Download as pdf or txt
Download as pdf or txt
You are on page 1of 35

Journal Pre-proof

Crosstalk between Endoplasmic Reticulum stress and oxidative stress:Focus on


Protein Disulfide Isomerase and Endoplasmic Reticulum Oxidase 1

Paul Victor, Dronamraju Sarada, Kunka Mohanram Ramkumar

PII: S0014-2999(20)30841-4
DOI: https://doi.org/10.1016/j.ejphar.2020.173749
Reference: EJP 173749

To appear in: European Journal of Pharmacology

Received Date: 28 June 2020


Revised Date: 17 November 2020
Accepted Date: 19 November 2020

Please cite this article as: Victor, P., Sarada, D., Ramkumar, K.M., Crosstalk between Endoplasmic
Reticulum stress and oxidative stress:Focus on Protein Disulfide Isomerase and Endoplasmic Reticulum
Oxidase 1 European Journal of Pharmacology, https://doi.org/10.1016/j.ejphar.2020.173749.

This is a PDF file of an article that has undergone enhancements after acceptance, such as the addition
of a cover page and metadata, and formatting for readability, but it is not yet the definitive version of
record. This version will undergo additional copyediting, typesetting and review before it is published
in its final form, but we are providing this version to give early visibility of the article. Please note that,
during the production process, errors may be discovered which could affect the content, and all legal
disclaimers that apply to the journal pertain.

© 2020 Elsevier B.V. All rights reserved.


Crosstalk between Endoplasmic Reticulum stress and oxidative stress:

Focus on Protein Disulfide Isomerase and Endoplasmic Reticulum Oxidase 1

Paul Victora, Dronamraju Saradaa, Kunka Mohanram Ramkumar a,b *

a. Department of Biotechnology, School of Bio-engineering, SRM Institute of Science and


Technology, Kattankulathur, Chennai-603 203, Tamil Nadu, India
b. Life Science Division, SRM Research Institute, SRM Institute of Science and Technology,
Kattankulathur, Chennai-603 203, Tamil Nadu, India

of
ro
-p
re
lP

*Correspondence to
na

Dr. K.M. Ramkumar


Life Science division, SRM Research Institute, SRM Institute of Science and Technology
ur

Kattankulathur- 603203, Tamil Nadu, India.


Jo

Tel- +91 9940737854


Mail: ramkumak@srmist.edu.in

1
ABSTRACT

Cellular stress and inflammation, establishing as disease pathology, have reached


great heights in the last few decades. Stress conditions such as hyperglycemia,
hyperlipidemia and lipoproteins are known to disturb proteostasis resulting in the
accumulation of unfolded or misfolded proteins, alteration in calcium homeostasis
culminating in unfolded protein response. Protein disulfide isomerase and endoplasmic
reticulum oxidase-1 are the key players in protein folding. The protein folding process
assisted by endoplasmic reticulum oxidase-1 results in the production of reactive oxygen
species in the lumen of the endoplasmic reticulum. Production of reactive oxygen species
beyond the quenching capacity of the antioxidant systems perturbs ER homeostasis.

of
Endoplasmic reticulum stress also induces the production of cytokines leading to

ro
inflammatory responses. This has been proven to be the major causative factor for various

-p
pathophysiological states compared to other cellular triggers in diseases, which further
manifests to increased oxidative stress, mitochondrial dysfunction, and altered inflammatory
re
responses, deleterious to cellular physiology and homeostasis. Numerous studies have drawn
lP

correlations between the progression of several diseases in association with endoplasmic


reticulum stress, redox protein folding, oxidative stress and inflammatory responses. This
na

review aims to provide an insight into the role of protein disulfide isomerase and endoplasmic
reticulum oxidase-1 in endoplasmic reticulum stress, unfolded protein response,
ur

mitochondrial dysfunction, and inflammatory responses, which exacerbate the progression of


Jo

various diseases.

Keywords: ER Stress; Oxidative Stress; Protein Disulphide Isomerase; Endoplasmic


Reticulum Oxidase; Crosstalk.

2
1. INTRODUCTION

Endoplasmic reticulum (ER) is an intricate, well-orchestrated centrally located


cytoplasmic organelle involved in biogenesis, folding of proteins and trafficking in the cell
known as proteostasis i.e. (Liu and Li, 2019). ER-resident molecular chaperones
meticulously fold a proportion of the newly formed proteins to their native conformations.
The protein folding process is primarily assisted by the two ER-resident enzymes, protein
disulfide isomerase (PDI) and ER oxidase-1 (ERO1). PDI caters to redox protein folding
through oxidation, disulfide bond formation, and isomerization. Reactive oxygen species are
generated in the ER lumen largely due to ERO1 mediated electron transfer from PDI to
molecular oxygen (Zeeshan et al., 2016). Stress conditions such as hyperglycemia, elevated

of
levels of free fatty acids, and lipoproteins disturb proteostasis resulting in the accumulation of

ro
unfolded/misfolded proteins, alteration in Calcium homeostasis and initiation of unfolded

-p
protein response (UPR) (Iwawaki and Oikawa, 2013). In mammals, the UPR signaling
cascade is triggered by three ER -transmembrane proteins, namely activating transcription
re
factor-6 (ATF6), inositol requiring enzyme-α (IRE1α) and protein kinase RNA-like
lP

endoplasmic reticulum kinase (PERK). Endoplasmic reticulum stress creates an imbalance


between the protein client load and folding capacity, ultimately resulting in the accumulation
na

of misfolded /unfolded proteins. ER stress induces cytokines involved in inflammatory


reactions through UPR resident sensors and signaling proteins of the TLR pathways,
ur

including NF-κB, MAPK, and GSK-3β. Though numerous studies have drawn a synergistic
Jo

correlation between ERS, redox protein folding, oxidative stress and inflammatory responses
and the progression of several diseases, the underlying molecular mechanisms remain unclear
(Amodio et al., 2018; Estébanez et al., 2018). In the upcoming sections, we unravel the
interplay that manifests between the pathways involved in UPR, mitochondrial dysfunction,
and inflammatory responses by elucidating the role of PDI and ERO1 in various diseases.

2. UNFOLDED PROTEIN RESPONSE


ER homeostasis is maintained by the regulation between protein synthesis, folding and
trafficking in a sequential manner. Under physiological conditions, proper protein folding is
carried out with the help of ER chaperons and PDI family of enzymes in the ER lumen (Zou
et al., 2018). However, pathophysiological stresses like hyperglycemia, hyperlipidemia,
hypoxia, redox, and Calcium imbalance perturb the ER machinery (Chaudhari et al., 2014).

3
ER chaperon glucose-regulated protein (GRP)-78 (GRP78/Bip) acts as an ER stress sensor of
accumulated un/misfolded proteins in the lumen and thereby elicits the UPR (Casas, 2017).
Un/misfolded proteins trigger intracellular stress signaling or UPR pathways involving three
known transmembrane anchored proteins, namely, ATF6, IRE1α and PERK (Gardner et al.,
2013) to restore homeostasis.

Under acute stress, the aforementioned transmembrane proteins of ER are activated.


The mRNA of X-box binding protein (XBP1) is cleaved by a bifunctional transmembrane
protein called IRE1α (Blazanin et al., 2017). IRE1α and PERK form homodimers after
detaching from GRP78/BIP, to activate self-phosphorylation (Gao et al., 2019). Then, in the
Golgi apparatus, site- proteases 1 (S1P) and 2 (S2P) cleave ATF6 such that a soluble

of
cytosolic portion is released, which readily translocates into the nucleus (Ye et al., 2000). As

ro
ER continues to combat the stress, phosphorylation of a eukaryotic translation initiation

-p
eIF2α by PERK, not only halts de novo translation but also activates ATF4 (Rozpedek et al.,
2016). ATF4 and XBP1 translocate to the nucleus and facilitate degradation through ER-
re
associated degradation pathway (Gao et al., 2019). However, under chronic stress, ER fails to
lP

degrade the un/misfolded proteins via ERAD pathway, thus activates the apoptosis pathway
(Chaudhari et al., 2014), mediated by induction of C/EBP homologous binding protein
na

(CHOP or DDIT3) by PERK and ATF4 (Yang et al., 2017). Elevated CHOP expression
drives the cells to apoptosis by reducing the anti-apoptotic Bcl2 and promoting pro-apoptotic
ur

Bax proteins (Paula-Neto et al., 2019) (Fig. 1).


Jo

3. CROSSTALK BETWEEN ER AND MITOCHONDRIA IN REDOX


HOMEOSTASIS

ER and mitochondria are bridged together physically and functionally through


mitochondrial associated membranes (MAMs), which facilitate Calcium-mediated signaling,
energy metabolism and cell survival (Vannuvel et al., 2013). Activities of calmodulin,
calretinin and sarcoplasmic reticulum Calcium ATPase, all Calcium regulating proteins are
impaired due to chronic ER stress (Primeau et al., 2018). Prolonged ER stress releases Ca2+
ions mediated by inositol-1,4,5-triphosphate receptors (IP3R) and voltage-gated Calcium
channels into the mitochondria. Recently, it has been reported that the IP3R receptors
isoforms (IP3R1, IP3R2, IP3R3) are responsible for differentially regulating Ca2+ and show
different activation and deactivation patterns. Evidence also suggests that IP3R1 and IP3R3

4
mediate ER-mitochondrial Ca2+ transfer (De Stefani et al., 2012) (Mendes et al., 2005),
while IP3R2 is closely associated with the rise in mitochondrial Ca2+ compared with other two
isoforms in DT40 cells (Bartok et al., 2019).

The imbalance between the mitochondrial and cytosolic Ca2+ levels alters the pH,
permeability and electrical potential of the inner mitochondrial membrane resulting in ATP
depletion and increased reactive oxygen species production. Imbalance in the Ca2+
homeostasis alters the mitochondrial permeability through the activation of mitochondrial
calcium uniporter. Prolonged increase in the cytosolic Ca2+ triggered the opening of
mitochondrial permeability transition pore by allosteric modulation of the channels leading to
mitochondrial swelling, cytochrome-c release and programmed cell death (Santo-Domingo

of
and Demaurex, 2010). Electron leak from complex I and III of the electron transport chain

ro
results in partial reduction of oxygen to form superoxide, contributing to mitochondrial

-p
reactive oxygen species production. Emerging evidences shed light on the impaired redox
homeostasis affecting the release of Calcium into the cytosol and further augments
re
mitochondrial reactive oxygen species production. The enzymatic and non-enzymatic
lP

antioxidants quench the released reactive oxygen species. Low levels of mitochondrial
reactive oxygen species release are beneficial for a metabolic niche, as seen in hypoxia by
na

stabilizing HIF1α (Chandel et al., 2000). High levels however create an imbalance between
free radicals and scavenging capacity resulting in mitochondrial dysfunction and cell death by
ur

activating autophagy/apoptotic pathways (Li et al., 2016). Thus, crosstalk between Ca2+ and
Jo

reactive oxygen species signaling is likely to act as a molecular switch in various functions at
the ER-mitochondria associations (Eisner et al., 2013).

4. ER STRESS INDUCED BY OXIDATIVE STRESS

ER is therefore exposed to an oxidative insult to a greater or lesser extent as the result of


oxidative protein folding (Cao and Kaufman, 2014). Under normal physiological conditions,
the intracellular microenvironment favors the ER protein folding and biogenesis. Among the
several types of chemical bonds that stabilize protein conformation, the disulfide bonds
formed between cysteine sulfhydryl groups determine the overall structure of several
proteins. ER oxidase 1, introduces a disulfide bond in PDI and its family members using
molecular oxygen as the oxidant, and in conjunction with consumption of molecular oxygen,
catalyzes disulfide formation in nascent proteins via PDI in the ER (Araki et al., 2013).
Hydrogen peroxide is produced as a byproduct in this oxidative protein folding process (Heo

5
et al., 2020). Cellular enzymatic and non-enzymatic antioxidants counteract the free radical
production during redox protein folding, thus maintaining ER redox homeostasis.

Under chronic stress conditions the vicious nexus accelerates protein misfolding,
aggregation of misfolded proteins and perturbs ER homeostasis. The redox folding enzymes-
PDI and ERO1 under stressed condition accelerate free radicals production and misfolded
proteins in the ER further perturb Redox balance of the ER. While ERO1 utilizes molecular
oxygen as electron acceptor to catalyse the reaction, peroxiredoxins utilize peroxide to
oxidize the sulfhydryls in the PDI (Fujii et al., 2018). Erp46, a member of PDI family is
favored target of peroxiredoxins for disulfide formation in proteins. Reports also substantiate
that overexpression of peroxiredoxin 4 (PRDX4) shows protection in streptozotocin induced

of
diabetes in transgenic mice by reducing oxidative stress and levels of inflammatory cytokines

ro
(Cao and Kaufman, 2014). Since, reactive oxygen species production also sensitizes ER

-p
calcium channels, upregulates ERO1 and enhances mitochondrial reactive oxygen species
production (Anelli et al., 2012). Recent experimental reports conclusively prove the excessive
re
oxidative stress further worsen the ER stress in aged mice even after the administration of
lP

antioxidants (Liu et al., 2019). This vicious cycle exacerbates the redox homeostasis,
proteostasis and initiate apoptotic signalling cascade.
na
ur

5. CROSSTALK BETWEEN ER AND OXIDATIVE STRESS RESPONSE


MECHANISMS- ROLE OF REDOX PROTEIN FOLDING ENZYMES
Jo

The integrated stress response that occurs due to ER stress and oxidative stress is
evidenced in several pathophysiological circumstances. Several chaperones and enzymes
responsible for proper folding of synthesized proteins and translocation to the Golgi
apparatus reside in the ER lumen. The ER chaperones include GRP78/Bip, GRP94, GRP170,
Lectin chaperones like calnexin and calreticulin and a family of redox folding proteins
(Behnke and Hendershot, 2014). As mentioned, PDI and ERO1 are the two major redox
folding enzymes involved in proper polypeptide rearrangements and native disulfide bridges
between the cysteine residues.

PDI is a 55 KDa, 508 amino acids, 21 membered families of enzymes (Kozlov et al.,
2010) that establishes disulfide bridges between the cysteine residues and a few of its
members (PDIA1, PDIA2, PDIA3, Erp72, Erp29, Erp44) (Parakh and Atkin, 2015) involved
in redox folding of proteins. Amongst the array of PDI types, PDIA1 is a predominant player

6
in ER oxidative protein folding, and this is carried out by redox reactions of substrate S-S
bonds. PDIA2 serves as a chaperone that inhibits aggregation of misfolded proteins and has
autonomous isomerase activity (Fu and Zhu, 2010). Peptide loading onto MHC-I molecules is
primarily fostered by PDIA3 and assisted by ERp57/GRP58, a PDI family member present in
the peptide loading complex (Dong et al., 2009).

PDI is harbored with the help of ERO1, an important glycoprotein, which exists in
two isoforms: ERO1α and ERO1β. Under physiological conditions, ERO1 transfers
electrons from cysteine residues to molecular oxygen and contributes to reactive oxygen
species formation (Sevier and Kaiser, 2008). Nevertheless, the liberated reactive oxygen
species is counteracted by the glutathione (GSH) and other antioxidants, thus curtailing OS.

of
(van der Vlies et al., 2003) (Fig. 2). Formation of non-native disulfide bridges between the

ro
cysteine residues, under chronic stress, results in the accumulation of the un/misfolded

-p
proteins. Given that normal protein folding consumes a large amount of energy, the
misfolding of proteins results in ATP depletion. This energy deprivation coaxes the
re
mitochondria to enhance ATP synthesis through oxidative phosphorylation (Liemburg-Apers
lP

et al., 2015). Impaired Ca2+ homeostasis and glucose-stimulated reactive oxygen species
production increase overall oxidative stress, as evidenced in diabetes and its associated
na

complications. (Gorlach et al., 2006; Pi et al., 2007) (Susztak et al., 2006). CHOP, a pro-
apoptotic protein, also activates ERO1α resulting in Ca2+ release through IP3R and induces
ur

apoptosis through high peroxide concentration in the ER lumen (Hu et al., 2018) (Li et al.,
Jo

2009). Reports also indicate that CHOP degradation results in repression of ERO1α and
ameliorates reactive oxygen species-induced apoptosis in-vitro and in-vivo (Iurlaro and
Muñoz-Pinedo, 2016; Li et al., 2014). In CHOP deficient mice, overexpression of ERO1α is
reported to reduce hepatocellular injury induced by galactosamine (GaIN)/lipopolysaccharide
(Rao et al., 2015).

6. CROSSTALK BETWEEN UPR AND INFLAMMATORY RESPONSES

Evidence substantiating the interplay between ER stress and inflammatory response in


various pathological conditions like neurodegenerative disease, cancer, arthritis, diabetes is
reported (Verfaillie et al., 2013; Wang et al., 2016; Zhang and Kaufman, 2008). In addition to
hyperglycemia and hyperlipidemia, elevated levels of cytokines trigger UPR mediated
inflammatory stress signaling leading to perturbation of cellular homeostasis. Ca2+ and

7
reactive oxygen species liberated from the ER trigger the master inflammatory regulator,
nuclear factor-κB (NF-κB), mitogen-activated protein kinase (MAPK) called JNK (JUN N-
terminal kinase) and Glycogen synthase kinase-3β (GSK-3β) activating an inflammatory
response. Phosphorylation of IκB releases NF-κB, which translocates to the nucleus and
transcribes a number of inflammatory genes (Liu et al., 2017a). The activation of NF-κB is
also mediated by three major ER transmembrane proteins PERK, ATF6 and IRE1α, involved
in signal transduction leading to either ERAD or autophagy/apoptosis pathways, thus
connecting ER stress and inflammatory responses. During ER stress, the TIR domain-
containing adapter inducing-β (TRIF)-dependent and caspase-8 pathways are said to activate
toll-like receptor-4 (TLR) and promote the production of mature interleukin (IL) IL-1β by

of
macrophages (Shenderov et al., 2014). Evidence shows that PERK mediates NF-κB

ro
activation regardless of IκB phosphorylation through the induction of signal transducer and
activator of transcription-3 (STAT 3). Estrogen triggered apoptosis of breast cancer cells,
-p
where in the absence of PERK kinase, prevents STAT3 from binding to NF-κB and resulting
re
in repression of inflammatory genes (Fan et al., 2018). Reports also support the link between
the UPR pathway and TLR signaling cascade, resulting in pro-inflammatory production of
lP

cytokines. Increased expression of IL-6 and IL-8 in primary bronchial epithelial cells under
ER stress is attributed to phosphorylation of ERK, p38, and JNK involved in PERK and
na

ATF6 pathways (Mijosek et al., 2016).


ur

Under a pathophysiological state, IRE1α plays a vital role in modulating


Jo

macrophages, lymphocytes and endothelial cells responsible for producing cytokines. Even in
the absence of ER stress inducers, TLR-triggered genes IRE1α and XBP are induced and lead
to the production of IL-6 instead of activating UPR (Martinon et al., 2010). Expression of
pro-inflammatory cytokines is mediated through an IRE1α-TRAF2-JNK pathway, which
ultimately activates activator protein-1. The β-cell apoptosis in diabetes is triggered by UPR
activated pro-inflammatory cytokines IL-β, TNF-α, and IL-6 (Allagnat et al., 2010). In
animal models of rheumatoid arthritis, IRE1α triggered by TLR signaling where MyD88 and
TNF receptor-associated factor 6 (TRAF6) plays a crucial role in the production of various
pro-inflammatory cytokines in macrophages and neutrophils (Qiu et al., 2013). Studies by
Zhang et al., proposed that cyclic adenosine monophosphate (cAMP)-responsive element-
binding protein H (CREBH) and ATF6 may dimerize and simultaneously induce systemic
inflammatory response by the activation of acute-phase response genes (Zhang et al., 2006).

8
Accumulating evidences suggest the integrated UPR and inflammatory cytokine
production contribute to the progression of the disease, however, PDI showed an inverse
correlation regulation with activation of NF-κB. Intriguingly, the formation of NF-κB/DNA
complex and phosphorylation was intact in PDI expressing cells following stimulation with
lipopolysaccharide. This affirms that PDI is a negative regulator of NF-κB and may act as a
promising target in inflammatory diseases (Higuchi et al., 2004). On the contrary, studies by
Neill et al., showed that overnight exposure to IL-6 and IL-1β alters ER Calcium content and
glucose-stimulated insulin secretion inducing ER stress inducer leading to β-cell apoptosis in
db/db mice (O'Neill et al., 2013).

7. PDI and ERO1 as therapeutic targets

of
Various pathophysiological complications in human diseases are associated with altered

ro
functions of PDI and ERO1. Several factors are known to contribute to the improper
-p
functioning of PDI and ERO1, leading to misfolding and aggregation of proteins,
respectively. However, the exact mechanism in PDI and ERO1 contribution to the various
re
disease pathogenesis is still unclear. Reports also shed light on the application of natural and
lP

synthetic compounds to act as an antagonists or inhibitors for PDI and ERO1. In the
following sections, we review the recent findings on the role of impaired redox folding
na

enzymes related to ER stress, oxidative stress and inflammation in various diseases and shed
light on the importance of PDI and ERO1 as a novel therapeutic target for chronic diseases
ur

(Fig. 3). A few known pharmacological agents which act as a PDI/ERO1 modulators
Jo

particularly focusing on the therapeutic benefits to ameliorate the complications in various


diseases are also listed (Table-1).

8. DISEASES ASSOCIATED WITH REDOX PROTEIN FOLDING ENZYMES

8.1 CANCER

It has been a long-term conundrum as to why tumors with high ERO1α expression have a
poor prognosis. This has been looked into from an angiogenic perspective in MDA-MB-231
cells. It is proven that ERO1α establishes post-transcriptional protein oxidative folding of
vascular endothelial growth factor and indirectly promotes the expression of VEGF mRNA
through the reactive oxygen species-induced stabilization of HIF1α protein. This shows that
targeting of ERO1α can be developed as a potential therapy for arresting the progression of
tumors (Tanaka et al., 2016). The interaction between programmed death ligand-1 and
9
programmed cell death protein-1 (PD-L1/PD-1) is known to bring about T-cell tolerance and
apoptosis in tumor cells to avoid immunosurveillance. The aforementioned mechanism was
proven in triple-negative breast cancer that ERO1α facilitates the oxidative protein folding of
PD-L1 and the overexpression of the former upregulates HIF1-α and cascades into induction
and expression of PD-L1 (Tanaka et al., 2017).

A second mechanism by which tumors evade immune checks is through modulation


of the susceptibility of MHC-1 molecules expressed on their cell surface. In hypoxic colon
cancer tumor cells, the overexpressed ERO1α with PDI and calnexin modify redox state and
cell surface expression of MHC-1 through the oxidative folding of its immature form, thus
avoiding recognition by CD8+ T cells (Kukita et al., 2015). ERO1α not only works in concert

of
with PDI but also largely involves in regulation. Cigarette smoke-induced dysfunction of PDI

ro
is said to promote UPR and ER stress and studies have reported overexpression of both the

-p
enzymes in the case of tumors. A study showed PDI and ERO1 in non-small cell lung cancers
resulted in reduced lifespan and hence proposed to be used as a unique prognostic marker
re
(Kim et al., 2018). Recent reports shed light on the complex formation of PDI-ERO1α in
lP

angiogenesis, cell migration, and tumorigenesis. Zhang et al., showed that the conserved
Valine 101 of ERO1α is responsible for catalytic domain identification in PDI and redox
na

activity. Interestingly, the mutation in Val 101 results in dysfunctional PDI-ERO1α catalytic
activity resulting in reduced peroxide production (Zhang et al., 2019).
ur

A glioblastoma is an aggressive form of brain cancer having no specific cure. As in all


Jo

cancer types, overexpression of PDI plays a critical role in its rapid progression. Xu et al.,
listed several potential PDI inhibitors amongst which BAP2 significantly altered the
transcription of genes involved in UPR, ER stress, apoptotis, and DNA repair responses, thus
reducing cell proliferation and angiogenesis. BAP2 specifically and allosterically binds to
His256 in the b’ domain of PDIA1 and PDIp and inhibits its activity resulting in the arrest of
tumor growth (Xu et al., 2019). Numerous studies have reported the anti-tumor activity and
the potential to activate pro-apoptotic genes as a therapeutic application by targeting PDI and
ERO1 in various cancer cells. Hashida et al., reported that SiRNA mediated PDI knockdown
induced cell death in MCF-7 cells through caspase-dependent apoptosis (caspase-9, caspase-
6, caspase-7) (Hashida et al., 2011). Several potent natural and synthetic compounds activate
or inhibit UPR, mitochondrial dysfunction through several targets. In this regard, Xu et al.,
demonstrated the pharmacological role of PDI using propionic acid carbamoyl methyl amides
(PACMAs). PACMA-31 suppresses cellular proliferation and colony formation by binding to

10
the active site of PDI, both in-vitro and in-vivo. Interestingly, the administration of PACMA-
31 to mouse xenograft model of human OVCAR-8 ovarian cancer did not develop any
adverse side effects and organ damage, indicating PDI to be a promising target for cancer
treatment (Xu et al., 2012).

8.2 DIABETES

Evidence substantiates the deleterious effects of un/misfolded protein in the ER give


rise to various diseases. In addition to ER stress and oxidative stress mediated endothelial
dysfunction, vascular inflammation and pancreatic β-cell apoptosis contribute to diabetes, its

of
progression, and its associated complications (Suganya et al., 2018a; Suganya et al., 2018b).

ro
Under the hyperglycemic condition, there is an imbalance in the redox homeostasis due to
several mechanisms, including ER-Ca2+ targeting the mitochondrial electron transport chain
-p
and redox folding enzymes' activity. This is because of the demand for the synthesis of
re
enzymatic machinery to utilize excess glucose through glycolysis and oxidative
phosphorylation. (Maamoun et al., 2019). Oxidative stress and ER stress are closely entwined
lP

phenomena because oxidative stress can induce protein misfolding by disturbing the ER
na

redox state and disrupting disulfide bond formation and protein misfolding itself, leading to
the production of reactive oxygen species (Cao & Kaufman 2014). This leads to a decline in
ur

the levels of antioxidants such as heme oxygenase, glutathione, superoxide dismutase etc.,
leading to endothelial dysfunction, the progression of diabetes and its associated
Jo

complications, and generation of advanced glycation end products (Du et al., 2003). Recent
evidence shed light on the contribution of PDI and ERO1 towards the pathogenesis of
diabetes. According to Zito et al., the oxidoreductases, PDI and ERO1 are abundant and
facilitate oxidative folding. The significance of ERO1β in oxidative protein folding in insulin
producing cells and glucose homeostasis was demonstrated in-vivo (Zito et al., 2010).
Glucose intolerance was promoted in mutant mice of ERO1β, but knockout mice
compromised the oxidative folding of proinsulin. Insulin folding and secretion improved in
cells expressing ERO1α isoform consequently decreased ER stress. Thus, prevention of ER
stress can be achieved by solely altering ER redox state, even in β‐cells expressing
misfolded proinsulin (Riahi et al., 2018). Studies by Zhang et al. showed that PDI
overexpression causes misfolding of insulin and increased intracellular proinsulin levels in
INS-1 832/13 cells (Zhang et al., 2009). Studies from our laboratory reported amelioration of

11
oxidative stress induced DNA damage by morin through redox regulator Nrf2 signaling in
INS-1E cells and glucose-stimulated insulin secretion by streptozotocin. In mature
adipocytes, glucose- induced toxicity increased CHOP expression. (Manuel et al., 2017).

Morin enabled the nuclear translocation of Nrf2 and upregulated the downstream
targets, which was confirmed by Nrf2 phosphorylation in the nucleus (Vanitha et al., 2017).
Morin exhibited anti-apoptotic activity, and ER stress mediated diabetic liver injury via the
activation of PERK/ATF4/CHOP pathway as observed through immunoblotting studies.
Also, it enhanced the PDI activity and GSH/GSSG levels in the ER lumen in streptozotocin-
induced diabetic rats (Pandey et al., 2019). However, studies suggest that PDI may participate
in multiple phases of ER stress. Initial responses are protective and defined by processing

of
accumulated levels of unfolded proteins, while subsequent effects are pro-apoptotic. It has

ro
also been speculated that subcellular localization of PDI and/or protein aggregates, redox

-p
environment, and post-translational modification of susceptible thiols may contribute to
defining and regulating the protective or apoptotic roles of PDI (Grek and Townsend, 2014).
re
lP

8.3 CARDIOVASCULAR DISEASE


na

The pathological manifestation of the cardiac muscle is predominantly due to reactive oxygen
species generated in vascular cells by NADPH oxidase-1 (Nox1) protein activation. It has
ur

been reported that atherosclerotic aortas of non-human primates have elevated levels of PDI.
Jo

These PDI molecules associate with p47pHox through disulfide bonds between Cys400 and
Cys196, respectively, enhancing phosphorylation of the latter and finally activates Nox1 and
thus facilitating vascular smooth muscle cell (VSMCs) migration. Thus, PDI can be a
potential drug target for vascular disease treatments (Gimenez et al., 2019). Vascular smooth
muscle cells' fate is primarily decided by PDI activity, but the exact process is unclear. A
study showed that different levels of PDI expression elicit different cell fates in VSMCs. The
synergistic effect of stretch stress and advanced glycation end products, mostly in diabetic
groups, induced the co-expression of PDI and SM-α-actin in the proliferating VSMCs. PDI-
Nox1- reactive oxygen species production could also take part in increased proliferation as
well as apoptosis of the cells. The inhibition of PDI largely reduced the proliferation and cell
death of VSMCs and has been suggested as a therapeutic target (Ping et al., 2017).

Normally, PDI protects the heart from cardiomyopathy. The role of PDIA6 remains
highly obscure under hypoxic conditions. Kiouptsi et al. investigated the role of gut

12
microbiota on the expression of PDI and PDIA6 under hypoxic conditions and their effects
on the cardiac tissue. PDI and PDIA6 are highly expressed in the ischemic cardiac tissue of
the left anterior descending artery ligated mice model. In the absence of gut microbiota, there
was a drastic fall in the PDIA6 expression displaying the importance of the germ layer on
UPR mechanisms during myocardial infarction conditions (Kiouptsi et al., 2019). Evidence
substantiating the protective function of PDI under hypoxia condition was given by the
studies of Toldo et al., who showed that increased expression of PDI in cardiomyocyte cell
enhanced superoxide dismutase-1 activity and improved the protein folding (Toldo et al.,
2011).

Endothelial dysfunction and elevated homocysteine levels, a non-proteinogenic α-

of
amino acid, contribute to pathophysiological abnormalities such as blood clot and arterial

ro
damage. Recent evidence showed that homocysteine induces Ero1α expression with enhanced

-p
ER stress and inflammation in human umbilical vein endothelial cells. Intriguingly,
homocysteine alters the GSH/GSSG ratio in the ER lumen, leads to aberrant peroxide
re
generation, and perturbs glutathione peroxidase 7 redox defense system in
lP

hyperhomocysteinemia mice. This finding put forth a promising therapeutic strategy by


targeting the ERO1α and ER redox homeostasis for hyperhomocysteinemia related vascular
na

diseases (Wu et al., 2019). Studies by Jasuja et al., investigated the inhibitory role of PDI by
administering quercetin-3-rutinoside in human endothelial cells. The in-vitro analysis showed
ur

quercetin-3-rutinoside inhibited the fibrin and platelet aggregation in endothelial cells. Also,
Jo

its impasse the thrombus formation by inhibiting PDI in-vivo (Jasuja et al., 2012). Recent
reports on applying synthetic derivative of β-nitrostyrene- HPW-RX40 as a novel
antithrombotic agent by targeting PDI.

Interestingly, HPW-RX40 binds to PDI's active site and inhibited platelet aggregation
and P-selectin formation in human platelets by forming hydrogen bonds to the active site of
PDI, (Erp5, Erp57). Also, the administration of HPW-RX40 to A549 and MDA-MB-231
cells exert non-cytotoxic behaviour and ameliorate ER stress together put forth a potential
PDI inhibitor (Kung et al., 2017). This finding above sheds light on PDI inhibitors
pharmacological role that can be used as an antithrombotic agent to alleviate cardiovascular
outcomes.

13
8.4 ALZHEIMER’S DISEASE

Dementia, depression, and disorientation are the major symptoms exhibited by patients with
this progressive neurodegenerative disease. Dysfunction of PDI and ERO1 enzymes leads to
increased amyloid plaque (Aβ) accumulation, further cascading into calcium depletion of ER
(mutation of PS1), mitochondrial membrane depolarization, and finally neuronal cell death
(Fonseca et al., 2015; Morris et al., 2018). A study showed that S-nitrosylation of cysteine
residues (in thioredoxin-like domains) of PDI by endogenous nNOS activity results in a
reduced neuroprotective activity of the former during ER stress in neurodegenerative diseases
(Hashimoto et al., 2018). As mentioned, PDI plays a very important role as a chaperone,
facilitating the phase separation of the accumulated tau proteins, thus evading mitochondrial

of
stress (Uehara et al., 2006). Alzheimer's disease is said to affect the entorhinal cortex of the

ro
cerebrum initially. The effect of ER stress on the elevated levels of GRP78, CHOP, PDI,

-p
GSH levels, and cognitive memory impairment caused by entorhinal cortex amyloidopathy
could be circumvented by treatment with L-type calcium channel blockers (Ghanbari-Maman
re
et al., 2019).
lP
na

8.5 PARKINSON’S DISEASE

Dopaminergic neuronal damage is primarily caused by the accumulation of insoluble


ur

misfolded α-Synuclein (α-Syn), also termed as Lewy bodies in the soma of neurons. This
Jo

protein aggregation is said to interrupt Ca2+ homeostasis, tamper intracellular protein transit
and synaptic vesicle shipment and ultimately leads to ER stress (Colla, 2019). Cheng et al.,
reported that PDI's domain specifically binds to the protein fibril α-Syn aggregates and
impedes its formation and further dissemination. Hence, PDI can be taken to be an effective
treatment modality for the amelioration of Parkinson's disease (PD) (Lehtonen et al., 2016).
To investigate the role of PDI, C. elegans was treated with the neurotoxin 1-methyl-4-
phenylpyridinium and analyzed. Inhibition of PDI or ERO1α with the help of bacitracin in
these PD models largely suppressed the neurotoxicity and accumulation of α-Syn in the ER
through redirecting the cells into autophagy (Lehtonen et al., 2016). The most common form
of PD is the GBA risk variant (GBAN370S), and under this condition, there is an elevated ER
stress due to the protein aggregation. One of the studies reported Histone Deacetylase 4
(HDAC4) to be a critical modulator of PD progression. Under physiological conditions, the
phosphorylated enzyme is localized in the cytosol, under diseased conditions, it is

14
dephosphorylated by protein phosphatase-2 and is translocated to the nucleus where it
induces the expression of PDI, ERO1α, and FK506-binding protein 9. Treatment of GBA
variant iPSC-derived dopaminergic neurons with tasquinimod, an allosteric inhibitor of
HDAC4 or cantharidin, a protein phosphatase-2 inhibitor, substantially reduced the α-Syn
released into the extracellular matrix, promotes autophagy, ameliorates ER stress, and hence
used as a treatment strategy for PD (Lang et al., 2019).

8.6 CATARACT

A cataract is an opacification in the lens of an eye. 33% of blindness and visual impairment is
caused due to cataract worldwide (Pascolini and Mariotti, 2012). Pathophysiological

of
abnormalities include sensitivity to glare, double vision in the affected eye, blurry vision and
insensitivity to night vision. Causative factors include age, metabolic and genetic diseases,

ro
exposure to ultraviolet radiation, and prolonged intake of steroidal drugs (Liu et al., 2017b).
-p
Emerging pieces of evidence shed light on the contribution of protein misfolding and
aggregation of light scattering proteins to cataract formation. Crystallin is a water-soluble,
re
molecular weight approximately 20 KDa protein with three subunits -alpha, beta and gamma
lP

crystallin (Santhoshkumar et al., 2008). Misfolding and formation of insoluble aggregates of


crystallin cause opacification and perturbs signal transmission to the retina. Studies by
na

Palsamy et al., showed that in sodium selenite administered human lens epithelial cells, Ero1-
Lα was overexpressed while PDI and Ero1-Lβ repressed. This has been attributed to the
ur

overproduction of reactive oxygen species, initiation of UPR and ER-Ca2+ to the cytosol,
Jo

mitochondrial dysfunction induction of cataract in suckling rats (Palsamy et al., 2014a). On


the other hand, PDI and Ero1-Lβ were elevated, and overproduction of reactive oxygen
species observed in homocysteine and valproic acid-induced human lens epithelial cells
(Elanchezhian et al., 2012) (Palsamy et al., 2014b).

8.7 EXFOLIATION GLAUCOMA

Exfoliation glaucoma is an age-related disease characterized by misfolding and accumulation


of Lysyl oxidase-like 1 enzyme (LOX1). This enzyme is required for elastin biogenesis and
plays a major role in elastin homeostasis and matrix remodeling during injury, fibrosis, and
cancer development (Greene et al., 2020). Apart from protein misfolding, genetic
susceptibility of LOX1 gene polymorphism contributes to exfoliation glaucoma. Three single
nucleotide polymorphisms of rs1048661 (R141L), rs3825942 (G153D), and rs2165241
(V385V) were genotyped in exfoliation glaucoma of American and European subjects

15
(Aragon-Martin et al., 2008). In another study substantiating the PDI gene polymorphism for
adult-onset primary open-angle glaucoma reported three single nucleotide polymorphisms of
PDIA5, rs11720822, rs2241962, and rs2667465 in Salt Lake City and San Diego populations
(Carbone et al., 2011). Also, myocilin, a 504 amino acid glycoprotein widely present in
ocular tissues and trabecular meshwork responsible for maintaining the intraocular pressure,
is reported to contribute to the pathogenesis of exfoliation glaucoma (Wang et al., 2019).

ER stress, oxidative stress, and mutation cause the progression of exfoliation


glaucoma. Adenoviral expression of various myocilin mutants (G364V, Q368X, K423E,
Y437H, and I477N) in human trabecular meshwork cells tend to trigger UPR and increases
the activity of PDI and GRP78 in ER. This leads to an altered structure of trabecular cells

of
and arrest their proliferation resulting in glaucoma (Joe et al., 2003). Elevated levels of ER

ro
stress markers such as CHOP, ERO1α, GRP78 and ATF4 were significantly increased in

-p
primary trabecular meshwork cells cultured from eyes of donors with glaucoma compared to
normal donors signifying impaired proteostasis in glaucomatous trabecular meshwork tissues
re
and cells (Peters et al., 2015).
lP
na

CONCLUSION

While the roles of PDI and ERO1 in chronic diseases like cancer, diabetes, cardiovascular,
ur

neurodegenerative diseases, age-related ocular disease, and inflammatory responses are


Jo

established, the application of these two ER-resident chaperons as potential therapeutic


targets is limited. Further insights in signaling mechanisms that connect UPR, oxidative stress
and mitochondrial dysfunction are essential to underpin the crosstalk between ER stress and
oxidative stress in order to progress towards development of better strategies to overcome
several associated pathophysiological conditions.

CONFLICT OF INTEREST

The authors have no conflict of interest.

ACKNOWLEDGEMENT

This work was, in part, supported by the SRM Institute of Science and Technology,
Kattankulathur, Chennai, Tamil Nadu 603 203.

16
REFERENCES

Allagnat, F., Christulia, F., Ortis, F., Pirot, P., Lortz, S., Lenzen, S., Eizirik, D.L., Cardozo,
A.K., 2010. Sustained production of spliced X-box binding protein 1 (XBP1) induces
pancreatic beta cell dysfunction and apoptosis. Diabetologia 53, 1120-1130.
Amodio, G., Moltedo, O., Faraonio, R., Remondelli, P., 2018. Targeting the Endoplasmic
Reticulum Unfolded Protein Response to Counteract the Oxidative Stress-Induced
Endothelial Dysfunction, Oxidative medicine and cellular longevity, p. 4946289.
Anelli, T., Bergamelli, L., Margittai, E., Rimessi, A., Fagioli, C., Malgaroli, A., Pinton, P.,

of
Ripamonti, M., Rizzuto, R., Sitia, R., 2012. Ero1alpha regulates Ca(2+) fluxes at the
endoplasmic reticulum-mitochondria interface (MAM). Antioxidants & redox signaling 16,

ro
1077-1087.
-p
Aragon-Martin, J.A., Ritch, R., Liebmann, J., O'Brien, C., Blaaow, K., Mercieca, F., Spiteri,
A., Cobb, C.J., Damji, K.F., Tarkkanen, A., Rezaie, T., Child, A.H., Sarfarazi, M., 2008.
re
Evaluation of LOXL1 gene polymorphisms in exfoliation syndrome and exfoliation
lP

glaucoma. Molecular vision 14, 533-541.


Araki, K., Iemura, S., Kamiya, Y., Ron, D., Kato, K., Natsume, T., Nagata, K., 2013. Ero1-
na

alpha and PDIs constitute a hierarchical electron transfer network of endoplasmic reticulum
oxidoreductases. The Journal of cell biology 202, 861-874.
ur

Banerjee, R., Pace, N.J., Brown, D.R., Weerapana, E., 2013. 1,3,5-Triazine as a modular
Jo

scaffold for covalent inhibitors with streamlined target identification. Journal of the American
Chemical Society 135, 2497-2500.
Bartok, A., Weaver, D., Golenar, T., Nichtova, Z., Katona, M., Bansaghi, S., Alzayady, K.J.,
Thomas, V.K., Ando, H., Mikoshiba, K., Joseph, S.K., Yule, D.I., Csordas, G., Hajnoczky,
G., 2019. IP3 receptor isoforms differently regulate ER-mitochondrial contacts and local
calcium transfer. Nature communications 10, 3726.
Behnke, J., Hendershot, L.M., 2014. The large Hsp70 Grp170 binds to unfolded protein
substrates in vivo with a regulation distinct from conventional Hsp70s. The Journal of
biological chemistry 289, 2899-2907.
Blais, J.D., Chin, K.T., Zito, E., Zhang, Y., Heldman, N., Harding, H.P., Fass, D., Thorpe, C.,
Ron, D., 2010. A small molecule inhibitor of endoplasmic reticulum oxidation 1 (ERO1) with
selectively reversible thiol reactivity. The Journal of biological chemistry 285, 20993-21003.

17
Blazanin, N., Son, J., Craig-Lucas, A.B., John, C.L., Breech, K.J., Podolsky, M.A., Glick,
A.B., 2017. ER stress and distinct outputs of the IRE1alpha RNase control proliferation and
senescence in response to oncogenic Ras. Proceedings of the National Academy of Sciences
of the United States of America 114, 9900-9905.
Cao, S.S., Kaufman, R.J., 2014. Endoplasmic reticulum stress and oxidative stress in cell fate
decision and human disease. Antioxidants & redox signaling 21, 396-413.
Carbone, M.A., Chen, Y., Hughes, G.A., Weinreb, R.N., Zabriskie, N.A., Zhang, K., Anholt,
R.R., 2011. Genes of the unfolded protein response pathway harbor risk alleles for primary
open angle glaucoma. PloS one 6, e20649.
Casas, C., 2017. GRP78 at the Centre of the Stage in Cancer and Neuroprotection. Frontiers

of
in neuroscience 11, 177.

ro
Chandel, N.S., McClintock, D.S., Feliciano, C.E., Wood, T.M., Melendez, J.A., Rodriguez,
A.M., Schumacker, P.T., 2000. Reactive oxygen species generated at mitochondrial complex
-p
III stabilize hypoxia-inducible factor-1alpha during hypoxia: a mechanism of O2 sensing.
re
The Journal of biological chemistry 275, 25130-25138.
Chaudhari, N., Talwar, P., Parimisetty, A., Lefebvre d'Hellencourt, C., Ravanan, P., 2014. A
lP

molecular web: endoplasmic reticulum stress, inflammation, and oxidative stress. Frontiers in
cellular neuroscience 8, 213.
na

Chin, K.T., Kang, G., Qu, J., Gardner, L.B., Coetzee, W.A., Zito, E., Fishman, G.I., Ron, D.,
ur

2011. The sarcoplasmic reticulum luminal thiol oxidase ERO1 regulates cardiomyocyte
excitation-coupled calcium release and response to hemodynamic load. FASEB journal :
Jo

official publication of the Federation of American Societies for Experimental Biology 25,
2583-2591.
Colla, E., 2019. Linking the Endoplasmic Reticulum to Parkinson's Disease and Alpha-
Synucleinopathy. Frontiers in neuroscience 13, 560.
De Stefani, D., Bononi, A., Romagnoli, A., Messina, A., De Pinto, V., Pinton, P., Rizzuto, R.,
2012. VDAC1 selectively transfers apoptotic Ca2+ signals to mitochondria. Cell death and
differentiation 19, 267-273.
Dong, G., Wearsch, P.A., Peaper, D.R., Cresswell, P., Reinisch, K.M., 2009. Insights into
MHC class I peptide loading from the structure of the tapasin-ERp57 thiol oxidoreductase
heterodimer. Immunity 30, 21-32.
Du, X., Matsumura, T., Edelstein, D., Rossetti, L., Zsengeller, Z., Szabo, C., Brownlee, M.,
2003. Inhibition of GAPDH activity by poly(ADP-ribose) polymerase activates three major

18
pathways of hyperglycemic damage in endothelial cells. The Journal of clinical investigation
112, 1049-1057.
Eisner, V., Csordas, G., Hajnoczky, G., 2013. Interactions between sarco-endoplasmic
reticulum and mitochondria in cardiac and skeletal muscle - pivotal roles in Ca(2)(+) and
reactive oxygen species signaling. Journal of cell science 126, 2965-2978.
Elanchezhian, R., Palsamy, P., Madson, C.J., Lynch, D.W., Shinohara, T., 2012. Age-related
cataracts: homocysteine coupled endoplasmic reticulum stress and suppression of Nrf2-
dependent antioxidant protection. Chemico-biological interactions 200, 1-10.
Estébanez, B., de Paz, J.A., Cuevas, M.J., González-Gallego, J., 2018. Endoplasmic
Reticulum Unfolded Protein Response, Aging and Exercise: An Update. Frontiers in

of
physiology 9.

ro
Fan, P., Tyagi, A.K., Agboke, F.A., Mathur, R., Pokharel, N., Jordan, V.C., 2018.
Modulation of nuclear factor-kappa B activation by the endoplasmic reticulum stress sensor
-p
PERK to mediate estrogen-induced apoptosis in breast cancer cells. Cell death discovery 4,
re
15.
Fonseca, A.C., Moreira, P.I., Oliveira, C.R., Cardoso, S.M., Pinton, P., Pereira, C.F., 2015.
lP

Amyloid-beta disrupts calcium and redox homeostasis in brain endothelial cells. Molecular
neurobiology 51, 610-622.
na

Fu, X.M., Zhu, B.T., 2010. Human pancreas-specific protein disulfide-isomerase (PDIp) can
ur

function as a chaperone independently of its enzymatic activity by forming stable complexes


with denatured substrate proteins. The Biochemical journal 429, 157-169.
Jo

Fujii, J., Homma, T., Kobayashi, S., Seo, H.G., 2018. Mutual interaction between oxidative
stress and endoplasmic reticulum stress in the pathogenesis of diseases specifically focusing
on non-alcoholic fatty liver disease. World journal of biological chemistry 9, 1-15.
Gao, Y., Kim, S., Lee, Y.I., Lee, J., 2019. Cellular Stress-Modulating Drugs Can Potentially
Be Identified by in Silico Screening with Connectivity Map (CMap). International journal of
molecular sciences 20.
Gardner, B.M., Pincus, D., Gotthardt, K., Gallagher, C.M., Walter, P., 2013. Endoplasmic
reticulum stress sensing in the unfolded protein response. Cold Spring Harbor perspectives in
biology 5, a013169.
Ghanbari-Maman, A., Ghasemian-Roudsari, F., Aliakbari, S., Gholamipour-Badie, H.,
Khodagholi, F., Shaerzadeh, F., Daftari, M., 2019. Calcium Channel Blockade Ameliorates
Endoplasmic Reticulum Stress in the Hippocampus Induced by Amyloidopathy in the
Entorhinal Cortex. Iranian Journal of Pharmaceutical Research 18, 1466-1476.

19
Gimenez, M., Verissimo-Filho, S., Wittig, I., Schickling, B.M., Hahner, F., Schurmann, C.,
Netto, L.E.S., Rosa, J.C., Brandes, R.P., Sartoretto, S., De Lucca Camargo, L., Abdulkader,
F., Miller, F.J., Jr., Lopes, L.R., 2019. Redox Activation of Nox1 (NADPH Oxidase 1)
Involves an Intermolecular Disulfide Bond Between Protein Disulfide Isomerase and
p47(phox) in Vascular Smooth Muscle Cells. Arteriosclerosis, thrombosis, and vascular
biology 39, 224-236.
Gorlach, A., Klappa, P., Kietzmann, T., 2006. The endoplasmic reticulum: folding, calcium
homeostasis, signaling, and redox control. Antioxidants & redox signaling 8, 1391-1418.
Greene, A.G., Eivers, S.B., Dervan, E.W.J., O'Brien, C.J., Wallace, D.M., 2020. Lysyl
Oxidase Like 1: Biological roles and regulation. Experimental eye research 193, 107975.

of
Grek, C., Townsend, D.M., 2014. Protein Disulfide Isomerase Superfamily in Disease and the

ro
Regulation of Apoptosis. Endoplasmic reticulum stress in diseases 1, 4-17.
Hashida, T., Kotake, Y., Ohta, S., 2011. Protein disulfide isomerase knockdown-induced cell
-p
death is cell-line-dependent and involves apoptosis in MCF-7 cells. The Journal of
re
toxicological sciences 36, 1-7.
Hashimoto, S., Ishii, A., Kamano, N., Watamura, N., Saito, T., Ohshima, T., Yokosuka, M.,
lP

Saido, T.C., 2018. Endoplasmic reticulum stress responses in mouse models of Alzheimer's
disease: Overexpression paradigm versus knockin paradigm. The Journal of biological
na

chemistry 293, 3118-3125.


ur

Heo, S., Kim, S., Kang, D., 2020. The Role of Hydrogen Peroxide and Peroxiredoxins
throughout the Cell Cycle. Antioxidants 9.
Jo

Higuchi, T., Watanabe, Y., Waga, I., 2004. Protein disulfide isomerase suppresses the
transcriptional activity of NF-kappaB. Biochemical and biophysical research communications
318, 46-52.
Hoffstrom, B.G., Kaplan, A., Letso, R., Schmid, R.S., Turmel, G.J., Lo, D.C., Stockwell,
B.R., 2010. Inhibitors of protein disulfide isomerase suppress apoptosis induced by misfolded
proteins. Nature chemical biology 6, 900-906.
Hu, H., Tian, M., Ding, C., Yu, S., 2018. The C/EBP Homologous Protein (CHOP)
Transcription Factor Functions in Endoplasmic Reticulum Stress-Induced Apoptosis and
Microbial Infection. Frontiers in immunology 9, 3083.
Iurlaro, R., Muñoz-Pinedo, C., 2016. Cell death induced by endoplasmic reticulum stress.
The FEBS Journal 283, 2640-2652.
Iwawaki, T., Oikawa, D., 2013. The role of the unfolded protein response in diabetes
mellitus. Seminars in immunopathology 35, 333-350.

20
Jasuja, R., Passam, F.H., Kennedy, D.R., Kim, S.H., van Hessem, L., Lin, L., Bowley, S.R.,
Joshi, S.S., Dilks, J.R., Furie, B., Furie, B.C., Flaumenhaft, R., 2012. Protein disulfide
isomerase inhibitors constitute a new class of antithrombotic agents. The Journal of clinical
investigation 122, 2104-2113.
Joe, M.K., Sohn, S., Hur, W., Moon, Y., Choi, Y.R., Kee, C., 2003. Accumulation of mutant
myocilins in ER leads to ER stress and potential cytotoxicity in human trabecular meshwork
cells. Biochemical and biophysical research communications 312, 592-600.
Kaplan, A., Gaschler, M.M., Dunn, D.E., Colligan, R., Brown, L.M., Palmer, A.G., 3rd, Lo,
D.C., Stockwell, B.R., 2015. Small molecule-induced oxidation of protein disulfide
isomerase is neuroprotective. Proceedings of the National Academy of Sciences of the United

of
States of America 112, E2245-2252.

ro
Khan, M.M., Simizu, S., Lai, N.S., Kawatani, M., Shimizu, T., Osada, H., 2011. Discovery of
a small molecule PDI inhibitor that inhibits reduction of HIV-1 envelope glycoprotein gp120.
ACS chemical biology 6, 245-251.
-p
re
Kim, K.M., An, A.R., Park, H.S., Jang, K.Y., Moon, W.S., Kang, M.J., Lee, Y.C., Ku, J.H.,
Chung, M.J., 2018. Combined expression of protein disulfide isomerase and endoplasmic
lP

reticulum oxidoreductin 1-alpha is a poor prognostic marker for non-small cell lung cancer.
Oncology letters 16, 5753-5760.
na

Kiouptsi, K., Finger, S., Garlapati, V.S., Knorr, M., Brandt, M., Walter, U., Wenzel, P.,
ur

Reinhardt, C., 2019. Hypoxia evokes increased PDI and PDIA6 expression in the infarcted
myocardium of ex-germ-free and conventionally raised mice. Biology open 8.
Jo

Kozlov, G., Maattanen, P., Thomas, D.Y., Gehring, K., 2010. A structural overview of the
PDI family of proteins. The FEBS journal 277, 3924-3936.
Kukita, K., Tamura, Y., Tanaka, T., Kajiwara, T., Kutomi, G., Saito, K., Okuya, K., Takaya,
A., Kanaseki, T., Tsukahara, T., Hirohashi, Y., Torigoe, T., Furuhata, T., Hirata, K., Sato, N.,
2015. Cancer-Associated Oxidase ERO1-alpha Regulates the Expression of MHC Class I
Molecule via Oxidative Folding. Journal of immunology 194, 4988-4996.
Kung, P.H., Hsieh, P.W., Lin, Y.T., Lee, J.H., Chen, I.H., Wu, C.C., 2017. HPW-RX40
prevents human platelet activation by attenuating cell surface protein disulfide isomerases.
Redox biology 13, 266-277.
Kyani, A., Tamura, S., Yang, S., Shergalis, A., Samanta, S., Kuang, Y., Ljungman, M.,
Neamati, N., 2018. Discovery and Mechanistic Elucidation of a Class of Protein Disulfide
Isomerase Inhibitors for the Treatment of Glioblastoma. ChemMedChem 13, 164-177.

21
Lang, C., Campbell, K.R., Ryan, B.J., Carling, P., Attar, M., Vowles, J., Perestenko, O.V.,
Bowden, R., Baig, F., Kasten, M., Hu, M.T., Cowley, S.A., Webber, C., Wade-Martins, R.,
2019. Single-Cell Sequencing of iPSC-Dopamine Neurons Reconstructs Disease Progression
and Identifies HDAC4 as a Regulator of Parkinson Cell Phenotypes. Cell stem cell 24, 93-
106 e106.
Lehtonen, S., Jaronen, M., Vehvilainen, P., Lakso, M., Rudgalvyte, M., Keksa-Goldsteine,
V., Wong, G., Courtney, M.J., Koistinaho, J., Goldsteins, G., 2016. Inhibition of Excessive
Oxidative Protein Folding Is Protective in MPP(+) Toxicity-Induced Parkinson's Disease
Models. Antioxidants & redox signaling 25, 485-497.
Li, G., Mongillo, M., Chin, K.T., Harding, H., Ron, D., Marks, A.R., Tabas, I., 2009. Role of

of
ERO1-alpha-mediated stimulation of inositol 1,4,5-triphosphate receptor activity in

ro
endoplasmic reticulum stress-induced apoptosis. The Journal of cell biology 186, 783-792.
Li, X., Fang, P., Li, Y., Kuo, Y.M., Andrews, A.J., Nanayakkara, G., Johnson, C., Fu, H.,
-p
Shan, H., Du, F., Hoffman, N.E., Yu, D., Eguchi, S., Madesh, M., Koch, W.J., Sun, J., Jiang,
re
X., Wang, H., Yang, X., 2016. Mitochondrial Reactive Oxygen Species Mediate
Lysophosphatidylcholine-Induced Endothelial Cell Activation. Arteriosclerosis, thrombosis,
lP

and vascular biology 36, 1090-1100.


Li, Y., Guo, Y., Tang, J., Jiang, J., Chen, Z., 2014. New insights into the roles of CHOP-
na

induced apoptosis in ER stress. Acta biochimica et biophysica Sinica 46, 629-640.


ur

Liemburg-Apers, D.C., Willems, P.H., Koopman, W.J., Grefte, S., 2015. Interactions
between mitochondrial reactive oxygen species and cellular glucose metabolism. Archives of
Jo

toxicology 89, 1209-1226.


Lin, L., Gopal, S., Sharda, A., Passam, F., Bowley, S.R., Stopa, J., Xue, G., Yuan, C., Furie,
B.C., Flaumenhaft, R., Huang, M., Furie, B., 2015. Quercetin-3-rutinoside Inhibits Protein
Disulfide Isomerase by Binding to Its b'x Domain. The Journal of biological chemistry 290,
23543-23552.
Liu, L., Li, J., 2019. Communications Between the Endoplasmic Reticulum and Other
Organelles During Abiotic Stress Response in Plants. Frontiers in plant science 10, 749.
Liu, T., Zhang, L., Joo, D., Sun, S.C., 2017a. NF-kappaB signaling in inflammation. Signal
transduction and targeted therapy 2.
Liu, X., Zhang, R., Huang, L., Zheng, Z., Vlassara, H., Striker, G., Zhang, X., Guan, Y.,
Zheng, F., 2019. Excessive Oxidative Stress Contributes to Increased Acute ER Stress
Kidney Injury in Aged Mice. Oxidative medicine and cellular longevity 2019, 2746521.

22
Liu, Y.C., Wilkins, M., Kim, T., Malyugin, B., Mehta, J.S., 2017b. Cataracts. Lancet 390,
600-612.
Maamoun, H., Benameur, T., Pintus, G., Munusamy, S., Agouni, A., 2019. Crosstalk
Between Oxidative Stress and Endoplasmic Reticulum (ER) Stress in Endothelial
Dysfunction and Aberrant Angiogenesis Associated With Diabetes: A Focus on the
Protective Roles of Heme Oxygenase (HO)-1. Frontiers in physiology 10, 70.
Manuel, A.M., Walla, M.D., Faccenda, A., Martin, S.L., Tanis, R.M., Piroli, G.G., Adam, J.,
Kantor, B., Mutus, B., Townsend, D.M., Frizzell, N., 2017. Succination of Protein Disulfide
Isomerase Links Mitochondrial Stress and Endoplasmic Reticulum Stress in the Adipocyte
During Diabetes. Antioxidants & redox signaling 27, 1281-1296.

of
Martinon, F., Chen, X., Lee, A.H., Glimcher, L.H., 2010. TLR activation of the transcription

ro
factor XBP1 regulates innate immune responses in macrophages. Nature immunology 11,
411-418.
-p
Mendes, C.C., Gomes, D.A., Thompson, M., Souto, N.C., Goes, T.S., Goes, A.M.,
re
Rodrigues, M.A., Gomez, M.V., Nathanson, M.H., Leite, M.F., 2005. The type III inositol
1,4,5-trisphosphate receptor preferentially transmits apoptotic Ca2+ signals into
lP

mitochondria. The Journal of biological chemistry 280, 40892-40900.


Mijosek, V., Lasitschka, F., Warth, A., Zabeck, H., Dalpke, A.H., Weitnauer, M., 2016.
na

Endoplasmic Reticulum Stress Is a Danger Signal Promoting Innate Inflammatory Responses


ur

in Bronchial Epithelial Cells. Journal of innate immunity 8, 464-478.


Morris, G., Puri, B.K., Walder, K., Berk, M., Stubbs, B., Maes, M., Carvalho, A.F., 2018.
Jo

The Endoplasmic Reticulum Stress Response in Neuroprogressive Diseases: Emerging


Pathophysiological Role and Translational Implications. Molecular neurobiology 55, 8765-
8787.
Muchowicz, A., Firczuk, M., Chlebowska, J., Nowis, D., Stachura, J., Barankiewicz, J.,
Trzeciecka, A., Klossowski, S., Ostaszewski, R., Zagozdzon, R., Pu, J.X., Sun, H.D., Golab,
J., 2014. Adenanthin targets proteins involved in the regulation of disulphide bonds.
Biochemical pharmacology 89, 210-216.
O'Neill, C.M., Lu, C., Corbin, K.L., Sharma, P.R., Dula, S.B., Carter, J.D., Ramadan, J.W.,
Xin, W., Lee, J.K., Nunemaker, C.S., 2013. Circulating levels of IL-1B+IL-6 cause ER stress
and dysfunction in islets from prediabetic male mice. Endocrinology 154, 3077-3088.
Okumura, M., Kadokura, H., Hashimoto, S., Yutani, K., Kanemura, S., Hikima, T., Hidaka,
Y., Ito, L., Shiba, K., Masui, S., Imai, D., Imaoka, S., Yamaguchi, H., Inaba, K., 2014.
Inhibition of the functional interplay between endoplasmic reticulum (ER) oxidoreduclin-

23
1alpha (Ero1alpha) and protein-disulfide isomerase (PDI) by the endocrine disruptor
bisphenol A. The Journal of biological chemistry 289, 27004-27018.
Palsamy, P., Bidasee, K.R., Shinohara, T., 2014a. Selenite cataracts: activation of
endoplasmic reticulum stress and loss of Nrf2/Keap1-dependent stress protection. Biochimica
et biophysica acta 1842, 1794-1805.
Palsamy, P., Bidasee, K.R., Shinohara, T., 2014b. Valproic acid suppresses Nrf2/Keap1
dependent antioxidant protection through induction of endoplasmic reticulum stress and
Keap1 promoter DNA demethylation in human lens epithelial cells. Experimental eye
research 121, 26-34.
Pandey, V.K., Mathur, A., Khan, M.F., Kakkar, P., 2019. Activation of PERK-eIF2alpha-

of
ATF4 pathway contributes to diabetic hepatotoxicity: Attenuation of ER stress by Morin.

ro
Cellular signalling 59, 41-52.
Parakh, S., Atkin, J.D., 2015. Novel roles for protein disulphide isomerase in disease states: a
-p
double edged sword? Frontiers in cell and developmental biology 3, 30.
re
Pascolini, D., Mariotti, S.P., 2012. Global estimates of visual impairment: 2010. The British
journal of ophthalmology 96, 614-618.
lP

Paula-Neto, H.A., Pereira, R.M., Carneiro, L.A.M., 2019. Editorial: Producing, Sensing and
Responding to Cellular Stress in Immunity. Frontiers in immunology 10, 2053.
na

Peters, J.C., Bhattacharya, S., Clark, A.F., Zode, G.S., 2015. Increased Endoplasmic
ur

Reticulum Stress in Human Glaucomatous Trabecular Meshwork Cells and Tissues.


Investigative ophthalmology & visual science 56, 3860-3868.
Jo

Pi, J., Bai, Y., Zhang, Q., Wong, V., Floering, L.M., Daniel, K., Reece, J.M., Deeney, J.T.,
Andersen, M.E., Corkey, B.E., Collins, S., 2007. Reactive oxygen species as a signal in
glucose-stimulated insulin secretion. Diabetes 56, 1783-1791.
Ping, S., Liu, S., Zhou, Y., Li, Z., Li, Y., Liu, K., Bardeesi, A.S., Wang, L., Chen, J., Deng,
L., Wang, J., Wang, H., Chen, D., Zhang, Z., Sheng, P., Li, C., 2017. Protein disulfide
isomerase-mediated apoptosis and proliferation of vascular smooth muscle cells induced by
mechanical stress and advanced glycosylation end products result in diabetic mouse vein
graft atherosclerosis. Cell death & disease 8, e2818.
Primeau, J.O., Armanious, G.P., Fisher, M.E., Young, H.S., 2018. The SarcoEndoplasmic
Reticulum Calcium ATPase. Sub-cellular biochemistry 87, 229-258.
Qiu, Q., Zheng, Z., Chang, L., Zhao, Y.S., Tan, C., Dandekar, A., Zhang, Z., Lin, Z., Gui, M.,
Li, X., Zhang, T., Kong, Q., Li, H., Chen, S., Chen, A., Kaufman, R.J., Yang, W.L., Lin,
H.K., Zhang, D., Perlman, H., Thorp, E., Zhang, K., Fang, D., 2013. Toll-like receptor-

24
mediated IRE1alpha activation as a therapeutic target for inflammatory arthritis. The EMBO
journal 32, 2477-2490.
Rao, J., Zhang, C., Wang, P., Lu, L., Qian, X., Qin, J., Pan, X., Li, G., Wang, X., Zhang, F.,
2015. C/EBP homologous protein (CHOP) contributes to hepatocyte death via the promotion
of ERO1alpha signalling in acute liver failure. The Biochemical journal 466, 369-378.
Riahi, Y., Israeli, T., Cerasi, E., Leibowitz, G., 2018. Effects of proinsulin misfolding on
beta-cell dynamics, differentiation and function in diabetes. Diabetes, obesity & metabolism
20 Suppl 2, 95-103.
Rozpedek, W., Pytel, D., Mucha, B., Leszczynska, H., Diehl, J.A., Majsterek, I., 2016. The
Role of the PERK/eIF2alpha/ATF4/CHOP Signaling Pathway in Tumor Progression During

of
Endoplasmic Reticulum Stress. Current molecular medicine 16, 533-544.

ro
Santhoshkumar, P., Udupa, P., Murugesan, R., Sharma, K.K., 2008. Significance of
interactions of low molecular weight crystallin fragments in lens aging and cataract
-p
formation. The Journal of biological chemistry 283, 8477-8485.
re
Santo-Domingo, J., Demaurex, N., 2010. Calcium uptake mechanisms of mitochondria.
Biochimica et biophysica acta 1797, 907-912.
lP

Sevier, C.S., Kaiser, C.A., 2008. Ero1 and redox homeostasis in the endoplasmic reticulum.
Biochimica et biophysica acta 1783, 549-556.
na

Shenderov, K., Riteau, N., Yip, R., Mayer-Barber, K.D., Oland, S., Hieny, S., Fitzgerald, P.,
ur

Oberst, A., Dillon, C.P., Green, D.R., Cerundolo, V., Sher, A., 2014. Cutting edge:
Endoplasmic reticulum stress licenses macrophages to produce mature IL-1beta in response
Jo

to TLR4 stimulation through a caspase-8- and TRIF-dependent pathway. Journal of


immunology 192, 2029-2033.
Shergalis, A., Xue, D., Gharbia, F.Z., Driks, H., Shrestha, B., Tanweer, A., Cromer, K.,
Ljungman, M., Neamati, N., 2020. Characterization of Aminobenzylphenols as Protein
Disulfide Isomerase Inhibitors in Glioblastoma Cell Lines. Journal of medicinal chemistry
63, 10263-10286.
Suganya, N., Dornadula, S., Chatterjee, S., Mohanram, R.K., 2018a. Quercetin improves
endothelial function in diabetic rats through inhibition of endoplasmic reticulum stress-
mediated oxidative stress. European journal of pharmacology 819, 80-88.
Suganya, N., Mani, K.P., Sireesh, D., Rajaguru, P., Vairamani, M., Suresh, T., Suzuki, T.,
Chatterjee, S., Ramkumar, K.M., 2018b. Establishment of pancreatic microenvironment
model of ER stress: Quercetin attenuates beta-cell apoptosis by invoking nitric oxide-cGMP
signaling in endothelial cells. The Journal of nutritional biochemistry 55, 142-156.

25
Susztak, K., Raff, A.C., Schiffer, M., Bottinger, E.P., 2006. Glucose-induced reactive oxygen
species cause apoptosis of podocytes and podocyte depletion at the onset of diabetic
nephropathy. Diabetes 55, 225-233.
Tanaka, T., Kutomi, G., Kajiwara, T., Kukita, K., Kochin, V., Kanaseki, T., Tsukahara, T.,
Hirohashi, Y., Torigoe, T., Okamoto, Y., Hirata, K., Sato, N., Tamura, Y., 2016. Cancer-
associated oxidoreductase ERO1-alpha drives the production of VEGF via oxidative protein
folding and regulating the mRNA level. British journal of cancer 114, 1227-1234.
Tanaka, T., Kutomi, G., Kajiwara, T., Kukita, K., Kochin, V., Kanaseki, T., Tsukahara, T.,
Hirohashi, Y., Torigoe, T., Okamoto, Y., Hirata, K., Sato, N., Tamura, Y., 2017. Cancer-
associated oxidoreductase ERO1-alpha promotes immune escape through up-regulation of

of
PD-L1 in human breast cancer. Oncotarget 8, 24706-24718.

ro
Toldo, S., Severino, A., Abbate, A., Baldi, A., 2011. The role of PDI as a survival factor in
cardiomyocyte ischemia. Methods in enzymology 489, 47-65.
-p
Uehara, T., Nakamura, T., Yao, D., Shi, Z.Q., Gu, Z., Ma, Y., Masliah, E., Nomura, Y.,
re
Lipton, S.A., 2006. S-nitrosylated protein-disulphide isomerase links protein misfolding to
neurodegeneration. Nature 441, 513-517.
lP

van der Vlies, D., Makkinje, M., Jansens, A., Braakman, I., Verkleij, A.J., Wirtz, K.W., Post,
J.A., 2003. Oxidation of ER resident proteins upon oxidative stress: effects of altering
na

cellular redox/antioxidant status and implications for protein maturation. Antioxidants &
ur

redox signaling 5, 381-387.


Vanitha, P., Senthilkumar, S., Dornadula, S., Anandhakumar, S., Rajaguru, P., Ramkumar,
Jo

K.M., 2017. Morin activates the Nrf2-ARE pathway and reduces oxidative stress-induced
DNA damage in pancreatic beta cells. European journal of pharmacology 801, 9-18.
Vannuvel, K., Renard, P., Raes, M., Arnould, T., 2013. Functional and morphological impact
of ER stress on mitochondria. Journal of cellular physiology 228, 1802-1818.
Vatolin, S., Phillips, J.G., Jha, B.K., Govindgari, S., Hu, J., Grabowski, D., Parker, Y.,
Lindner, D.J., Zhong, F., Distelhorst, C.W., Smith, M.R., Cotta, C., Xu, Y., Chilakala, S.,
Kuang, R.R., Tall, S., Reu, F.J., 2016. Novel Protein Disulfide Isomerase Inhibitor with
Anticancer Activity in Multiple Myeloma. Cancer research 76, 3340-3350.
Verfaillie, T., Garg, A.D., Agostinis, P., 2013. Targeting ER stress induced apoptosis and
inflammation in cancer. Cancer letters 332, 249-264.
Wang, H., Li, M., Zhang, Z., Xue, H., Chen, X., Ji, Y., 2019. Physiological function of
myocilin and its role in the pathogenesis of glaucoma in the trabecular meshwork (Review).
International journal of molecular medicine 43, 671-681.

26
Wang, Z., Huang, Y., Cheng, Y., Tan, Y., Wu, F., Wu, J., Shi, H., Zhang, H., Yu, X., Gao,
H., Lin, L., Cai, J., Zhang, J., Li, X., Cai, L., Xiao, J., 2016. Endoplasmic reticulum stress-
induced neuronal inflammatory response and apoptosis likely plays a key role in the
development of diabetic encephalopathy. Oncotarget 7, 78455-78472.
Weston, B.S., Wahab, N.A., Roberts, T., Mason, R.M., 2001. Bacitracin inhibits fibronectin
matrix assembly by mesangial cells in high glucose. Kidney international 60, 1756-1764.
Wu, X., Zhang, L., Miao, Y., Yang, J., Wang, X., Wang, C.C., Feng, J., Wang, L., 2019.
Homocysteine causes vascular endothelial dysfunction by disrupting endoplasmic reticulum
redox homeostasis. Redox biology 20, 46-59.
Xu, S., Butkevich, A.N., Yamada, R., Zhou, Y., Debnath, B., Duncan, R., Zandi, E., Petasis,

of
N.A., Neamati, N., 2012. Discovery of an orally active small-molecule irreversible inhibitor

ro
of protein disulfide isomerase for ovarian cancer treatment. Proceedings of the National
Academy of Sciences of the United States of America 109, 16348-16353.
-p
Xu, S., Liu, Y., Yang, K., Wang, H., Shergalis, A., Kyani, A., Bankhead, A., 3rd, Tamura, S.,
re
Yang, S., Wang, X., Wang, C.C., Rehemtulla, A., Ljungman, M., Neamati, N., 2019.
Inhibition of protein disulfide isomerase in glioblastoma causes marked downregulation of
lP

DNA repair and DNA damage response genes. Theranostics 9, 2282-2298.


Yang, Y., Liu, L., Naik, I., Braunstein, Z., Zhong, J., Ren, B., 2017. Transcription Factor
na

C/EBP Homologous Protein in Health and Diseases. Frontiers in immunology 8, 1612.


ur

Ye, J., Rawson, R.B., Komuro, R., Chen, X., Dave, U.P., Prywes, R., Brown, M.S.,
Goldstein, J.L., 2000. ER stress induces cleavage of membrane-bound ATF6 by the same
Jo

proteases that process SREBPs. Molecular cell 6, 1355-1364.


Zeeshan, H.M., Lee, G.H., Kim, H.R., Chae, H.J., 2016. Endoplasmic Reticulum Stress and
Associated ROS. International journal of molecular sciences 17, 327.
Zhang, K., Kaufman, R.J., 2008. From endoplasmic-reticulum stress to the inflammatory
response. Nature 454, 455-462.
Zhang, K., Shen, X., Wu, J., Sakaki, K., Saunders, T., Rutkowski, D.T., Back, S.H.,
Kaufman, R.J., 2006. Endoplasmic reticulum stress activates cleavage of CREBH to induce a
systemic inflammatory response. Cell 124, 587-599.
Zhang, L., Lai, E., Teodoro, T., Volchuk, A., 2009. GRP78, but Not Protein-disulfide
Isomerase, Partially Reverses Hyperglycemia-induced Inhibition of Insulin Synthesis and
Secretion in Pancreatic {beta}-Cells. The Journal of biological chemistry 284, 5289-5298.
Zhang, Y., Li, T., Zhang, L., Shangguan, F., Shi, G., Wu, X., Cui, Y., Wang, X., Wang, X.,
Liu, Y., Lu, B., Wei, T., Wang, C.C., Wang, L., 2019. Targeting the functional interplay

27
between endoplasmic reticulum oxidoreductin-1alpha and protein disulfide isomerase
suppresses the progression of cervical cancer. EBioMedicine 41, 408-419.
Zhou, X., Li, G., Kaplan, A., Gaschler, M.M., Zhang, X., Hou, Z., Jiang, M., Zott, R.,
Cremers, S., Stockwell, B.R., Duan, W., 2018. Small molecule modulator of protein disulfide
isomerase attenuates mutant huntingtin toxicity and inhibits endoplasmic reticulum stress in a
mouse model of Huntington's disease. Human molecular genetics 27, 1545-1555.
Zito, E., Chin, K.T., Blais, J., Harding, H.P., Ron, D., 2010. ERO1-beta, a pancreas-specific
disulfide oxidase, promotes insulin biogenesis and glucose homeostasis. The Journal of cell
biology 188, 821-832.
Zou, H., Wen, C., Peng, Z., Shao, Y., Hu, L., Li, S., Li, C., Zhou, H.H., 2018. P4HB and

of
PDIA3 are associated with tumor progression and therapeutic outcome of diffuse gliomas.

ro
Oncology reports 39, 501-510.

-p
FIGURE LEGENDS
re
Fig 1. Unfolded Protein Response in Mammals
lP

During ER stress, the ER sensors such as PERK, ATF6 and IRE1α get activated by the ER
chaperone GRP78 when bound to mis/unfolded proteins. Activation of ER stress sensors
na

leads to UPR through their respective downstream targets. PERK phosphorylates eIF2α, thus
leads to an increase in ATF4 expression. IRE1α and PERK form homodimers, after detaching
ur

from GRP78/BIP, to activate self-phosphorylation. On the other hand, after detaching from
Jo

GRP78/BIP, ATF6(p90) translocates to the Golgi apparatus, where it is activated by S1P and
S2P. S1P cleaves at the ER luminal domain of ATF6 while S2P does at the transmembrane
domain of ATF6. Active ATF6 (p50) translocates to the nucleus and activates UPR genes.
Lastly, IRE1 transcribes the XBP1 mRNA to an active XBP1 by its endoribonuclease
activity. XBP1 enters into the nucleus and activate UPR genes.

UPR -unfolded protein response; IRE-1α, inositol-requiring enzyme 1 α; PERK, Protein


Kinase RNA-like endoplasmic reticulum kinase; ATF6, activating transcription factor 6; BiP,
binding immunoglobulin protein; eIF-2a, Eukaryotic translation initiation factor 2a; GRP78,
78-kDa glucose regulated protein; XBP1, X-box binding protein 1; S1P - sphingosine-1-
phosphate; S2P- site-2 protease.

Fig 2. PDI and ERO1 mainly catalyse redox Protein folding by PDI and ERO1 Disulfide
bond formation in the ER. PDI accepts electrons from protein folding substrates, thus

28
oxidizing the thiol (SH) groups in the cysteine residues resulting in disulfide bonds. ERO1
uses a redox-active coenzyme-FAD to mediate electrons transfer from PDI to molecular
oxygen, resulting in ROS generation. Reduced glutathione (GSH) can assist in disulfide-bond
reduction, which balances between protein load and misfolded proteins, and results in
oxidized glutathione (GSSG). PDI, Protein disulfide isomerase; ERO1, Endoplasmic
reticulum oxidoreductase 1.
Fig 3. PDI-ERO1 dysfunction in various diseases
During Proteostasis, PDI and ERO1 are involved in forming native disulfide bridges between
the protein's cysteine residues. This leads to the release of functional proteins in the
cytoplasm and maintains the redox homeostasis in the ER lumen. Under ER stress, there is an

of
imbalance between the protein client load and folding capacity leads to the formation of non-

ro
native disulfide bridges, thus forming protein aggregates in the ER and cytosol. This leads to
functional protein aggregation resulting in diabetes, cardiovascular disease, cancer,
-p
neurogenerative and ocular degenerative diseases.
re
lP
na
ur
Jo

29
Table 1- List of PDI/ERO1 modulators and their effects in various diseases

Pharmacological
S. No Compound Target Model Outcome Reference
Action
OVCAR-8
PACMA-31
cells Forming a covalent
(propynoic acid Anti-tumor (Xu et al.,
1 PDI (Ovarian bond with the active
Carbamoyl activity 2012)
Cancer Cell site cysteines of PDI
methyl amides)
Lines)
Downregulation of ER Improved
(Zhou et al.,
stress proteins in HD. motor function,
N171-82Q 2018)
2 LOC14 PDI LOC14 forces PDI to ameliorated
HD mice (Kaplan et al.,

of
adopt an oxidized brain atrophy
2015)
conformation and cell survival

ro
Disulfide bond
formation between Reduced
Human
3 Bacitracin PDI mesangial cells
(HMC)
-p
thiazoline ring and
cysteines in the
mesangial
extracellular
(Weston et
al., 2001)
re
substrate-binding matrix
domain of PDI.
Inhibits
lP

Quercetin-3- C57BL/6J binds to the b' domain (Lin et al.,


4 PDI thrombus
rutinoside mice of PDI 2015)
formation
na

α-
aminobenzylphe Glioblastoma Covalent binding to Anticancer (Shergalis et
5 PDI
nol analogues cells the active site activity al., 2020)
ur

(AS15)
Human
Pyrimidotriazin Binds to the catalytic Anti- tumor (Kyani et al.,
Jo

6 PDI glioblastoma
edione 35G8 site of PDI activity 2018)
cell lines
Inhibit PDI
HS-5 bone Covalent binding in reductase
(Vatolin et
7 CCF642 PDI marrow stroma active-site activity,
al., 2016)
cells CGHCK motifs Anticancer
activity
Mouse
xenograft Binds to His 256 in Arrest Cell and (Xu et al.,
8 BAP2 PDI
model of the b' domain of PDI tumor growth 2019)
Glioblastoma
Mouse
Anticancer
embryonic Prevents re-oxidation (Blais et al.,
9 EN460 ERO1 activity and
fibroblast of ERO1 2010)
induce apoptosis
(MEF) cells
Disrupts PDI-ERO1
ERO1/P Anticancer (Okumura et
10 Bisphenol A HeLa cells interaction by binding
DI activity al., 2014)
to the b’ domain of PDI
Recombinant Binding to the Reduction of (Khan et al.,
11 Juniferdin PDI
HIV-1IIIB catalytic site HIV-1 envelope 2011)
30
gp120 glycoprotein
produced in gp120.
CHO cells
Inhibits (Muchowicz
12 Adenanthin PDI HEK cells Binds to the b’ domain
thioredoxins et al., 2014)
Binds to the active site Inhibits cell (Banerjee et
13 RB-11-ca PDI HeLa Cells
in PDI proliferation al., 2013)
mutant
huntingtin
Binds to the b’ domain Antiapoptotic (Hoffstrom et
14 16F16 PDI protein
of PDI activity al., 2010)
transfected in
PC12 cells
AC16 Human
Traps ERO1 in its Calcium (Chin et al.,
15 QM295 ERO1 Cardiomyocy
reduced state homeostasis 2011)
te Cell Line

of
ro
-p
re
lP
na
ur
Jo

31
Jo
ur
na
lP
re
-p
ro
of
Jo
ur
na
lP
re
-p
ro
of
Jo
ur
na
lP
re
-p
ro
of

You might also like