves The proteins Ero1p and DsbB specifically oxidize a thioredoxin-
as a major thio-disulfide redox buffer in cells and the ratio of like protein (PDI in eukaryotes, DsbA in bacteria) that further
GSH:GSSG is used as an index of the redox state. While the serves as an intermediate in electron transfer. In both prokary-
ratio of reduced glutathione to oxidized glutathione is (>50:1) otes and eukaryotes molecular oxygen serves as the terminal
in the cytoplasm, this ratio is (1:1 to 3:1) in the ER lumen [112]. electron acceptor for disulfide bond formation. Ero1p uses a
The oxidizing environment of the ER lumen promotes disulfide flavin-dependent reaction to pass electrons directly to molecu-
bond formation. In addition, the greater oxidizing environment lar oxygen. This reaction has the potential to generate ROS that
of ER was suggested to contribute to the preferred oxidation would contribute to cellular oxidative stress. The role of Ero1p in
and inactivation of ER resident proteins, thereby contributing to electron transfer suggests that the activity of Ero1p is tightly cou-
unfolded protein accumulation [111]. pled with the protein-folding load in the ER [121]. In mammals,
Proteins that transit the secretory pathway frequently require there are two ERO1 genes hERO1-L␣ and hERO1-L␤ [122,123]
disulfide bond formation for their maturation, stability, and/or that differ in their tissue distribution and transcriptional regu-
function. Mispairing of cysteine residues and formation of inap- lation. Only ERO1-L␤ is induced by the UPR [123], whereas
propriate disulfide bonds prevents proteins from attaining their ERO1-L␣ is induced during hypoxia [124]. Further studies in
native conformation and leads to misfolding. Although it is yeast, and later in metazoan cells, identified a critical role for
likely that GSH reduces non-native disulfide bonds in mis- flavin adenine dinucleotide (FAD) in oxidative protein folding.
folded proteins, this is likely not the major pathway utilized As Ero1p is a novel FAD-binding protein [121], the FAD require-
in cells. The ER lumen maintains redox conditions that enable ment in oxidative folding may reflect its function in Ero1p. These
a distinct set of folding catalysts to facilitate the formation and studies suggest that the versatile redox molecule FAD functions
isomerization of disulfide bonds [112]. The process of disulfide in disulfide bond formation in the ER lumen.
bond-dependent protein folding is slow due to its dependence It has been estimated that approximately 25% of the ROS gen-
on a redox reaction, which requires an electron acceptor. During erated in a cell may result from formation of disulfide bonds in
this folding process a protein may be oxidized to form disul- the ER during oxidative protein folding (Fig. 3A). Two mech-
fide bonds, isomerized to allow polypeptide rearrangement, or anisms have been proposed for how disulfide bond formation
reduced to allow unfolding and subsequent degradation [113]. generates ROS. During formation of disulfide bonds, ROS are
The idea that disulfide bond formation is an assisted process a byproduct formed as ERO1 and PDI act in concert to trans-
in vivo is supported by the discovery of DsbA mutants in fer electrons from thiol groups in proteins to molecular oxygen.
Escherichia coli that display compromised disulfide bond for- In addition, ROS may be formed as a consequence of the glu-
mation [114]. tathione depletion that occurs as glutathione reduces unstable
In eukaryotes, oxidative protein folding, i.e., disulfide bond and improper disulfide bonds. The consumption of GSH would
formation, is catalyzed by a family of ER oxidoreductases, return thiols involved in non-native disulfide bonds to their
including PDI (protein disulphide isomerase), ERp57, ERp72, reduced form so they may again interact with ERO1/PDI1 to
PDIR, PDIp and P5. When chaperone assisted disulfide bond be reoxidized. This would generate a futile cycle of disulfide
formation occurs, cysteine residues within the PDI active site bond formation and breakage in which each cycle would gen-
[–C–X–X–C–] accept two electrons from the polypeptide chain erate ROS and consume GSH (Fig. 3B). As a consequence, it
substrate. This electron transfer results in the oxidation of the is expected that proteins that have multiple disulfide bonds may
substrate and the reduction of the PDI active site. Despite the be more prone to generating oxidative stress.
ability of PDI to enhance the rate of disulfide-linked folding, It is not known whether ROS are generated as a consequence
the mechanisms by which the ER disposes of electrons as a of unfolded protein accumulation in the ER. There several possi-
result of the oxidative disulfide bond formation has remained ble mechanisms by which unfolded protein in the ER could lead
an enigma. A number of different factors have been proposed to ROS production. First, as described above, protein misfold-
to maintain the oxidizing environment of the ER, including ing may be associated with inappropriate pairing and bonding
the preferential secretion of reduced thiols and uptake of oxi- of cysteine residues. A futile cycle of disulfide bond break-
dized thiols, and a variety of different redox enzymes and small age and formation would deplete GSH reducing equivalents,
molecule oxidants. However, there is a lack of genetic evidence thereby leading to oxidative stress. It order to test this hypoth-
demonstrating that these factors are physiologically important esis, it will be important to determine whether misfolding of a
[115–117]. It was believed for many years that the low molec- protein that has no disulfide bonds can generate ROS. Alterna-
ular mass thiol glutathione is responsible for oxidizing the PDI tively, unfolded protein accumulation in the ER may elicit Ca2+
active sites. This was contrary to observations in yeast where leak into the cytosol to increase ROS production in mitochon-
J.D. Malhotra, R.J. Kaufman / Seminars in Cell & Developmental Biology 18 (2007) 716–731 725

dria. Finally, since both protein folding and refolding in the ER although the precise relationship and mechanism(s) remain to
lumen are highly energy-dependent processes, ATP depletion as be elucidated.
a consequence of protein misfolding could stimulate mitochon- A unique requirement for UPR signaling in ␤ cell func-
drial oxidative phosphorylation to increase ATP production, and tion was first suggested by the identification of PERK as the
consequently increase ROS production. gene defective in the human disease Wolcott-Rallison syndrome
(WRS) [132]. Individuals with WRS and Perk−/− mice develop
6. ER stress and disease pathogenesis ␤ cell apoptosis with early onset insulin-dependent diabetes
[10]. In addition, mice with homozygous Ser51Ala mutation
The UPR has evolved as a series of signaling pathways to at the PERK phosphorylation site in eIF2␣ display even greater
ensure the rate of protein synthesis, the capacity for chaperone- ␤ cell loss that appears in utero [13]. Finally, although mice
assisted protein folding, and the ERAD potential are coupled with heterozygous Ser51Ala mutation in eIF2␣ do not display a
with environmental, genetic, and nutritional influences to pre- detectable phenotype, upon feeding a high-fat diet, they develop
vent the accumulation of unfolded protein in the ER lumen. insulin resistance and a failure in the ␤ cells to produce insulin,
Increasing evidence suggests that protein misfolding in the typical of type 2 diabetes. The insulin secretion defect in the
ER lumen and alterations in UPR signaling play important high-fat-fed heterozygous Ser51Ala eIF2␣ mutant mice was due
roles in the etiology of numerous disease states, including to an increased rate of glucose-stimulated proinsulin translation
metabolic disease, atherosclerosis, and neurodegenerative dis- which overwhelmed the protein folding machinery of the ER and
ease [1,125]. led to (1) a distended ER compartment, (2) prolonged association
of proinsulin with the ER chaperone BiP, (3) reduced process-
6.1. ER stress contributes to metabolic disease ing of proinsulin to insulin, and (4) reduced granule biogenesis
[133]. Thus, regulation of translation initiation through eIF2␣
The development of type 2 diabetes is associated with a phosphorylation is required for ER stress signaling to prevent ␤
combination of insulin resistance in fat, muscle, and liver and cell dysfunction when the demand for insulin is increased due to
a failure of pancreatic ␤-cells to adequately compensate to a highfat diet and insulin resistance. These findings indicate that
increase insulin production [126,127]. Insulin signaling is very ␤ cells display a unique requirement for PERK/eIF2␣-regulated
sensitive to alterations in ER homeostasis and redox status. ER translation.
stress and oxidative stress, as well as inflammatory cytokines There are several mechanisms that may explain why ␤
and free fatty acids, inhibit insulin signaling through activation cells uniquely require the PERK/eIF2␣ pathway. First, ␤ cells
of the protein kinase JNK. JNK phosphorylation of IRS-1 on may require PERK/eIF2␣ signaling because they are sensi-
Ser307 reduces insulin receptor-stimulated Tyr phosphorylation tive to physiological fluctuations in blood glucose. In ␤ cells,
and insulin signaling [128]. Induction of ER stress may suppress the generation of ATP fluctuates with blood glucose concen-
insulin receptor signaling via IRE1␣-dependent activation of the trations because glycolysis is controlled by glucokinase that
JNK pathway. Indeed, suppressing the JNK pathway can ame- has a low affinity for glucose. Periodic decreases blood glu-
liorate insulin resistance [129], possibly by counterbalancing cose levels reduce the ATP/ADP ratio and would compromise
the deleterious effects of ER stress, oxidative stress, free fatty protein folding in the ER so that UPR may be frequently acti-
acids, and proinflammatory cytokines. The role of ER stress in vated. Through this mechanism, PERK/eIF2␣ signaling would
insulin signaling was also suggested by the finding that ectopic be required in ␤ cells to couple protein synthesis with energy
expression of the molecular chaperone ORP150/GRP170 in hep- available for protein folding reactions in the ER lumen. Alterna-
atocytes improved insulin sensitivity [130]. It is possible that tively, glucose stimulates insulin transcription, translation and
elevated levels of ORP150 expression improve the protein fold- secretion. PERK phosphorylation of eIF2␣ may be required for
ing capacity of the ER and reduce UPR signaling. The ability ␤ cells to attenuate protein synthesis so that insulin produc-
for ER stress signaling to cause insulin resistance was also sug- tion does not exceed the protein folding capacity of the ER.
gested by recent observations showing that treatment of mice Results from the high-fat-fed heterozygous Ser51Ala eIF2␣
with chemical chaperones that can improve protein folding in mutant mice would support this hypothesis [133]. Finally, as
the ER and reduce ER stress and UPR signaling, can increase the PERK/eIF2␣ pathway is known to reduce oxidative stress,
insulin sensitivity [35]. Alternatively, there are observations that it is possible that ␤ cells require PERK/eIF2␣ to minimize
suggest ER stress and UPR signaling through IRE1 can actually oxidative stress [134]. Two mechanisms have been proposed
improve insulin sensitivity. Heterozygous Xbp1+/− mice devel- to account for the role of the PERK/eIF2␣ in limiting oxidative
oped insulin resistance compared to control mice when fed a stress. First, the PERK/eIF2␣ pathway can prevent oxidative
high-fat diet [131]. It is possible that reduced XBP1 signaling stress through inhibition of translation initiation when pro-
impairs the ER protein folding capacity, thereby activating the tein folding in the ER lumen is disturbed [63]. Alternatively,
UPR, which may lead to JNK activation. Therefore, ER stress the PERK/eIF2␣/ATF4 pathway induces expression of antiox-
signaling through IRE1-mediated XBP1 mRNA splicing may idative stress response genes [10,63]. It is likely that both
increase the ER protein folding capacity to improve insulin mechanisms contribute to the protective role for PERK/eIF2␣
signaling, whereas IRE1-mediated JNK activation could cause in limiting ROS accumulation. There is increasing evidence
insulin resistance. The sum of these observations indicates that that suggests oxidative stress contributes to the ␤ cell failure
there exists a link between insulin resistance and ER stress, in diabetes [128,135]. ␤ cells express low levels of catalase
726 J.D. Malhotra, R.J. Kaufman / Seminars in Cell & Developmental Biology 18 (2007) 716–731

and glutathione peroxidase, two enzymes that protect from expression of ER chaperones such as PDI is up-regulated in
ROS [136]. Therefore, oxidative stress would preferentially the brain of PD patients, and PDI is accumulated in Lewy
perturb ␤ cell function due to their reduced capacity to neu- bodies [148]. The identification of PDI family member PDIp
tralize ROS. Further studies are required to elucidate why the in experimental Parkinson’s disease and Lewy bodies suggest
PERK/eIF2␣ pathway is essential for ␤ cell function and sur- that oxidative protein folding in the ER may be perturbed in
vival. PD.
In humans, mutations in SIL1, which encodes an adenine
6.2. ER stress contributes to neurodegenerative disease nucleotide exchange factor for BiP cause Marinesco-Sjögren
syndrome, a rare disease associated with cerebellar ataxia, pro-
Neurodegenerative diseases such as Alzheimer’s disease gressive myopathy and cataracts [149]. In mice homozygous for
(AD) and Parkinson’s disease (PD), represent a large class a spontaneously occurring mutation in the Sil1 transcript, cere-
of conformational diseases associated with accumulation of bellar Purkinje cell degeneration and subsequent ataxia occurs
abnormal protein aggregates in and around affected neurons. [150]. Analysis of Sil1 mutant mice demonstrated that affected
Oxidative stress and protein misfolding play critical roles in Purkinje cells have ubiquitinated nuclear- and ER-associated
the pathogenesis of these neurodegenerative diseases [137] that protein aggregates and also display upregulation of several ER
are characterized by fibrillar aggregates composed of misfolded stress markers BiP, CHOP, and ORP150 [150]. It seems likely
proteins [138]. At the cellular level, neuronal death or apoptosis that the protein chaperone BiP uses ATP/ADP exchange that
may be mediated by oxidative stress and/or ER stress. Upregula- is essential to preserve ER function and prevent activation of
tion of ER stress markers has been demonstrated in postmortem the UPR. A reduced efficiency of ATP-dependent BiP-mediated
brain tissues and cell culture models of many neurodegenerative chaperone function may predispose to unfolded protein accu-
disorders, including PD, AD, amyotropic lateral sclerosis (ALS) mulation in the ER, activate the UPR, and contribute to Purkinje
and expanded polyglutamine diseases such as Huntingtons’s dis- cell degeneration.
ease and spinocerebellar ataxias [139]. Recent studies indicate Oxidative stress is implicated in the pathogenesis of neu-
that oligomeric forms of polypeptides predisposed to ␤ sheet rodegenerative diseases. A group of neurodegenerative diseases
polymerization and fibril formation may be the toxic forms that including Alzheimer’s disease is characterized pathologically by
cause neuronal death. The impact of these oligomeric, poten- the deposition of intracellular aggregates containing abnormally
tially toxic species on ER function and generation of ROS is phosphorylated forms of the microtubule-binding protein Tau
presently not understood. [151]. Using a Drosophila model relevant to human neurodegen-
In vitro studies suggest these aggregates can inhibit the erative diseases, including Alzheimer’s disease, it was demon-
proteasome and ERAD. For example, in the Machado-Joseph strated that oxidative stress plays a casual role in neurotoxicity
syndrome, the polyglutamine repeats present in spinocerebrocel- and promotes Tau-phoshorylation. In this model, activation of
lular atrophy protein (SCA3) form cytosolic aggregates that can the JNK pathway correlated with the degree of tau-induced neu-
inhibit the proteasome. Proteasome inhibition in the cytosol can rodegeneration [152]. Although oxidative stress and ER stress
interfere with ERAD to elicit UPR activation, caspase 12 activa- have been linked to neurodegenerative diseases, at this point it is
tion, and apoptosis [79,140]. Deletion of the ER stress-induced not possible to conclude these processes are the primary cause of
pro-apoptotic transcription factor CHOP preserved neuronal neuron death. However, it is possible that these stresses modify
function, suggesting the importance of UPR signaling in this the progression and severity of these complex diseases.
model. Nitric oxide (NO) is a second messenger for signaling path-
PD is the second most common neurodegenerative disease ways that regulate a variety of physiological processes. In the
and is characterized by a loss of dopaminergic neurons. Analyses brain, NO is implicated in neurotransmission, neuromodula-
of familial PD revealed involvement of three genes encod- tion, and synaptic plasticity. However, excessive generation of
ing ␣-synuclein, Parkin, and ubiquitin C-terminal esterase L1 NO and NO-derived reactive nitrogen species are implicated in
(UCH-L1). ␣-synuclein is a cytoplasmic protein that forms the pathogenesis of neurodegenerative disorders, including AD
aggregates, called Lewy bodies which are characteristic of PD. and PD [153]. Studies now indicate ER stress and apoptosis are
Athough the link between ␣-synuclein and ER stress is unclear, critical features underlying these disorders [154]. Uehara et al.
Parkin is a ubiquitin-protein ligase (E3) involved in ERAD [141]. [155] elegantly demonstrated that NO-mediated S-nitrosylation
One of the substrates of ERAD ubiquitinated by Parkin is the of protein disulfide isomerase (PDI) inhibits PDI function, leads
Pael receptor, a homolog of endothelin receptor type B [142]. to dysregulated protein folding within the ER, elicits ER stress,
Interestingly, expression of Parkin is induced by ER stress, and and initiates neuronal cell death. A causal role for this sequence
neuronal cells overexpressing Parkin are resistant to ER stress of events in neurodegenerative disease was supported by the
[143]. UCH-L1 is an abundant protein in neurons and stabilizes demonstration that PDI is S-nitrosylated in the brains of patients
a monomeric ubiquitin to ubiquitinate unfolded proteins and suffering from PD or AD, but not in normal brains. Thus, these
might be involved in ERAD [144–146]. These findings strongly findings provide additional evidence of a role for dysregulated
suggest the involvement of ER stress in PD. In addition, there protein S-nitrosylation (oxidative stress) in neurodegenerative
are several additional reports supporting the link between ER disease and indicate that ER dysfunction may serve as a criti-
stress with PD. First, PD mimetics, such as 6-hydroxydopamine, cal common factor that couples NO-induced cellular stress to
specifically induce ER stress in neuronal cells [147]. Second, neurodegeneration.
J.D. Malhotra, R.J. Kaufman / Seminars in Cell & Developmental Biology 18 (2007) 716–731 727

6.3. ER stress contributes to hyperhomocysteinemia and take up oxidized lipoprotein particles and become laden with
atherosclerosis cholesterol. The cholesterol is stored as esters within large lipid
vesicles producing a foamy appearance, and hence their name.
Elevated plasma levels of homocysteine (Hcy), a sulfur- Overload of cholesterol in macrophages elicits apoptosis. Excess
containing amino acid, are linked to the development of ischemic cholesterol must accumulate in specific pools within the cell to
heart disease, stroke and peripheral vascular disease are asso- elicit cytotoxicity. Intracellular cholesterol is known to traffic to
ciated with elevated plasma levels of homocysteine (Hcy). the plasma membrane, mitochondria and the ER. Although the
However, it is not known whether Hcy is a primary cause of ER membrane has low levels of free cholesterol, it is particularly
atherosclerosis and thromobosis. Hcy may mediate vascular sensitive to cholesterol loading. Recent findings suggest that free
toxicity through dysregulation of cholesterol and triglyceride cholesterol requires trafficking to the ER to produce its toxic
biosynthesis. Hyperhomocysteinemia activates lipogenic signal- effects [165]. This trafficking results in activation of UPR sig-
ing via the sterol regulated element-binding proteins (SREBPs), naling and caspase activation, and ultimately in macrophage cell
leading to intracellular accumulation of cholesterol [156]. Sur- death/apoptosis. Macrophages from Perk−/− mice are hypersen-
prisingly, ER stress appears to contribute to the activation of sitive to cholesterol-induced cell death whereas macrophages
SREBP by homocysteine. Livers of homocysteine-fed mice from Chop−/− mice are highly protected. Recent findings
contain elevated levels of ER chaperones. In addition, over- also suggest that defective insulin signaling and reduced AKT
expression of BiP prevented SREBP induction in response to activity impair the ability of macrophages to deal with ER stress-
homocysteine [156]. induced apoptosis within atherosclerotic plaques [166]. This
Under normal circumstances, Hcy is converted to cysteine mechanism may contribute to the association between insulin
and partly remethylated to methionine by vitamin B12 and resistance in metabolic syndrome and atherogenesis [167].
folate. When normal metabolism is disturbed due to deficiency These findings suggest that the UPR plays an important role in
of cystathionine-␤ synthase (CBS) that requires vitamin B6 for the progression of the atherogenic disease process [165]. In addi-
activation, Hcy accumulates in blood and results in severe hyper- tion, free cholesterol loading of macrophages increases levels of
homocysteinemia. CBS condenses homocysteine and serine cell-surface Fas ligand, activated proapoptotic Bax protein, and
to form cystathionine. The harmful effects of hyperhomocys- increases mitochondrial-dependent apoptosis. Though both the
teinemia may be mediated through several processes. First, a Fas death pathway and the mitochondrial cell death pathway may
decrease in cysteine may cause disease, due to reduced synthe- contribute to macrophage apoptosis, there is accumulating evi-
sis of glutathione (antioxidant). Thrombotic and cardiovascular dence suggesting that depletion of calcium stores in the ER and
diseases may also be encountered. Second, ROS generated subsequent activation of the UPR is the dominant driving force
during oxidation of Hcy to homocystine and disulfides may in cholesterol-induced macrophage death. Finally, ER stress
oxidize membrane lipids and proteins. Third, Hcy can react caused by free cholesterol loading in macrophages promotes
with thiols within proteins and form disulfides (thiolation) to chemokine secretion, and this may contribute to the formation
interfere with protein folding, structure, and function. Finally, of vulnerable atherosclerotic lesions. These lesions lead to an
Hcy can be converted to highly reactive thiolactone that can inflammatory condition with further infiltration of macrophages
react with proteins forming –NH–CO– adducts, thus affecting and lymphocytes from the blood and subsequent release of
protein structure and function. In cultured vascular endothelial hydrolytic enzymes, cytokines, chemokines and growth factors
cells, Hcy induces protein misfolding in the ER by interfering that can inflict more damage and eventually lead to focal necrosis
with disulfide bond formation [157] and activates the UPR to [168,169].
induce expression of several ER stress response proteins, such
as BiP, GRP94, CHOP and HERP [158–161]. Hcy can also trig- 7. Future perspectives
ger apoptosis by a signaling pathway that requires intact IRE1
[160]. These studies support the notion that Hcy can disrupt ER Tremendous progress has been made in understanding the
homoeostasis to cause UPR induction [158–160]. This is con- mechanisms underlying the cause of ER stress and cellular adap-
sistent with the observed activation of UPR markers in livers of tive responses. Future studies are required to understand the
normal or Cbs+/− mice in response to hyperhomocysteinemia physiological significance of ER stress and UPR signaling in
[162]. disease pathogenesis. The relationships between ER stress and
Atherosclerosis is caused by the abnormal deposition of apoptosis also remain to be defined. Further studies are also
cholesterol in the coronary arteries. Cholesterol accumula- required to elucidate how ER stress and UPR signaling in inte-
tion in macrophages plays a critical role in the progression grated with other stress signaling pathways, particularly those
of atherosclerosis. Macrophages have multiple mechanisms to related to oxidative stress. A greater understanding of the com-
prevent excess cholesterol accumulation including an increase plex interrelationship between protein misfolding and oxidative
in cholesterol esterification, induction of cellular cholesterol stress may lead to the development of general pharmacolog-
efflux, and the repression of lipoprotein receptor and choles- ical agents, such as chemical chaperones to improve protein
terol biosynthetic enzymes [163,164]. Upon formation of an folding and/or antioxidants to reduce oxidative stress, for the
initial atherosclerotic lesion these mechanisms are dysregulated, treatment of human disease. A coherent mechanistic under-
thereby leading to the characteristic appearance of foam cells standing of the mechanisms and pathways that signal ER stress
within the vessel intima. The macrophage-derived foam cells responses should contribute to the development of more selec-
728 J.D. Malhotra, R.J. Kaufman / Seminars in Cell & Developmental Biology 18 (2007) 716–731

tive and specific-acting therapeutic agents targeted for diseases [21] Wang W, Chan JY. Nrf1 is targeted to the endoplasmic reticulum mem-
associated with ER pathologies. brane by an N-terminal transmembrane domain. Inhibition of nuclear
translocation and transacting function. J Biol Chem 2006;281:19676–87.
[22] Cox JS, Walter P. A novel mechanism for regulating activity of a
Acknowledgments transcription factor that controls the unfolded protein response. Cell
1996;87:391–404.
[23] Mori K, Ogawa N, Kawahara T, Yanagi H, Yura T. mRNA splicing-
R.J. Kaufman is an Investigator of the Howard Hughes Med- mediated C-terminal replacement of transcription factor Hac1p is required
ical Institute and is supported by NIH grants RO1 DK042394 for efficient activation of the unfolded protein response. Proc Natl Acad
and R01 HL052173. Sci USA 2000;97:4660–5.
[24] Travers KJ, Patil CK, Wodicka L, Lockhart DJ, Weissman JS, Wal-
ter P. Functional and genomic analyses reveal an essential coordination
References between the unfolded protein response and ER-associated degradation.
Cell 2000;101:249–58.
[1] Kaufman RJ. Orchestrating the unfolded protein response in health and [25] Tirasophon W, Welihinda AA, Kaufman RJ. A stress response pathway
disease. J Clin Invest 2002;110:1389–98. from the endoplasmic reticulum to the nucleus requires a novel bifunc-
[2] Patil C, Walter P. Intracellular signaling from the endoplasmic reticulum tional protein kinase/endoribonuclease (Ire1p) in mammalian cells. Genes
to the nucleus: the unfolded protein response in yeast and mammals. Curr Dev 1998;12:1812–24.
Opin Cell Biol 2001;13:349–55. [26] Wang XZ, Harding HP, Zhang Y, Jolicoeur EM, Kuroda M, Ron D.
[3] Mori K, Ma W, Gething MJ, Sambrook J. A transmembrane protein with Cloning of mammalian Ire1 reveals diversity in the ER stress responses.
a cdc2+/CDC28-related kinase activity is required for signaling from the Embo J 1998;17:5708–17.
ER to the nucleus. Cell 1993;74:743–56. [27] Niwa M, Sidrauski C, Kaufman RJ, Walter P. A role for presenilin-1 in
[4] Harding HP, Calfon M, Urano F, Novoa I, Ron D. Transcriptional and nuclear accumulation of Ire1 fragments and induction of the mammalian
translational control in the mammalian unfolded protein response. Annu unfolded protein response. Cell 1999;99:691–702.
Rev Cell Dev Biol 2002;18:575–99. [28] Yoshida H, Haze K, Yanagi H, Yura T, Mori K. Identification of the
[5] Schroder M, Kaufman RJ. The mammalian unfolded protein response. cis-acting endoplasmic reticulum stress response element responsible
Annu Rev Biochem 2005;74:739–89. for transcriptional induction of mammalian glucose-regulated proteins.
[6] Wu J, Kaufman RJ. From acute ER stress to physiological roles of the Involvement of basic leucine zipper transcription factors. J Biol Chem
unfolded protein response. Cell Death Differ 2006;13:374–84. 1998;273:33741–9.
[7] Molinari M. N-glycan structure dictates extension of protein folding or [29] Calfon M, Zeng H, Urano F, Till JH, Hubbard SR, Harding HP, et al. IRE1
onset of disposal. Nat Chem Biol 2007;3:313–20. couples endoplasmic reticulum load to secretory capacity by processing
[8] Sidrauski C, Walter P. The transmembrane kinase Ire1p is a site-specific the XBP-1 mRNA. Nature 2002;415:92–6.
endonuclease that initiates mRNA splicing in the unfolded protein [30] Lee K, Tirasophon W, Shen X, Michalak M, Prywes R, Okada T, et
response. Cell 1997;90:1031–9. al. IRE1-mediated unconventional mRNA splicing and S2P-mediated
[9] Yoshida H, Matsui T, Yamamoto A, Okada T, Mori K. XBP1 mRNA is ATF6 cleavage merge to regulate XBP1 in signaling the unfolded protein
induced by ATF6 and spliced by IRE1 in response to ER stress to produce response. Genes Dev 2002;16:452–66.
a highly active transcription factor. Cell 2001;107:881–91. [31] Shen X, Ellis RE, Lee K, Liu CY, Yang K, Solomon A, et al. Comple-
[10] Harding HP, Zhang Y, Bertolotti A, Zeng H, Ron D. Perk is essential mentary signaling pathways regulate the unfolded protein response and
for translational regulation and cell survival during the unfolded protein are required for C. elegans development. Cell 2001;107:893–903.
response. Mol Cell 2000;5:897–904. [32] Yoshida H, Matsui T, Hosokawa N, Kaufman RJ, Nagata K, Mori K. A
[11] Yoshida H, Okada T, Haze K, Yanagi H, Yura T, Negishi M, et al. ATF6 time-dependent phase shift in the mammalian unfolded protein response.
activated by proteolysis binds in the presence of NF-Y (CBF) directly to Dev Cell 2003;4:265–71.
the cis-acting element responsible for the mammalian unfolded protein [33] Reimold AM, Etkin A, Clauss I, Perkins A, Friend DS, Zhang J, et al. An
response. Mol Cell Biol 2000;20:6755–67. essential role in liver development for transcription factor XBP-1. Genes
[12] Kaufman RJ. Regulation of mRNA translation by protein folding in the Dev 2000;14:152–7.
endoplasmic reticulum. Trends Biochem Sci 2004;29:152–8. [34] Bertolotti A, Wang X, Novoa I, Jungreis R, Schlessinger K, Cho JH, et
[13] Scheuner D, Song B, McEwen E, Liu C, Laybutt R, Gillespie P, et al. al. Increased sensitivity to dextran sodium sulfate colitis in IRE1beta-
Translational control is required for the unfolded protein response and in deficient mice. J Clin Invest 2001;107:585–93.
vivo glucose homeostasis. Mol Cell 2001;7:1165–76. [35] Ozcan U, Cao Q, Yilmaz E, Lee AH, Iwakoshi NN, Ozdelen E, et al.
[14] Harding HP, Zhang Y, Zeng H, Novoa I, Lu PD, Calfon M, et al. An Endoplasmic reticulum stress links obesity, insulin action, and type 2
integrated stress response regulates amino acid metabolism and resistance diabetes. Science 2004;306:457–61.
to oxidative stress. Mol Cell 2003;11:619–33. [36] Reimold AM, Iwakoshi NN, Manis J, Vallabhajosyula P, Szomolanyi-
[15] Harding HP, Zhang Y, Ron D. Protein translation and folding are coupled Tsuda E, Gravallese EM, et al. Plasma cell differentiation requires the
by an endoplasmic-reticulum-resident kinase. Nature 1999;397:271–4. transcription factor XBP-1. Nature 2001;412:300–7.
[16] Harding HP, Novoa I, Zhang Y, Zeng H, Wek R, Schapira M, et al. Reg- [37] Iwakoshi NN, Lee AH, Vallabhajosyula P, Otipoby KL, Rajewsky
ulated translation initiation controls stress-induced gene expression in K, Glimcher LH. Plasma cell differentiation and the unfolded pro-
mammalian cells. Mol Cell 2000;6:1099–108. tein response intersect at the transcription factor XBP-1. Nat Immunol
[17] Ron D. Translational control in the endoplasmic reticulum stress response. 2003;4:321–9.
J Clin Invest 2002;110:1383–8. [38] Zhang K, Wong HN, Song B, Miller CN, Scheuner D, Kaufman RJ. The
[18] Ma Y, Brewer JW, Diehl JA, Hendershot LM. Two distinct stress signal- unfolded protein response sensor IRE1alpha is required at 2 distinct steps
ing pathways converge upon the CHOP promoter during the mammalian in B cell lymphopoiesis. J Clin Invest 2005;115:268–81.
unfolded protein response. J Mol Biol 2002;318:1351–65. [39] Lee AH, Chu GC, Iwakoshi NN, Glimcher LH. XBP-1 is required for
[19] Cullinan SB, Zhang D, Hannink M, Arvisais E, Kaufman RJ, Diehl JA. biogenesis of cellular secretory machinery of exocrine glands. Embo J
Nrf2 is a direct PERK substrate and effector of PERK-dependent cell 2005;24:4368–80.
survival. Mol Cell Biol 2003;23:7198–209. [40] Haze K, Yoshida H, Yanagi H, Yura T, Mori K. Mammalian transcription
[20] Nguyen T, Sherratt PJ, Pickett CB. Regulatory mechanisms controlling factor ATF6 is synthesized as a transmembrane protein and activated by
gene expression mediated by the antioxidant response element. Annu Rev proteolysis in response to endoplasmic reticulum stress. Mol Biol Cell
Pharmacol Toxicol 2003;43:233–60. 1999;10:3787–99.
J.D. Malhotra, R.J. Kaufman / Seminars in Cell & Developmental Biology 18 (2007) 716–731 729

[41] Yoshida H, Okada T, Haze K, Yanagi H, Yura T, Negishi M, et al. [60] Ron D, Habener JF. CHOP, a novel developmentally regulated nuclear
Endoplasmic reticulum stress-induced formation of transcription factor protein that dimerizes with transcription factors C/EBP and LAP and
complex ERSF including NF-Y (CBF) and activating transcription factors functions as a dominant-negative inhibitor of gene transcription. Genes
6alpha and 6beta that activates the mammalian unfolded protein response. Dev 1992;6:439–53.
Mol Cell Biol 2001;21:1239–48. [61] Zinszner H, Kuroda M, Wang X, Batchvarova N, Lightfoot RT,
[42] Li M, Baumeister P, Roy B, Phan T, Foti D, Luo S, et al. ATF6 as a Remotti H, et al. CHOP is implicated in programmed cell death in
transcription activator of the endoplasmic reticulum stress element: thap- response to impaired function of the endoplasmic reticulum. Genes Dev
sigargin stress-induced changes and synergistic interactions with NF-Y 1998;12:982–95.
and YY1. Mol Cell Biol 2000;20:5096–106. [62] Novoa I, Zeng H, Harding HP, Ron D. Feedback inhibition of the unfolded
[43] Ye J, Rawson RB, Komuro R, Chen X, Dave UP, Prywes R, et al. ER protein response by GADD34-mediated dephosphorylation of eIF2alpha.
stress induces cleavage of membrane-bound ATF6 by the same proteases J Cell Biol 2001;153:1011–22.
that process SREBPs. Mol Cell 2000;6:1355–64. [63] Marciniak SJ, Yun CY, Oyadomari S, Novoa I, Zhang Y, Jungreis R, et
[44] Goldstein JL, DeBose-Boyd RA, Brown MS. Protein sensors for mem- al. CHOP induces death by promoting protein synthesis and oxidation in
brane sterols. Cell 2006;124:35–46. the stressed endoplasmic reticulum. Genes Dev 2004;18:3066–77.
[45] Okada T, Yoshida H, Akazawa R, Negishi M, Mori K. Distinct roles [64] Yamaguchi H, Wang HG. CHOP is involved in endoplasmic reticulum
of activating transcription factor 6 (ATF6) and double-stranded RNA- stress-induced apoptosis by enhancing DR5 expression in human carci-
activated protein kinase-like endoplasmic reticulum kinase (PERK) in noma cells. J Biol Chem 2004;279:45495–502.
transcription during the mammalian unfolded protein response. Biochem [65] Ohoka N, Yoshii S, Hattori T, Onozaki K, Hayashi H. TRB3, a novel
J 2002;366:585–94. ER stress-inducible gene, is induced via ATF4-CHOP pathway and is
[46] Zhang K, Shen X, Wu J, Sakaki K, Saunders T, Rutkowski DT, et al. involved in cell death. Embo J 2005;24:1243–55.
Endoplasmic reticulum stress activates cleavage of CREBH to induce a [66] Sok J, Wang XZ, Batchvarova N, Kuroda M, Harding H, Ron D.
systemic inflammatory response. Cell 2006;124:587–99. CHOP-dependent stress-inducible expression of a novel form of carbonic
[47] Kondo S, Murakami T, Tatsumi K, Ogata M, Kanemoto S, Otori K, et anhydrase VI. Mol Cell Biol 1999;19:495–504.
al. OASIS, a CREB/ATF-family member, modulates UPR signalling in [67] McCullough KD, Martindale JL, Klotz LO, Aw TY, Holbrook NJ.
astrocytes. Nat Cell Biol 2005;7:186–94. Gadd153 sensitizes cells to endoplasmic reticulum stress by down-
[48] Kondo S, Saito A, Hino S, Murakami T, Ogata M, Kanemoto S, et regulating Bcl2 and perturbing the cellular redox state. Mol Cell Biol
al. BBF2H7, a novel transmembrane bZIP transcription factor, is a 2001;21:1249–59.
new type of endoplasmic reticulum stress transducer. Mol Cell Biol [68] Rutkowski DT, Arnold SM, Miller CN, Wu J, Li J, Gunnison KM, et
2007;27:1716–29. al. Adaptation to ER stress is mediated by differential stabilities of pro-
[49] Nagamori I, Yabuta N, Fujii T, Tanaka H, Yomogida K, Nishimune Y, survival and pro-apoptotic mRNAs and proteins. PLoS Biol 2006;4:e374.
et al. Tisp40, a spermatid specific bZip transcription factor, functions by [69] Morishima N, Nakanishi K, Tsuchiya K, Shibata T, Seiwa E. Translo-
binding to the unfolded protein response element via the Rip pathway. cation of Bim to the endoplasmic reticulum (ER) mediates ER stress
Genes Cells 2005;10:575–94. signaling for activation of caspase-12 during ER stress-induced apoptosis.
[50] Liang G, Audas TE, Li Y, Cockram GP, Dean JD, Martyn AC, et al. J Biol Chem 2004;279:50375–81.
Luman/CREB3 induces transcription of the endoplasmic reticulum (ER) [70] Scorrano L, Oakes SA, Opferman JT, Cheng EH, Sorcinelli MD, Pozzan
stress response protein Herp through an ER stress response element. Mol T, et al. BAX and BAK regulation of endoplasmic reticulum Ca2+ : a
Cell Biol 2006;26:7999–8010. control point for apoptosis. Science 2003;300:135–9.
[51] Raggo C, Rapin N, Stirling J, Gobeil P, Smith-Windsor E, O’Hare P, [71] Zong WX, Li C, Hatzivassiliou G, Lindsten T, Yu QC, Yuan J, et al. Bax
et al. Luman, the cellular counterpart of herpes simplex virus VP16, and Bak can localize to the endoplasmic reticulum to initiate apoptosis.
is processed by regulated intramembrane proteolysis. Mol Cell Biol J Cell Biol 2003;162:59–69.
2002;22:5639–49. [72] Nakagawa T, Yuan J. Cross-talk between two cysteine protease fam-
[52] Stirling J, O’Hare P. CREB4, a transmembrane bZip transcription factor ilies. Activation of caspase-12 by calpain in apoptosis. J Cell Biol
and potential new substrate for regulation and cleavage by S1P. Mol Biol 2000;150:887–94.
Cell 2006;17:413–26. [73] Zong WX, Lindsten T, Ross AJ, MacGregor GR, Thompson CB. BH3-
[53] Bertolotti A, Zhang Y, Hendershot LM, Harding HP, Ron D. Dynamic only proteins that bind pro-survival Bcl-2 family members fail to induce
interaction of BiP and ER stress transducers in the unfolded-protein apoptosis in the absence of Bax and Bak. Genes Dev 2001;15:1481–6.
response. Nat Cell Biol 2000;2:326–32. [74] Li J, Lee B, Lee AS. Endoplasmic reticulum stress-induced apoptosis:
[54] Morris JA, Dorner AJ, Edwards CA, Hendershot LM, Kaufman RJ. multiple pathways and activation of p53-up-regulated modulator of apop-
Immunoglobulin binding protein (BiP) function is required to protect cells tosis (PUMA) and NOXA by p53. J Biol Chem 2006;281:7260–70.
from endoplasmic reticulum stress but is not required for the secretion of [75] Chae HJ, Kim HR, Xu C, Bailly-Maitre B, Krajewska M, Krajewski S, et
selective proteins. J Biol Chem 1997;272:4327–34. al. BI-1 regulates an apoptosis pathway linked to endoplasmic reticulum
[55] Ng DT, Watowich SS, Lamb RA. Analysis in vivo of GRP78- stress. Mol Cell 2004;15:355–66.
BiP/substrate interactions and their role in induction of the GRP78-BiP [76] Mathai JP, Germain M, Shore GC. BH3-only BIK regulates BAX,
gene. Mol Biol Cell 1992;3:143–55. BAK-dependent release of Ca2+ from endoplasmic reticulum stores and
[56] Graham KS, Le A, Sifers RN. Accumulation of the insoluble PiZ variant mitochondrial apoptosis during stress-induced cell death. J Biol Chem
of human alpha 1-antitrypsin within the hepatic endoplasmic reticu- 2005;280:23829–36.
lum does not elevate the steady-state level of grp78/BiP. J Biol Chem [77] Hetz C, Bernasconi P, Fisher J, Lee AH, Bassik MC, Antonsson B, et al.
1990;265:20463–8. Proapoptotic BAX and BAK modulate the unfolded protein response by
[57] Shen J, Snapp EL, Lippincott-Schwartz J, Prywes R. Stable binding of a direct interaction with IRE1alpha. Science 2006;312:572–6.
ATF6 to BiP in the endoplasmic reticulum stress response. Mol Cell Biol [78] Urano F, Wang X, Bertolotti A, Zhang Y, Chung P, Harding HP, et al.
2005;25:921–32. Coupling of stress in the ER to activation of JNK protein kinases by
[58] Credle JJ, Finer-Moore JS, Papa FR, Stroud RM, Walter P. On the mecha- transmembrane protein kinase IRE1. Science 2000;287:664–6.
nism of sensing unfolded protein in the endoplasmic reticulum. Proc Natl [79] Nishitoh H, Matsuzawa A, Tobiume K, Saegusa K, Takeda K, Inoue K,
Acad Sci USA 2005;102:18773–84. et al. ASK1 is essential for endoplasmic reticulum stress-induced neu-
[59] Zhou J, Liu CY, Back SH, Clark RL, Peisach D, Xu Z, et al. The crystal ronal cell death triggered by expanded polyglutamine repeats. Genes Dev
structure of human IRE1 luminal domain reveals a conserved dimerization 2002;16:1345–55.
interface required for activation of the unfolded protein response. Proc [80] Mauro C, Crescenzi E, De Mattia R, Pacifico F, Mellone S, Salzano S,
Natl Acad Sci USA 2006;103:14343–8. et al. Central role of the scaffold protein tumor necrosis factor receptor-
730 J.D. Malhotra, R.J. Kaufman / Seminars in Cell & Developmental Biology 18 (2007) 716–731

associated factor 2 in regulating endoplasmic reticulum stress-induced [102] Berridge MJ, Bootman MD, Roderick HL. Calcium signalling: dynamics,
apoptosis. J Biol Chem 2006;281:2631–8. homeostasis and remodelling. Nat Rev Mol Cell Biol 2003;4:517–29.
[81] Yoneda T, Imaizumi K, Oono K, Yui D, Gomi F, Katayama T, et al. Acti- [103] Gorlach A, Klappa P, Kietzmann T. The endoplasmic reticulum: fold-
vation of caspase-12, an endoplastic reticulum (ER) resident caspase, ing, calcium homeostasis, signaling, and redox control. Antioxid Redox
through tumor necrosis factor receptor-associated factor 2-dependent Signal 2006;8:1391–418.
mechanism in response to the ER stress. J Biol Chem 2001;276:13935–40. [104] Eckert A, Keil U, Kressmann S, Schindowski K, Leutner S, Leutz S, et
[82] Zhang C, Kawauchi J, Adachi MT, Hashimoto Y, Oshiro S, Aso T, al. Effects of EGb 761 Ginkgo biloba extract on mitochondrial function
et al. Activation of JNK and transcriptional repressor ATF3/LRF1 and oxidative stress. Pharmacopsychiatry 2003;36(Suppl. 1):S15–23.
through the IRE1/TRAF2 pathway is implicated in human vascular [105] St-Pierre J, Buckingham JA, Roebuck SJ, Brand MD. Topology of
endothelial cell death by homocysteine. Biochem Biophys Res Commun superoxide production from different sites in the mitochondrial electron
2001;289:718–24. transport chain. J Biol Chem 2002;277:44784–90.
[83] Yang Q, Kim YS, Lin Y, Lewis J, Neckers L, Liu ZG. Tumour necrosis fac- [106] Jacobson J, Duchen MR. Mitochondrial oxidative stress and cell death
tor receptor 1 mediates endoplasmic reticulum stress-induced activation in astrocytes—requirement for stored Ca2+ and sustained opening of the
of the MAP kinase JNK. EMBO Rep 2006;7:622–7. permeability transition pore. J Cell Sci 2002;115:1175–88.
[84] Hu P, Han Z, Couvillon AD, Kaufman RJ, Exton JH. Autocrine tumor [107] Berridge MJ. The endoplasmic reticulum: a multifunctional signaling
necrosis factor alpha links endoplasmic reticulum stress to the mem- organelle. Cell Calcium 2002;32:235–49.
brane death receptor pathway through IRE1alpha-mediated NF-kappaB [108] Viner RI, Williams TD, Schoneich C. Nitric oxide-dependent modifica-
activation and down-regulation of TRAF2 expression. Mol Cell Biol tion of the sarcoplasmic reticulum Ca-ATPase: localization of cysteine
2006;26:3071–84. target sites. Free Radic Biol Med 2000;29:489–96.
[85] Tobiume K, Saitoh M, Ichijo H. Activation of apoptosis signal-regulating [109] Favero TG, Zable AC, Abramson JJ. Hydrogen peroxide stimulates the
kinase 1 by the stress-induced activating phosphorylation of pre-formed Ca2+ release channel from skeletal muscle sarcoplasmic reticulum. J Biol
oligomer. J Cell Physiol 2002;191:95–104. Chem 1995;270:25557–63.
[86] Oono K, Yoneda T, Manabe T, Yamagishi S, Matsuda S, Hitomi J, et [110] Xu L, Eu JP, Meissner G, Stamler JS. Activation of the cardiac calcium
al. JAB1 participates in unfolded protein responses by association and release channel (ryanodine receptor) by poly-S-nitrosylation. Science
dissociation with IRE1. Neurochem Int 2004;45:765–72. 1998;279:234–7.
[87] Cheung HH, Lynn Kelly N, Liston P, Korneluk RG. Involvement of [111] van der Vlies D, Makkinje M, Jansens A, Braakman I, Verkleij AJ, Wirtz
caspase-2 and caspase-9 in endoplasmic reticulum stress-induced apop- KW, et al. Oxidation of ER resident proteins upon oxidative stress: effects
tosis: a role for the IAPs. Exp Cell Res 2006;312:2347–57. of altering cellular redox/antioxidant status and implications for protein
[88] Dahmer MK. Caspases-2, -3, and -7 are involved in thapsigargin- maturation. Antioxid Redox Signal 2003;5:381–7.
induced apoptosis of SH-SY5Y neuroblastoma cells. J Neurosci Res [112] Hwang C, Sinskey AJ, Lodish HF. Oxidized redox state of glutathione in
2005;80:576–83. the endoplasmic reticulum. Science 1992;257:1496–502.
[89] Di Sano F, Ferraro E, Tufi R, Achsel T, Piacentini M, Cecconi F. Endo- [113] Chakravarthi S, Bulleid NJ. Glutathione is required to regulate the for-
plasmic reticulum stress induces apoptosis by an apoptosome-dependent mation of native disulfide bonds within proteins entering the secretory
but caspase 12-independent mechanism. J Biol Chem 2006;281:2693– pathway. J Biol Chem 2004;279:39872–9.
700. [114] Bardwell JC, McGovern K, Beckwith J. Identification of a protein
[90] Hitomi J, Katayama T, Eguchi Y, Kudo T, Taniguchi M, Koyama Y, et required for disulfide bond formation in vivo. Cell 1991;67:581–9.
al. Involvement of caspase-4 in endoplasmic reticulum stress-induced [115] Carelli S, Ceriotti A, Cabibbo A, Fassina G, Ruvo M, Sitia R. Cysteine
apoptosis and Abeta-induced cell death. J Cell Biol 2004;165:347–56. and glutathione secretion in response to protein disulfide bond formation
[91] Tan Y, Dourdin N, Wu C, De Veyra T, Elce JS, Greer PA. Ubiquitous in the ER. Science 1997;277:1681–4.
calpains promote caspase-12 and JNK activation during endoplasmic [116] Frand AR, Kaiser CA. Two pairs of conserved cysteines are required for
reticulum stress-induced apoptosis. J Biol Chem 2006;281:16016–24. the oxidative activity of Ero1p in protein disulfide bond formation in the
[92] Morishima N, Nakanishi K, Takenouchi H, Shibata T, Yasuhiko Y. endoplasmic reticulum. Mol Biol Cell 2000;11:2833–43.
An endoplasmic reticulum stress-specific caspase cascade in apoptosis. [117] Frand AR, Kaiser CA. The ERO1 gene of yeast is required for oxidation of
Cytochrome c-independent activation of caspase-9 by caspase-12. J Biol protein dithiols in the endoplasmic reticulum. Mol Cell 1998;1:161–70.
Chem 2002;277:34287–94. [118] Jessop CE, Chakravarthi S, Watkins RH, Bulleid NJ. Oxidative protein
[93] Nakagawa T, Zhu H, Morishima N, Li E, Xu J, Yankner BA, et al. Caspase- folding in the mammalian endoplasmic reticulum. Biochem Soc Trans
12 mediates endoplasmic-reticulum-specific apoptosis and cytotoxicity 2004;32:655–8.
by amyloid-beta. Nature 2000;403:98–103. [119] Cuozzo JW, Kaiser CA. Competition between glutathione and protein
[94] Fischer H, Koenig U, Eckhart L, Tschachler E. Human caspase 12 thiols for disulphide-bond formation. Nat Cell Biol 1999;1:130–5.
has acquired deleterious mutations. Biochem Biophys Res Commun [120] Pollard MG, Travers KJ, Weissman JS. Ero1p: a novel and ubiquitous pro-
2002;293:722–6. tein with an essential role in oxidative protein folding in the endoplasmic
[95] Kim SJ, Zhang Z, Hitomi E, Lee YC, Mukherjee AB. Endoplasmic reticulum. Mol Cell 1998;1:171–82.
reticulum stress-induced caspase-4 activation mediates apoptosis and [121] Tu BP, Weissman JS. Oxidative protein folding in eukaryotes: mecha-
neurodegeneration in INCL. Hum Mol Genet 2006;15:1826–34. nisms and consequences. J Cell Biol 2004;164:341–6.
[96] Brookes PS, Darley-Usmar VM. Role of calcium and superoxide dismu- [122] Cabibbo A, Pagani M, Fabbri M, Rocchi M, Farmery MR, Bulleid NJ, et
tase in sensitizing mitochondria to peroxynitrite-induced permeability al. ERO1-L, a human protein that favors disulfide bond formation in the
transition. Am J Physiol Heart Circ Physiol 2004;286:H39–46. endoplasmic reticulum. J Biol Chem 2000;275:4827–33.
[97] Stadtman ER. Importance of individuality in oxidative stress and aging. [123] Pagani M, Fabbri M, Benedetti C, Fassio A, Pilati S, Bulleid NJ, et al.
Free Radic Biol Med 2002;33:597–604. Endoplasmic reticulum oxidoreductin 1-lbeta (ERO1-Lbeta), a human
[98] Cai H, Harrison DG. Endothelial dysfunction in cardiovascular diseases: gene induced in the course of the unfolded protein response. J Biol Chem
the role of oxidant stress. Circ Res 2000;87:840–4. 2000;275:23685–92.
[99] Finkel T. Reactive oxygen species and signal transduction. IUBMB Life [124] Gess B, Hofbauer KH, Wenger RH, Lohaus C, Meyer HE, Kurtz A. The
2001;52:3–6. cellular oxygen tension regulates expression of the endoplasmic oxidore-
[100] Brigelius-Flohe R, Banning A, Schnurr K. Selenium-dependent enzymes ductase ERO1-Lalpha. Eur J Biochem 2003;270:2228–35.
in endothelial cell function. Antioxid Redox Signal 2003;5:205–15. [125] Marciniak SJ, Ron D. Endoplasmic reticulum stress signaling in disease.
[101] Lizak B, Czegle I, Csala M, Benedetti A, Mandl J, Banhegyi G. Translo- Physiol Rev 2006;86:1133–49.
con pores in the endoplasmic reticulum are permeable to small anions. [126] Saltiel AR, Kahn CR. Insulin signalling and the regulation of glucose and
Am J Physiol Cell Physiol 2006;291:C511–7. lipid metabolism. Nature 2001;414:799–806.
J.D. Malhotra, R.J. Kaufman / Seminars in Cell & Developmental Biology 18 (2007) 716–731 731

[127] Shulman GI. Cellular mechanisms of insulin resistance. J Clin Invest [150] Zhao L, Longo-Guess C, Harris BS, Lee JW, Ackerman SL. Protein accu-
2000;106:171–6. mulation and neurodegeneration in the woozy mutant mouse is caused by
[128] Kaneto H, Nakatani Y, Kawamori D, Miyatsuka T, Matsuoka TA, Mat- disruption of SIL1, a cochaperone of BiP. Nat Genet 2005;37:974–9.
suhisa M, et al. Role of oxidative stress, endoplasmic reticulum stress, and [151] Lee VM, Goedert M, Trojanowski JQ. Neurodegenerative tauopathies.
c-Jun N-terminal kinase in pancreatic beta-cell dysfunction and insulin Annu Rev Neurosci 2001;24:1121–59.
resistance. Int J Biochem Cell Biol 2006;38:782–93. [152] Dias-Santagata D, Fulga TA, Duttaroy A, Feany MB. Oxidative stress
[129] Kaneto H, Nakatani Y, Miyatsuka T, Kawamori D, Matsuoka TA, Mat- mediates tau-induced neurodegeneration in Drosophila. J Clin Invest
suhisa M, et al. Possible novel therapy for diabetes with cell-permeable 2007;117:236–45.
JNK-inhibitory peptide. Nat Med 2004;10:1128–32. [153] Calabrese V, Bates TE, Stella AM. NO synthase and NO-dependent sig-
[130] Nakatani Y, Kaneto H, Kawamori D, Yoshiuchi K, Hatazaki M, Mat- nal pathways in brain aging and neurodegenerative disorders: the role of
suoka TA, et al. Involvement of endoplasmic reticulum stress in insulin oxidant/antioxidant balance. Neurochem Res 2000;25:1315–41.
resistance and diabetes. J Biol Chem 2005;280:847–51. [154] Paschen W, Mengesdorf T. Endoplasmic reticulum stress response and
[131] Ozcan U, Yilmaz E, Ozcan L, Furuhashi M, Vaillancourt E, Smith RO, et neurodegeneration. Cell Calcium 2005;38:409–15.
al. Chemical chaperones reduce ER stress and restore glucose homeostasis [155] Uehara T, Nakamura T, Yao D, Shi ZQ, Gu Z, Ma Y, et al. S-nitrosylated
in a mouse model of type 2 diabetes. Science 2006;313:1137–40. protein-disulphide isomerase links protein misfolding to neurodegenera-
[132] Delepine M, Nicolino M, Barrett T, Golamaully M, Lathrop GM, tion. Nature 2006;441:513–7.
Julier C. EIF2AK3, encoding translation initiation factor 2-alpha kinase [156] Werstuck GH, Lentz SR, Dayal S, Hossain GS, Sood SK, Shi YY, et
3 is mutated in patients with Wolcott-Rallison syndrome. Nat Genet al. Homocysteine-induced endoplasmic reticulum stress causes dysreg-
2000;25:406–9. ulation of the cholesterol and triglyceride biosynthetic pathways. J Clin
[133] Scheuner D, Patel R, Wang F, Lee K, Kumar K, Wu J, et al. Double- Invest 2001;107:1263–73.
stranded RNA-dependent protein kinase phosphorylation of the alpha- [157] Lentz SR, Sadler JE. Homocysteine inhibits von Willebrand factor pro-
subunit of eukaryotic translation initiation factor 2 mediates apoptosis. J cessing and secretion by preventing transport from the endoplasmic
Biol Chem 2006;281:21458–68. reticulum. Blood 1993;81:683–9.
[134] Marciniak SJ, Garcia-Bonilla L, Hu J, Harding HP, Ron D. Activation- [158] Outinen PA, Sood SK, Pfeifer SI, Pamidi S, Podor TJ, Li J, et al.
dependent substrate recruitment by the eukaryotic translation initiation Homocysteine-induced endoplasmic reticulum stress and growth arrest
factor 2 kinase PERK. J Cell Biol 2006;172:201–9. leads to specific changes in gene expression in human vascular endothelial
[135] Robertson RP. Oxidative stress and impaired insulin secretion in type 2 cells. Blood 1999;94:959–67.
diabetes. Curr Opin Pharmacol 2006;6:615–9. [159] Huang RF, Huang SM, Lin BS, Wei JS, Liu TZ. Homocysteine thiolac-
[136] Sigfrid LA, Cunningham JM, Beeharry N, Hakan Borg LA, Rosales Her- tone induces apoptotic DNA damage mediated by increased intracellular
nandez AL, Carlsson C, et al. Antioxidant enzyme activity and mRNA hydrogen peroxide and caspase 3 activation in HL-60 cells. Life Sci
expression in the islets of Langerhans from the BB/S rat model of type 1 2001;68:2799–811.
diabetes and an insulin-producing cell line. J Mol Med 2004;82:325–35. [160] Zhang C, Cai Y, Adachi MT, Oshiro S, Aso T, Kaufman RJ, et al. Homo-
[137] Forman MS, Lee VM, Trojanowski JQ. ‘Unfolding’ pathways in neu- cysteine induces programmed cell death in human vascular endothelial
rodegenerative disease. Trends Neurosci 2003;26:407–10. cells through activation of the unfolded protein response. J Biol Chem
[138] Selkoe DJ. Folding proteins in fatal ways. Nature 2003;426:900–4. 2001;276:35867–74.
[139] Lindholm D, Wootz H, Korhonen L. ER stress and neurodegenerative [161] Austin RC, Lentz SR, Werstuck GH. Role of hyperhomocysteinemia in
diseases. Cell Death Differ 2006;13:385–92. endothelial dysfunction and atherothrombotic disease. Cell Death Differ
[140] Bence NF, Sampat RM, Kopito RR. Impairment of the ubiquitin- 2004;11(Suppl. 1):S56–64.
proteasome system by protein aggregation. Science 2001;292:1552–5. [162] Hofmann MA, Lalla E, Lu Y, Gleason MR, Wolf BM, Tanji N, et
[141] Shimura H, Hattori N, Kubo S, Mizuno Y, Asakawa S, Minoshima S, et al. Hyperhomocysteinemia enhances vascular inflammation and accel-
al. Familial Parkinson disease gene product, parkin, is a ubiquitin-protein erates atherosclerosis in a murine model. J Clin Invest 2001;107:675–
ligase. Nat Genet 2000;25:302–5. 83.
[142] Imai Y, Soda M, Inoue H, Hattori N, Mizuno Y, Takahashi R. An unfolded [163] Tabas I. Consequences of cellular cholesterol accumulation: basic con-
putative transmembrane polypeptide, which can lead to endoplasmic cepts and physiological implications. J Clin Invest 2002;110:905–11.
reticulum stress, is a substrate of Parkin. Cell 2001;105:891–902. [164] Tabas I. Cholesterol in health and disease. J Clin Invest 2002;110:583–
[143] Imai Y, Soda M, Takahashi R. Parkin suppresses unfolded protein stress- 90.
induced cell death through its E3 ubiquitin-protein ligase activity. J Biol [165] Feng B, Yao PM, Li Y, Devlin CM, Zhang D, Harding HP, et al. The
Chem 2000;275:35661–4. endoplasmic reticulum is the site of cholesterol-induced cytotoxicity in
[144] Osaka H, Wang YL, Takada K, Takizawa S, Setsuie R, Li H, et al. Ubiqui- macrophages. Nat Cell Biol 2003;5:781–92.
tin carboxy-terminal hydrolase L1 binds to and stabilizes monoubiquitin [166] Han S, Liang CP, DeVries-Seimon T, Ranalletta M, Welch CL, Collins-
in neuron. Hum Mol Genet 2003;12:1945–58. Fletcher K, et al. Macrophage insulin receptor deficiency increases
[145] Saigoh K, Wang YL, Suh JG, Yamanishi T, Sakai Y, Kiyosawa H, et ER stress-induced apoptosis and necrotic core formation in advanced
al. Intragenic deletion in the gene encoding ubiquitin carboxy-terminal atherosclerotic lesions. Cell Metab 2006;3:257–66.
hydrolase in gad mice. Nat Genet 1999;23:47–51. [167] Pirro M, Mauriege P, Tchernof A, Cantin B, Dagenais GR, Despres JP,
[146] Liu Y, Fallon L, Lashuel HA, Liu Z, Lansbury Jr PT. The UCH-L1 et al. Plasma free fatty acid levels and the risk of ischemic heart dis-
gene encodes two opposing enzymatic activities that affect alpha- ease in men: prospective results from the Quebec cardiovascular study.
synuclein degradation and Parkinson’s disease susceptibility. Cell Atherosclerosis 2002;160:377–84.
2002;111:209–18. [168] Li Y, Schwabe RF, DeVries-Seimon T, Yao PM, Gerbod-Giannone
[147] Holtz WA, O’Malley KL. Parkinsonian mimetics induce aspects of MC, Tall AR, et al. Free cholesterol-loaded macrophages are an abun-
unfolded protein response in death of dopaminergic neurons. J Biol Chem dant source of tumor necrosis factor-alpha and interleukin-6: model
2003;278:19367–77. of NF-kappaB- and map kinase-dependent inflammation in advanced
[148] Conn KJ, Gao W, McKee A, Lan MS, Ullman MD, Eisenhauer PB, et al. atherosclerosis. J Biol Chem 2005;280:21763–72.
Identification of the protein disulfide isomerase family member PDIp in [169] Ross R. Atherosclerosis—an inflammatory disease. N Engl J Med
experimental Parkinson’s disease and Lewy body pathology. Brain Res 1999;340:115–26.
2004;1022:164–72. [170] Puthalakath H, O’Reilly LA, Gunn P, Lee L, Kelly PN, Huntington ND,
[149] Anttonen AK, Mahjneh I, Hamalainen RH, Lagier-Tourenne C, Kopra Hughes PD, Michalak EM, McKimm-Breschkin J, Motovama N, Gotoh
O, Waris L, et al. The gene disrupted in Marinesco-Sjogren syndrome T, Akirs S, Bouillet P, Strasser A. ER stress triggers apoptosis by activating
encodes SIL1, an HSPA5 cochaperone. Nat Genet 2005;37:1309–11. BH3-only protein Bim. Cell 2007;129:1337–49.

Você também pode gostar