A Guide To Assessing Endoplasmic Reticulum Homeostasis

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 16

A GUIDE TO. . .

A guide to assessing endoplasmic reticulum homeostasis


and stress in mammalian systems
s Delaunay-Moisan3, Laurent Combettes4,5, Eric Chevet1,2
Daria Sicari1,2, Agne and
1,2
Aeid Igbaria
1 Inserm U1242, University of Rennes, France
2 Centre de lutte contre le cancer Euge ne Marquis, Rennes, France

3 Institute for Integrative Biology of the Cell (I2BC), CEA-Saclay, CNRS, ISVJC/SBIGEM, Laboratoire Stress Oxydant et Cancer, Universite
Paris-Saclay, Gif-sur-Yvette, France
4 UMRS1174, Universite  Paris Sud, Orsay, France
5 UMRS1174, Institut National de la Sante  et de la Recherche Me dicale (Inserm), Orsay, France

Keywords The endoplasmic reticulum (ER) is a multifunctional organelle that consti-


Calcium distribution; ER redox state; ER tutes the entry into the secretory pathway. The ER contributes to the
stress; ER structure; UPR
maintenance of cellular calcium homeostasis, lipid synthesis and productive
secretory, and transmembrane protein folding. Physiological, chemical, and
Correspondence
E. Chevet or A. Igbaria, Chemistry, pathological factors that compromise ER homeostasis lead to endoplasmic
Oncogenesis, stress, Signaling-(COSS), reticulum stress (ER stress). To cope with this situation, cells activate an
Inserm U1242, Universite de Rennes-1, adaptive signaling pathway termed the unfolded protein response (UPR)
CLCC Eugene Marquis, Rue de la Bataille that aims at restoring ER homeostasis. The UPR is transduced through
Flandres Dunkerque. Bat D, 1er etage post-translational, translational, post-transcriptional, and transcriptional
35042 Rennes, France
mechanisms initiated by three ER-resident sensors, inositol-requiring pro-
Tel: +33 2 23237258
tein 1a, activating transcription factor 6a, and PRKR-like endoplasmic
E-mails: eric.chevet@inserm.fr (EC) or
aeid.igbaria@gmail.com (AI) reticulum kinase. Determining the in and out of ER homeostasis control
and UPR activation still represents a challenge for the community. Hence,
(Received 25 July 2019, revised 10 standardized criteria and methodologies need to be proposed for monitor-
September 2019, accepted 23 October ing ER homeostasis and ER stress in different model systems. Here, we
2019) summarize the pathways that are activated during ER stress and provide
approaches aimed at assess ER homeostasis and stress in vitro and in vivo
doi:10.1111/febs.15107
mammalian systems that can be used by researchers to plan and interpret
experiments. We recommend the use of multiple assays to verify ER stress
because no individual assay is guaranteed to be the most appropriate one.

Introduction
The endoplasmic reticulum (ER) represents a complex secretory proteins. The ER is a multifunctional orga-
membranous network that beyond ensuring cell cal- nelle crowded with many enzymes, foldases, and chap-
cium and lipid homeostasis and mediates the produc- erones that assist oxidative protein folding and other
tive folding and trafficking of transmembrane and post-translational modifications, which ensure the

Abbreviations
AM, acetomethoxy; ATF6, activating transcription factor 6; EM, electron microscopy; ER stress, endoplasmic reticulum stress; ERSE-I/II,
endoplasmic reticulum stress element I/II; FP, fluorescent protein; FRET, Fo €rster resonance energy transfer; GECIs, genetically encoded
calcium indicators; GSH, glutathione; IRE1, inositol-requiring protein 1; PERK, PRKR-like endoplasmic reticulum kinase; RIDD, RNA IRE1a-
dependent decay; roGFP, reduction- to oxidation-sensitive GFP; rxYFP, redox-sensitive YFP; sfGFP, superfolder green fluorescent protein;
Tg, thapsigargin; ThT, thioflavin; Tm, tunicamycin; TXN, thioredoxin; UPR, unfolded protein response; UPRE, UPR element; XBP1, X-box-
binding protein 1.

The FEBS Journal 287 (2020) 27–42 ª 2019 Federation of European Biochemical Societies 27
Tools to measure ER homeostasis in mammals D. Sicari et al.

maintenance of ER homeostasis [1]. In addition, cells Approaches to assess ER stress


have developed different quality control systems in
order to ensure that only properly folded proteins are The unfolded protein response
exported through the other compartments of the secre-
tory pathway, the improperly folded proteins being The UPR is transduced by three ER transmembrane
degraded by the proteasome in a process called ER-as- sensor proteins (PRKR-like endoplasmic reticulum
sociated degradation [2]. Intrinsic (genetic mutations, kinase (PERK), activating transcription factor 6
aneuploidy) and extrinsic (environmental disturbances) (ATF6)a, and inositol-requiring protein 1 (IRE1)a),
factors compromise ER homeostasis leading to a situa- through their luminal domains they are able to moni-
tion called endoplasmic reticulum stress (ER stress) tor and sense the protein folding status of the ER, and
that occurs when the balance between the protein fold- then, they signal through their cytosolic domain either
ing demand exceeds that of the ER. In this situation, directly through specific catalytic activities or select
cells adapt by adjusting their folding capacity through post-translational processing (Fig. 1). UPR signaling
activation of an intracellular signaling pathway known aims at alleviating stress; however, if homeostasis can-
as the unfolded protein response (UPR) (Fig. 1). Once not be restored, the UPR activates cell death pathways
activated, the UPR outputs can be measured by differ- [3]. Hence, ER stress and the UPR play an important
ent methods that are discussed below. Monitoring the role in physiology and in the development of numer-
UPR outputs through measuring the activation of the ous diseases.
different signaling pathways of the UPR represents a
powerful and easy tool but may not be enough to mir- The IRE1a arm
ror ER proteostasis. In this article, we also review the
available methods to assess ER homeostasis and stress IRE1a (referred to IRE1 hereafter) is the most con-
as well as their use in biological research. Our recom- served sensor of the UPR, present from yeast to
mendation is to test different outputs including signal- human. IRE1 is a type I transmembrane protein with
ing pathways, ER morphology, and ER calcium flux a lumenal domain that senses misfolded proteins and a
in addition to assessing changes in the ER redox state cytosolic domain comprising two catalytic activities, a
to ensure the production of a proper representation of serine/threonine kinase, and an endoribonuclease
the ER functionality. (RNase) (Fig. 1). Accumulation of misfolded proteins

eroGFP [1] ThT 3


ER-Redox State rxYFP [2] S 1 SH
1 S
ThT [3] S Aggregated protein eroGFP
ER-structure eroGFP rxYFP S SH SH
ER 405nm 488nm
CEPIA [4] 2 rxYFP SH
Calcium Status 2
PERK
ATF6
[5]
[6]
5 Ca+2 CEPIA
IRE1
7
UPR PERK 4 6 IRE1 IRE1
IRE1 [7] PERK ATF6

P K mRNA
K K K K RIDD miRNA
Cytoplasm P P P P
R R R
eIF2α Chem. Golgi
P Dyes AT
F6 Xbp1 Xbp1s-Venus/Luc
.
GECIs TRAF2
TED ASK1
Global Protein Selective Translation SERCaMP s1p/s2p
Synthesis (μORFs) Xbp1s Xbp1s-Venus/Luc
.
P JNK
NRF2 ATF6-f
Xbp1/
ATF4 Xbp1s-Venus
Target
genes Target
genes
ERSE-I UPRE
Nucleus
(CCAAT-N9-CCACG/A) (TGACGTGG/A)
ERSE-II
(ATTGG-N1-CCACG)

Fig. 1. Summary of the activation of the different signaling outputs during ER stress. in green redox state: eroGFP, rxYFP (1,2); in black ER
structure: ThT (3); in red calcium status: CEPIA (4); UPR: in orange PERK pathway (5), in violet ATF6 pathway (6); in blue IRE1a pathway (7).

28 The FEBS Journal 287 (2020) 27–42 ª 2019 Federation of European Biochemical Societies
D. Sicari et al. Tools to measure ER homeostasis in mammals

in the ER leads to IRE1 dimerization and trans-au- ligase RTCB [6]. This nonconventional splicing shifts
tophosphorylation. IRE1 activation can be directly the open reading frame on the mRNA resulting in the
measured by assessing the phosphorylation state at ser- translation into an active transcription factor XPB1s
ines 724 or 729, which are phosphorylated upon its [7]. The abundance of XBP1s can be measured by
activation, and antibodies specific for the phosphory- using PCR with primers flanking the intron (Table 3).
lated IRE1 are available commercially (S724) or upon XBP1u (unspliced) amplicons migrate on agarose gel
request (S729) (Table 1). Because IRE1 is poorly (3% or 4%) around 476 bp, while XBP1s amplicons’
abundant, it is useful to concentrate the protein by mobility is faster (450 bp). Alternatively, because the
immunoprecipitation (IP) prior to immunoblotting 26nt intron amplicon contains a Pst1 cleavage site,
(IB) (IP-IB). Alternatively, a Phostag-based western digestion with Pst1 after PCR yields better separation
blot approach can be used. In particular, this method of XBP1u/s with XBP1u producing two fragment of
allows a direct visualization and quantitation of IRE1 ~186 and ~290 bp, whereas the XBP1s corresponds to
signaling in various conditions [4] (Data S1). More- an amplicon of ~450bp (Data S2). In addition, it is
over, IRE1 oligomerization state can be tested to mea- also possible to measure XBP1s mRNA by qPCR
sure its activity, for instance using the superfolder using primers that only recognize XBP1 spliced version
green fluorescent protein (sfGFP)-IRE1 construct in (Table 3) [8].
which the sfGFP is N terminally fused to the luminal A genetic method to monitor XBP1 mRNA splicing
domain of IRE1 (Table 2) [5]. has been developed by fusing XBP1 sequence to venus,
IRE1a phosphorylation causes a conformational a GFP variant or to luciferase (Table 2). Venus/lu-
change leading to the activation of the RNase domain, ciferase was cloned downstream of the 26-nt ER
which in turn leads to the excision of a 26-nt intron stress-specific intron of human XBP1. Similar to
from the X-box-binding protein 1 (XBP1) mRNA. endogenous XBP1, upon ER stress the frame shift
The 50 and 30 fragments are then ligated by the tRNA allows expression of the chimeric XBP1-venus/

Table 1. List of antibodies used to assess ER stress by WB, IF, IHC, and IP. CST, cell signaling technology; SCBT, Santa Cruz
Biotechnology; Ptg, ProteinTech; H, human; M, mouse; R, R. Application score: 1 = good, 2 = medium, 3 = bad.

Antibody Source Reactivity Application Company/Cat#


1 1 1 1
IRE1a Rabbit H, M IP , IF , WB , IHC CST/3294S
IRE1a (B12) Mouse M,R,H IP1, IF1, WB1, IHC1 SCBT/sc390960
p-IRE1 pS724 Rabbit M,R,H WB2 Abcam/ab48187
p-IRE1 pS729 Rabbit M WB1 Tang et al [83]
XBP1s (9D11A43) Mouse H,M IF1, WB1 Biolegends/658802
XBP1s (D2C1F) Mouse H WB2 CST/12782S
RTCB Rabbit H,M IP1, IF1, WB1, IHC1 Ptg/19809-1-AP
PERK (C33E10) Rabbit H,M,R,Mk WB1 CST/3192S
PERK (D11A8) Rabbit H IP2, IF2, WB1 CST/5683S
p-PERK (16F8) (Thr980) Rabbit R WB3 CST/3179S
p-PERK (Thr982) Rabbit H,M IP2, IF1, WB2, IHC1 Abcam/Ab192591
eIF2a (L57A5) Mouse H,M,R,Mk WB1 CST/2103S
p-eIF2a(119A11) (S51) Rabbit H,M,R,Mk WB2 CST/3597S
ATF4 Mouse M,R,H WB1 Sigma-aldrich/ WH0000468M1
ATF6 (1-7) Mouse H IP1, IF1, WB1 Abcam/ab122897
CHOP (L63F7) Mouse H,M,R IP1, IF1, WB1 CST/2895S
BIP (C50B12) Rabbit H,M WB1 CST/3177S
PDIA3 (3G4G7) Mouse H,M,R IP2, IF1, WB1, IHC1 Ptg/66423-1-Ig
SAPK/JNK Rabbit H,M,R WB2 CST/9252S
p-SAPK/JNK(81E11) (Thr183/Tyr185) Rabbit H,M,R WB2 CST/4668S
p-SAPK/JNK(G9) (Thr183/Tyr185) Mouse H,M,R WB2 CST/9255S
TXNIP Mouse H,M,R WB1 MBL/ K0205-3
Insulin (2D11-H5) Mouse H,M,R IP1, IF1, WB1, IHC1 SCBT/8033
PRDX4 Rabbit H,M,R IP1, IF1, WB1, IHC1 Ptg/10703-1-AP
BLOS1 Rabbit H,M,R IP1, IF1, WB1, IHC1 Ptg/19687-1-AP
MANF Rabbit H,M WB1 Bethyl/A305-572A
GADD34/PPP1R15A Rabbit H,M,R IP1, IF1, WB1, IHC1 Ptg/10449-1-AP

The FEBS Journal 287 (2020) 27–42 ª 2019 Federation of European Biochemical Societies 29
Tools to measure ER homeostasis in mammals D. Sicari et al.

Table 2. Chemical and genetic tools to assess ER stress in mammalian cells.

Method Notes Ref

The UPR assays


IRE1a
XBP1s PCR/qPCR/WB/ DnaJ/Hsp40-like genes, EDEM, p58IPK, [7,9,84–88]
Florescence ERdj4, HEDJ, protein disulfide isomerase-P5,
and RAMP4
IRE1a Phosphorylation WB/Phostag pIRE1a antibodies, Phostag Gels –
RIDD WB/qPCR RTN4, PRDX4, Galnt2, Gyltl1b, Pdia4, and [5,10,11,89,90]
Blos1 (see Refs for list of genes)
miRNA qPCR miRNA-17, miRNA-34a, miRNA-96, and [91,92]
miRNA-125b
IRE1a Oligomerization WB/Florescence sfGFP-IRE1a construct [5,93,94]
EGFP-IRE1a construct
Cell death WB/Annexin V CHOP/ pJNK/ TXNIP-NLRP3 [5,13,91,92]
‘ER stress-activated Florescence Transgenic mouse [9]
indicator’ (ERAI)
IRE1a Inhibitors
Kinase and RNAse Inhibitor WB/qPCR on IRE1a and Compound 18, MKC-3946, MKC-8866, STF- [95–98]
IRE1a targets. 083010, 4l8c.
PERK
PERK phosphorylation WB/Phostag p-PERK antibodies, Phostag Gels [16,99]
ATF4 target genes WB/qPCR CHOP/GADD153 and Noxa [19–21]
eIF2a phosphorylation WB peIF2a antibodies [16,99]
NRF2-KEAP1 WB/qPCR of the NRF2 GCLC, NQO1 [22]
target genes
PERK inhibitors
Phosphorylation inhibitors WB/qPCR on PERK and GSK-2606414*, GSK2656157* [102,103]
its targets ISRIB
*also known to inhibit RIPK1 and/or cKIT
[100,101]
ATF6
ATF6f localization WB/Luciferase assay p5xATF6-GL3, Plasmid #11976 [25]
ATF6f target genes WB/qPCR GRP78, GRP94, ERp72, PDIA4, SEL1L, OS9, [104]
HerpUD, and Hyou1
ERSE-LacZ model mouse Florescence Transgenic mouse [29]
model
ATF6 Inhibitor
ATF6-f transport inhibitor, WB/qPCR on ATF6 protein Ceapins, Nelfinavir [105–107]
S1p protease inhibitors and ATF6 targets PF-429242
Assessing the redox state of the ER
Redox state of the ER lumen
Assaying the redox state of Non reducing WB/ eroGFP, ER-rxYFGP, Grx-rxYFP, PDI redox [37–41,43,45–47,51,53]
redox sensitive FPs Fluorescence/FACS/ state, ERO1 redox state
Microscopy
Measuring calcium distribution
Chemical Indicators Fluorescent dyes Fura-2, Fluo3, Fluo4, Calcium Green-1, Indo1, [62–64,108]
Cytosol: Cal-520, etc.
ER lumen Mag-Fura2, Mag-Fluo4, Fluo-4FF, Fluo-5N,
Calcium Green-5N, Magnesium Green NTA
GECIs Florescence/Microscopy CEPIA family, YC family, D1ER and CatchER [68–70]
(TED) Florescence/Microscopy GFPCES3, Fluo 5N/Ca + 2 [109]
Membrane-targeted GECIs Florescence/Microscopy OER-GCaMP6f, Lck-GCaMP6f, LcK-RCaMP2 [73]
Gaussia Luciferase SERCaMP Luminescence SERCaMP [110]

30 The FEBS Journal 287 (2020) 27–42 ª 2019 Federation of European Biochemical Societies
D. Sicari et al. Tools to measure ER homeostasis in mammals

Table 2. (Continued).

Method Notes Ref

Functional changes
Morpho
ER size and shape EM, Fluorescent EM, ER-TrackerTM, DiIC16(3) and DiIC18(3), [74,75]
Microscopy PDI, SERCA, GPR78, etc.
Immunofluorescence GFP-Sec63, Calnexin IF
Function
Secretion Luciferase assay Gaussia luciferase [111]
Misfolded protein aggregation
Protein aggregation Fluorescence ThT [78–82]
The UPR genetics tools
IRE1a
Xbp1s XBP1-venus Monitoring the fluorescence activity of venus [7,9,84–88]
F-XBP1DDBD-venus in the nucleus
IRE1a RNAse activity RFP-GFP fusion Flow cytometry [112]
IRE1a Oligomerization WB/Florescence sfGFP-IRE1a construct [5,93,94]
EGFP-IRE1a construct
IRE1a Transfection IRE1a KA-pcDNA3.EGFP [113]
IRE1a-pcDNA3.EGFP
PERK
NRF2-KEAP1 Luciferase assay Neh2-luciferase to monitor drug-induced Nrf2 [114]
stabilization
ATF6
ATF6f localization Luciferase assay p5xATF6-GL3, Plasmid #11976 [25]
ATF6 localization Immunofluorescence p5xFLAG-ATF6, Plasmid #11974 [115]
ATF6f localization Immunofluorescence pCGN-ATF6 (1-373), Plasmid #27173 [25]
pCGN-ATF6 (1-373)m, Plasmid #27174
ATF6 nuclear localization Western blot/ DHFR:ATF6 [116]
Immunofluorescence,
selective ATF6f induction
Assessing the redox state of the ER
Redox state of the ER lumen
Assaying the redox state of Fluorescence/FACS/ eroGFP, ER-rxYFGP, Grx-rxYFP, [37–41,43,45–47,51,53]
redox sensitive FPs Microscopy/western blot
Measuring calcium distribution
GECIs Florescence/Microscopy CEPIA family, YC family, D1ER and CatchER [68–70]
(TED) Florescence/Microscopy GFPCES3, Fluo 5N/Ca + 2 [109]
Membrane-targeted GECIs Florescence/Microscopy OER-GCaMP6f, Lck-GCaMP6f, LcK-RCaMP2 [73]
Gaussia Luciferase SERCaMP Luminescence SERCaMP [110]

luciferase mRNA [9], which can in turn be detected Once in the nucleus, XBP1s regulate the transcrip-
using either microscopy or luciferase-based light emis- tion of subset of genes that have a cis-acting element
sion, thus making it a suitable method to monitor called UPR element (UPRE/[TGACGTGG/A]). The
Xbp1 splicing in vitro and in vivo models including expression of these target genes can be monitored
mouse, worms, and flies. Another Xbp1-Venus variant using real-time PCR, and primers can be found in
was developed (F-XBP1DDBD-venus), which lacks the Table 3. IRE1 RNase can also regulate the stability of
DNA binding domain of Xbp1. Because it is unable to different RNA through endonucleolytic cleavage of
bind DNA and mainly localize to the cytosol, F- either mRNA encoding for ER targeted proteins,
XBP1DDBD-venus is recommended in experiments mRNA that localize to the ER membrane, or miRNA
that requires overexpression of the reporter without in a mechanism called regulated IRE1-dependent
interfering in a dominant negative way with induction decay (RIDD) of RNA [10–12]. RNA stability can be
of UPR-target genes. At last, the expression of the assayed by measuring the IRE1-dependent decay and
XBP1s protein can be measured using western blotting is usually assessed after pulsing cells with the tran-
approaches (Table 1). scription inhibitor actinomycin D in the presence and

The FEBS Journal 287 (2020) 27–42 ª 2019 Federation of European Biochemical Societies 31
Tools to measure ER homeostasis in mammals D. Sicari et al.

Table 3. List of sequences of published primers used to assess ER stress and the UPR by RT-qPCR and PCR products in human and
mouse samples.

Pathway Gene Human Mouse (50 –30 )

IRE1a
RIDD BLOC1S1 50 -CCCAATTTGCCAAGCAGACA-30 50 -CAAGGAGCTGCAGGAGAAGA-30
50 -CATCCCCAATTTCCTTGAGTGC-30 [117] 50 -GCCTGGTTGAAGTTCTCCAC-30 [11]
SCARA3 50 - CGCTGCCAGAAGAACCTATC-30 50 -TGCATGGATACTGACCCTGA-30
50 - AACCAGAGAGGCCAACACAG-30 [118] 50 -GCCGTGTTACCAGCTTCTTC-30 [11]
PDGFRB TCCATCCCTCTGTTCTCCTG 50 -AACCCCCTTACAGCTGTCCT-30
CTGCCCTCTCCCAGTTATCA [118] 50 -TAATCCCGTCAGCATCTTCC-30 [11]
PMP22 50 -CTGGTCTGTGCGTGATGAGTG-30 50 -TGCGATACAGCAGAATGGAG-30
50 -ATGTAGGCGAAACCGTAGGAG-30 [119] 50 -TTGGTGGCCAATACAAGTCA-30 [11]
Col6A1 50 - CCCTCGTGGACAAAGTCAAG-30 50 -TGCTCAACATGAAGCAGACC-30
50 - GTTTCGGTCACAGCGGTAGT-30 [118] 50 -TTGAGGGAGAAAGCTCTGGA-30 [11]
GALNT10 50 -ACAGCCAGGTAATGGGTGAG-30 50 -CCTTAGAGATGCTGGGATCG-30
50 -GAAGATGGGATGGCTTTTCA-30 [120] 50 -TGAGGACTCAACTCCCCTTG-30 [11]
XBP1s XBP1spl. (PCR) 50 -GGAGTTAAGACAGCGCTTGGGGA-30 50 -AGGAAACTGAAAAACAGAGTAGCAGC-30
50 -TGTTCTGGAGGGGTGACAACTGGG-30 [121] 50 -TCCTTCTGGGTAGACCTCTGG-30 [122]
XBP1s XBP1spl. (qPCR) 50 -TGCTGAGTCCGCAGCAGGTG-30 50 -GAGTCCGCAGCAGGTG-30
miR-17 50 - GCTGGCAGGCTCTGGGGAAG-30 [123] 50 -GTGTCAGAGTC-CATGGGA-30 [124]
MANF 50 -TCACATTCTCACCAGCCACT-30 50 -AGGTCCACTGTGCTCAGGTC-30
50 -CAGGTCGATCTGC TTGTCATAC-30 [125] 50 -CCACCATATCCCTGTGGAAA-30 [126]
TXNIP 50 -CTTGCGGAGTGGCTAAAGTG-30 50 -TCAAGGGCCCCTGGGAACATC-30
50 -TTGAAGGATGTTCCCAGAGG-30 [127] 50 -GACACTGGTGCCATTAAGTCAG-30 [91]
ATF6
ATF6 BIP 50 - TGTTCAACCAATTATCAGCAAACTC-30 50 -TCAGCATCAAGCAAGGATTG-30
50 - TTCTGCTGTATCCTCTTCACCAGT-30 [123] 50 -AAGCCGTGGAGAAGATCTGA-30 [122]
PDIA4 50 -AGTGGGGAGGATGTCAATGC-30 50 -GGGCTCTTTCAGGGAGATGG-30
50 -TGGCTGGGATTTGATGACTG-30 [116] 50 -GGGAGACTTTCAGGAACTTGGC-30 [83]
HYOU1 50 -GCAGACCTGTTGGCACTGAG-30 50 -GTGATAGTGCAGCCGGCAT-30
50 -TCACGATCACCGGTGTTTTC-30 [116] 50 -AACGGAGCGTAGCCTTTGG-30 [104]
SEL1L 50 -ATCTCCAAAAGGCAGCAAGC-30 50 -TGGGTTTTCTCTCTCTCCTCTG-30
50 -TGGGAGAGCCTTCCTCAGTC-30 [116] 50 -CCTTTGTTCCGGTTACTTCTTG-30 [128]
EDEM1 50 -TTCCCTCCTGGTGGAATTTG-30 50 -GGGACCAAGAGGAAAAGTTTG-30
50 -AGGCCACTCTGCTTTCCAAC-30 [116] 50 -GAGGTGAGCAGGTCAAATCAA-30 [128]
HERPUD1 50 - CGTTGTTATGTACCTGCATC-30 50 -CAGTTGGAGTGTGAGT-30
50 - TCAGGAGGAGGACCATCATTT-30 [129] 50 -CAACAGCAGCTTCCCAGAATA-30 [129]
XBP1 50 -TAAGACAGCGCTTGGGGATG-30 50 -AAGAACACGCTTGGGAATGG-30
50 -GCACGTAGTCTGAGTCGTGC-30 [123] 50 -ACTCCCCTTGGCCTCCAC-30 [124]
PERK
ATF4 CHOP 50 - CAGAACCAGCAGAGGTCACA-30 50 -CACATCCCAAAGCCCTCGCTCTC-30
50 - AGCTGTGCCACTTTCCTTTC-30 [123] 50 -TCATGCTTGGTGCAGGCTGACCAT-30 [122]
GADD34 50 - CCTCTACTTCTGCCTTGTCTCCAG-30 50 - CTTTTGGCAACCAGAACCG-30
50 -TTTTCCTCCTTCTTCTCGGACG-30 [118] 50 -CAGAGCCGCAGCTTCTATCT-30 [130]
NOXA 50 -TTTCTTCGGTCACTACACAACG-30 50 -GACAAAGTGAATTTACGGCAGA-30
50 -GAGCATTTTCCGAACCTTTAGA--30 [131] 50 -GGTTTCACGTTATCACAGCTCA-30 [131]
NRF2 HMOX1 50 -ACTGCGTTCCTGCTCAACATC-30 50 -GCCGAGAATGCTGAGTTCATG-30
50 -GCTCTGGTCCTTGGTGTCATG-30 [132] 50 -TGGTACAAGGAAGCCATCACC-30 [132]
NQO1 50 -ATGTATGACAAAGGACCCTTCC-30 50 -CGCCTGAGCCCAGATATTGT-30
50 -TCCCTTGCAGAGAGTACATGG-30 [133] 50 -GCACTCTCTCAAACCAGCCT-30 [132]

absence of ER stressor. As a control, RNAi-mediated total RNA for 2hrs. One good negative control for
silencing of IRE1 is usually used in cells lacking func- this reaction is the use of heat-denatured IRE1. This is
tional XBP1 [11], or alternatively, the IRE1 RNase followed by qRT-PCR with primers flanking regions
activity can be blocked using select pharmacological of potential cleavage sites, and then, the relative
inhibitors (Table 2). For assaying the cleavage of cellu- mRNA levels of each reaction are normalized to
lar RNA in vitro, recombinant IRE1 is incubated with housekeeping gene such as GAPDH. Examples of

32 The FEBS Journal 287 (2020) 27–42 ª 2019 Federation of European Biochemical Societies
D. Sicari et al. Tools to measure ER homeostasis in mammals

RIDD target mRNA and corresponding primers are In addition, since it is well established that other
listed in Table 1. eIF2a kinases exist, other controls should be included
Finally, IRE1 was shown to lead to the activation to confirm PERK-dependent eIF2a phosphorylation.
of JNK1 through interacting with TRAF2, an adaptor Other methods to measure PERK activity could be
protein that couples plasma membrane receptors to c- testing both mRNA and protein levels of downstream
Jun NH2-terminal kinase (JNK) activation. JNK1 targets such as ATF4 and CHOP. PERK activation
phosphorylation can be tested using western blot, as a induces the phosphorylation of the transcription factor
surrogate marker of its activation, using phospho- NRF2. Normally, NRF2 is kept in check by interac-
JNK1 antibodies (Table 1) [13]. tion with the protein KEAP1. Upon phosphorylation
by PERK, NRF2 is liberated from KEAP1 and
translocates to the nucleus where it activates the tran-
The PERK arm
scription of genes encoding antioxidant proteins [22].
PERK is a type I transmembrane protein, which is Thus, NRF2 protein levels and its target genes could
functionally and structurally related to IRE1 (Fig. 1). represent a good strategy to test PERK activity.
The ER luminal portion of PERK contains a stress- Finally, translational attenuation can be useful tool to
sensing domain, and the cytoplasmic portion of PERK test PERK activation by taking advantages from meta-
has a protein kinase domain that is activated when bolic pulse labeling of newly synthesized proteins and
PERK dimerize and/or oligomerize in stressed cells polyribosome profiling [16] (Data S3). Pharmacological
[14,15]. PERK activity is mainly linked to global trans- inhibition of PERK can be achieved using specific
lation inhibition, antioxidative response, and mitochon- compounds as described in Table 2.
drial bioenergetics demands [16–18]. Once PERK is
activated, it phosphorylates the a-subunit of the
The ATF6 arm
eukaryotic translation initiation factor 2 alpha (eIF2a)
to attenuate global translation and to reduce ER-lumen ATF6 is a 90 kDa type II single-pass transmembrane
protein load [16]. To measure PERK activation, PERK protein that is present in two variants in mammalian
and eIF2a phosphorylation status can be detected cells -ATF6a and ATF6b (Fig. 1). During ER stress,
using antibodies for total and phospho-specific proteins ATF6 translocate from the ER to Golgi where it gets
(Table 1). We must note that proper controls (such as cleaved by two serine proteases (S1P and S2P) to gen-
the use of PERK inhibitors) should be carried when erate a 50kDa active basic-leucine zipper (bZip) tran-
assaying eIF2a phosphorylation as it can also be phos- scriptional factor (ATF6f) [23]. ATF6a activation can
phorylated in PERK-independent way through GCN2, be monitored using western blot and specific antibod-
PKR, and HRI. Notably, since phospho-PERK-speci- ies to the C terminus of the protein (Table 1). Upon
fic antibodies do not work well in many cell types and ER stress, ATF6 and ATF6f can be resolved as 90
because of the low abundancy of PERK protein, the and 55 kDa proteins by SDS/PAGE, respectively.
antitotal PERK can be used to measure the band shift Alternatively, cells can be transfected with a 3xFLAG-
obtained upon thapsigargin (Tg), tunicamycin (Tm), or human ATF6a plasmid (Addgene, Cambridge, MA,
dithiothreitol treatments using western blot [4]. Alter- USA, Plasmid #11975) (Table 2) and the activation
natively, immunoprecipitating PERK before IB (IP-IB) (processing) detected using anti-FLAG antibodies
with anti-phospho-PERK antibodies increases the effi- (Table 2). Of note, it is important to ensure the
ciency of this method. amounts of plasmid transfected are appropriate for
Although cap-dependent translation is inhibited, each cell type studied as too much of ATF6 expression
translation of certain transcripts is increased. In partic- promotes its activation.
ular, transcription factor such as ATF4 is selectively In the nucleus, ATF6f promotes transcription of
induced in response to eIF2a phosphorylation [19,20]. UPR-target genes through binding of three different
ATF4 acts as a transcriptional activator of various consensus sequences: (endoplasmic reticulum stress ele-
pro-survival genes including many ER chaperones and ment, ERSE)-I (CCAAT-N9-CCACG/A), ERSE-II
antioxidative factors [21]. Interestingly, ATF4 target (ATTGG-N1-CCACG), and the UPRE [24,25]. ATF6f
genes also include well-known factors that are associ- transcriptional activity can be assayed by measuring
ated with ER stress-induced apoptosis such as CHOP/ the expression and transcription of most of its cognate
GADD153 and NOXA. The expression of ATF4 can targets [26–28]. The expression of these target genes
be measured using western blotting (Table 1), and the can be monitored at the transcript level using real-time
expression of its target genes can be evaluated using PCR (Table 3) or using western blot with specific anti-
real-time PCR (Table 3). bodies (Table 1). Another way to evaluate the ATF6f

The FEBS Journal 287 (2020) 27–42 ª 2019 Federation of European Biochemical Societies 33
Tools to measure ER homeostasis in mammals D. Sicari et al.

activity is by introducing cells with a luciferase con- roGFPs (for reduction- to oxidation-sensitive GFP)
struct that was made by adding five repetitions of a [37,43]. Mechanistically, both rxYFP and roGFPs sur-
specific binding sequence for ATF6f upstream to the face-expose two redox-active cysteines located close to
luciferase gene (p5xATF6-GL3; Addgene, Plasmid the fluorophore whose protonation status is then
#11976) [25]. This construct is highly specific for affected by the structural modifications brought about
ATF6 activity allowing researchers to measure ATF6 by the reversible cysteine oxidation. Consequently, in
activation both in untreated conditions and during ER the oxidized probe, the excitation peak at 400 nm
stress induction. In vivo, the ATF6 activity can be increases at the expense of the peak at 490 nm. The
monitored using the ERSE-LacZ mouse model that ratio of fluorescence after excitation at these two wave-
was constructed by using LacZ reporter gene driven lengths reports on the redox state of the probe and
by the GRP78 promoter [29]. As a control and in thus on the redox potential of the equilibrating couple,
order to control specificity of this system, it is impor- somehow independently of the probe concentration.
tant to use the ERSE model lacking the GRP78 pro- Ratiometric measurement of roGFPs redox status can
moter [29]. The selective pharmacological inhibition or be determined in living cells by FACS, confocal micro-
activation of ATF6 can be achieved using specific scopes, or plate readers and by nonreducing western
drugs as described in Table 2. blotting, after quenching of free cysteines with alkylat-
ing agents and cell lysis (Tables 1 and 2).
rxYFP/roGFPs equilibrates with the GSH/GSSG
Monitoring changes in the ER redox state
couple, a reaction enhanced by fusing the probes to
ER unique redox metabolism supports disulfide bond the glutaredoxin-1 (GRX) enzyme [44–46]. Targeted to
formation in protein substrates. These are catalyzed by the ER, (GRX1-) rxYFP/roGFPs locally reports on
the FAD containing ER oxidase (ERO1), and proteins the reduction potential of GSH (EGSH), which inte-
from the disulfide isomerase (PDI) family, yielding to grates both the extent of ERO1-PDI-dependent oxida-
H2O2 as a by-product [30–34]. Poorly defined ER tion of GSH and of its ER transport. Nevertheless,
reducing pathways are also needed to reduce non-na- measuring EGSH in the ER with rxYFP/roGFPs
tive disulfides and terminally unfolded proteins, thus should be taken cautiously. First, as targets of ERO1-
maintaining/restoring ER homeostasis. To function, PDI, rxYFP/roGFPs may also directly report on their
these are linked to the cytosolic reduction pathways, activity independently of GSH [47,48]. Second, the
namely the glutathione (GSH) and the thioredoxin probe may wrongly report on ER redox modulation if
(TXN) pathways. While the way TXN pathway oper- mistargeted or refluxed from the ER to the cytosol
ates in ER reduction remains enigmatic, GSH is upon stress [49]. A solution is to tether the probe to
imported in the ER where it can reduce PDI and reset the ER membrane [40]. Third, it is critical to consider
ERO1 activity, both of which shall help relieve ER the range of redox potentials the probe linearly reports
stress conditions. Yet, irremediable misfolded protein on, which typically lies within +/ 40 mV from the
accumulation may engage futile cycles of oxidation probe midpoint redox potential [50]. As the ER is
and reduction, triggering H2O2 production and more oxidizing than the cytosol, ER-specific probes
exhausting GSH pools [35]. The UPR control of few with shifted midpoint redox potentials have been
redox enzymes and the sensitivity of the UPR to redox developed [43] by inserting either a leucine or a glu-
cues further highlight the reciprocal interconnection tamic acid at GFP position 147. Because these probes
linking ER stress to redox metabolism [36]. have an intrinsically lower fluorescence, imaging, based
Monitoring ER redox parameters shall thus be on the distinct fluorescence life time between the
informative to rationalize if and how they deviate dur- reduced and oxidized probe [51], and insertion of addi-
ing ER stress, which could then be used as indepen- tional GFP superfolder mutations have been worked
dent measures of ER health. As nonproteinaceous out for these variants [52,53]. Fusing roGFP probes to
molecules, monitoring GSH and H2O2 ER redox state peroxide sensors instead of GRX1 shifts the probe
and levels is challenging. It benefited from the develop- specificity toward H2O2 detection [54–56]. Targeted to
ment of genetically encoded redox sensitive fluorescent the ER, these probes, however, also behave as PDI
proteins (FP), which dynamically measure, in living substrate and thus do not strictly report on ER H2O2
cells and in a given compartment, the redox state of production [57]. To overcome this caveat, several mod-
specific redox couples [37–41]. Stemming from the ini- ifications of disulfide-based reporters and protocols
tial work on the first redox sensitive YFP, redox-sensi- were suggested [57,58]. A better way to solve this issue
tive YFP (rxYFP) [42], the repertoire of these redox would be the development of disulfide independent ER
biosensors now includes a family of ratiometric probes that are actively being searched for [59,60].

34 The FEBS Journal 287 (2020) 27–42 ª 2019 Federation of European Biochemical Societies
D. Sicari et al. Tools to measure ER homeostasis in mammals

Peroxide probes will however never report on H2O2 color palette of these calcium indicators available now
levels as dynamically and accurately as GSH/GSSG makes it easy to observe at the same time, variations
reporting probes. Indeed, the H2O2/ H2O redox couple in calcium in different compartments.
being unproductive in the reduction reaction, the There are plenty of genetically encoded calcium indi-
probe redox state will also depend on in situ reductase cators (GECIs), which consist in general of a calcium
activities, which will differ among cellular compart- binding domain (calmodulin or troponin C), fused to
ments. Alternatively, irreversibly oxidized probe oxida- one or two FP. These markers can be thus divided
tion will not provide dynamic monitoring. Quantifying into two main groups: single-FP vs. double-FP GECIs
H2O2 ER production thus faces important intrinsic (see Ref. [67], for review). In the first case, the fluores-
challenges. Finally, directly reporting the redox state cence intensity of a circularly permuted or split FP is
of ER proteins, especially endogenous ones, shall pro- modulated by calcium binding-dependent changes in
vide a complementary and necessary approach to map the chromophore environment. Single-FP-type GECIs
redox events triggered by ER stress. Different tech- show generally a high dynamic range because they
niques are currently carried out ranging from nonre- show narrow excitation and emission spectra and
ducing western blotting to elaborated mass allow to use them with other fluorescent molecules.
spectrometry analyses [61]. Proteins to be followed Double-FP GECIs, rely on the principle of F€ orster
should include ERO1 and PDI family members, but resonance energy transfer (FRET) is that the fluores-
also disulfide containing substrates to scrutinize oxida- cence variation of the two FPs allows ratiometric mea-
tive folding efficiency. surements. Indeed, Ca2+ binding to the Ca2+ sensitive
element induces a conformational change in the indica-
tor, which modifies the FRET efficiency between the
Measuring calcium distribution
two FPs, resulting in a change in FP fluorescence
Although it is considered mainly as a factory for pro- intensity. Note that certain single-FP indicators can be
tein folding, the ER is the largest Ca2+ store in the used also for ratiometric imaging. The first ER GECIs,
cell. Calcium release from this compartment can be such as the YC3er developed as early as 1997, were
monitored using several chemical and genetic tools. based on FRET and were subsequently improved to
Since the synthesis more than 30 years ago of quin2 generate D1ER family and others YCer [68–70]. More
then fura2, tens of chemical fluorescent markers of cal- recently, several groups introducing mutations in cal-
cium that vary in their Ca2+ dissociation constants cium binding sites of calmodulin have developed dif-
(Kd) or Ca2+ response range, excitation/emission ferent types of single-FP GECIer [71]. Among them,
wavelengths, and spectral shift have been proposed our preference is for the CEPIA family (Table 2). In
[62–64]. Acetomethoxy (AM) derivatives of these our hands, these GECIs show a large dynamic and a
probes allow the dyes to readily enter cells. Once very good signal-to-noise ratio.
inside, nonspecific intracellular esterases cleave the Although it is interesting to measure calcium in the
AM groups off, thus trapping the dyes within the cells ER, it is also important to visualize the calcium com-
[65]. These markers have also a very good signal/noise ing out of the ER. Experiments with chemical fluo-
ratio and a large dynamic range. Moreover, they are rophores have shown that calcium release from ER
easy to calibrate and some of them are ratiometric, occurred at specific ER sites rich in RInsP3 [72] partic-
that is, that upon binding to calcium, they undergo a ularly in areas of contact with mitochondria called
shift either in excitation wavelength, emission wave- MAMs (mitochondria associated membranes). Thus,
length, or both, therefore avoid artifacts, for example, in order to measure Ca2+ release from the ER at sub-
related to the geometry of the cells. The most used cellular resolution, several GECI have been developed,
dyes are the fura2, which allows ratiometric measure- targeted either to the outer ER membrane, or to the
ments, the fluo4 which shows a very good signal/noise outer mitochondria membrane (Table 2) [72,73].
ratio and a newer marker the cal520, which is retained
longer in the cells (Table 2). In the context of the
Assessing ER structure and functions
UPR, the genetic indicators of calcium are probably a
better choice for calcium measurements since they The ER is an organelle of many different functions
allow to observe calcium variations over long periods that must be tightly regulated to carry out the proper
of time, compatible with kinetics of the expression pat- functions. When there is an increase in protein folding
terns of UPR-target genes induced by UPR-inducing demand, translational inhibition, degradation of
compounds such as Tg or Tm, which are observed at unfolded or misfolded proteins, and an increase in the
least 4 h after treatment [66]. In addition, the large production of chaperones and folding enzymes are

The FEBS Journal 287 (2020) 27–42 ª 2019 Federation of European Biochemical Societies 35
Tools to measure ER homeostasis in mammals D. Sicari et al.

part of the adaptive response. In this case, the cell models. We believe that the experimental approaches
must be capable to adjust the morphology, architec- reviewed here should help researcher to better evaluate
ture, dimension, and molecular composition of specific ER protein folding stress in vitro and in vivo. As men-
organelle to meet the increase in protein folding tioned before, we highly recommend not to rely on
demand. The ER has the plasticity to expand its sur- one assay for this task and to try and test as many
face in response to cargo load and in order to increase outputs as possible.
the expression of resident chaperones and other factors
to assist protein folding. The same observation was
Acknowledgements
also documented in the case of B-lymphocytes differ-
entiating into plasma B cells that need to secrete huge This work was funded by grants from Institut
amounts of immunoglobulin. Using electron micro- National du Cancer (INCa PLBIO), Fondation pour
scopy (EM), it was shown that the ER in this case fills la Recherche Medicale (FRM, equipe labellisee 2018),
all the cytoplasm [74,75]. The use of EM or the use of ANR (ERAAT), MSCA RISE-734749 (INSPIRED)
immunofluorescence against ER membrane proteins grants to EC. DS was supported by an AIRC fellow-
including calnexin and Sec63, which are powerful ship for Abroad.
methods to assess the size and shape of the ER, does
not allow to follow the kinetics of the morphological
Conflict of interest
change of this organelle [76]. Fluorescent dyes such as
the long-chain carbocyanines DiIC16(3) and DiIC18(3) EC is a founding member of Cell Stress Discoveries,
have long been used to monitor the ER morphology Ltd. (https://cellstressdiscoveries.com/).
in living cells [77]. In most of the above cases, measur-
ing ER stress needs either introducing of exogenic pro-
Author contributions
teins or assaying mRNA/protein levels indirectly. In
order to be able to look at protein aggregation during DS, ADM, LC, EC, and AI conceptualized and wrote
ER stress, a small molecule called thioflavin T (ThT) the manuscript.
was developed. ThT can give us a direct quantifiable
measure of ER stress in living cell in real time. ThT
References
exhibits enhanced fluorescence when it binds to protein
aggregates—particularly b-sheets—and it correlates 1 Rapoport TA (2007) Protein translocation across the
with the induction of the UPR. ThT can be used in eukaryotic endoplasmic reticulum and bacterial plasma
different cell types and responds to different ER stress membranes. Nature 450, 663–669.
stimulators. It has an advantage that can be assayed 2 Vembar SS & Brodsky JL (2008) One step at a time:
in vivo and in mouse tissue samples without the need endoplasmic reticulum-associated degradation. Nat Rev
to in vivo introduction of an exogenous transgene Mol Cell Biol 9, 944–957.
reporter like measuring the decrease in activity of 3 Oyadomari S, Koizumi A, Takeda K, Gotoh T, Akira
secreted alkaline phosphatase (SEAP), XBP1-Venus, S, Araki E & Mori M (2002) Targeted disruption of
the Chop gene delays endoplasmic reticulum stress-
or the Ca+2 indicator GLuc. Thus, it makes ThT a
mediated diabetes. J Clin Invest 109, 525–532.
more handy technique to rapidly detect and quantify
4 Qi L, Yang L & Chen H (2011) Detecting and
misfolded proteins [78–82]. At last, the ER function in
quantitating physiological endoplasmic reticulum
protein biogenesis can be monitored either by measur-
stress. Methods Enzymol 490, 137–146.
ing the secretion of cargo proteins in the culture med-
5 Ghosh R, Wang L, Wang ES, Perera BG, Igbaria A,
ium using western blot (Table 1) or by using a Morita S, Prado K, Thamsen M, Caswell D, Macias H
secretion reporter with Gaussia luciferase (Table 2). et al. (2014) Allosteric inhibition of the IRE1alpha
RNase preserves cell viability and function during
Conclusion endoplasmic reticulum stress. Cell 158, 534–548.
6 Jurkin J, Henkel T, Nielsen AF, Minnich M, Popow J,
No doubt that the ER plays an important function in Kaufmann T, Heindl K, Hoffmann T, Busslinger M &
development and in different diseases. Monitoring the Martinez J (2014) The mammalian tRNA ligase
folding state of the ER and to be able to measure and complex mediates splicing of XBP1 mRNA and
quantify the stress within this organelle is an impor- controls antibody secretion in plasma cells. EMBO J
tant feature that will help many laboratories that are 33, 2922–2936.
interested to monitor the function of the ER and the 7 Calfon M, Zeng H, Urano F, Till JH, Hubbard SR,
subsequent ER stress response in different disease Harding HP, Clark SG & Ron D (2002) IRE1 couples

36 The FEBS Journal 287 (2020) 27–42 ª 2019 Federation of European Biochemical Societies
D. Sicari et al. Tools to measure ER homeostasis in mammals

endoplasmic reticulum load to secretory capacity by 20 Lu PD, Jousse C, Marciniak SJ, Zhang Y, Novoa I,
processing the XBP-1 mRNA. Nature 415, 92–96. Scheuner D, Kaufman RJ, Ron D & Harding HP
8 van Schadewijk A, van’t Wout EFA, Stolk J & (2004) Cytoprotection by pre-emptive conditional
Hiemstra PS (2012) A quantitative method for phosphorylation of translation initiation factor 2.
detection of spliced X-box binding protein-1 (XBP1) EMBO J 23, 169–179.
mRNA as a measure of endoplasmic reticulum (ER) 21 Avivar-Valderas A, Salas E, Bobrovnikova-Marjon E,
stress. Cell Stress Chaperones 17, 275–279. Diehl JA, Nagi C, Debnath J & Aguirre-Ghiso JA
9 Iwawaki T, Akai R, Kohno K & Miura M (2004) A (2011) PERK integrates autophagy and oxidative stress
transgenic mouse model for monitoring endoplasmic responses to promote survival during extracellular
reticulum stress. Nat Med 10, 98–102. matrix detachment. Mol Cell Biol 31, 3616–3629.
10 Hollien J & Weissman JS (2006) Decay of endoplasmic 22 Cullinan SB & Diehl JA (2004) PERK-dependent
reticulum-localized mRNAs during the unfolded activation of Nrf2 contributes to redox homeostasis
protein response. Science 313, 104–107. and cell survival following endoplasmic reticulum
11 Hollien J, Lin JH, Li H, Stevens N, Walter P & stress. J Biol Chem 279, 20108–20117.
Weissman JS (2009) Regulated Ire1-dependent decay 23 Ye J, Rawson RB, Komuro R, Chen X, Dave UP,
of messenger RNAs in mammalian cells. J Cell Biol Prywes R, Brown MS & Goldstein JL (2000) ER stress
186, 323–331. induces cleavage of membrane-bound ATF6 by the
12 Maurel M, McGrath EP, Mnich K, Healy S, Chevet E same proteases that process SREBPs. Mol Cell 6,
& Samali A (2015) Controlling the unfolded protein 1355–1364.
response-mediated life and death decisions in cancer. 24 Kokame K, Kato H & Miyata T (2001) Identification
Semin Cancer Biol 33, 57–66. of ERSE-II, a new cis-acting element responsible for
13 Urano F, Wang X, Bertolotti A, Zhang Y, Chung P, the ATF6-dependent mammalian unfolded protein
Harding HP & Ron D (2000) Coupling of stress in the response. J Biol Chem 276, 9199–9205.
ER to activation of JNK protein kinases by 25 Wang Y, Shen J, Arenzana N, Tirasophon W,
transmembrane protein kinase IRE1. Science 287, 664– Kaufman RJ & Prywes R (2000) Activation of ATF6
666. and an ATF6 DNA binding site by the endoplasmic
14 Carrara M, Prischi F, Nowak PR, Kopp MC & Ali reticulum stress response. J Biol Chem 275, 27013–
MMU (2015) Noncanonical binding of BiP ATPase 27020.
domain to Ire1 and Perk is dissociated by unfolded 26 Adachi Y, Yamamoto K, Okada T, Yoshida H,
protein CH1 to initiate ER stress signaling. eLife 4, Harada A & Mori K (2008) ATF6 is a transcription
e03522. factor specializing in the regulation of quality control
15 Carrara M, Prischi F, Nowak PR & Ali MM (2015) proteins in the endoplasmic reticulum. Cell Struct
Crystal structures reveal transient PERK luminal Funct 33, 75–89.
domain tetramerization in endoplasmic reticulum stress 27 Yamamoto K, Takahara K, Oyadomari S, Okada T,
signaling. EMBO J 34, 1589–1600. Sato T, Harada A & Mori K (2010) Induction of liver
16 Harding HP, Zhang Y & Ron D (1999) Protein steatosis and lipid droplet formation in ATF6alpha-
translation and folding are coupled by an knockout mice burdened with pharmacological
endoplasmic-reticulum-resident kinase. Nature 397, endoplasmic reticulum stress. Mol Biol Cell 21, 2975–
271–274. 2986.
17 Bobrovnikova-Marjon E, Grigoriadou C, Pytel D, 28 Maiuolo J, Bulotta S, Verderio C, Benfante R &
Zhang F, Ye J, Koumenis C, Cavener D & Diehl JA Borgese N (2011) Selective activation of the
(2010) PERK promotes cancer cell proliferation and transcription factor ATF6 mediates endoplasmic
tumor growth by limiting oxidative DNA damage. reticulum proliferation triggered by a membrane
Oncogene 29, 3881–3895. protein. Proc Natl Acad Sci USA 108, 7832–7837.
18 Balsa E, Soustek MS, Thomas A, Cogliati S, Garcıa- 29 Mao C, Tai WC, Bai Y, Poizat C & Lee AS (2006) In
Poyatos C, Martın-Garcıa E, Jedrychowski M, Gygi vivo regulation of Grp78/BiP transcription in the
SP, Enriquez JA & Puigserver P (2019) ER nutrient embryonic heart: role of the endoplasmic reticulum
stress promote assembly of respiratory chain stress response element and GATA-4. J Biol Chem
supercomplexes through the PERK-eIF2alpha Axis. 281, 8877–8887.
Mol Cell 74, 877–890 e6. 30 Gross E, Sevier CS, Heldman N, Vitu E, Bentzur M,
19 Lu PD, Harding HP & Ron D (2004) Translation Kaiser CA, Thorpe C & Fass D (2006) Generating
reinitiation at alternative open reading frames regulates disulfides enzymatically: reaction products and electron
gene expression in an integrated stress response. J Cell acceptors of the endoplasmic reticulum thiol oxidase
Biol 167, 27–33. Ero1p. Proc Natl Acad Sci USA 103, 299–304.

The FEBS Journal 287 (2020) 27–42 ª 2019 Federation of European Biochemical Societies 37
Tools to measure ER homeostasis in mammals D. Sicari et al.

31 Tu BP & Weissman JS (2002) The FAD- and O(2)- Real-time imaging of the intracellular glutathione
dependent reaction cycle of Ero1-mediated oxidative redox potential. Nat Methods 5, 553–559.
protein folding in the endoplasmic reticulum. Mol Cell 45 Bjornberg O, Ostergaard H & Winther JR (2006)
10, 983–994. Mechanistic insight provided by glutaredoxin within a
32 Bertoli G, Simmen T, Anelli T, Molteni SN, Fesce R fusion to redox-sensitive yellow fluorescent protein.
& Sitia R (2004) Two conserved cysteine triads in Biochemistry 45, 2362–2371.
human Ero1alpha cooperate for efficient disulfide 46 Birk J, Ramming T, Odermatt A & Appenzeller-
bond formation in the endoplasmic reticulum. J Biol Herzog C (2013) Green fluorescent protein-based
Chem 279, 30047–30052. monitoring of endoplasmic reticulum redox poise.
33 Frand AR & Kaiser CA (1998) The ERO1 gene of Front Genet 4, 108.
yeast is required for oxidation of protein dithiols in the 47 Tsunoda S, Avezov E, Zyryanova A, Konno T,
endoplasmic reticulum. Mol Cell 1, 161–170. Mendes-Silva L, Pinho Melo E, Harding HP & Ron D
34 Pollard MG, Travers KJ & Weissman JS (1998) (2014) Intact protein folding in the glutathione-
Ero1p: a novel and ubiquitous protein with an depleted endoplasmic reticulum implicates alternative
essential role in oxidative protein folding in the protein thiol reductants. Elife 3, e03421.
endoplasmic reticulum. Mol Cell 1, 171–182. 48 Avezov E, Konno T, Zyryanova A, Chen W, Laine R,
35 Malhotra JD, Miao H, Zhang K, Wolfson A, Crespillo-Casado A, Melo EP, Ushioda R, Nagata K,
Pennathur S, Pipe SW & Kaufman RJ (2008) Kaminski CF et al. (2015) Retarded PDI diffusion and
Antioxidants reduce endoplasmic reticulum stress and a reductive shift in poise of the calcium depleted
improve protein secretion. Proc Natl Acad Sci USA endoplasmic reticulum. BMC Biol 13, 2.
105, 18525–18530. 49 Igbaria A, Merksamer PI, Trusina A, Tilahun F,
36 Eletto D, Chevet E, Argon Y & Appenzeller-Herzog C Johnson JR, Brandman O, Krogan NJ, Weissman JS
(2014) Redox controls UPR to control redox. J Cell & Papa FR (2019) Chaperone-mediated reflux of
Sci 127 (Pt 17), 3649–3658. secretory proteins to the cytosol during endoplasmic
37 Dooley CT, Dore TM, Hanson GT, Jackson WC, reticulum stress. bioRxiv 562306. “[PREPRINT]”
Remington SJ & Tsien RY (2004) Imaging dynamic 50 Meyer AJ & Dick TP (2010) Fluorescent protein-based
redox changes in mammalian cells with green redox probes. Antioxid Redox Signal 13, 621–650.
fluorescent protein indicators. J Biol Chem 279, 22284– 51 Avezov E, Cross BCS, Kaminski Schierle GS, Winters
22293. M, Harding HP, Melo E, Kaminski CF & Ron D
38 Hanson GT, Aggeler R, Oglesbee D, Cannon M, (2013) Lifetime imaging of a fluorescent protein sensor
Capaldi RA, Tsien RY & Remington SJ (2004) reveals surprising stability of ER thiol redox. J Cell
Investigating mitochondrial redox potential with Biol 201, 337–349.
redox-sensitive green fluorescent protein indicators. J 52 Zhang J, Zhu Q, Wang X, Yu J, Chen X, Wang J,
Biol Chem 279, 13044–13053. Wang X, Xiao J, Wang CC & Wang L (2018)
39 Merksamer PI, Trusina A & Papa FR (2008) Real- Secretory kinase Fam20C tunes endoplasmic reticulum
time redox measurements during endoplasmic redox state via phosphorylation of Ero1alpha. EMBO
reticulum stress reveal interlinked protein folding J 37, e98699
functions. Cell 135, 933–947. 53 Hoseki J, Oishi A, Fujimura T& Sakai Y (2016)
40 Ponsero AJ, Igbaria A, Darch MA, Miled S, Outten Development of a stable ERroGFP variant suitable for
CE, Winther JR, Palais G, D’Autreaux B, Delaunay- monitoring redox dynamics in the ER. Biosci Rep 36,
Moisan A & Toledano MB (2017) Endoplasmic e00316.
reticulum transport of glutathione by Sec61 Is 54 Bilan DS & Belousov VV (2017) New tools for redox
regulated by Ero1 and Bip. Mol Cell 67, 962–973 e5. biology: from imaging to manipulation. Free Radic
41 Ezerina D, Morgan B & Dick TP (2014) Imaging Biol Med 109, 167–188.
dynamic redox processes with genetically encoded 55 Morgan B, Van Laer K, Owusu TNE, Ezerin ß a D,
probes. J Mol Cell Cardiol 73, 43–49. Pastor-Flores D, Amponsah PS, Tursch A & Dick TP
42 Ostergaard H, Tachibana C & Winther JR (2004) (2016) Real-time monitoring of basal H2O2 levels with
Monitoring disulfide bond formation in the eukaryotic peroxiredoxin-based probes. Nat Chem Biol 12, 437–
cytosol. J Cell Biol 166, 337–345. 443.
43 Lohman JR & Remington SJ (2008) Development of a 56 Gutscher M, Sobotta MC, Wabnitz GH, Ballikaya S,
family of redox-sensitive green fluorescent protein Meyer AJ, Samstag Y & Dick TP (2009) Proximity-
indicators for use in relatively oxidizing subcellular based protein thiol oxidation by H2O2-scavenging
environments. Biochemistry 47, 8678–8688. peroxidases. J Biol Chem 284, 31532–31540.
44 Gutscher M, Pauleau A-L, Marty L, Brach T, 57 Melo EP, Lopes C, Gollwitzer P, Lortz S, Lenzen S,
Wabnitz GH, Samstag Y, Meyer AJ & Dick TP (2008) Mehmeti I, Kaminski CF, Ron D & Avezov E (2017)

38 The FEBS Journal 287 (2020) 27–42 ª 2019 Federation of European Biochemical Societies
D. Sicari et al. Tools to measure ER homeostasis in mammals

TriPer, an optical probe tuned to the endoplasmic 71 Suzuki J, Kanemaru K, Ishii K, Ohkura M, Okubo Y &
reticulum tracks changes in luminal H2O2. BMC Biol Iino M (2014) Imaging intraorganellar Ca2+ at
15, 24. subcellular resolution using CEPIA. Nat Commun 5, 4153.
58 Ramming T, Hansen HG, Nagata K, Ellgaard L & 72 Csordas G, Varnai P, Golenar T, Roy S, Purkins G,
Appenzeller-Herzog C (2014) GPx8 peroxidase Schneider TG, Balla T & Hajn oczky G (2010) Imaging
prevents leakage of H2O2 from the endoplasmic interorganelle contacts and local calcium dynamics at
reticulum. Free Radic Biol Med 70, 106–116. the ER-mitochondrial interface. Mol Cell 39, 121–132.
59 Gao C, Tian Y, Zhang R, Jing J & Zhang X (2017) 73 Bannai H,Hirose M, Niwa F & Mikoshiba K (2019)
Endoplasmic reticulum-directed ratiometric fluorescent Dissection of local Ca2+ signals in cultured cells by
probe for quantitive detection of basal H2O2. Anal membrane-targeted Ca2+ indicators. J Vis Exp 145,
Chem 89, 12945–12950. e59246. https://doi.org/10.3791/59246
60 Xiao H, Li P, Hu X, Shi X, Zhang W & Tang B 74 van Anken E, Romijn EP, Maggioni C, Mezghrani A,
(2016) Simultaneous fluorescence imaging of hydrogen Sitia R, Braakman I & Heck AJR (2003) Sequential
peroxide in mitochondria and endoplasmic reticulum waves of functionally related proteins are expressed
during apoptosis. Chem Sci 7, 6153–6159. when B cells prepare for antibody secretion. Immunity
61 Chiappetta G, Ndiaye S, Igbaria A, Kumar C, Vinh J 18, 243–253.
& Toledano MB (2010) Proteome screens for Cys 75 Federovitch CM, Ron D & Hampton RY (2005) The
residues oxidation: the redoxome. Methods Enzymol dynamic ER: experimental approaches and current
473, 199–216. questions. Curr Opin Cell Biol 17, 409–414.
62 Bootman MD, Rietdorf K, Collins T, Walker S & 76 Schuck S, Prinz WA, Thorn KS, Voss C& Walter P
Sanderson M (2013) Ca2+-sensitive fluorescent dyes (2009) Membrane expansion alleviates endoplasmic
and intracellular Ca2+ imaging. Cold Spring Harb reticulum stress independently of the unfolded protein
Protoc 2013, 83–99. response. J Cell Biol 187, 525–536.
63 Minta A, Kao JP & Tsien RY (1989) Fluorescent 77 Terasaki M, Chen LB & Fujiwara K (1986)
indicators for cytosolic calcium based on rhodamine Microtubules and the endoplasmic reticulum are highly
and fluorescein chromophores. J Biol Chem 264, 8171– interdependent structures. J Cell Biol 103, 1557–1568.
8178. 78 Sigurdsson EM (2005) Histological staining of
64 Paredes RM, Etzler JC, Watts LT, Zheng W & amyloid-beta in mouse brains. Methods Mol Biol 299,
Lechleiter JD (2008) Chemical calcium indicators. 299–308.
Methods 46, 143–151. 79 Lindgren M, Sorgjerd K & Hammarstrom P (2005)
65 Oakes SG, Martin WJ, Lisek CA & Powis G (1988) Detection and characterization of aggregates,
Incomplete hydrolysis of the calcium indicator prefibrillar amyloidogenic oligomers, and protofibrils
precursor fura-2 pentaacetoxymethyl ester (fura-2 AM) using fluorescence spectroscopy. Biophys J 88, 4200–
by cells. Anal Biochem 169, 159–166. 4212.
66 Shinjo S, Mizotani Y, Tashiro E& Imoto M (2013) 80 Naiki H, Higuchi K, Hosokawa M & Takeda T (1989)
Comparative analysis of the expression patterns of Fluorometric determination of amyloid fibrils in vitro
UPR-target genes caused by UPR-inducing using the fluorescent dye, thioflavin T1. Anal Biochem
compounds. Biosci Biotechnol Biochem 77, 729–735. 177, 244–249.
67 Suzuki J, Kanemaru K & Iino M (2016) Genetically 81 Vassar PS & Culling CF (1959) Fluorescent stains,
encoded fluorescent indicators for organellar calcium with special reference to amyloid and connective
imaging. Biophys J 111, 1119–1131. tissues. Arch Pathol 68, 487–498.
68 Palmer AE, Jin C, Reed JC & Tsien RY (2004) Bcl-2- 82 Beriault DR & Werstuck GH (2013) Detection and
mediated alterations in endoplasmic reticulum Ca2+ quantification of endoplasmic reticulum stress in living
analyzed with an improved genetically encoded cells using the fluorescent compound, Thioflavin T.
fluorescent sensor. Proc Natl Acad Sci USA 101, Biochim Biophys Acta 1833, 2293–2301.
17404–17409. 83 Tang C-HA, Chang S, Paton AW, Paton JC,
69 Tang S, Wong H-C, Wang Z-M, Huang Y, Zou J, Gabrilovich DI, Ploegh HL, Del Valle JR & Hu C-CA
Zhuo Y, Pennati A, Gadda G, Delbono O & Yang JJ (2018) Phosphorylation of IRE1 at S729 regulates
(2011) Design and application of a class of sensors to RIDD in B cells and antibody production after
monitor Ca2+ dynamics in high Ca2+ concentration immunization. J Cell Biol 217, 1739–1755.
cellular compartments. Proc Natl Acad Sci USA 108, 84 Brunsing R, Omori SA, Weber F, Bicknell A, Friend
16265–16270. L, Rickert R & Niwa M (2008) B- and T-cell
70 Whitaker M (2010) Genetically encoded probes for development both involve activity of the unfolded
measurement of intracellular calcium. Methods Cell protein response pathway. J Biol Chem 283, 17954–
Biol 99, 153–182. 17961.

The FEBS Journal 287 (2020) 27–42 ª 2019 Federation of European Biochemical Societies 39
Tools to measure ER homeostasis in mammals D. Sicari et al.

85 Acosta-Alvear D, Zhou Y, Blais A, Tsikitis M, Lents selective inhibition of unconventional mRNA splicing
NH, Arias C, Lennon CJ, Kluger Y & Dynlacht BD by an IRE1-binding small molecule. Proc Natl Acad
(2007) XBP1 controls diverse cell type- and condition- Sci USA 109, E869–E878.
specific transcriptional regulatory networks. Mol Cell 97 Papandreou I, Denko NC, Olson M, Van Melckebeke
27, 53–66. H, Lust S, Tam A, Solow-Cordero DE, Bouley DM,
86 Lee AH, Iwakoshi NN & Glimcher LH (2003) XBP-1 Offner F, Niwa M et al. (2011) Identification of an
regulates a subset of endoplasmic reticulum resident Ire1alpha endonuclease specific inhibitor with cytotoxic
chaperone genes in the unfolded protein response. Mol activity against human multiple myeloma. Blood 117,
Cell Biol 23, 7448–7459. 1311–1314.
87 Shen X, Ellis E, Lee K, Liu C-Y, Yang K, Solomon 98 Mimura N, Fulciniti M, Gorgun G, Tai YT, Cirstea
A, Yoshida H, Morimoto R, Kurnit DM, Mori K D, Santo L, Hu Y, Fabre C, Minami J, Ohguchi H
et al. (2001) Complementary signaling pathways et al. (2012) Blockade of XBP1 splicing by inhibition
regulate the unfolded protein response and are of IRE1alpha is a promising therapeutic option in
required for C. elegans development. Cell 107, 893– multiple myeloma. Blood 119, 5772–5781.
903. 99 Harding HP, Zhang Y, Bertolotti A, Zeng H & Ron
88 Yoshida H, Matsui T, Yamamoto A, Okada T & D (2000) Perk is essential for translational regulation
Mori K (2001) XBP1 mRNA is induced by ATF6 and and cell survival during the unfolded protein response.
spliced by IRE1 in response to ER stress to produce a Mol Cell 5, 897–904.
highly active transcription factor. Cell 107, 881–891. 100 Rojas-Rivera D, Delvaeye T, Roelandt R, Nerinckx
89 Han D, Lerner AG, Vande Walle L, Upton JP, Xu W, W, Augustyns K, Vandenabeele P& Bertrand MJM
Hagen A, Backes BJ, Oakes SA & Papa FR (2009) (2017) When PERK inhibitors turn out to be new
IRE1alpha kinase activation modes control alternate potent RIPK1 inhibitors: critical issues on the
endoribonuclease outputs to determine divergent cell specificity and use of GSK2606414 and GSK2656157.
fates. Cell 138, 562–575. Cell Death Differ 24, 1100–1110.
90 Moore K & Hollien J (2015) Ire1-mediated decay in 101 Mahameed M, Wilhelm T, Darawshi O, Obiedat A,
mammalian cells relies on mRNA sequence, structure, Tommy W-S, Chintha C, Schubert T, Samali A,
and translational status. Mol Biol Cell 26, 2873–2884. Chevet E, Eriksson LA et al. (2019) The unfolded
91 Lerner AG, Upton JP, Praveen PV, Ghosh R, protein response modulators GSK2606414 and KIRA6
Nakagawa Y, Igbaria A, Shen S, Nguyen V, Backes are potent KIT inhibitors. Cell Death Dis 10, 300.
BJ, Heiman M et al. (2012) IRE1alpha induces 102 Sidrauski C, Acosta-Alvear D, Khoutorsky A,
thioredoxin-interacting protein to activate the NLRP3 Vedantham P, Hearn BR, Li H, Gamache K,
inflammasome and promote programmed cell death Gallagher CM, Ang KK-H, Wilson C et al. (2013)
under irremediable ER stress. Cell Metab 16, 250–264. Pharmacological brake-release of mRNA translation
92 Upton JP, Wang L, Han D, Wang ES, Huskey NE, enhances cognitive memory. Elife 2, e00498.
Lim L, Truitt M, McManus MT, Ruggero D, Goga A 103 Axten JM, Medina JR, Feng Y, Shu A, Romeril SP,
et al. (2012) IRE1alpha cleaves select microRNAs Grant SW, Li WH, Heerding DA, Minthorn E,
during ER stress to derepress translation of Mencken T et al. (2012) Discovery of 7-methyl-5-(1-
proapoptotic Caspase-2. Science 338, 818–822. {[3-(trifluoromethyl)phenyl]acetyl}-2,3-dihydro-1H-
93 Korennykh AV, Egea PF, Korostelev AA, Finer-Moore indol-5-yl)-7H-p yrrolo[2,3-d]pyrimidin-4-amine
J, Zhang C, Shokat KM, Stroud RM & Walter P (GSK2606414), a potent and selective first-in-class
(2009) The unfolded protein response signals through inhibitor of protein kinase R (PKR)-like endoplasmic
high-order assembly of Ire1. Nature 457, 687–693. reticulum kinase (PERK). J Med Chem 55, 7193–7207.
94 Li H, Korennykh AV, Behrman Sl & Walter P (2010) 104 Belmont PJ, Tadimalla A, Chen WJ, Martindale JJ,
Mammalian endoplasmic reticulum stress sensor IRE1 Thuerauf DJ, Marcinko M, Gude N, Sussman MA &
signals by dynamic clustering. Proc Natl Acad Sci USA Glembotski CC (2008) Coordination of growth and
107, 16113–16118. endoplasmic reticulum stress signaling by regulator of
95 Harrington PE, Biswas K, Malwitz D, Tasker AS, calcineurin 1 (RCAN1), a novel ATF6-inducible gene.
Mohr C, Andrews KL, Dellamaggiore K, Kendall R, J Biol Chem 283, 14012–14021.
Beckmann H, Jaeckel P et al. (2015) Unfolded protein 105 Guan M, Fousek K & Chow WA (2012) Nelfinavir
response in cancer: IRE1alpha inhibition by selective inhibits regulated intramembrane proteolysis of sterol
kinase ligands does not impair tumor cell viability. regulatory element binding protein-1 and activating
ACS Med Chem Lett 6, 68–72. transcription factor 6 in castration-resistant prostate
96 Cross BC, Bond PJ, Sadowski PG, Jha BK, Zak J, cancer. FEBS J 279, 2399–2411.
Goodman JM, Silverman RH, Neubert TA, Baxendale 106 Hay BA, Abrams B, Zumbrunn AY, Valentine JJ,
IR, Ron D et al. (2012) The molecular basis for Warren LC, Petras SF, Shelly LD, Xia A, Varghese

40 The FEBS Journal 287 (2020) 27–42 ª 2019 Federation of European Biochemical Societies
D. Sicari et al. Tools to measure ER homeostasis in mammals

AH, Hawkins JL et al. (2007) Aminopyrrolidineamide Papadodima O, Jouan F et al. (2018) Dual IRE1
inhibitors of site-1 protease. Bioorg Med Chem Lett RNase functions dictate glioblastoma development.
17, 4411–4414. EMBO Mol Med 10, e7929.
107 Gallagher CM, Garri C, Cain EL, Ang KK, Wilson 119 Cai W, Chen G, Luo Q, Liu J, Guo X, Zhang T, Ma
CG, Chen S, Hearn BR, Jaishankar P, Aranda-Diaz F, Yuan L, Li B & Cai J (2017) PMP22 regulates self-
A, Arkin MR et al. (2016) Ceapins are a new class of renewal and chemoresistance of gastric cancer cells.
unfolded protein response inhibitors, selectively Mol Cancer Ther 16, 1187–1198.
targeting the ATF6alpha branch. eLife 5, e11878. 120 Wu Q, Liu HO, Liu YD, Liu WS, Pan D, Zhang WJ,
108 Grynkiewicz G, Poenie M & Tsien RY (1985) A new Yang L, Fu Q, Xu JJ & Gu JX (2015) Decreased
generation of Ca2+ indicators with greatly improved expression of hepatocyte nuclear factor 4alpha
fluorescence properties. J Biol Chem 260, 3440–3450. (Hnf4alpha)/microRNA-122 (miR-122) axis in
109 Rehberg M, Lepier A, Solchenberger B, Osten P & hepatitis B virus-associated hepatocellular carcinoma
Blum R (2008) A new non-disruptive strategy to target enhances potential oncogenic GALNT10 protein
calcium indicator dyes to the endoplasmic reticulum. activity. J Biol Chem 290, 1170–1185.
Cell Calcium 44, 386–399. 121 Kaser A, Lee A-H, Franke A, Glickman JN, Zeissig
110 Henderson MJ, Wires ES, Trychta KA, Yan X & S, Tilg H, Nieuwenhuis EES, Higgins DE, Schreiber S,
Harvey BK (2015) Monitoring endoplasmic reticulum Glimcher LH et al. (2008) XBP1 links ER stress to
calcium homeostasis using a Gaussia Luciferase intestinal inflammation and confers genetic risk for
SERCaMP. J Vis Exp 53199. human inflammatory bowel disease. Cell 134, 743–756.
111 Badr CE, Hewett JW, Breakefield XO & Tannous BA 122 Thamsen M, Ghosh R, Auyeung VC, Brumwell A,
(2007) A highly sensitive assay for monitoring the Chapman HA, Backes BJ, Perara G, Maly DJ,
secretory pathway and ER stress. PLoS ONE 2, e571. Sheppard D & Papa FR (2019) Small molecule
112 Roy G, Zhang S, Li L, Higham E, Wu H, Marelli M inhibition of IRE1alpha kinase/RNase has anti-fibrotic
& Bowen MA (2017) Development of a fluorescent effects in the lung. PLoS ONE 14, e0209824.
reporter system for monitoring ER stress in Chinese 123 Sicari D, Fantuz M, Bellazzo A, Valentino E,
hamster ovary cells and its application for therapeutic Apollonio M, Pontisso I, Di Cristino F, Dal Ferro M,
protein production. PLoS ONE 12, e0183694. Bicciato S, Del Sal G et al. (2019) Mutant p53
113 Lipson KL, Fonseca SG, Ishigaki S, Nguyen LX, Foss improves cancer cells’ resistance to endoplasmic
E, Bortell R, Rossini AA & Urano F (2006) reticulum stress by sustaining activation of the UPR
Regulation of insulin biosynthesis in pancreatic beta regulator ATF6. Oncogene 38, 6184–6195
cells by an endoplasmic reticulum-resident protein 124 Zhang K & Kaufman RJ (2008) Identification and
kinase IRE1. Cell Metab 4, 245–254. characterization of endoplasmic reticulum stress-
114 Smirnova NA, Haskew-Layton RE, Basso M, induced apoptosis in vivo. Methods Enzymol 442, 395–
Hushpulian DM, Payappilly JB, Speer RE, Ahn Y-H, 419.
Rakhman I, Cole PA, Pinto JT et al. (2011) 125 Xu S, Di Z, He Y, Wang R, Ma Y, Sun R, Li J,
Development of Neh2-luciferase reporter and its Wang T, Shen Y, Fang S et al. (2019) Mesencephalic
application for high throughput screening and real- astrocyte-derived neurotrophic factor (MANF)
time monitoring of Nrf2 activators. Chem Biol 18, protects against Abeta toxicity via attenuating Abeta-
752–765. induced endoplasmic reticulum stress. J
115 Zhu C, Johansen FE & Prywes R (1997) Interaction of Neuroinflammation 16, 35.
ATF6 and serum response factor. Mol Cell Biol 17, 126 Morita S, Villalta SA, Feldman HC, Register AC,
4957–4966. Rosenthal W, Hoffmann-Petersen IT, Mehdizadeh M,
116 Shoulders MD, Ryno LM, Genereux JC, Moresco JJ, Ghosh R, Wang L, Colon-Negron K et al. (2017)
Tu PG, Wu C, Yates JR, Su AI, Kelly JW & Targeting ABL-IRE1alpha signaling spares ER-
Wiseman RL (2013) Stress-independent activation of stressed pancreatic beta cells to reverse autoimmune
XBP1s and/or ATF6 reveals three functionally diverse diabetes. Cell Metab 25, 883–897 e8.
ER proteostasis environments. Cell Rep 3, 1279–1292. 127 Wei M, Jiao D, Han D, Wu J, Wei F, Zheng G, Guo
117 Bright MD, Itzhak DN, Wardell CP, Morgan GJ & Z, Xi W, Yang F, Xie P et al. (2017) Knockdown of
Davies FE (2015) Cleavage of BLOC1S1 mRNA by RNF2 induces cell cycle arrest and apoptosis in
IRE1 Is sequence specific, temporally separate from prostate cancer cells through the upregulation of
XBP1 splicing, and dispensable for cell viability under TXNIP. Oncotarget 8, 5323–5338.
acute endoplasmic reticulum stress. Mol Cell Biol 35, 128 Sun S, Shi G, Han X, Francisco AB, Ji Y, Mendoncßa
2186–2202. N, Liu X, Locasale JW, Simpson KW, Duhamel GE
118 Lhomond S, Avril T, Dejeans N, Voutetakis K, et al. (2014) Sel1L is indispensable for mammalian
Doultsinos D, McMahon M, Pineau R, Obacz J, endoplasmic reticulum-associated degradation,

The FEBS Journal 287 (2020) 27–42 ª 2019 Federation of European Biochemical Societies 41
Tools to measure ER homeostasis in mammals D. Sicari et al.

endoplasmic reticulum homeostasis, and survival. Proc Dalgaard K et al. (2014) Heme oxygenase-1 drives
Natl Acad Sci USA 111, E582– E591. metaflammation and insulin resistance in mouse and
129 Paredes F, Parra V, Torrealba N, Navarro-Marquez man. Cell 158, 25–40.
M, Gatica D, Bravo-Sagua R, Troncoso R, Pennanen 133 Lisek K, Campaner E, Ciani Y, Walerych D& Del Sal
C, Quiroga C, Chiong M et al. (2016) HERPUD1 G (2018) Mutant p53 tunes the NRF2-dependent
protects against oxidative stress-induced apoptosis antioxidant response to support survival of cancer
through downregulation of the inositol 1,4,5- cells. Oncotarget 9, 20508–20523.
trisphosphate receptor. Free Radic Biol Med 90, 206–
218.
130 Nishio N & Isobe K (2015) GADD34-deficient mice
Supporting information
develop obesity, nonalcoholic fatty liver disease, Additional supporting information may be found
hepatic carcinoma and insulin resistance. Sci Rep 5, online in the Supporting Information section at the end
13519. of the article.
131 Wang Q, Mora-Jensen H, Weniger M a, Perez-Galan
P, Wolford C, Hai T, Ron D, Chen W, Trenkle W, Data S1. Protocol to assess IRE1a and PERK phos-
Wiestner A et al. (2009) ERAD inhibitors integrate
phorylation using PhosTag technique.
ER stress with an epigenetic mechanism to activate
Data S2. Protocol to assess Xbp1 splicing using qPCR
BH3-only protein NOXA in cancer cells. Proc Natl
and semi-quantitative RT-PCR.
Acad Sci USA 106, 2200–2205.
Data S3. Protocol to assess PERK-dependent transla-
132 Jais A, Einwallner E, Sharif O, Gossens K, Lu TT-H,
tional control using 35S-Met incorporation.
Soyal SM, Medgyesi D, Neureiter D, Paier-Pourani J,

42 The FEBS Journal 287 (2020) 27–42 ª 2019 Federation of European Biochemical Societies

You might also like