Introduction
The nuclear factor (erythroid 2)-like (NRF)
5The abbreviations used are:
NRF
nuclear factor (erythroid 2)-like
ARE
antioxidant-response element
bZip
basic leucine zipper
ChIP
chromatin immunoprecipitation
GO
gene ontology
LRE
likely regulatory element
NQO1
NAD(P)H quinone dehydrogenase 1
sMaf
small musculoaponeurotic fibrosarcoma
TSS
transcription start site
ANOVA
analysis of variance
qPCR
quantitative PCR
ER
endoplasmic reticulum
GAPDH
glyceraldehyde-3-phosphate dehydrogenase
RNA-Seq
RNA-sequencing
exo
exonuclease
CNC
cap'n'collar
ChIP-Seq
ChIP-sequencing
GPI
glycosylphosphatidylinositol
DST
dystonin.
transcription factors are a subset of basic leucine zipper (bZip) cap’n’collar (CNC) transcription factors sharing close homology. There are three NRF family members: NRF1, NRF2, and NRF3 (
1- Motohashi H.
- O'Connor T.
- Katsuoka F.
- Engel J.D.
- Yamamoto M.
Integration and diversity of the regulatory network composed of Maf and CNC families of transcription factors.
). The discovery of different NRF family members followed an effort to find transcription factors controlling expression of β-globin, a gene required for hemoglobin production and proper oxygen transport in vertebrates. NRF1 was the first discovered NRF family member and was identified in a cDNA library screen for binding to the erythroid-specific NF-E2/AP1 consensus sequence found in the 5′ β-globin locus control region, 5′-GCTGAGTCA-3′ (
2- Chan J.Y.
- Han X.-L.
- Kan Y.W.
Cloning of Nrf1, an NF-E2-related transcription factor, by genetic selection in yeast.
). Shortly thereafter, NRF2 was identified in a comparable cDNA library screen (
3- Moi P.
- Chan K.
- Asunis I.
- Cao A.
- Kan Y.W.
Isolation of NF-E2-related factor 2 (Nrf2), a NF-E2-like basic leucine zipper transcriptional activator that binds to the tandem NF-E2/AP1 repeat of the β-globin locus control region.
). Intriguingly, the isolation and characterization of NRF3 arose a few years later following a logical deduction: as the genomic locations of
nrf1,
nrf2, and the NF-E2 subunit
p45 (NF-E2p45) were found near
hoxB,
hoxD, and
hoxC genes, respectively,
nrf3 was predicted and shown to map close to the
hoxA gene (
4- Kobayashi A.
- Ito E.
- Toki T.
- Kogame K.
- Takahashi S.
- Igarashi K.
- Hayashi N.
- Yamamoto M.
Molecular cloning and functional characterization of a new Cap'n'collar family transcription factor Nrf3.
). Thus, the CNC families of NF-E2, NRF1, NRF2, and NRF3 were all predicted to have arisen from a common genetic lineage, diverging after chromosomal duplication.
Almost immediately after the discovery of NF-E2, NRF1, and NRF2, investigators began dissecting the differing roles of these three transcription factors. As they had all been discovered as mediators of β-globin expression, initial investigations looked into their roles in erythropoiesis and hematology.
NF-E2p45 knockout mice exhibited an absence of platelets and hemorrhaging, but had very little effect on erythroid cell lineages and were able to survive to adulthood (
5- Shivdasani R.A.
- Orkin S.H.
Erythropoiesis and globin gene expression in mice lacking the transcription factor NF-E2.
). In contrast,
NRF1 knockout mice displayed severe anemia due to impaired fetal liver erythropoiesis and died
in utero (
6- Chan J.Y.
- Kwong M.
- Lu R.
- Chang J.
- Wang B.
- Yen T.S.
- Kan Y.W.
Targeted disruption of the ubiquitous CNC-bZIP transcription factor, Nrf-1, results in anemia and embryonic lethality in mice.
,
7- Farmer S.C.
- Sun C.W.
- Winnier G.E.
- Hogan B.L.
- Townes T.M.
The bZIP transcription factor LCR-F1 is essential for mesoderm formation in mouse development.
). Homozygous
Nrf2 knockout mice, however, displayed no obvious defects in embryogenesis, fertility, or litter sizes (
8- Chan K.
- Lu R.
- Chang J.C.
- Kan Y.W.
NRF2, a member of the NFE2 family of transcription factors, is not essential for murine erythropoiesis, growth, and development.
). The role of NRF2 became more apparent following challenge with carcinogens, where it was initially shown to protect mice against chemical carcinogenesis. Similarly,
Nrf3 knockout mice displayed no overt phenotype (
9- Derjuga A.
- Gourley T.S.
- Holm T.M.
- Heng H.H.
- Shivdasani R.A.
- Ahmed R.
- Andrews N.C.
- Blank V.
Complexity of CNC transcription factors as revealed by gene targeting of the Nrf3 locus.
). Further insight into the role of Nrf3 may become more apparent after appropriate challenges: for example,
Nrf3−/− animals were more likely to develop cell lymphoblastic lymphoma following benzo-
a-pyrene exposure (
10- Chevillard G.
- Paquet M.
- Blank V.
Nfe2l3 (Nrf3) deficiency predisposes mice to T-cell lymphoblastic lymphoma.
). The NRF transcription factors also differed in their tissue localization;
Nrf1 and
Nrf2 are expressed relatively ubiquitously, whereas
Nrf3 expression is highly localized to placental tissue (
2- Chan J.Y.
- Han X.-L.
- Kan Y.W.
Cloning of Nrf1, an NF-E2-related transcription factor, by genetic selection in yeast.
,
3- Moi P.
- Chan K.
- Asunis I.
- Cao A.
- Kan Y.W.
Isolation of NF-E2-related factor 2 (Nrf2), a NF-E2-like basic leucine zipper transcriptional activator that binds to the tandem NF-E2/AP1 repeat of the β-globin locus control region.
4- Kobayashi A.
- Ito E.
- Toki T.
- Kogame K.
- Takahashi S.
- Igarashi K.
- Hayashi N.
- Yamamoto M.
Molecular cloning and functional characterization of a new Cap'n'collar family transcription factor Nrf3.
).
Although these early studies into the differential underpinnings of the NRF members showed stark contrasts in expression and mediation of downstream responses between family members, mechanistic investigations into their transcriptional modes of action indicated a more conserved process. To control transcription, all CNC members utilize a similar mechanism. Nuclear CNC transcription factors dimerize with the small musculoaponeurotic fibrosarcoma, or sMaf proteins (sMafG, sMafK, and sMafF), to bind to DNA. The resulting sMaf/bZip factor complexes have the capacity to bind to several related target DNA sequences: the palindromic Maf recognition element (5′-TGCTGAC(G)TCAGCA-3′), the NF-E2 recognition element (5′-TGCTGACTCAT-3′), and the antioxidant-response element (ARE, 5′-TGABNNNGC-3′) (
1- Motohashi H.
- O'Connor T.
- Katsuoka F.
- Engel J.D.
- Yamamoto M.
Integration and diversity of the regulatory network composed of Maf and CNC families of transcription factors.
,
11- Toki T.
- Itoh J.
- Kitazawa J.
- Arai K.
- Hatakeyama K.
- Akasaka J.
- Igarashi K.
- Nomura N.
- Yokoyama M.
- Yamamoto M.
- Ito E.
Human small Maf proteins form heterodimers with CNC family transcription factors and recognize the NF-E2 motif.
,
12Small Maf proteins in mammalian gene control: mere dimerization partners or dynamic transcriptional regulators?.
). Historically, genes under ARE control were of particular interest; many of these genes were transcriptionally inducible by electrophilic xenobiotics and consisted predominately of detoxifying enzymes like GSH–
S-transferase or NAD(P)H quinone dehydrogenase 1 (NQO1) (
13- Rushmore T.H.
- King R.G.
- Paulson K.E.
- Pickett C.B.
Regulation of glutathione S-transferase Ya subunit gene expression: identification of a unique xenobiotic-responsive element controlling inducible expression by planar aromatic compounds.
,
14- Rushmore T.H.
- Morton M.R.
- Pickett C.B.
The antioxidant responsive element. Activation by oxidative stress and identification of the DNA consensus sequence required for functional activity.
15Transcriptional regulation of the rat glutathione S-transferase Ya subunit gene. Characterization of a xenobiotic-responsive element controlling inducible expression by phenolic antioxidants.
). In a paradigm-shifting work, ARE-containing genes were discovered to be under the transcriptomic control of NRF members, particularly NRF2 (
16- Itoh K.
- Chiba T.
- Takahashi S.
- Ishii T.
- Igarashi K.
- Katoh Y.
- Oyake T.
- Hayashi N.
- Satoh K.
- Hatayama I.
- Yamamoto M.
- Nabeshima Y.
An Nrf2/small Maf heterodimer mediates the induction of phase II detoxifying enzyme genes through antioxidant-response elements.
).
Given the distinct, and occasionally conflicting, roles that have been attributed to the NRF family members, we combined ChIP and RNA-Seq analyses to catalog the overlapping and distinct genes regulated by each NRF family member, providing insights into the transcriptional program governing the NRF family members.
Discussion
Here, we present a comprehensive comparison of NRF1, NRF2, and NRF3 transcriptional targets by comparing differentially-regulated genes determined from RNA-Seq with genes harboring nearby NRF-binding sites as determined by ChIP-exo sequencing. Although several genes were up-regulated or down-regulated by all three members, each member also individually regulated its own subset of genes. Indeed, 235 of the 262 high-confidence genes were regulated differently by the NRF members. Based on this, it is likely that the observed transcriptional differences were determined by disparities in DNA-binding sequence preference, upstream-signaling pathways, and specific cellular contexts (i.e. biochemical/xenobiotic/nutritional status, cofactor availability, competitive transcriptional programs, etc.). These features dictate which NRF family member becomes activated, where it binds in the genome, and what binding partners are available for determining gene induction or repression.
Supporting this, the known regulatory networks governing the different NRF family members appear to vary significantly. This is due in part to the dominant role that the discovery of NRF2 mediation of the antioxidant response had on the field, as investigations on NRF members subsequently diverged into disparate studies that generally focused on individual members. Furthermore, localization of each NRF member also plays a key role in dictating to which context each member is activated. For example, Nrf1 resides in an inactive, glycosylated state in the endoplasmic reticulum (
22Nrf1 is targeted to the endoplasmic reticulum membrane by an N-terminal transmembrane domain Inhibition of nuclear translocation and transacting function.
,
24- Zhang Y.
- Lucocq J.M.
- Yamamoto M.
- Hayes J.D.
The NHB1 (N-terminal homology box 1) sequence in transcription factor Nrf1 is required to anchor it to the endoplasmic reticulum and also to enable its asparagine-glycosylation.
,
25- Zhang Y.
- Crouch D.H.
- Yamamoto M.
- Hayes J.D.
Negative regulation of the Nrf1 transcription factor by its N-terminal domain is independent of Keap1: Nrf1, but not Nrf2, is targeted to the endoplasmic reticulum.
); once activated, Nrf1 translocates to the cytosol where it is deglycosylated and processed into several active isoforms, each of which harbors differential capacity to regulate transcription (
26- Wang M.
- Qiu L.
- Ru X.
- Song Y.
- Zhang Y.
Distinct isoforms of Nrf1 diversely regulate different subsets of its cognate target genes.
). Thus, glucose homeostasis, proteostasis, and other cellular conditions control Nrf1 activity (
27- Bugno M.
- Daniel M.
- Chepelev N.L.
- Willmore W.G.
Changing gears in Nrf1 research, from mechanisms of regulation to its role in disease and prevention.
). To control protein levels, Nrf1 is constantly degraded by the ubiquitin–proteasome system by several ubiquitin ligase systems, including HRD1 and β-TrCP complexes (
28- Biswas M.
- Phan D.
- Watanabe M.
- Chan J.Y.
The Fbw7 tumor suppressor regulates nuclear factor E2-related factor 1 transcription factor turnover through proteasome-mediated proteolysis.
,
29- Tsuchiya Y.
- Morita T.
- Kim M.
- Iemura S.
- Natsume T.
- Yamamoto M.
- Kobayashi A.
Dual regulation of the transcriptional activity of Nrf1 by β-TrCP- and Hrd1-dependent degradation mechanisms.
). Moreover, NRF1 is a well-described regulator of proteasomal function (
30- Steffen J.
- Seeger M.
- Koch A.
- Krüger E.
Proteasomal degradation is transcriptionally controlled by TCF11 via an ERAD-dependent feedback loop.
,
31- Radhakrishnan S.K.
- Lee C.S.
- Young P.
- Beskow A.
- Chan J.Y.
- Deshaies R.J.
Transcription factor Nrf1 mediates the proteasome recovery pathway after proteasome inhibition in mammalian cells.
).
The main negative regulator of NRF2 is KEAP1, which resides in the cytosol. KEAP1 is part of an E3 ubiquitylation complex that constantly ubiquitylates NRF2, marking it for proteasomal degradation (
32- Itoh K.
- Wakabayashi N.
- Katoh Y.
- Ishii T.
- Igarashi K.
- Engel J.D.
- Yamamoto M.
Keap1 represses nuclear activation of antioxidant responsive elements by Nrf2 through binding to the amino-terminal Neh2 domain.
). KEAP1 binds to the Neh2 domain of NRF2 and mediates NRF2 ubiquitylation. Following adduction of reactive cysteines on KEAP1 by electrophiles, the complex no longer effectively ubiquitylates NRF2, and newly-synthesized NRF2 can accumulate, translocate to the nucleus, and facilitate or repress transcription (
33Distinct cysteine residues in Keap1 are required for Keap1-dependent ubiquitination of Nrf2 and for stabilization of Nrf2 by chemopreventive agents and oxidative stress.
). This mechanism makes NRF2 the primary responder to chemical stressors. Intriguingly, KEAP1 stabilizes NRF1 when it binds to its Neh2 domain, contrasting the regulation of NRF2 from NRF1 (
34- Tian W.
- Rojo de la Vega M.
- Schmidlin C.J.
- Ooi A.
- Zhang D.D.
Kelch-like ECH-associated protein 1 (KEAP1) differentially regulates nuclear factor erythroid-2–related factors 1 and 2 (NRF1 and NRF2).
). As more mechanistic insights into NRF2 regulation comes to light, the importance of NRF2 in maintaining cellular homeostasis, even in the absence of chemical stressors, continues to expand. For example, other ubiquitylation pathways, including HRD1- and β-TrCP–dependent mechanisms, also control the NRF2 protein level under specific physiological conditions (
35- Rada P.
- Rojo A.I.
- Chowdhry S.
- McMahon M.
- Hayes J.D.
- Cuadrado A.
SCF/β-TrCP promotes glycogen synthase kinase 3-dependent degradation of the Nrf2 transcription factor in a Keap1-independent manner.
,
36- Wu T.
- Zhao F.
- Gao B.
- Tan C.
- Yagishita N.
- Nakajima T.
- Wong P.K.
- Chapman E.
- Fang D.
- Zhang D.D.
Hrd1 suppresses Nrf2-mediated cellular protection during liver cirrhosis.
). Additionally, we recently discovered that NRF2 is negatively regulated by many autophagy-related genes, implicating cellular proteostasis and metabolism also play important roles in NRF2 regulation (
37- Kerins M.J.
- Liu P.
- Tian W.
- Mannheim W.
- Zhang D.D.
- Ooi A.
Genome wide CRISPR screen reveals autophagy disruption as the convergence mechanism that regulates the NRF2 transcription factor.
).
Less is definitively known about NRF3 regulation; however, studies have shown that NRF3 is controlled in part by a PEST degron sequence and that NRF3 has three isoforms with varying subcellular localizations, including isoform “A” in the ER, isoform “B” in the cytosol, and isoform “C” in the nucleus (
38- Zhang Y.
- Kobayashi A.
- Yamamoto M.
- Hayes J.D.
The Nrf3 transcription factor is a membrane-bound glycoprotein targeted to the endoplasmic reticulum through its N-terminal homology box 1 sequence.
). Similar to NRF1, NRF3 is targeted to and eventually cleaved from the ER (
40- Nouhi Z.
- Chevillard G.
- Derjuga A.
- Blank V.
Endoplasmic reticulum association and N-linked glycosylation of the human Nrf3 transcription factor.
). NRF3 degradation is also proteasome-dependent (
40- Nouhi Z.
- Chevillard G.
- Derjuga A.
- Blank V.
Endoplasmic reticulum association and N-linked glycosylation of the human Nrf3 transcription factor.
). Hence, the different mechanisms controlling the signaling and processing of NRF transcription factors, despite their structural homology, highlight the functional differences among them. Reviews are available describing the different signaling nuances of the NRF members (
27- Bugno M.
- Daniel M.
- Chepelev N.L.
- Willmore W.G.
Changing gears in Nrf1 research, from mechanisms of regulation to its role in disease and prevention.
,
41- Yamamoto M.
- Kensler T.W.
- Motohashi H.
The KEAP1-NRF2 system: a thiol-based sensor-effector apparatus for maintaining redox homeostasis.
,
42NFE2L3 (NRF3): the Cinderella of the Cap'n'Collar transcription factors.
43- Kim H.M.
- Han J.W.
- Chan J.Y.
Nuclear factor erythroid-2 like 1 (NFE2L1): structure, function and regulation.
).
Accordingly, a better understanding of the functional differences between NRF members can further our understanding of the different roles each plays in physiology and pathology. We identified that some processes, like antioxidant response, are under the control of all three members. Other processes, like maintenance of cellular structure, may be limited to just a subset, like NRF1 and NRF3. Still, other processes may be completely under the jurisdiction of just one member. For the less well-studied NRF members, NRF1 and NRF3, this study provides unique target genes and their associated biological processes that could form the groundwork for further elucidation of their biological functions.
Functionally, NRF1 has been shown to be involved in various metabolic processes, including insulin resistance, diabetes, and inflammation (
44- Hirotsu Y.
- Hataya N.
- Katsuoka F.
- Yamamoto M.
NF-E2-related factor 1 (Nrf1) serves as a novel regulator of hepatic lipid metabolism through regulation of the Lipin1 and PGC-1β genes.
45- Hirotsu Y.
- Higashi C.
- Fukutomi T.
- Katsuoka F.
- Tsujita T.
- Yagishita Y.
- Matsuyama Y.
- Motohashi H.
- Uruno A.
- Yamamoto M.
Transcription factor NF-E2-related factor 1 impairs glucose metabolism in mice.
,
46- Tsujita T.
- Peirce V.
- Baird L.
- Matsuyama Y.
- Takaku M.
- Walsh S.V.
- Griffin J.L.
- Uruno A.
- Yamamoto M.
- Hayes J.D.
Transcription factor Nrf1 negatively regulates the cystine/glutamate transporter and lipid-metabolizing enzymes.
47- Xu Z.
- Chen L.
- Leung L.
- Yen T.S.
- Lee C.
- Chan J.Y.
Liver-specific inactivation of the Nrf1 gene in adult mouse leads to nonalcoholic steatohepatitis and hepatic neoplasia.
). This is, in part, due to Nrf1-validated control of enzymes involved in glycolysis, gluconeogenesis, and lipid metabolism (
43- Kim H.M.
- Han J.W.
- Chan J.Y.
Nuclear factor erythroid-2 like 1 (NFE2L1): structure, function and regulation.
,
48Role of Nrf1 in antioxidant-response element-mediated gene expression and beyond.
); some of these regulated genes appear to be NRF1-specific and not shared by other NRF members like NRF2. NRF1 also regulates genes involved in cell differentiation, proteostasis, cell survival, and mitosis (
43- Kim H.M.
- Han J.W.
- Chan J.Y.
Nuclear factor erythroid-2 like 1 (NFE2L1): structure, function and regulation.
); however, NRF2 shares several of these functions, such as energy metabolism and cell differentiation (
49- Chartoumpekis D.V.
- Wakabayashi N.
- Kensler T.W.
Keap1/Nrf2 pathway in the frontiers of cancer and non-cancer cell metabolism.
50- Holmström K.M.
- Baird L.
- Zhang Y.
- Hargreaves I.
- Chalasani A.
- Land J.M.
- Stanyer L.
- Yamamoto M.
- Dinkova-Kostova A.T.
- Abramov A.Y.
Nrf2 impacts cellular bioenergetics by controlling substrate availability for mitochondrial respiration.
,
51- Ludtmann M.H.
- Angelova P.R.
- Zhang Y.
- Abramov A.Y.
- Dinkova-Kostova A.T.
Nrf2 affects the efficiency of mitochondrial fatty acid oxidation.
,
52- Mitsuishi Y.
- Taguchi K.
- Kawatani Y.
- Shibata T.
- Nukiwa T.
- Aburatani H.
- Yamamoto M.
- Motohashi H.
Nrf2 redirects glucose and glutamine into anabolic pathways in metabolic reprogramming.
,
53- Kuosmanen S.M.
- Kansanen E.
- Kaikkonen M.U.
- Sihvola V.
- Pulkkinen K.
- Jyrkkänen H.-K.
- Tuoresmäki P.
- Hartikainen J.
- Hippeläinen M.
- Kokki H.
- Tavi P.
- Heikkinen S.
- Levonen A.L.
NRF2 regulates endothelial glycolysis and proliferation with miR-93 and mediates the effects of oxidized phospholipids on endothelial activation.
,
54Roles of Nrf2 in cell proliferation and differentiation.
,
55- Homma S.
- Ishii Y.
- Morishima Y.
- Yamadori T.
- Matsuno Y.
- Haraguchi N.
- Kikuchi N.
- Satoh H.
- Sakamoto T.
- Hizawa N.
- Itoh K.
- Yamamoto M.
Nrf2 enhances cell proliferation and resistance to anticancer drugs in human lung cancer.
56- Kerins M.J.
- Vashisht A.A.
- Liang B.X.
- Duckworth S.J.
- Praslicka B.J.
- Wohlschlegel J.A.
- Ooi A.
Fumarate mediates a chronic proliferative signal in fumarate hydratase inactivated cancer cells by increasing transcription and translation of ferritin genes.
), while still controlling several unique genes in its own right. Metabolically, a recent work (
57- Schneider K.
- Valdez J.
- Nguyen J.
- Vawter M.
- Galke B.
- Kurtz T.W.
- Chan J.Y.
Increased energy expenditure, Ucp1 expression, and resistance to diet-induced obesity in mice lacking nuclear factor-erythroid-2-related transcription factor-2 (Nrf2).
) has shown that loss of NRF2 in a rodent model can result in a lean phenotype. Beyond the aforementioned processes, a main function of NRF2 has been its role in antioxidation and detoxification, and while NRF1 and NRF3 have been shown to regulate several detoxification genes (
58- Ohtsuji M.
- Katsuoka F.
- Kobayashi A.
- Aburatani H.
- Hayes J.D.
- Yamamoto M.
Nrf1 and Nrf2 play distinct roles in activation of antioxidant-response element-dependent genes.
,
59- Raghunath A.
- Sundarraj K.
- Nagarajan R.
- Arfuso F.
- Bian J.
- Kumar A.P.
- Sethi G.
- Perumal E.
Antioxidant response elements: discovery, classes, regulation and potential applications.
), NRF2 has been designated the master regulator of the antioxidant response, due in part to the KEAP1 electrophile sensory mechanism and several studies indicating NRF2 induction of this subset of genes to be the most robust. Specific roles of NRF3 have remained the most enigmatic; although several target genes have been identified, little clear evidence has determined the
in vivo relevance of those genes (
42NFE2L3 (NRF3): the Cinderella of the Cap'n'Collar transcription factors.
). Indeed, a consistent lack of information exists on NRF3, particularly its physiological roles, again highlighting the need for more studies clarifying the role of the lesser-studied NRF family members.
To uncover potential unique roles of the different NRF members, we first conducted RNA-Seq to establish which genes each member regulates. This was then combined with ChIP-exo sequencing to identify LREs near those genes that were either similarly or differentially regulated. Many of the genes that were differentially regulated at the transcription level by an NRF member did not harbor an LRE. Similarly, many of the genes with ChIP-exo peaks were not annotated to a differentially-regulated gene. One possible explanation for these discrepancies could be distant enhancer regions. Enhancers have been shown to interact with promoters over 150–200 kb or even span several megabases. Enhancers can even effect transcription across chromosomes due to the inherent flexibility in chromatin (
60- Kulaeva O.I.
- Nizovtseva E.V.
- Polikanov Y.S.
- Ulianov S.V.
- Studitsky V.M.
Distant activation of transcription: mechanisms of enhancer action.
,
61- Li G.
- Ruan X.
- Auerbach R.K.
- Sandhu K.S.
- Zheng M.
- Wang P.
- Poh H.M.
- Goh Y.
- Lim J.
- Zhang J.
- Sim H.S.
- Peh S.Q.
- Mulawadi F.H.
- Ong C.T.
- Orlov Y.L.
- et al.
Extensive promoter-centered chromatin interactions provide a topological basis for transcription regulation.
). To further complicate the situation, genes regulated by certain enhancers can vary from tissue to tissue and cell to cell depending on the chromatin state. Furthermore, we also recently reported that NRF2 can repress gene expression by interacting with replication protein A1 (RPA1) to bind 5′-TGABNNNGCAAACTTCA-3′ consensus sequence (
62- Liu P.
- De La Vega M.R.
- Sammani S.
- Mascarenhas J.B.
- Kerins M.
- Dodson M.
- Sun X.
- Wang T.
- Ooi A.
- Garcia J.G.
- Garcia J.G.
- Zhang D.D.
RPA1 binding to NRF2 switches ARE-dependent transcriptional activation to ARE-NRE–dependent repression.
). Although several NRF2–RPA1 target genes were indeed down-regulated in the RNA-Seq data, no ChIP peaks were identified for those target genes possibly due to the lack of statistical power in the genome-wide approach or to cell type–specific effects. Regardless of these constraints, this remains the first study to systematically investigate NRF member dynamics at both a transcriptional and DNA-binding level simultaneously.
The ChIP-exo results showed a majority of NRF1- and NRF3-binding sites harbored canonical AREs. Interestingly, more than 50% of the binding sites for NRF2 did not contain a canonical ARE. This indicates NRF2 may be less specific than NRF1 or NRF3. Although regulating the antioxidant response is one of the primary functions of NRF2, and NRF2-based literature has often focused on this role, the 50% of NRF2-binding sites not harboring an ARE supports a role for NRF2 outside of xenobiotic elimination. Indeed, Gene Ontology for genes up-regulated by NRF1, NRF2, and NRF3 centered on the stress-response pathways, and 100% of those genes harbored an ARE. In contrast, the processes enriched from other gene groups associated with NRF2 did not contribute to antioxidation or stress-response processes, participating instead in areas like protein quality control and trafficking. Future investigations on NRF2 and protein quality control could uncover more specific roles of NRF2 in the process of ER protein folding and subsequent targeting to the membrane. In the past decade, new roles are increasingly being uncovered for NRF2 in areas such as metabolism, proliferation and differentiation, iron homeostasis, and inflammation (
49- Chartoumpekis D.V.
- Wakabayashi N.
- Kensler T.W.
Keap1/Nrf2 pathway in the frontiers of cancer and non-cancer cell metabolism.
50- Holmström K.M.
- Baird L.
- Zhang Y.
- Hargreaves I.
- Chalasani A.
- Land J.M.
- Stanyer L.
- Yamamoto M.
- Dinkova-Kostova A.T.
- Abramov A.Y.
Nrf2 impacts cellular bioenergetics by controlling substrate availability for mitochondrial respiration.
,
51- Ludtmann M.H.
- Angelova P.R.
- Zhang Y.
- Abramov A.Y.
- Dinkova-Kostova A.T.
Nrf2 affects the efficiency of mitochondrial fatty acid oxidation.
,
52- Mitsuishi Y.
- Taguchi K.
- Kawatani Y.
- Shibata T.
- Nukiwa T.
- Aburatani H.
- Yamamoto M.
- Motohashi H.
Nrf2 redirects glucose and glutamine into anabolic pathways in metabolic reprogramming.
,
53- Kuosmanen S.M.
- Kansanen E.
- Kaikkonen M.U.
- Sihvola V.
- Pulkkinen K.
- Jyrkkänen H.-K.
- Tuoresmäki P.
- Hartikainen J.
- Hippeläinen M.
- Kokki H.
- Tavi P.
- Heikkinen S.
- Levonen A.L.
NRF2 regulates endothelial glycolysis and proliferation with miR-93 and mediates the effects of oxidized phospholipids on endothelial activation.
,
54Roles of Nrf2 in cell proliferation and differentiation.
,
55- Homma S.
- Ishii Y.
- Morishima Y.
- Yamadori T.
- Matsuno Y.
- Haraguchi N.
- Kikuchi N.
- Satoh H.
- Sakamoto T.
- Hizawa N.
- Itoh K.
- Yamamoto M.
Nrf2 enhances cell proliferation and resistance to anticancer drugs in human lung cancer.
56- Kerins M.J.
- Vashisht A.A.
- Liang B.X.
- Duckworth S.J.
- Praslicka B.J.
- Wohlschlegel J.A.
- Ooi A.
Fumarate mediates a chronic proliferative signal in fumarate hydratase inactivated cancer cells by increasing transcription and translation of ferritin genes.
,
63The roles of NRF2 in modulating cellular iron homeostasis.
64- Kasai S.
- Mimura J.
- Ozaki T.
- Itoh K.
Emerging regulatory role of Nrf2 in iron, heme, and hemoglobin metabolism in physiology and disease.
,
65- Mills E.L.
- Ryan D.G.
- Prag H.A.
- Dikovskaya D.
- Menon D.
- Zaslona Z.
- Jedrychowski M.P.
- Costa A.S.H.
- Higgins M.
- Hams E.
- Szpyt J.
- Runtsch M.C.
- King M.S.
- McGouran J.F.
- Fischer R.
- et al.
Itaconate is an anti-inflammatory metabolite that activates Nrf2 via alkylation of KEAP1.
66- Kansanen E.
- Kuosmanen S.M.
- Leinonen H.
- Levonen A.-L.
The Keap1-Nrf2 pathway: mechanisms of activation and dysregulation in cancer.
). Perhaps some of these newer roles require NRF2 binding to less restrictive ARE sequences and may be an avenue of future investigations.
After combining the ChIP-exo and RNA-Seq data, we identified 262 high-confidence genes that fell into varying regulatory categories: some genes were regulated similarly by all NRF family members, whereas others were differentially regulated. We hypothesize that genes regulated similarly by all NRF members may be compensatory,
i.e. deficiency in one gene could be compensated by up-regulation of another. For example, the following genes were up-regulated by all three NRF members and are lethal upon knockout in mice:
VCP,
GCLC,
TRIO,
TBK1, and
TXNRD1 (
67- Hemmi H.
- Takeuchi O.
- Sato S.
- Yamamoto M.
- Kaisho T.
- Sanjo H.
- Kawai T.
- Hoshino K.
- Takeda K.
- Akira S.
The roles of two IκB kinase-related kinases in lipopolysaccharide and double stranded RNA signaling and viral infection.
68- Jakupoglu C.
- Przemeck G.K.
- Schneider M.
- Moreno S.G.
- Mayr N.
- Hatzopoulos A.K.
- de Angelis M.H.
- Wurst W.
- Bornkamm G.W.
- Brielmeier M.
- Conrad M.
Cytoplasmic thioredoxin reductase is essential for embryogenesis but dispensable for cardiac development.
,
69- Bonnard M.
- Mirtsos C.
- Suzuki S.
- Graham K.
- Huang J.
- Ng M.
- Itié A.
- Wakeham A.
- Shahinian A.
- Henzel W.J.
- Elia A.J.
- Shillinglaw W.
- Mak T.W.
- Cao Z.
- Yeh W.C.
Deficiency of T2K leads to apoptotic liver degeneration and impaired NF-κB-dependent gene transcription.
,
70- Dalton T.P.
- Dieter M.Z.
- Yang Y.
- Shertzer H.G.
- Nebert D.W.
Knockout of the mouse glutamate cysteine ligase catalytic subunit (Gclc) gene: embryonic lethal when homozygous, and proposed model for moderate glutathione deficiency when heterozygous.
,
71- Zong W.
- Liu S.
- Wang X.
- Zhang J.
- Zhang T.
- Liu Z.
- Wang D.
- Zhang A.
- Zhu M.
- Gao J.
Trio gene is required for mouse learning ability.
72- Müller J.
- Deinhardt K.
- Rosewell I.
- Warren G.
- Shima D.T.
Targeted deletion of p97 (VCP/CDC48) in mouse results in early embryonic lethality.
). Perhaps the loss of some of these transcription targets is partly to blame for the lethality seen with
Nrf1 knockout animals as
Nrf2 and
Nrf3 may not be expressed at the right space and time to compensate during development. However, because Nrf1, Nrf2, and Nrf3 share several transcription targets, could activation of Nrf2 or Nrf3 in an
Nrf1 knockout mouse rescue the embryonic lethal phenotype? It is important to note that under different cellular contexts, the jurisdiction of different NRF members may be swapped according to upstream signaling, cross-talk with other pathways, and proficiency/abundance of negative regulators. For example, some genes show cross-talk with aryl hydrocarbon receptor transcriptional programs (
73- Yeager R.L.
- Reisman S.A.
- Aleksunes L.M.
- Klaassen C.D.
Introducing the “TCDD-inducible AhR-Nrf2 gene battery”.
). The relative location of each NRF member on the genome may dictate their roles during development as is the case with
KRAS and
HRAS genes, whereby knocking-in
HRAS into the
KRAS locus circumvents the embryonic lethality seen with
KRAS loss (
74- Potenza N.
- Vecchione C.
- Notte A.
- De Rienzo A.
- Rosica A.
- Bauer L.
- Affuso A.
- De Felice M.
- Russo T.
- Poulet R.
- Cifelli G.
- De Vita G.
- Lembo G.
- Di Lauro R.
Replacement of K-Ras with H-Ras supports normal embryonic development despite inducing cardiovascular pathology in adult mice.
). Assessing whether embryonic lethality is maintained in
NRF1−/− animals by substituting
NRF2 or
NRF3 at the
NRF1 locus would allow us to identify functional redundancy of NRF members and the importance of their genomic locations in their function. A putative understanding of the epigenetic and signaling pathways determining NRF member activation during development would help clarify their roles. Importantly, consensus sequence analyses also identified sequence preferences for NRF1 and NRF2. Thus, certain target genes may be preferentially regulated by NRF1 and NRF2, implicating that the two genes are not always interchangeable.
From a gene regulatory standpoint, an intriguing area to consider were those genes and GO processes that were regulated differentially by the NRF1, NRF2, and NRF3 members. Dystonin (
DST) was the single gene that was up-regulated by NRF1 and NRF3 but down-regulated by NRF2. DST is a cytoskeletal linker whose loss–of–function is responsible for dystonia musculorum. Defects in DST are associated with malformations in the ER (
75Dystonin/Bpag1 is a necessary endoplasmic reticulum/nuclear envelope protein in sensory neurons.
,
76- Ryan S.D.
- Ferrier A.
- Sato T.
- O'Meara R.W.
- De Repentigny Y.
- Jiang S.X.
- Hou S.T.
- Kothary R.
Neuronal dystonin isoform 2 is a mediator of endoplasmic reticulum structure and function.
). Considering NRF1 and NRF3 are targeted to the ER whereas NRF2 is not, the up-regulation of
DST by NRF1 and NRF3 may be necessary for ER organization and thus proper functioning of NRF1 and NRF3. Interestingly, NRF2 is activated following ER stress and contributes to cell survival (
77- Mukaigasa K.
- Tsujita T.
- Nguyen V.T.
- Li L.
- Yagi H.
- Fuse Y.
- Nakajima-Takagi Y.
- Kato K.
- Yamamoto M.
- Kobayashi M.
Nrf2 activation attenuates genetic endoplasmic reticulum stress induced by a mutation in the phosphomannomutase 2 gene in zebrafish.
,
78- Cullinan S.B.
- Zhang D.
- Hannink M.
- Arvisais E.
- Kaufman R.J.
- Diehl J.A.
Nrf2 is a direct PERK substrate and effector of PERK-dependent cell survival.
). Why
DST is not up-regulated by NRF2 when DST could help restore ER homeostasis remains enigmatic, but NRF2 did exclusively up-regulate the genes
ARFGAP1,
ERGIC1,
TOR1B, and
VAPA, which are involved in “endoplasmic reticulum organization” and “retrograde vesicle-mediated transport, Golgi to ER.” More inquiry into the divergent roles of NRF members in ER biology would be valuable in unraveling the differing transcriptional programs.
BRD2, TXN, and
TNFRSF1A were genes up-regulated by both NRF1 and NRF2 but down-regulated by NRF3. Bromodomain-containing 2 (
BRD2) is a transcriptional regulator known to inhibit NRF member activity (
79- Michaeloudes C.
- Mercado N.
- Clarke C.
- Bhavsar P.K.
- Adcock I.M.
- Barnes P.J.
- Chung K.F.
Bromodomain and extraterminal proteins suppress NF-E2–related factor 2–mediated antioxidant gene expression.
). Thus, for NRF1 and NRF2 it may serve as a negative feedback mechanism, whereas for NRF3 it is a feed-forward pathway. Thioredoxin (
TXN) is a
bona fide NRF2 target gene; the contrarian role of NRF3 in regulating this gene and the others in this group is intriguing, but not unheard of, as NRF3 has been shown to negatively regulate levels of other
bona fide antioxidant genes before, such as
NQO1 and
PRDX6 by “sitting” on the ARE sequence without activating transcription (
80- Chowdhury I.
- Mo Y.
- Gao L.
- Kazi A.
- Fisher A.B.
- Feinstein S.I.
Oxidant stress stimulates expression of the human peroxiredoxin 6 gene by a transcriptional mechanism involving an antioxidant-response element.
,
81- Pepe A.E.
- Xiao Q.
- Zampetaki A.
- Zhang Z.
- Kobayashi A.
- Hu Y.
- Xu Q.
Crucial role of nrf3 in smooth muscle cell differentiation from stem cells.
82- Sankaranarayanan K.
- Jaiswal A.K.
Nrf3 negatively regulates antioxidant-response element-mediated expression and antioxidant induction of NAD(P)H:quinone oxidoreductase1 gene.
). Additionally, all three of these genes are involved in the viral infection process, which is supported from the GO analysis for “viral process.”
The NRF transcription factors differ in their tissue localization;
Nrf1 and
Nrf2 are expressed across almost all tissues, whereas the
Nrf3 expression is predominately placental (
2- Chan J.Y.
- Han X.-L.
- Kan Y.W.
Cloning of Nrf1, an NF-E2-related transcription factor, by genetic selection in yeast.
,
3- Moi P.
- Chan K.
- Asunis I.
- Cao A.
- Kan Y.W.
Isolation of NF-E2-related factor 2 (Nrf2), a NF-E2-like basic leucine zipper transcriptional activator that binds to the tandem NF-E2/AP1 repeat of the β-globin locus control region.
4- Kobayashi A.
- Ito E.
- Toki T.
- Kogame K.
- Takahashi S.
- Igarashi K.
- Hayashi N.
- Yamamoto M.
Molecular cloning and functional characterization of a new Cap'n'collar family transcription factor Nrf3.
). NRF2-specific processes (see
Fig. 9) included several metabolic processes, such as fatty acid catabolism. It has been well-described that maternal–to–fetal nutrient exchange is a critical function of the placenta, and this includes fatty acid transport and catabolism (
83The role of the placenta in fetal nutrition and growth.
).
Perhaps the more intriguing proposition is the developmental activation and inactivation of NRF members over time.
Nrf1 transcripts have been shown to be expressed in all tissues and at all stages of development investigated (
84Expression of the bZIP transcription factor TCF11 and its potential dimerization partners during development.
). In contrast, NRF2 is expressed solely in fetal liver and muscle and was absent in all other embryonic tissues investigated (
3- Moi P.
- Chan K.
- Asunis I.
- Cao A.
- Kan Y.W.
Isolation of NF-E2-related factor 2 (Nrf2), a NF-E2-like basic leucine zipper transcriptional activator that binds to the tandem NF-E2/AP1 repeat of the β-globin locus control region.
).
Nrf3 transcript expression has been investigated in avian species, where it was first noticed in heart myocardium (
85The cap'n'collar family member NF-E2-related factor 3 (Nrf3) is expressed in mesodermal derivatives of the avian embryo.
). Nrf3 transcripts then disappear from the heart and are next observed in the myotomal compartment of maturing somites. Central nervous system expression appears gradually and persists at low levels in ventricular neuroepithelial cells until at least embryonic day 6. Strong expression is observed in the early epiphysis, in the collecting ducts of the developing kidney, and in individual cells of the yolk sac (
85The cap'n'collar family member NF-E2-related factor 3 (Nrf3) is expressed in mesodermal derivatives of the avian embryo.
). Studies in zebrafish have indicated that the differing expressions of
Nrf transcripts in various tissue and developmental stages are due to the varying needs of oxidative stress regulation during development (
86- Hahn M.E.
- Timme-Laragy A.R.
- Karchner S.I.
- Stegeman J.J.
Nrf2 and Nrf2-related proteins in development and developmental toxicity: insights from studies in zebrafish (Danio rerio).
). Particularly, the absence of NRF2 during development may be to facilitate oxidative stress, which is required for some developmental processes (
87Oxidative stress in development: nature or nurture?.
).
Nrf3 presents a second interesting case study; it is expressed early in avian heart development, and it is also critical in smooth muscle differentiation (
81- Pepe A.E.
- Xiao Q.
- Zampetaki A.
- Zhang Z.
- Kobayashi A.
- Hu Y.
- Xu Q.
Crucial role of nrf3 in smooth muscle cell differentiation from stem cells.
,
85The cap'n'collar family member NF-E2-related factor 3 (Nrf3) is expressed in mesodermal derivatives of the avian embryo.
). As we saw here, biological processes related to vascularization and muscle development such as “M Band” and “endothelial cell proliferation” also suggest a role for NRF3 in circulatory system development.
However, until a detailed description of NRF1, NRF2, and NRF3 expressions at all different stages of development in the same model system is completed, we can only guess whether the different genes specific to each NRF member are relevant for a developmental function. Much of the available data on the developmental patterns of NRF member expression has utilized transcript levels instead of protein levels, and thus the preponderance of post-translational regulations of NRF members (i.e. KEAP1-mediated degradation of NRF2 or ER compartmentalization of NRF1) lessens the data's utility.
To help understand some of these differences between NRF members, we investigated whether any genomic sequences flanking the AREs were consistent among particular NRF groups. While looking for motifs flanking the AREs, we identified that NRF2 prefers GC-rich flanking bases, whereas NRF1 prefers AT-rich flanking bases. Although these moderate preferences may help determine differences between NRF1 and NRF2, there were no other trends among the NRF members or their overlaps.
Perhaps NRF members differentially regulate genes to some degree based on their relative promiscuities for the ARE. As has been shown, mutations to several bases of the canonical 5′-TGABNNNGC-3′ do not always abolish NRF2 binding, and moreover, they can actually enhance binding. Particularly, mutations to the distal GC have a wide array of effects, but none abolish NRF2 binding (
89- Kuosmanen S.M.
- Viitala S.
- Laitinen T.
- Peräkylä M.
- Pölönen P.
- Kansanen E.
- Leinonen H.
- Raju S.
- Wienecke-Baldacchino A.
- Närvänen A.
- Poso A.
- Heinäniemi M.
- Heikkinen S.
- Levonen A.L.
The effects of sequence variation on genome-wide NRF2 binding—new target genes and regulatory SNPs.
). If NRF2 binding and lack of NRF1/NRF3 binding to noncanonical AREs could be validated on a host of NRF2-specific genes, it may explain some of the differential regulatory capacities between the three. For example, the NRF2-specific up-regulated genes
SYT14, SIAH1, and
UBE2E2 harbor 5′-TGACCACAT-3′, 5′-TGAGTGCGG-3′, and 5′-TGAGTGCGG-3′ sequences near the ChIP-exo peaks that closely mimic the canonical ARE and could still retain binding activity. An exhaustive comparison of binding capabilities for each NRF member to ARE derivatives may inform the differential binding capacities more completely.
Additionally, it is unclear how differing binding affinities may play a role. As has been shown previously (
4- Kobayashi A.
- Ito E.
- Toki T.
- Kogame K.
- Takahashi S.
- Igarashi K.
- Hayashi N.
- Yamamoto M.
Molecular cloning and functional characterization of a new Cap'n'collar family transcription factor Nrf3.
), NRF2 binds more loosely to canonical AREs compared with NRF1 and NRF3. It is possible that NRF2 binding to some ARE sequences was blocked due to the presence of different ARE-binding proteins, such as dimers of Maf proteins and either p45, NRF1, NRF3, or BACH1, even with NRF2 overexpression. Similarly, the different affinities of NRF members to the sMAF proteins are not known. Thus, the context of other ARE-binding proteins and their binding sites may determine NRF2 binding as well, given it has less affinity in some cases.
In addition to a more refined understanding of binding affinities and promiscuities for each NRF member, a complete understanding of upstream events leading to transcriptional activation by NRFs could also uncover mechanisms governing their similarities and differences. For example, SETD6 was shown to inhibit NRF2 after ARE binding (
90- Chen A.
- Feldman M.
- Vershinin Z.
- Levy D.
SETD6 is a negative regulator of oxidative stress response.
). As mentioned above, RPA1 is an additional inhibitor of NRF2 activity (
62- Liu P.
- De La Vega M.R.
- Sammani S.
- Mascarenhas J.B.
- Kerins M.
- Dodson M.
- Sun X.
- Wang T.
- Ooi A.
- Garcia J.G.
- Garcia J.G.
- Zhang D.D.
RPA1 binding to NRF2 switches ARE-dependent transcriptional activation to ARE-NRE–dependent repression.
). In the context of the project presented here, the following questions arise. Can inhibitory proteins like SETD6 and RPA1 impede NRF1 and NRF3 activity, or just NRF2 activity? Does SETD6 bind to DNA near all AREs or just select ones? Understanding the interactions between NRF members and other transcription modulators is critical for interpreting differences between NRF members.
Here, we provide a compendium of the similarities and differences between the NRF1, NRF2, and NRF3 transcriptional programs. All three NRF members identify and bind to the ARE, indicating upstream signaling and cellular context, as well as other factors such as competitive binding of other transcription factors and ARE promiscuity, could play a major role in dictating the observed phenotypic differences among them. As such, determining the pathological and physiological relevance of the overlap and the disparities among the NRF family member transcriptional programs are key steps in targeting these pathways in human disease.
Experimental procedures
Cell culture and reagents
All cell lines were obtained from American Tissue Culture Collection (ATCC), and grown in Dulbecco's modified Eagle's medium with high glucose (4.5 g/liter) and no pyruvate and supplemented with heat-inactivated 10% fetal bovine serum. Cells were cultured in atmospheric air enriched with 5% CO2 in an incubator maintained at 37 °C. Zeocin, blasticidin, hygromycin, and doxycycline were obtained from Thermo Fisher Scientific and used at concentrations of 400, 10, 300, and 0.5 μg/ml, respectively. Tunicamycin was purchased from Sigma, and sulforaphane was from LKT Labs (S8044).
Generation of U2OSTetR–lacZ cells
All plasmids were obtained from Thermo Fisher Scientific (Flp-In T-REx
TM core kit, K6500-01). pcDNA5/FRT/TO/3×FLAG vector was generated by cloning a triple FLAG sequence into the pcDNA5/FRT/TO vector (Thermo Fisher Scientific) after multiple cloning sequences. NRF member open reading frames were reverse-transcribed and amplified by PCR from U2OS total RNA and modified using site-directed mutagenesis as outlined in
Fig. 1B. U2OS
TetR–lacZ cells were generated according to the manufacturer’s protocol. Briefly, U2OS cells were transfected with pFRTlacZ–Zeocin; cells with stable incorporation of this plasmid were selected with Zeocin and termed U2OS–lacZ. U2OS–lacZ cells were transfected with pCDNA/TetR; cells with stable incorporation of this plasmid were selected for with blasticidin and termed U2OS
TetR–lacZ. While U2OS
TetR–lacZ cells were being generated, the modified ORFs of NRF members NRF1, NRF2, and NRF3 (collectively NRFX) were each cloned into pcDNA5/FRT/TO/3×FLAG vector (see
Fig. 1B for modifications to NRF ORFs). The pcDNA5/FRT/TO/NRFX/3×FLAG vectors were co-transfected with pOG44 into U2OS
TetR–lacZ to generate U2OS
TetR–NRFX. U2OS
TetR–NRFX cells with proper incorporation of pcDNA5/FRT/TO/NRF/3×FLAG at the Flp recombinase site were selected with hygromycin. Thus, U2OS
TetR–NRFX cells expressed the hygromycin resistance gene, and NRFX was tagged with triple FLAG under tet-operon control. See
Fig. S1 for a schematic.
Cell line validations
U2OSTetR–NRF1, U2OSTetR–NRF2, and U2OSTetR–NRF3 cells were treated with vehicle or doxycycline for 16 h and harvested for immunoblotting and immunofluorescence.
Western blotting
Immunoblotting was performed according to standard procedures. Antibodies used in this study include anti-FLAG (Sigma, catalog no. 3165), anti-GAPDH (Santa Cruz Biotechnologies, sc-32233), anti-NRF1 (Cell Signaling Technologies, no. 8052), and anti-NRF2 (Santa Cruz Biotechnologies, sc-13032).
ADGRG7 and PHF20L1 validation
Four cell lines (MDA-MB-231, HK2, BEAS-2B, and H1299) were treated with 5 μg/ml tunicamycin, 5 μ
m sulforaphane, or vehicle control. After 4 h, cells were harvested for protein and prepared for immunoblotting. After 16 h, cells were harvested in TRIzol (Thermo Fisher Scientific). cDNA was synthesized using equal amounts of mRNA and a transcriptor first-strand cDNA synthesis kit (Roche Applied Science). Real-time quantitative PCR (qRT-PCR) was then performed. The
GAPDH gene was used for RT-qPCR normalization according to the 2
−ΔΔCt method (
91- Livak K.J.
- Schmittgen T.D.
Analysis of relative gene expression data using real-time quantitative PCR and the 2− ΔΔCT method.
). All experiments were performed in triplicate. Primers were as follows:
GAPDH (forward, 5′-CTG ACT TCA ACA GCG ACA CC-3′, and reverse, 5′-TGC TGT AGC CAA ATT CGT TGT-3′);
ADGRG7 (forward, 5′-GAC AAG GAA TAA GTC TCC ATT TGC-3′, and reverse, 5′-TGC CAT AGT AAC AGT GAG TCA AC-3′);
FTL (forward, 5′-ATT TCG ACC GCG ATG ATG TG-3′, and reverse, 5′-CAT GGC GTC TGG GGT TTT AC-3′);
GCLM (forward, 5′-GAC AAA ACA CAG TTG GAA CAG C-3′, and reverse, 5′-CAG TCA AAT CTG GTG GCA TC-3′);
NQO1 (forward, 5′-ATG TAT GAC AAA GGA CCC TTC C-3′, and reverse, 5′-TCC CTT GCA GAG AGT ACA TGG-3′); and
PHF20L1 (forward, 5′-CAT CTT GCC CTC CTC ATA GTC-3′, and reverse, 5′-AAA TCG CCC TGG AAT CAC TT-3′).
Immunofluorescence
For immunofluorescence assays, cells were grown on glass coverslips in 35-mm dishes. Cells were fixed in ice-cold methanol for 20 min and then blocked in 5% BSA for 1 h. FLAG–NRFX was detected using an anti-FLAG antibody (Sigma, catalog no. 3165) and an Alexa Fluor 488-conjugated anti-mouse secondary antibody. Images were taken with a Zeiss Observer.Z1 microscope. Images were acquired with Slidebook 4.2.0.11 software.
Luciferase assay—synthetic sequence
The dominating base pair sequences containing the NRF1- and NRF2-flanking regions on either side of a core ARE sequence (5′-GTT AGT TGC ATA GAG TGT GTT AAT TTC TAG CCA ATC GAC TAG GAA AGA CA TGACTCAGC AAT TAC GTA ACA GTA TGA AAG GGA AAC ATA AAA AAA GTC TCA ATT GAA TA-3′ and 5′-GCC GGG GGC GGG GGG GGG GCC GCG GGG GGG CGG GCG GGG CGG GCG GGG GC TGAGGCGGC CGG GCG GCG TGC GGC CGC GCG GCG GGC GGG GCC GCG GGC GCC GCT GCG GC-3′, respectively) were cloned into pGL 4.37 vector (Promega) in place of the commercially-available endogenous ARE sequence, to make NRF1–AREflank and NRF2–AREflank. U2OSTetR–NRF1 and U2OSTetR–NRF2 cells were co-transfected with a Renilla luciferase control vector (Promega E2261) and the respective NRF1-flank or NRF2-flank luciferase constructs. Cells were treated with doxycycline as described above. After 24 h, a dual-luciferase assay (Promega E1910) was conducted according to the manufacturer’s instructions. Readouts from U2OSTetR–NRF1 and U2OSTetR–NRF2 were compared using Student’s t test.
Luciferase assay–ADGRG7 and PHF20L1
Promoter regions were amplified by PCR from U2OS genomic DNA. Target regions included chr8:132775076–132777861 (PHF20L1) and chr3:100630731–100637341 (ADGRG7). The amplicons were cloned 5′ to the firefly luciferase ORF within the pGL4.21 vector (Promega) using Gibson cloning. Using these vectors and Renilla luciferase control vector, cells were subjected to dual-luciferase assay as with the synthetic vector above. All experiments were conducted in triplicate. Readouts were compared using ANOVA with post hoc Tukey’s highly-significant difference test.
RNA sequencing
U2OSTetR–NRFX cells expressing NRF1, NRF2, and NRF3 were treated with doxycycline. The NRF1 and NRF3 open reading frames had their ER targeting sequences removed in U2OSTetR–NRF1 and U2OSTetR–NRF3, respectively. U2OSTetR–NRF2 cells were treated with 10 μm sulforaphane (LKT Laboratories) for 10 h. An empty vector (U2OSTetR–FLAG) was used as a control. Total RNA from duplicate samples of the above cells was isolated using TRIzol and a column-based RNeasy kit (Qiagen). rRNA was depleted using Ribo-Zero Gold rRNA removal kit (Illumina) and prepared for sequencing using the Ultra Directional RNA library prep kit from Illumina (New England Biolabs). Samples were run on a HiSeq3000 Illumina Sequencing Platform as 50-bp single-end read runs. FASTQ files from RNA-Seq were mapped to the reference genome (UCSC hg38) using TopHat2, a splicing aligner for RNA-Seq data. Resultant binary equivalent files were sorted and indexed for quicker access using SAMtools. The number of reads per gene was annotated using a custom in-house generated script. Differentially expressed genes between control and NRF-activated samples were identified using the DESeq2 package in the R statistical environment on the read count data, which utilizes the negative binomial test to assess statistical significance. Differentially-expressed genes were defined as all genes with multiple testing corrected and adjusted p value of <0.05.
ChIP-exo sequencing
U2OSTetR–NRFX cells expressing NRF1, NRF2, and NRF3 were treated with doxycycline. NRF1 ORFs and NRF3 ORFs had their ER-targeting sequences removed. The NRF2 ORF had its two degron motifs, DLG and ETGE, mutated to AAA and AAAA, respectively. ChIP assay was performed according to the manufacturer’s instructions (EZ-CHIPTM, Merck, Germany). In brief, U2OSTetR–NRFX cells were seeded in a 150-mm dish. Upon ∼95% confluence, DNA–protein complexes were cross-linked by adding formaldehyde to a final concentration of 1%. The cells were washed with cold PBS and suspended in 1 ml of SDS lysis buffer containing 1 mm phenylmethylsulfonyl fluoride and 1% protease inhibitor mixture. Next, the samples were sonicated (nine times for 20 s and performed on ice) and centrifuged at 15,000 rpm for 30 min at 4 °C. The solubilized chromatin was diluted with ChIP dilution buffer (1:9 ratio) for ChIP assays. 0.1 ml of diluted solubilized chromatin was saved for total chromatin input. The chromatin was pre-cleared using protein G–agarose beads for 1 h at 4 °C and then incubated with 4 μg of anti-FLAG antibody or mouse normal IgG overnight at 4 °C with rotation. The cross-linked immunoprecipitates and total chromatin input were reverse cross-linked, and the DNA was isolated via phenol chloroform extraction ethanol precipitation. The library was created using adapter ligation methods, and each library was sequenced along with an input control library on a HiSeq3000 Illumina Sequencing Platform as 50-bp single-end read runs.
Sequencing FASTQ files for each NRF treatment group were mapped to UCSC hg38 using Bowtie2, a genomic aligner for DNA sequences. The resulting binary alignment sequence files were sorted, indexed, and purged of duplicate reads using SAMtools. ChIP peaks were identified using two algorithms: the Genome-wide Event finding and Motif discovery (GEM) algorithm and the Model-based Analysis for ChIP-Seq (MACS) algorithm. GEM was chosen for its utility in resolving ChIP-exo data, which usually has narrow, sharp peaks; the analysis was complemented with MACS, which can better resolve wider peaks in data with high background. Overlapping binding sites were calculated using Bedtools (
92BEDTools: a flexible suite of utilities for comparing genomic features.
). Peaks were annotated by their location to nearby genes using a custom script in the R programming environment that identified all peaks that fell within genes’ exons, introns, or within 5 kb of the transcription start and end sites. We defined peaks that fell within the first exon, first intron, or within 5 kb of the transcription start site as LREs, and genes harboring LREs were used for further analyses. ChIP peaks and genomic regions were visualized using the ChIPSeeker package, Gviz package, or in-house scripts in the R statistical environment (
88Visualizing genomic data using gviz and bioconductor.
,
93R Core Team
R: A Language and Environment for Statistical Computing.
,
94- Yu G.
- Wang L.-G.
- He Q.-Y.
ChIPseeker: an R/Bioconductor package for ChIP peak annotation, comparison and visualization.
).
Combining ChIP-exo and RNA sequencing
Only genes harboring both LREs and differential RNA expression were taken forward for further analysis as high-confidence genes likely to be regulated by one or more NRF family members. If genes were inconsistent between any NRF members then that gene was excluded from analysis. Inconsistent genes include genes that had LREs but did not have differential RNA expression or genes that had differential RNA expression but did not harbor an LRE. Genes were then categorized by their regulatory network (i.e. NRF1 and NRF2 up-regulate Gene A).
Gene ontology annotation
High-confidence genes within each gene grouping or NRF categorization were subjected to a Gene Ontology (GO) analysis. GO terms were tested for statistically-significant enrichment by hypergeometric test (threshold p value <0.05 was considered significant).
Consensus sequence flanking analysis
Across the whole genome, the previously-identified LREs from high-confidence genes were scoured for sequences conforming to the core ARE sequence, 5′-TGABNNNGC-3′. All core sequences plus 50 flanking base pairs were extracted. For the different sets of genes presented, position weight matrices harboring the relative proportions of each base pair (A, T, G, and C) at each position were evaluated. Relative proportions of GC and AT base pairs were compared between gene sets and between flanking positions using the χ2 test. p values were corrected for multiple testing using the Benjamini-Hotchberg methodology. Position weight matrices were visualized using the ggseqlogo package in the R environment.
ChIP peak sequence conservation
To evaluate conservation of ChIP-peaks' sequences across different members of the subphylum vertebrata, we retrieved phastCons scores for genomic regions covered by the high-confidence ChIP-peaks directly from the UCSC genome browser's Cons 7 Verts track. This track compares sequences from seven different species within the subphylum vertebrata by multiple alignment. The seven species being compared are human, chimp, rhesus, mouse, rat, dog, and opossum.
Data and materials availability
Datasets from the sequencing studies provided here are available at the NCBI short read archive (BioProject 541591).