bioRxiv preprint doi: https://doi.org/10.1101/2021.03.15.435342; this version posted November 14, 2021. The copyright holder for this preprint
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
available under aCC-BY 4.0 International license.
bioRxiv, November 14, 2021
Diversification of Transcription Factor NF-κB in Protists
Leah M. Williams, Sainetra Sridhar, Jason Samaroo, Ebubechi K. Adindu,
Anvitha Addanki, BB522 Molecular Biology Laboratory,1 Pablo J. Aguirre Carrión,
Christopher J. DiRusso, Nahomie Rodriguez-Sastre, Trevor Siggers, Thomas D.
Gilmore,*
Department of Biology, Boston University, Boston, MA 02215, USA
*Dr. Thomas D. Gilmore, Biology Department, Boston University, 5 Cummington Mall,
Boston, MA 02215, USA; 617-353-5444 (phone); 617-353-6340 (fax); email:
gilmore@bu.edu
1
BB522 Molecular Biology Laboratory: Dana H.M. Alburi, Ludmila Anisimov, Aria Y.
Armstrong, Sydney J. Badger, Elham Banaie, Joana A. Barbosa Teixeira, Madeleine T.
Billingsley, Anoush Calikyan, Yinxing Chen, Aidan B. Coia, Daniel Cutillo, Breanna R.
Dooling, Parth P. Doshi, Kyra R. Dubinsky, Berta Escude Velasco, Jabari R. Evans,
Jasmine Gordon, Huibo Guan, Spiro N. Haliotis, Niccolas T. Hood, Yen-Chun Huang,
Wenjing Jiang, Isabelle C. Kreber, Ekin B. Kurak, Cheng-Che Lee, Tanner M. Lehmann,
Savina J.W. Lin, Edward Liu, Kevin Liu, Yen-Yu Liu, Alexandra L. Luther, Alexa A.
Macgranaky-Quaye, Daniel J. Magat, Lauren E. Malsick, Parmida Masoudi, Parsida
Masoudi, Chad R.H. Max, Ethan Z. McCaslin, Eleanor T. McGeary, Kathleen M.
McLaughlin, Victoria S.A. Momyer, Lake D. Murphy, Sonny V. Nguyen, Kareemah Ni,
Leon Novak, Roberto Nunes Campos E Santos, Yemi D. Osayame, Jun Bai Park
Chang, Harshal M. Patel, Tony V. Pham, Sheila M. Phillips, Jhonathan Perea Piedrahita,
Tricia L. Post, Rebecca A. Prather, Pauline I. Reck, Jaime A. Rodriguez, Kirquenique A.
Rolle, Joseph A. Salzo, Kathryn M. Satko, Davis G. Settipane, Kara J. Sevola, Mithil V.
Shah, Viktoriya Skidanova, Georgia M. Snyder, Rebecca J. Sprague, Ryan A. Stagg,
Danielle Tong, Andreas A. Towers, Nicholas W. Turgiss, Natalie S. Wheeler, Ann S.
Yung
Keywords: NF-κB, Protists, Choanoflagellates, Capsaspora, Signal Transduction
1
bioRxiv preprint doi: https://doi.org/10.1101/2021.03.15.435342; this version posted November 14, 2021. The copyright holder for this preprint
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
available under aCC-BY 4.0 International license.
Significance
These results represent the first functional characterization of the biologically important
transcription factor NF-κB in a taxonomic kingdom other than Animalia. As such, they
provide information on the evolutionary origins and basal diversification of NF-κB outside
of metazoans. These results suggest that NF-κB plays life stage-specific roles in
Capsaspora, the closest unicellular ancestor to all metazoans. Finally, the analysis of
three NF-κB proteins in a single choanoflagellate indicates that choanoflagellates have
subclasses of NF-κBs, which can form heterodimers, suggesting that NF-κB subunit
expansion and diversification has occurred at least twice in evolution.
2
bioRxiv preprint doi: https://doi.org/10.1101/2021.03.15.435342; this version posted November 14, 2021. The copyright holder for this preprint
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
available under aCC-BY 4.0 International license.
Abstract
In this report, we investigate the evolution of transcription factor NF-κB by examining its
structure, activity, and regulation in two protists using phylogenetic, cellular, and
biochemical techniques. In Capsaspora owczarzaki (Co), we find that full-length NF-κB
has an N-terminal DNA-binding domain and a C-terminal Ankyrin (ANK) repeat inhibitory
domain, and its DNA-binding activity is more similar to metazoan NF-κB rather than Rel
proteins. As with mammalian NF-κB proteins, removal of the ANK repeats is required for
Co-NF-κB to enter the nucleus, bind DNA, and activate transcription. However, Cterminal processing of Co-NF-κB is not induced by co-expression of IKK in human cells.
Exogenously expressed Co-NF-κB localizes to the nucleus in Co cells. NF-κB mRNA
and DNA-binding levels differ across three life stages of Capsaspora, suggesting distinct
roles for NF-κB in these life stages. RNA-seq and GO analyses identify possible gene
targets and biological functions of Co-NF-κB. We also show that three NF-κB-like
proteins from the choanoflagellate Acanthoeca spectabilis (As) all consist of primarily the
N-terminal conserved Rel Homology domain sequences of NF-κB, and lack C-terminal
ANK repeats. All three As-NF-κB proteins constitutively enter the nucleus of human and
Co cells, but differ in their DNA-binding and transcriptional activation activities.
Furthermore, all three As-NF-κB proteins can form heterodimers, indicating that NF-κB
diversified into multi-subunit families at least two times during evolution. Overall, these
results present the first functional characterization of NF-κB in a taxonomic kingdom
other than Animalia and provide information about the evolution and diversification of this
biologically important transcription factor.
3
bioRxiv preprint doi: https://doi.org/10.1101/2021.03.15.435342; this version posted November 14, 2021. The copyright holder for this preprint
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
available under aCC-BY 4.0 International license.
Introduction
Transcription factor NF-κB (Nuclear Factor-κB) has been extensively studied for its roles
in development and immunity in animals from sponges to humans (1–3). Only within the
last few years has it been discovered that NF-κB’s appearance pre-dates metazoan life;
that is, that certain single-cell eukaryotes, namely some choanoflagellates and the
holozoan Capsaspora owczarzaki, also contain genes encoding NF-κB-like proteins (4,
5). In this paper, we present the first functional characterization of this important
transcription factor in single-celled protists.
Protists comprise a large group of eukaryotes that are either unicellular or
multicellular with poorly differentiated tissue, and they make up one of the six major
Kingdoms (6). Presumably, protists have a common ancestor, but they are now known to
be an extensively diverse collection of organisms with several major supergroups. Two
protists that have been studied reasonably well are Capsaspora and the taxonomic class
of choanoflagellates, both of which are in the Opisthokonta subgroup.
Capsaspora is a single-celled eukaryote that is thought to be among the closest
unicellular relatives to animals (i.e., basal to sponges) and is the sister group to the
Filozoa (the clade comprising metazoans and choanoflagellates) (7). Capsaspora was
originally discovered as an amoeba-like symbiont in the hemolymph of the fresh-water
snail Biomphalaria glabrata. Capsaspora kills sporocysts of the flatworm Schistosoma
mansoli, the causative agent of schistosomiasis in humans, which also inhabits B.
glabrata (8). More recently, the life cycle of Capsaspora has been shown to contain three
different cell configurations (8). Under in vitro culture conditions, Capsaspora grow
primarily as filopodial cells, which attach to the substrate and undergo active replication
until the end of the exponential growth phase. Then, cells start to detach, retracting their
branching filopodia and encysting. During this cystic phase, cell division is stopped.
Alternatively, filopodia cells can form a multicellular aggregative structure by secreting an
4
bioRxiv preprint doi: https://doi.org/10.1101/2021.03.15.435342; this version posted November 14, 2021. The copyright holder for this preprint
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
available under aCC-BY 4.0 International license.
unstructured extracellular matrix that promotes aggregation but prevents direct cell-cell
contact (9). The genome of Capsaspora contains many genes involved in metazoan
multicellular processes including integrins, protein tyrosine kinases, and transcription
factors, including NF-κB (4). Furthermore, RNA-sequencing has revealed that each life
stage contains distinct transcriptomic profiles (9). However, the details of how these lifestage processes and transitions are carried out on the molecular level in a unicellular
organismal context are still unclear.
A second class of protists of interest for evolutionary biologists includes the
choanoflagellates. These flagellated eukaryotes comprise over 125 species of free-living
unicellular and colonial organisms distributed in nearly every aquatic environment (10).
Choanoflagellates are also widely regarded as being close living relatives to the animals,
and they are capable of asexual and sexual reproduction (10). The feeding of
choanoflagellates on bacteria provides a critical ecological role within the global carbon
cycle by linking trophic levels. Until recently, little was known about the genomic diversity
of choanoflagellates, with only two published genomes of Monosiga brevicollis and
Salpingoeca rosetta (11, 12). Neither of these species contains homologs to NF-κB, and
it was thought for nearly a decade that this transcription factor had been lost in the
evolutionary branch containing choanoflagellates. However, in 2018, Richter et al. (5)
reported the transcriptomes of 19 additional choanoflagellates, and 12 of these
choanoflagellates expressed NF-κB-like genes, and several of these species that
contained multiple NF-κB-like transcripts. Amazingly, sequence comparisons have
revealed that choanoflagellates are generally as genetically distant from each other as a
mouse is from a sea sponge, a testimony to the modern day diversity among this taxa
(5).
Proteins in the NF-κB superfamily are related by a conserved N-terminal Rel
Homology Domain (RHD) containing sequences important for dimerization, DNA binding,
5
bioRxiv preprint doi: https://doi.org/10.1101/2021.03.15.435342; this version posted November 14, 2021. The copyright holder for this preprint
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
available under aCC-BY 4.0 International license.
and nuclear localization (1). All NF-κB proteins bind to a collection of related DNA sites
known as κB sites, and NF-κB proteins do so as either homodimers or heterodimers. In
animals from insects to humans, there are two subgroups of NF-κB proteins that differ in
their C-terminal domain sequences and DNA-binding site preferences (1). One subgroup
includes the traditional NF-κB proteins (p52/p100, p50/p105, Relish) that contain Cterminal inhibitory repeats known as Ankyrin repeats (ANK), whereas the second group
consists of the Rel proteins (RelA, RelB, cRel, Dorsal, Dif) that contain C-terminal
transactivation domains. Among basal organisms---including cnidarians, poriferans, and
some protists---only NF-κB-like proteins have been found (3). Indeed, no Rel proteins
have been identified in any organism basal to flies.
In most metazoans, the activity of NF-κB proteins is controlled by subcellular
localization, wherein an inactive NF-κB dimer is sequestered in the cytoplasm due to
interaction with inhibitory IκB sequences (including the C-terminal ANK repeats of NF-κB
proteins). Many upstream signals, including the binding of various ligands to conserved
upstream receptors (e.g., Toll-like Receptors (TLRs), Interluekin-1 receptors (IL-1Rs),
and tumor necrosis factor receptors (TNFRs)), lead to the initiation of a signal
transduction pathway culminating in activation and nuclear translocation of NF-κB (1, 2).
In the non-canonical pathway, the translocation of NF-κB from the cytoplasm to the
nucleus is initiated through the phosphorylation of serine residues C-terminal to the ANK
repeats, which leads to removal of the C-terminal ANK repeats by a proteasomal
processing that begins at the C terminus and stops within a glycine-rich region (GRR)
near the end of the RHD (13). Relieved of inhibition, the NF-κB dimer is able to
translocate to the nucleus, bind DNA, and activate its transcriptional targets for a given
biological outcome.
Transcriptomic and genomic sequencing has revealed that NF-κB and homologs
of many of its upstream regulators are present in most eukaryotes from protists to
6
bioRxiv preprint doi: https://doi.org/10.1101/2021.03.15.435342; this version posted November 14, 2021. The copyright holder for this preprint
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
available under aCC-BY 4.0 International license.
vertebrates (3, 14). However, the numbers and structures of these signaling proteins
vary across species, and generally become more complex and numerous through
evolutionary time (14). Furthermore, in the most basal groups of metazoans (cnidarians
and sponges), only single homologs to NF-κB exist within their genomes (3).
Herein, we have characterized molecular functions of transcription factor NF-κB
in two unicellular protists using phylogenetic, cellular, and biochemical techniques. We
find that like the human p100 protein, some unicellular NF-κB proteins require removal of
C-terminal ANK repeats to enter the nucleus and bind DNA. However, Co-NF-κB does
not undergo IKK-mediated processing, and homologs to IKK do not exist in Capsaspora
or choanoflagellates. Furthermore, we show that the multiple NF-κBs of a single
choanoflagellate can form heterodimers, a first finding in an organism outside of the
kingdom Animalia, suggesting that choanoflagellates contain their own subclasses of
NF-κBs, much like in vertebrates and flies. These results are the first functional
characterization of NF-κB in a taxonomic kingdom other than Animalia.
Results
Protist NF-κB proteins vary in domain structure and choanoflagellates show
evidence of gene duplication
Suga et al. (4) reported the presence of a single gene encoding an NF-κB-like protein in
C. owczarzaki (Co). The overall protein structure of Co-NF-κB is similar to most other
basal NF-κB proteins known to date, in that it has an N-terminal RHD, followed by a
glycine-rich region (GRR), and five C-terminal ANK repeats (15–18). However, Co-NFκB is larger than other NF-κB homologs, due primarily to additional residues C-terminal
to the ANK repeats (Fig. 1A), Recently, Richter et al. (5) showed that the transcriptomes
of several choanoflagellates had NF-κB-like genes, even though NF-κB-like genes are
7
bioRxiv preprint doi: https://doi.org/10.1101/2021.03.15.435342; this version posted November 14, 2021. The copyright holder for this preprint
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
available under aCC-BY 4.0 International license.
not present in two commonly studied choanoflagellates (M. brevicollis and S. rosetta)
(11, 12). Overall, 12 of the 21 choanoflagellates are now known to contain NF-κB-like
genes, and among those 12, there are one to three NF-κB transcripts present. A
phylogenetic comparison suggests that many of these NF-κBs arose from gene
duplications within a given species because the multiple NF-κBs from a given species
often cluster closely to each other (for example, Diaphanoeca grandis and Salpingoeca
helianthica) (Fig. 1B). Nevertheless, there are some choanoflagellates that have multiple
NF-κBs that cluster separately with the NF-κBs of other choanoflagellate (e.g.,
Acanthoeca spectabilis and Savillea parva). In contrast to what is seen in most basal
metazoans, these choanoflagellates express transcripts that primarily encode RHD
sequences, with no C-terminal GRRs or ANK repeats. However, some choanoflagellate
NF-κBs do contain extended N-termini with homology to sequences not normally
associated with NF-κBs in vertebrates (Fig. 1A, pink bar).
DNA binding, nuclear translocation, and transactivation by Co-NF-κB
To investigate the overall DNA binding-site specificity of Co-NF-κB, we first
characterized the activity of a bacterially expressed Co-NF-κB RHD-only protein by
protein binding microarray (PBM) analysis on an array containing 2592 κB-like sites and
1159 random background sequences (for array probe sequences, see (17)). By
comparison of the z-scores for binding to DNA sites on the PBM, the DNA-binding profile
of Co-NF-κB is most similar to NF-κBs from the sea anemone N. vectensis and human
p52, and it is distinct from human c-Rel and RelA (Fig. 2A).
To investigate properties of Co-NF-κB in cells, we created pcDNA-FLAG vectors
for full-length Co-NF-κB and two truncation mutants, one (FLAG-Co-RHD) containing the
N-terminal RHD sequences including the NLS and the GRR, and a second (FLAG-CoCterm) consisting of the C-terminal ANK repeat sequences and downstream residues
8
bioRxiv preprint doi: https://doi.org/10.1101/2021.03.15.435342; this version posted November 14, 2021. The copyright holder for this preprint
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
available under aCC-BY 4.0 International license.
(Fig. 2B). As a control, we used the active, naturally truncated N. vectensis (Nv) FLAGtagged Nv-NF-κB protein that we have characterized previously (19) (Fig. 2B). As shown
by anti-FLAG Western blotting, each plasmid expressed a protein of the appropriate size
when transfected into HEK 293T cells (Fig. 2C).
In sponge and some cnidarian NF-κBs, removal of C-terminal ANK-repeat
sequences are required for nuclear localization in vertebrate cell-based assays (15–17).
Based on those results, we next transfected each FLAG expression plasmid into DF-1
chicken fibroblast cells and performed indirect immunofluorescence using anti-FLAG
antiserum (Fig. 2D; Supplemental Table 1). Full-length Co-NF-κB and Co-Cterm were
both located primarily in the cytoplasm of these cells (99.9% and 94%, respectively). In
contrast, the Co-RHD and control Nv-NF-κB proteins were both primarily nuclear, as
evidenced by co-localization with the Hoechst-stained nuclei (Fig. 2D; Supplemental
Table 1). Thus, the removal of the ANK repeats allows Co-NF-κB to enter the nucleus,
consistent with what is seen with other metazoan RHD-ANK bipartite NF-κB proteins.
To further assess the DNA-binding activity of Co-NF-κB proteins, whole-cell
extracts from 293T cells transfected with each of the FLAG constructs were analyzed in
an electrophoretic mobility shift assay (EMSA) using a high affinity κB-site probe.
Extracts containing overexpressed Nv-NF-κB and Co-RHD bound the κB site avidly,
whereas extracts containing full-length Co-NF-κB and Co-Cterm showed essentially no
κB site-binding activity (Fig. 2E).
We also assessed the ability of Co-NF-κB proteins to activate transcription in
reporter gene assays in HEK 293 cells using a κB-site reporter. Co-RHD and Nv-NF-κB
activated transcription well above control levels (i.e., Co-RHD was ~60-fold above the
negative control; Fig. 2F). In contrast, full-length or Cterm Co-NF-κB proteins showed
little to no ability to activate transcription. From these data, the ability to activate
transcription of a κB site gene locus appears to be a property of sequences within the N-
9
bioRxiv preprint doi: https://doi.org/10.1101/2021.03.15.435342; this version posted November 14, 2021. The copyright holder for this preprint
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
available under aCC-BY 4.0 International license.
terminal half of Co-NF-κB. We also assessed the ability of Co-RHD to activate
transcription in reporter gene assays in yeast cells using a GAL4-site reporter. Indeed,
the N-terminal half (RHD) of Co-NF-κB activated transcription strongly, nearly 1000-fold
above the GAL4 (aa 1-147) alone negative control. The transactivation ability of the
GAL4-RHD sequences of Co-NF-κB in yeast suggests that this is an intrinsic property of
these sequences.
Taken together, the results in this section show that Co-RHD can bind DNA,
activate transcription, and localizes primarily to the nucleus, unlike the inactive fulllength Co-NF-κB protein, consistent with findings with most NF-κBs from sponges to
humans when assayed in vertebrate cells (15–17, 19).
IKK-mediated processing of NF-κB appears to have evolved with the rise of
multicellularity
As we describe above for Co-NF-κB, vertebrate NF-κB p100 requires the removal of its
C-terminal ANK repeats to enter the nucleus and activate transcription (13). This
proteasome-mediated processing of p100 is initiated by phosphorylation of a C-terminal
cluster of serine residues by an IκB kinase (IKK) (13). We have previously shown that
some basal organisms, including NF-κB proteins from two cnidarians and one sponge,
contain homologous C-terminal serines that can be phosphorylated by IKKs to initiate
proteasome-mediated processing in human cell culture assays (15–17). Examination of
the C-terminal sequences of Co-NF-κB failed to identify any C-terminal serine clusters
similar to other NF-κBs that undergo IKK-initiated processing. Nevertheless, we
performed a series of experiments that examined the ability of IKK to induce processing
of Co-NF-κB by co-transfecting HEK 293T cells with Co-NF-κB and several IKK proteins,
including two from humans and one from a sea anemone (15–17). In all cases, coexpression of the IKK did not induce processing of Co-NF-κB (Fig. 3A), beyond the small
10
bioRxiv preprint doi: https://doi.org/10.1101/2021.03.15.435342; this version posted November 14, 2021. The copyright holder for this preprint
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
available under aCC-BY 4.0 International license.
amount of constitutive processing of Co-NF-κB that occurs even in the absence of IKK
(Fig. 3A and Fig. 2C). Of note, the lower Co-NF-κB band seen in these extracts was
roughly the same size as the predicted RHD (Fig. 2C), and incubation of transfected
cells with the proteasome inhibitor MG132 reduced the appearance of the lower band,
suggesting that it arises by proteasomal processing of full-length Co-NF-κB
(Supplemental Fig. 1).
To determine whether Co-NF-κB could be processed by an IKK-dependent
mechanism, we created a mutant in which we replaced C-terminal sequences Co-NF-κB
(downstream of the ANK repeats) with C-terminal sequences of the sea anemone
Aiptasia (Ap)-NF-κB that contain conserved serines which can facilitate IKK-induced
processing of Ap-NF-κB (17). We termed this mutant Co-NF-κB-SER, and also created
the analogous protein (Co-NF-κB-ALA) in which the relevant serines were replaced by
alanines. Co-expression of Co-NF-κB-SER with constitutively active human IKK (IKK
SS/EE) resulted in increased amounts of the lower band, which was not seen with CoNF-κB-ALA (Fig. 3B). Thus, the Co-NF-κB protein (consisting of the RHD, GRR, and
ANK repeats) can undergo IKK-induced processing if supplied with a C terminus
containing the IKK target serine residues. However, the native Co-NF-κB protein does
not appear to be susceptible to IKK-induced processing, which is also consistent with the
lack of any IKK sequences in the genome of Capsaspora.
Exogenously expressed full-length and truncated versions of Co-NF-κB localize
primarily to the nucleus in Capsaspora cells
We were next interested in examining the subcellular localization of NF-κB in
Capsaspora cells. For these experiments, we transfected Capsaspora cells with our
FLAG-tagged Co-NF-κB constructs (Co-NF-κB, mutant Co-RHD, and mutant Co-Cterm,
see Fig. 2D) and then performed anti-FLAG indirect immunofluorescence. Consistent
11
bioRxiv preprint doi: https://doi.org/10.1101/2021.03.15.435342; this version posted November 14, 2021. The copyright holder for this preprint
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
available under aCC-BY 4.0 International license.
with results seen in DF-1 chicken fibroblast cells (Fig. 2D), FLAG-Co-RHD and FLAGCo-Cterm localize to the nucleus and cytoplasm, respectively (Fig. 4, middle and bottom
rows, Supplemental Video 1). Surprisingly, full-length FLAG-Co-NF-κB also appeared to
be fully nuclear, as judged by its co-localization with the Hoechst-stained nuclei (Fig. 4,
top row, and Supplemental Fig. 2 and Supplemental Video 2).
Co-NF-κB mRNA levels and DNA-binding activity vary coordinately across
different life stages and the identification of putative NF-κB target genes
Capsaspora has been shown to have three different life stages: aggregative, filopodia,
and cystic, and RNA-Seq of each life stage has been reported (9). We were interested in
whether Capsaspora NF-κB protein was active at different levels in these three life
stages and whether we could use the previous RNA-Seq data to identify genes whose
expression may be controlled by NF-κB. We first examined previous mRNA expression
data (9) for NF-κB mRNA, and found that NF-κB was expressed at the lowest level in the
aggregative stage and 2.3- and 5-fold higher in the filopodic and cystic stages,
respectively (Fig. 5A). We next generated cultures of Capsaspora at each life stage (Fig.
5A), made protein extracts, and performed an EMSA using the κB-site probe that we
showed can be bound by Co-NF-κB expressed in HEK 293T cells (Fig. 2E) and by
bacterially expressed Co-RHD in our PBM assays (Supplemental Table 2). Consistent
with the mRNA expression data, the κB-site probe was bound progressively stronger in
aggregative, filopodic, and cystic stages (Fig. 5B). To determine whether the EMSA
band indeed included Co-NF-κB, we incubated our protein extracts with 10X and 25X
excesses of unlabeled κB-site probe, and we saw a substantial decrease in binding in
the putative NF-κB band. In contrast, incubation with a 25X excess of an unlabeled IRFsite probe did not decrease the putative Co-NF-κB complex, indicating that the binding
was specific for the κB-site probe.
12
bioRxiv preprint doi: https://doi.org/10.1101/2021.03.15.435342; this version posted November 14, 2021. The copyright holder for this preprint
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
available under aCC-BY 4.0 International license.
We next sought to identify genes that might be influenced by the expression of
NF-κB in order to gain insight into potential functional roles for NF-κB in these life stages.
We examined the existing RNA-Seq data (9), which contains the mRNA expression of
8674 genes of Capsaspora during its three life stages. We first narrowed our gene list to
those genes that were differentially expressed in a manner similar to NF-κB mRNA
levels and DNA-binding activity during each life stage (i.e., progressively increased in
expression from aggregative, filopodic, and cystic stages). From this exercise, we
identified 1348 mRNAs that were expressed at low levels in the aggregative stage, and
successively higher levels in the filopodic and cystic stages.
Of the 1348 genes that we identified, 389 genes were annotated (which is
consistent with approximately 1/4 of Capsaspora’s total predicted protein-encoding
genes being annotated, Supplemental Table 3), and 305 of these genes had human
homologs (Supplemental Table 3). We then performed GO analysis to look at Biological
Processes overrepresented in these 305 genes. That analysis showed that this set of
305 genes was predicted to be involved in several biological processes, including 14
genes that encode proteins associated with developmental and immune system
processes (Fig. 5C), which are biological processes regulated by NF-κB in many more
complex organisms and suggested to be regulated by NF-κB in several basal organisms
(15–17, 20–23). Although 14 genes may seem low, the total database for human GO
analysis of immune system and developmental processes genes is approximately 2200
genes, but the number of annotated homologs that exist in Capsaspora in these two GO
categories is only 66 genes (Supplemental Table 4). Thus, about 20% (14/66) of the
Capsaspora genes in the GO category for the developmental and immune processes
subset are among the 305 homologous genes coordinately regulated with NF-κB. Other
broad categories overrepresented in these 305 genes included Signaling, Metabolic
Process, and Locomotion (Supplemental Fig. 3).
13
bioRxiv preprint doi: https://doi.org/10.1101/2021.03.15.435342; this version posted November 14, 2021. The copyright holder for this preprint
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
available under aCC-BY 4.0 International license.
We then looked for κB sites within 500 bp upstream of the transcription start sites
(TSS) for each of the 1348 genes with expression profiles that were similar to NF-κB.
192 of these 1348 gene upstream regions contained 1-3 κB sites within 500 bp of the
TSS, with the majority of these genes containing 1 κB site (Supplemental Table 5). Two
of the 14 genes that encode protein homologs associated with GO developmental and
immune system processes contained a κB site within the 500 bp upstream of their TSS
(Fig. 5C).
Choanoflagellate NF-κBs can form heterodimers and have different abilities to
bind DNA and activate transcription
Richter et al. (5) sequenced the transcriptomes of 19 choanoflagellates and identified
RHD-containing NF-κB-related proteins in 12 of these species. We chose to characterize
the NF-κB proteins from Acanthoeca spectabilis (As) because it has three NF-κB-like
proteins, which separated into multiple branches when phylogenetically compared to all
choanoflagellate NF-κBs (Fig. 1B). These three As-NF-κB proteins contained ostensibly
complete DNA-binding sequences, which were similar to other NF-κB proteins and a
putative NLS (3). These three proteins contained extended sequences N-terminal to the
RHD, but they contained few C-terminal residues beyond the RHD (and no GRRs or
ANK repeats). As a first step in characterizing these proteins, we created pcDNA FLAG
vectors for As-NF-κB1, As-NF-κB2, and As-NF-κB3 (Fig. 6A) and transfected them into
HEK 293T cells. As assessed by anti-FLAG Western blotting, each vector directed the
expression of appropriately sized FLAG-tagged proteins (Fig. 6B).
To determine the subcellular localization properties of the three As-NF-κBs, we
performed indirect immunofluorescence on DF-1 chicken fibroblasts and Capsaspora
cells transfected with each FLAG-tagged vector. All three As-NF-κB proteins co-localized
with Hoechst-stained nuclei in both DF-1 cells (Fig. 6C) and Capsaspora cells (Fig. 6D).
14
bioRxiv preprint doi: https://doi.org/10.1101/2021.03.15.435342; this version posted November 14, 2021. The copyright holder for this preprint
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
available under aCC-BY 4.0 International license.
We then performed a κB-site EMSA on whole cell extracts from HEK 293T cells
overexpressing each As-NF-κB, using Co-RHD as a positive control. As-NF-κB2 and 3
bound the κB-site probe to nearly the same extent as Co-RHD, but As-NF-κB1 only
weakly bound the probe (Fig. 6E). We also assessed the transactivating ability of each
As-NF-κB protein in a κB-site reporter assay in HEK 293T cells, using the strongly
activating Co-RHD protein as a positive control. Both As-NF-κB2 and 3 were able to
activate transcription of the luciferase reporter above vector control levels (~1.6- and 2.7fold, respectively) but As-NF-κB1 did not (Fig. 6F). We also assessed the intrinsic
transactivating ability of each As-NF-κB protein using a GAL4-fusion reporter assay in
yeast cells. In this assay, all three As-NF-κBs activated transcription over vector control
levels, although As-NF-κB1 and 3 activated to a much lesser degree than As-NF-κB2
(Fig. 6G). From these data, we hypothesized that the homodimeric version of As-NF-κB1
was not capable of binding DNA and could not activate transcription using κB sites, but
likely contained some intrinsic ability to activate transcription (as a GAL4-fusion protein),
whereas homodimeric As-NF-κB2 and 3 could activate transcription both in human cellbased and yeast GAL4-fusion reporter assays.
Since As-NF-κB1 did not substantially bind DNA or activate transcription when
transfected alone, we hypothesized that As-NF-κB1 ordinarily acts as a heterodimer with
the other As-NF-κBs. Therefore, we performed a series of co-immunoprecipitation
experiments to determine whether As-NF-κB1 could interact with As-NF-κB2 or As-NFκB3. To do this, we subcloned As-NF-κB2 and 3 into MYC-tagged vectors, cotransfected each with FLAG-As-NF-κB1 in HEK 293T cells, and first immunoprecipitated
cell extracts using anti-FLAG beads. We then performed anti-FLAG and anti-MYC
Western blotting on the immunoprecipitates to assess whether these NF-κBs could
interact. MYC-As-NF-κB2 and MYC-As-NF-κB3 were both co-immunoprecipitated with
15
bioRxiv preprint doi: https://doi.org/10.1101/2021.03.15.435342; this version posted November 14, 2021. The copyright holder for this preprint
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
available under aCC-BY 4.0 International license.
FLAG-As-NF-κB1, as well as with each other (Fig. 6H, IP). The MYC-As-NF-κB proteins
were not seen when they were co-transfected with the empty vector control (Fig. 6H, IP).
From these data, it appears that all three As-NF-κBs can enter the nucleus when
expressed in vertebrate and protist cells, but they bind DNA and activate transcription to
varying degrees. Furthermore, all three As-NF-κBs can form heterodimers with the other
As-NF-κBs. As we discuss below, we think that the reduced ability of As-NF-κB1 to bind
DNA and activate a reporter gene is due to a limited ability to form homodimers.
Discussion
In this manuscript, we have functionally characterized and compared, for the first time,
NF-κB proteins from two protists. Taken together, these results demonstrate that
although functional DNA-binding and transcriptional-activating NF-κB proteins exist in
these protists, the overall structures and regulation of these proteins varies considerably,
both among protists and when compared to animal NF-κBs.
In the vertebrate NF-κB proteins p100 and p105, C-terminal ANK repeats inhibit
the DNA-binding activity of the RHD, and proteasome-mediated processing of the ANK
repeats is terminated by and within the GRR (13). In the Drosophila Relish protein, the
C-terminal ANK repeats also inhibit DNA binding, but the C-terminal ANK repeats are
removed by an internal site-specific cleavage event, which does not involve the
proteasome, and Relish has no GRR (24). Thus, the presence of ANK repeats and a
GRR in Co-NF-κB suggests that proteasomal processing would lead to nuclear
translocation and activation of its DNA-binding activity. Indeed, removal of the C-terminal
residues of Co-NF-κB does allow it to enter the nucleus of vertebrate cells and
unleashes its DNA-binding activity (Figs. 2D and E). Moreover, treatment of cells with
the proteasome inhibitor MG132 blocks the basal processing of Co-NF-κB that is seen in
16
bioRxiv preprint doi: https://doi.org/10.1101/2021.03.15.435342; this version posted November 14, 2021. The copyright holder for this preprint
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
available under aCC-BY 4.0 International license.
transfected 293T cells (Supplemental Fig. 1). However, this basal processing of Co-NFκB in 293T cells is not enhanced by co-expression of IKK (unless C-terminal IKK target
sequences are added, see Fig. 3B) and there are no obvious IKK target serines in CoNF-κB nor are there any IKK homologues in the Capsaspora genome. Furthermore,
most of the Co-NF-κB appears to be in the nucleus when it is overexpressed in
Capsaspora cells (Fig. 4), suggesting that it is constitutively processed to its RHD. If CoNF-κB does undergo a signal-induced proteasomal processing in Capsaspora cells, then
it is unlikely to be dependent on an IKK-like kinase. Alternatively, but probably less likely,
full-length Co-NF-κB enters the nucleus of Capsaspora cells but not DF-1 chicken cells.
In the absence of a Co-NF-κB-specific antiserum, we cannot distinguish between these
possibilities.
In contrast to Capsaspora, the choanoflagellate NF-κB proteins lack C-terminal
ANK repeats and GRRs, and all three As-NF-κB proteins are constitutively in the nucleus
when overexpressed in vertebrate or Capsaspora cells (Fig. 6). We have not been able
to identify an IκB-like protein in the A. spectabilis genome. Thus, it is unclear whether
choanoflagellate NF-κB is regulated by an ANK-dependent cytoplasmic retention
mechanism, or whether, for example, choanoflagellate proteins are constitutively nuclear
in their native settings. Nevertheless, it is clear that the regulation of both Capsaspora
and choanoflagellate NF-κB proteins is distinct from what is seen with NF-κB proteins in
higher metazoans.
Of interest, constitutively nuclear localization of NF-κB proteins has also been
seen in other settings. That is, we have previously shown that in both the sea anemone
Aiptasia and sponges, most NF-κB staining is nuclear and the proteins are mostly
processed in their native settings (15–17). Thus, we have argued previously (3) that
these basal NF-κB proteins may be constitutively in an active state, perhaps due to
17
bioRxiv preprint doi: https://doi.org/10.1101/2021.03.15.435342; this version posted November 14, 2021. The copyright holder for this preprint
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
available under aCC-BY 4.0 International license.
continual interaction with upstream activating ligands or pathogens. Of note, most NF-κB
p100 is also in its processed form in mouse spleen tissue (25).
Among the 21 choanoflagellates for which there is sufficient
transcriptomic/genomic information, it appears that only 12 have any NF-κB genes.
Surprisingly, in seven of these 12 choanoflagellates there are multiple NF-κB genes.
Thus, it is clear that there have been gains and losses of NF-κB genes among the
choanoflagellates. We note that NF-κB has also been lost in other organisms including
C. elegans and ctenophores (14). The absence of NF-κB in some choanoflagellates and
its expansion in others (e.g., A. spectabilis) suggests that NF-κB has a specialized,
rather than a general, biological function in choanoflagellates.
The presence of three NF-κB-like heterodimerizing proteins in A. spectabilis is
the first example of an organism basal to flies with multiple NF-κB family proteins that
are capable of forming heterodimers. Thus, expansion of NF-κB genes has occurred
multiple times in evolution, i.e., at least once in the metazoan lineage and once within
choanoflagellates. Furthermore, since each As-NF-κB homodimer has a different ability
to bind DNA and activate transcription, it appears that there are subclasses of NF-κB
within A. spectabilis and likely within other choanoflagellates that have multiple NF-κBs.
It is interesting to note that As-NF-κB1, 2, and 3 are phylogenetically separate and
cluster most closely with NF-κBs from certain other choanoflagellate species that contain
multiple NF-κBs. For example, Savillea parva contains three NF-κBs, each of which
clusters with a separate As-NF-κB (Fig. 1B). Thus, we hypothesize that
choanoflagellates, like vertebrates, have evolved a mechanism for differential
transcriptional control of genes through the use of combinatorial dimer formation.
The differential mRNA expression and DNA-binding activity of NF-κB among
different life stages of Capsaspora suggest that NF-κB has life stage-specific functions. It
is notable that the DNA-binding activity of NF-κB in these different life stages correlates
18
bioRxiv preprint doi: https://doi.org/10.1101/2021.03.15.435342; this version posted November 14, 2021. The copyright holder for this preprint
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
available under aCC-BY 4.0 International license.
with differences in the levels of NF-κB mRNA, rather than as differences in induced
activity. That is, in most metazoans, the activity of NF-κB is regulated at the posttranscriptional level, whereas in Aiptasia and corals, we have found that NF-κB mRNA
levels and DNA-binding activity appear to be coordinately regulated, suggesting
transcriptional regulation. That is, in Aiptasia, thermal bleaching causes transcriptional
upregulation of NF-κB, which also results in increased protein expression of nuclear,
DNA binding-active NF-κB (17), which is similar to what we see with NF-κB across the
Capsaspora life stages. Similarly, treatment of the coral Orbicella faveolata with
lipopolysaccharide results in increased expression of NF-κB target genes, rather than
increased post-translational activation of NF-κB (15). Thus, it appears that in several
basal organisms NF-κB proteins are constitutively nuclear and that increases in their
activity is the result of transcriptional upregulation of NF-κB mRNA, rather than induced
proteolysis, which occurs in most mammalian and fly systems.
Activation of NF-κB by signal-induced degradation of IκB sequences is
essentially dogma in vertebrates. Although several basal NF-κBs, including Co-NF-κB,
can be formulated to undergo IKK-induced processing when expressed in human cells in
culture (15–17), there is much evidence that such regulation may not occur in the native
animals. For example, although sponge and sea anemone Aiptasia NF-κBs have Cterminal ANK repeats and GRRs, they are largely processed and nuclear in the animals
themselves (15–17). Moreover, induction of NF-κB DNA- binding activity and protein
levels by loss of symbiosis in Aiptasia appears to be primarily a result of increased
transcription of NF-κB and not due to induced ANK repeat degradation. Similarly, the
increased NF-κB DNA-binding activity seen in different life stages of Capsaspora is
paralleled by increased expression of NF-κB mRNA in these life stages. Furthermore,
when Co-NF-κB is overexpressed in Capsaspora cells it is primarily in the nucleus, likely
due to constitutive processing to the RHD form. Thus, a more relevant question for many
19
bioRxiv preprint doi: https://doi.org/10.1101/2021.03.15.435342; this version posted November 14, 2021. The copyright holder for this preprint
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
available under aCC-BY 4.0 International license.
basal NF-κB proteins may be what conditions stop them from being processed. Of note,
the three As-NF-κBs do not have C-terminal ANK repeats and we have not been able to
identify a putative IκB in A. spectabilis transcriptomic databases. Therefore, it is possible
that the activity of these constitutively nuclear choanoflagellate NF-κBs is fully regulated
by transcriptional control of their genes.
Of the nearly 1350 genes whose expression correlated with NF-κB expression
across different Capsaspora life stages, almost 20% contained κB binding sites within
500 base pairs upstream their TSS (Fig. 5). While this might be an overestimate of NFκB gene targets or genes indirectly influenced by expression of NF-κB, there are likely
additional NF-κB binding sites that could affect target gene expression. For example,
ATAC-seq data have suggested that the regulatory sites in the Capsaspora genome are
present in first introns, 5’ UTRs, as well as the proximal intergenic regions (26). Among
the ~1350 genes that we identified with expression patterns similar to NF-κB, there are
192 genes that contain NF-κB binding sites in their proximal promoters. Two of these
genes are homologs of SYK and SRC (CAOG_08354 and CAOG_00206), which are
known in higher organisms to be involved in immunity and development. However, the
list of ~1350 genes most certainly contains genes that are controlled by other
transcription factors or are regulated by signaling or developmental events that are
partially or not at all affected by NF-κB. Nevertheless, it is clear that NF-κB might play
several roles, and perhaps different roles, in each life stage of Capsaspora.
Our studies suggest that the regulation and associated biology of NF-κB in
single-celled organisms are different from what is seen in multicellular vertebrates and
flies. Perhaps, the concerted effort of aggregation in Capsaspora and the correlative
decrease in NF-κB in these aggregated cells reflect a need to suppress collective
immunity to form a symbiotic group. Alternatively, Capsaspora is normally a symbiont in
20
bioRxiv preprint doi: https://doi.org/10.1101/2021.03.15.435342; this version posted November 14, 2021. The copyright holder for this preprint
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
available under aCC-BY 4.0 International license.
the hemolymph of the snail B. glabrata, where NF-κB may play a role in maintaining
symbiosis, which has been suggested as one function of NF-κB in other organisms (27).
It is not clear what type of pathway might lead to activation of NF-κB in protists. In
animals across a broad swath of phyla, the binding of a ligand to receptors such as
TLRs/IL-1Rs or TNFRs initiates signaling pathways that converge on an IKK complex
which then activates NF-κB (1). However, in basal organisms, many of these
components are missing, few in number, or lack critical domains (Supplemental Fig. 4).
For example, while some cnidarians contain homologs to TLRs, other cnidarians, some
sponges, and choanoflagellates contain only TIR-domain proteins that lack the important
extracellular components of the TLR (5, 11, 15, 16, 28). Furthermore, Capsaspora does
not contain any homologs to TLRs, ILR-1 or TNFRs (4).
The diversification in NF-κB that we see here between Capsaspora and
choanoflagellates, members of the same supergroup of protists, suggests that the
diversification of NF-κB among all protists will be considerable. Overall, our results
contribute to an understanding of NF-κB across extant phyla. The continued study of the
evolution of NF-κB and other basally derived transcription factors will likely lead to an
understanding of where and how these factors originated, as well as the basal biological
functions they control.
Materials and methods
Phylogenetic analyses
The RHD sequences of NF-κB from C. owczarzaki were compared phylogenetically to
the NF-κB-like sequences present in the transcriptomes of sequenced choanoflagellates.
Details on databases and sequence acquisition can be found in Supplemental Table 6.
Sequences were aligned by Clustal Omega (29). A maximum likelihood phylogenetic
21
bioRxiv preprint doi: https://doi.org/10.1101/2021.03.15.435342; this version posted November 14, 2021. The copyright holder for this preprint
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
available under aCC-BY 4.0 International license.
tree was created using PAUP* (30) and was bootstrapped 1000 times. Bayesian
phylogenetic analyses showed similar results (data not shown).
Plasmids
Expression plasmids for FLAG-tagged human IKKβ, FLAG-Nv-NF-κB, HA-Hu-IKKβ–
SS/EE (constitutively active), FLAG-Ap-IKK, and the empty pcDNA-FLAG vector have
been described previously (17, 31). The cDNA of the codon-optimized NF-κB sequence
of Capsaspora was synthesized in a pUC57 plasmid by GenScript and was then
subcloned into pcDNA-FLAG. PCR-generated Co-NF-κB truncation mutants (Co-RHD
and Co-Cterm) were subcloned into pcDNA-FLAG or the yeast GAL4-fusion vector
pGBT9. Three human codon-optimized As-NF-κB (named 1, 2, and 3) cDNAs were
synthesized by GenScript based on sequences from the transcriptome of A. spectabilis.
These cDNAs were subcloned into pcDNA-FLAG. To create MYC-tagged versions of AsNF-κB2 and 3, the sequences were excised from their pUC57 vectors using EcoRI and
XhoI, and were then subcloned into a MYC-tagged expression vector (a gift of Shigeki
Miyamoto, University of Wisconsin). Details of primers and plasmid constructions can be
found in Supplemental Tables 7 and 8, respectively.
Capsaspora culture, transfection, and protein extraction
Capsaspora cell cultures (strain ATCC ®30864) were grown axenically in 25 cm2 culture
flasks (ThermoScientific Nunclon) with 10 ml ATCC medium 1034 (modified PYNFH
medium) at 23°C. Cultures for each Capsaspora life stage were generated as previously
described (9) and as instructed by ATCC. Filopodic cells were maintained in an actively
dividing adherent state by scraping and passaging 1/40-1/50 of the cultures every 6-8
days, before floating cells appeared. Floating cystic cells were collected from 14 day-old
filopodic cultures. Aggregative cells were created by actively scraping dividing filopodic
22
bioRxiv preprint doi: https://doi.org/10.1101/2021.03.15.435342; this version posted November 14, 2021. The copyright holder for this preprint
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
available under aCC-BY 4.0 International license.
cells and seeding them into a 25 cm2 culture flask, which was gently agitated at 60 RPM
for 4-5 days, and grown axenically at 23°C. Whole-cell lysates of cells from each life
stage were prepared in AT Lysis Buffer (20 mM HEPES, pH 7.9, 150 mM NaCl, 1 mM
EDTA, 1 mM EGTA, 20% w/v glycerol, 1% w/v Triton X-100, 20 mM NaF, 1 mM
Na4P2O7·10H2O, 1 mM dithiothreitol, 1 mM phenylmethylsulfonyl fluoride, 1 μg/ml
leupeptin, 1 μg/ml pepstatin A, 10 μg/ml aprotinin), as described previously (19).
Transfection of Capsaspora cells with expression plasmids was performed using
polyethylenimine (PEI) (Polysciences, Inc.). Briefly, the day before transfection, actively
dividing Capsaspora filopodic cells were plated at about 80-90% confluency. The next
day, cells were transfected by incubation with 5 μg of plasmid DNA and 25 μl of 1 mg/ml
PEI. Media was changed ~20 h post-transfection, and Capsaspora cells to be analyzed
by immunofluorescence were passaged onto poly-D-lysine (ThermoFisher)-treated glass
coverslips on the day prior to fixation.
Vertebrate cell culture and transfection
DF-1 chicken fibroblasts and human HEK 293 or 293T cells were grown in Dulbecco’s
modified Eagle’s Medium (DMEM) (Invitrogen) supplemented with 10% fetal bovine
serum (Biologos), 50 units/ml penicillin, and 50 μg/ml streptomycin as described
previously (19). Transfection of cells with expression plasmids was performed using
polyethylenimine (PEI) (Polysciences, Inc.) as described previously (19, 31). Briefly, on
the day of transfection, cells were incubated with plasmid DNA and PEI at a DNA:PEI
ratio of 1:6. Media was changed ~20 h post-transfection, and whole-cell lysates were
prepared 24 h later in AT Lysis Buffer (20 mM HEPES, pH 7.9, 150 mM NaCl, 1 mM
EDTA, 1 mM EGTA, 20% w/v glycerol, 1% w/v Triton X-100, 20 mM NaF, 1 mM
Na4P2O7·10H2O, 1 mM dithiothreitol, 1 mM phenylmethylsulfonyl fluoride, 1 μg/ml
23
bioRxiv preprint doi: https://doi.org/10.1101/2021.03.15.435342; this version posted November 14, 2021. The copyright holder for this preprint
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
available under aCC-BY 4.0 International license.
leupeptin, 1 μg/ml pepstatin A, 10 μg/ml aprotinin). DF-1 cells analyzed by
immunofluorescence were passaged onto glass coverslips on the day prior to fixation.
Western blotting, electrophoretic mobility shift assays (EMSAs), reporter gene
assays, and indirect immunofluorescence
Western blotting was performed as described previously (17, 19). Briefly, cell extracts
were first separated on 7.5% or 10% SDS-polyacrylamide gels. Proteins were then
transferred to nitrocellulose at 4C at 250 mA for 2 h followed by 170 mA overnight. The
membrane was blocked in TBST (10 mM Tris-HCl [pH 7.4], 150 mM NaCl, 0.1% v/v
Tween 20) containing 5% powered milk (Carnation) for 1 h at room temperature. Filters
were incubated at 4 C with FLAG primary antiserum (1:1000, Cell Signaling
Technology) diluted in 5% milk TBST. After extensive washing in TBST, filters were
incubated with anti-rabbit horseradish peroxidase-linked secondary antiserum (1: 4000,
Cell Signaling Technology). Immunoreactive proteins were detected with SuperSignal
West Dura Extended Duration Substrate (Pierce) and imaged on a Sapphire
Biomolecular Imager (Azure Biosystems).
EMSAs were performed using a 32P-labeled κB-site probe (GGGAATTCCC, see
Supplemental Table 6) and 293T or Capsaspora whole-cell extracts prepared in AT
buffer (17, 19). EMSA gels were exposed on a phosphor screen, and then imaged on a
Sapphire Biomolecular Imager (Azure Biosystems). Yeast GAL4-site LacZ and 293 cell
κB-site luciferase reporter gene assays were performed as described previously (19).
Transfection and indirect immunofluorescence of DF-1 and Capsaspora cells were
performed on methanol-fixed or paraformaldehyde-fixed, respectively, cells that were
probed with rabbit anti-FLAG primary antiserum (1:80, Cell Signaling Technology),
essentially as described previously (19). Nuclei were stained with Hoechst as indicated.
24
bioRxiv preprint doi: https://doi.org/10.1101/2021.03.15.435342; this version posted November 14, 2021. The copyright holder for this preprint
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
available under aCC-BY 4.0 International license.
RNA-sequencing analysis and annotation
RNA-sequencing data from each life stage of Capsaspora was obtained from SebéPedrós et al. (9). To identify potential target genes of NF-κB, we sorted all sequencing
data with genes that were differentially expressed in the same manner as NF-κB for
each life stage (lowest expression in aggregative, medium expression in filopodic,
highest expression in cystic). We discarded genes if they were not expressed at a life
stage (i.e., had an RPKM of 0). Of the 8674 total genes, 1348 genes were differentially
expressed in a manner similar to NF-κB. Of these 1348 genes, 389 genes had annotated
homologs, and 305 had human homologs (Supplemental Table 3). We then performed
GO analysis (http://pantherdb.org) by entering the UniProt ID of each gene and selecting
Homo sapiens. We then created and analyzed the Biological Processes that were
present in this gene list.
To identify genes with potential upstream NF-κB binding sites among these 1348
genes, we also extracted the 500 base pair sequence upstream of each gene. We then
imported these 1348 upstream regions into MEME-FIMO and scanned the sequences
for NF-κB motifs (but not Rel motifs) extracted from JASPAR (http://jaspar.genereg.net)
(Supplemental Table 4).
Immunoprecipitations
HEK 293T cells were transfected with MYC-As-NF-κB2 and either a pcDNA FLAG empty
vector, FLAG-As-NF-κB1, or FLAG-As-NF-κB3 as above (See “Vertebrate cell culture
and transfection”). Lysates were prepared 48 h later and were incubated with 50 μl of a
1X PBS-washed anti-FLAG bead slurry (Sigma) overnight at 4 C with gentle rocking.
The next day, the beads were washed three times with 1X PBS. The pellet was then
25
bioRxiv preprint doi: https://doi.org/10.1101/2021.03.15.435342; this version posted November 14, 2021. The copyright holder for this preprint
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
available under aCC-BY 4.0 International license.
boiled in 2X SDS sample buffer, and the supernatant was electrophoresed on a 7.5%
SDS-polyacrylamide gel, as above (see “Western blotting”). The membrane was then
probed with a rabbit anti-MYC (Cell Signaling Technologies, 1:1000) antiserum, then
with anti-rabbit horseradish peroxidase-linked secondary antiserum (1: 4000, Cell
Signaling Technology), and reacted with SuperSignal West Dura substrate (Pierce). An
image was then obtained on a Sapphire Biomolecular Imager (Azure Biosystems). The
membrane was stripped and probed with rabbit anti-FLAG antiserum as above. These
same transfection and IP experiments were repeated with MYC-As-NF-κB3 and either
pcDNA FLAG empty vector, FLAG-As-NF-κB1, or FLAG-As-NF-κB2.
Acknowledgments
This research was supported by following National Science Foundation grants (to
T.D.G.): IOS-1354935 and IOS-1937650. L.M.W. was supported by an NSF Graduate
Research Fellowship and a Warren-McLeod Graduate Fellowship in Marine Sciences
(Boston University). J.S. and E.K.A. were supported by NSF REU BIO-1659605
(T.D.G.). S.S. was supported by the Boston University (BU) Undergraduate Research
Opportunities Program, and A.A. was supported by the BU GROW program. Designated
authors performed research as part of the undergraduate Molecular Biology Laboratory
course BB522 (Spring, 2019, 2020), and were supported by funds from the BU Biology
Department.
Competing Interest Statement: We have no competing interests.
Author Contributions: L.M.W. and T.D.G planned the project and wrote the
manuscript, and L.M.W performed experiments except those that follow. S.S. cloned
plasmids used in Fig. 6 and performed Western blot in Sup. Fig. 1. J.S. and E.A. did
initial Western blots for Fig. 3 and Luciferase assay on Fig. 2. A.A. analyzed and
selected As-NF-κBs. BB522 Molecular Biology Laboratory, N.R-S. and C.J.D. created
plasmids for Figs. 2B and 6A, and C.J.D. took images for Fig. 6B. P.J.A.C. performed
EMSA on Fig. 2E. T.S. analyzed PBMs in Fig. 2A.
26
bioRxiv preprint doi: https://doi.org/10.1101/2021.03.15.435342; this version posted November 14, 2021. The copyright holder for this preprint
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
available under aCC-BY 4.0 International license.
References
1. T.D. Gilmore, Introduction to NF-κB: players, pathways, perspectives. Oncogene 25,
6680–6684 (2006).
2. S. Ghosh, M.S. Hayden, Celebrating 25 years of NF-κB research. Immunol. Rev.
246, 5–13 (2012).
3. L. M. Williams, T.D. Gilmore, Looking down on NF-κB. Mol. Cell. Biol. 40, e00104-20
(2020).
4. H. Suga, Z. Chen, A. de Mendoza, A. Sebé-Pedrós, M.W. Brown, E. Kramer, M.
Carr, P. Kerner, M. Vervoot, N. Sánchez-Pons, G. Torruella, R. Derelle, G. Manning,
B.F. Lang, C. Russ, B.J. Haas, A.J. Roger, C. Nusbaun, I. Ruiz-Trillo, The
Capsaspora genome reveals a complex unicellular prehistory of animals. Nat.
Commun. 4, 1–9 (2013).
5. D.J. Richter, P. Fozouni, M. B. Eisen, N. King, Gene family innovation, conservation
and loss on the animal stem lineage. Elife 7, e34226 (2018).
6. A.G.B. Simpson, C.H. Slamovits, J.M. Archibald, “Protist Diversity and Eukaryote
Phylogeny” in Handbook of the Protists, J.M. Archibald, et al., Eds. (Springer
International Publishing, 2017), pp. 1–21.
7. M. Ferrer-Bonet, I. Ruiz-Trillo, Capsaspora owczarzaki. Curr. Biol. 27, PR829-PR830
(2017).
8. H. Stibbs H., A. Owczarzak, C. J. Bayne, P. DeWan, Schistosome sporocyst-killing
Amoebae isolated from Biomphalaria glabrata. J. Invert. Pathol. 33, 159–170 (1979).
9. A. Sebé-Pedrós, M. Irimia, J. Del Campo, H. Parra-Acero, C. Russ, C. Nusbaum,
B.J. Biencowe, I. Ruiz-Trillo, Regulated aggregative multicellularity in a close
unicellular relative of metazoa. eLife 2, e01287 (2013).
10. N. King, Choanoflagellates. Curr. Biol. 15, R113-R114 (2005).
11. N. King, M.J. Westbrook, S.L. Young, A. Kuo, M. Abedin, J. Chapman, S. Fairclough,
U. Hellsten, Y. Isogai, I Letunic, M. Marr, D. Pincus, N. Putnam, A. Rokas, K.J.
Wright, R. Zuzow, W. Dirks, M. Good, D. Goodstein, D. Lemons, W. Li, J.B. Lyons, A
Morris, S. Nichols, D.J. Richter, A. Salamov, JGI Sequencing, P. Bork, W.A. Lim G.
Manning, W.T. Miller, W. McGinnis, H. Shapiro, R. Tjian, I.V. Grigorie, D. Rokhsar,
The genome of the choanoflagellate Monosiga brevicollis and the origin of
metazoans. Nature 451, 783–788 (2008).
12. S.R. Fairclough, Z. Chen, E. Kramer, Q. Zeng, S. Young, H.M. Robertson, E
Begovic, D.J. Richter, C. Russ, M.J. Westbrook, G. Manning, B.F. Lang, B Haas, C.
Nusbaum, N. King, Premetazoan genome evolution and the regulation of cell
differentiation in the choanoflagellate Salpingoeca rosetta. Genome Biol. 14, R15
(2013).
27
bioRxiv preprint doi: https://doi.org/10.1101/2021.03.15.435342; this version posted November 14, 2021. The copyright holder for this preprint
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
available under aCC-BY 4.0 International license.
13. S.-C. Sun, Non-canonical NF-κB signaling pathway. Cell Res. 21, 71–85 (2011).
14. T.D. Gilmore, F.S. Wolenski, NF-κB: where did it come from and why? Immunol.
Rev. 246, 14–35 (2012).
15. L.M. Williams, L.E. Fuess, J.J. Brennan, K.M. Mansfield, E. Salas-Rodriguez, J.
Welsh, J. Awtry, S. Banic, C. Chacko, A. Chezian, D. Dowers, F. Estrada, Y.-H.
Hsieh, J. Kang, W. Li, Z. Malchiodi, J. Malinowski, S. Matuszak, T. McTigue IV, D.
Mueller, B. Nguyen, M. Nguyen, P. Nguyen, S. Nguyen, N. Njoku, K. Patel, W.
Pellegrini, T. Pliakas, D. Qadir, E. Ryan, A. Schiffer, A. Thiel, S.A. Yunes, K.E.
Spilios, J.H. Pinzón C, L.D. Mydlarz, T.D. Gilmore, A conserved Toll-like receptor-toNF-κB signaling pathway in the endangered coral Orbicella faveolata. Dev. Comp,
Immunol. 79, 128–136 (2018).
16. L.M. Williams, M.M. Inge, K.M. Mansfield, A. Rasmussen, J. Afghani, M. Agrba, C.
Albert, N. Andersson, M. Babaei, M. Babaei, A. Bagdasaryants, A. Bonilla, A.
Browne, S. Carpenter, T. Chen, B. Christie, A. Cyr, K. Dam, N. Dulock, G. Erdene, L.
Esau, S Esonwune, A Hanchate, X Huang, T Jennings, A Kasabwala, L Kehoe, R
Kobayashi, M. Lee, A. LeVan, Y. Liu, E. Murphy, A. Nambiar, M. Olive, D. Patel, F.
Pavesi, C.A. Petty, Y. Samofalova, S. Sanchez, C. Stejskal, Y. Tang, A. Yapo, J.P.
Cleary, S.A. Yunes, T. Siggers, T.D. Gilmore, Transcription factor NF-κB in a basal
metazoan, the sponge, has conserved and unique sequences, activities, and
regulation. Dev. Comp. Immunol. 104, 103559 (2020).
17. K.M. Mansfield, N.M. Carter, L. Nguyen, P.A. Cleves, A. Alshanbayeva, L.M.
Williams, A. Penvose, J.R. Finnerty, V.M. Weis, T. Siggers, T.D. Gilmore,
Transcription factor NF-κB is modulated by symbiotic status in a sea anemone model
of cnidarian bleaching. Sci. Rep. 7, 16025 (2017).
18. F.S. Wolenski, C.A. Bradham, J.R. Finnerty, T.D. Gilmore, NF-κB is required for
cnidocyte development in the sea anemone Nematostella vectensis. Dev. Biol. 373,
205–215 (2013).
19. F.S. Wolenski, M.R. Garbati, T.J. Lubinski, N. Traylor-Knowles, D.J. Stefanik, E.
Dresselhaus, H. Goucher, J.R. Finnerty, T.D. Gilmore, Characterization of the core
elements of the NF-κB signaling pathway of the sea anemone Nematostella
vectensis. Mol. Cell. Biol. 31, 1076–1087 (2011).
20. R. Steward, Dorsal, an embryonic polarity gene in Drosophila, is homologous to the
vertebrate proto-oncogene, c-rel. Science 238, 692–694 (1987).
21. S. Roth, D. Stein, C. Nüsslein-Volhard, A gradient of nuclear localization of the
dorsal protein determines dorsoventral pattern in the Drosophila embryo. Cell 59,
1189–1202 (1989).
22. C. Kappler, M. Meister, M. Lagueux, E. Gateff, J.A. Hoffmann, J.M. Reichart, Insect
immunity. Two 17 bp repeats nesting a kappa B-related sequence confer inducibility
to the diptericin gene and bind a polypeptide in bacteria-challenged Drosophila.
EMBO J. 12, 1561–1568 (1993).
28
bioRxiv preprint doi: https://doi.org/10.1101/2021.03.15.435342; this version posted November 14, 2021. The copyright holder for this preprint
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
available under aCC-BY 4.0 International license.
23. J.M. Reichhart, P. Georgel. M. Meister, B. Lemaitre, C. Kappler, J.A. Hoffmann,
Expression and nuclear translocation of the rel/NF-κB-related morphogen dorsal
during the immune response of Drosophila. C. R. Acad. Sci. III 316, 1218–1224
(1993).
24. S. Stöven, N. Silverman, A. Junell, M. Hedengren-Olcott, D. Erturk, Y. Engström, T.
Maniatis, D. Hultmark, Caspase-mediated processing of the Drosophila NF-κB factor
Relish. Proc. Natl. Acad. Sci. U.S.A. 100, 5991–5996 (2003).
25. U. Senftleben, Y. Cao, G. Xiao, F.R. Greten, G. Krähn, G. Bonizzi, Y. Chen, Y. Hu,
A. Fong, S.-C. Sun, M. Karin, Activation by IKK of a second, evolutionary
conserved, NF-κB signaling pathway. Science 293, 1495–1499 (2001).
26. A. Sebé-Pedrós, C. Ballaré, H. Parra-Acero, C. Chiva, J.J. Tena, E Sabidó, J.L.
Gómez-Skarmeta, L. Di Croce, I. Ruiz-Trillo, The dynamic regulatory genome of
Capsaspora and the origin of animal multicellularity. Cell 165, 1224–1237 (2016).
27. K.M. Mansfield, T.D. Gilmore, Innate immunity and cnidarian-Symbiodiniaceae
mutualism. Dev. Comp. Immunol. 90, 199–209 (2019).
28. J.J. Brennan, T.D. Gilmore, Evolutionary origins of Toll-like receptor signaling. Mol.
Biol. Evol. 35, 1576–1587 (2018).
29. F. Sievers, A. Wilm, D Dineen, T.J. Gibson, K. Karplus, W. Li, R Lopez, H.
McWilliam, M. Remmert, J. Söding. J.D. Thompson, D.G. Higgins, Fast, scalable
generation of high-quality protein multiple sequence alignments using Clustal
Omega. Mol. Syst. Biol. 7, 539 (2011).
30. D. L. Swofford, PAUP*: Phylogenetic Analysis Using Parsimony (and other methods)
4.0.b5 (2001).
31. J.C. Sullivan, F.S. Wolenski, A.M. Reitzel, C.E. French, N. Traylor-Knowles, T.D.
Gilmore, J.R. Finnerty, Two alleles of NF-κB in the sea anemone Nematostella
vectensis are widely dispersed in nature and encode proteins with distinct activities.
PLoS ONE 4, e7311 (2009).
29
bioRxiv preprint doi: https://doi.org/10.1101/2021.03.15.435342; this version posted November 14, 2021. The copyright holder for this preprint
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
available under aCC-BY 4.0 International license.
Fig. 1. Protist NF-κB proteins differ in domain structure and choanoflagellates
show evidence of gene duplication. (A) The general domain structures of both
Capsaspora NF-κB and the choanoflagellate RHD-only proteins. Green, RHD (Rel
Homology Domain); Purple, nuclear localization sequence; Blue, GRR (gylcine-rich
region); Black bars, Ankryin repeats; Pink, sequences in choanoflagellates that are not
typically seen in other organisms. (B) A phylogenetic estimation using maximum
likelihood (bootstrapped 1000 times) of choanoflagellate and Capsaspora RHDs. The
NF-κB proteins used in these studies are highlighted in red.
30
bioRxiv preprint doi: https://doi.org/10.1101/2021.03.15.435342; this version posted November 14, 2021. The copyright holder for this preprint
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
available under aCC-BY 4.0 International license.
Fig. 2. DNA-binding and transcriptional activation activity of Co-NF-κB. (A) Protein
binding microarray (PBM) DNA-binding profiles of Co-NF-κB as compared to
Nematostella vectensis (Nv) NF-κB cysteine (cys) allele (top, left), human (Hu) RelA
31
bioRxiv preprint doi: https://doi.org/10.1101/2021.03.15.435342; this version posted November 14, 2021. The copyright holder for this preprint
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
available under aCC-BY 4.0 International license.
(top, right), human p52 (bottom, left), and human cRel (bottom, right). The axes are zscores. Red dots represent random background sequences, and blue dots represent
NF-κB binding sites. (B) FLAG-tagged expression vectors used in these experiments.
From top to bottom, the drawings depict the naturally shortened Nv-NF-κB, the fulllength Co-NF-κB protein, an N-terminal-only mutant containing the RHD and GRR (CoRHD), and a C-terminal-only mutant containing the ANK repeats and other C-terminal
sequences (Co-Cterm). (C) Anti-FLAG Western blot of lysates of HEK 293T cells
transfected with the indicated expression vectors. (D) Indirect immunofluorescence of
DF-1 chicken fibroblast cells transfected with the indicated expression vectors. Cells
were then stained with anti-FLAG antiserum (left panels) and HOESCHT (middle
panels), and then MERGED on the right panels. (E) A κB-site electromobility shift assay
(EMSA) using each of the indicated lysates from (C). The NF-κB complexes and free
probe are indicated by arrows. (F) A κB-site luciferase reporter gene assay was
performed with the indicated proteins in HEK 293 cells. Luciferase activity is relative
(Rel.) to that seen with the empty vector control (1.0), and values are averages of three
assays performed with triplicate samples with standard error. (G) A GAL4-site LacZ
reporter gene assay was performed in yeast Y190 cells. Values are average units from
seven assays performed with duplicate samples with standard error.
32
bioRxiv preprint doi: https://doi.org/10.1101/2021.03.15.435342; this version posted November 14, 2021. The copyright holder for this preprint
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
available under aCC-BY 4.0 International license.
Fig. 3. IKK-mediated processing of NF-κB arose with multicellularity. (A) Cotransfection with various kinases does not induce processing of FLAG-Co-NF-κB in HEK
293T cells. Arrows indicate the various FLAG- or HA-tagged kinases used in these
assays. Full-length Co-NF-κB and processed Co-NF-κB are also indicated. (B) Co-NFκB with C-terminal IKK target serines from Aiptasia NF-κB (Co-NF-κB-SER) or serine-toalanine mutants (Co-NF-κB-ALA) were co-transfected with constitutively active human
33
bioRxiv preprint doi: https://doi.org/10.1101/2021.03.15.435342; this version posted November 14, 2021. The copyright holder for this preprint
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
available under aCC-BY 4.0 International license.
HA-IKKβ (SS/EE). Transfecting Co-NF-κB-SER and HA-IKKβ (SS/EE) resulted in the
appearance of an increased amount of the lower band, but the alanine mutations (CoNF-κB-ALA) abolished that processing.
34
bioRxiv preprint doi: https://doi.org/10.1101/2021.03.15.435342; this version posted November 14, 2021. The copyright holder for this preprint
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
available under aCC-BY 4.0 International license.
Fig. 4. Transfection of FLAG-Co-NF-κB and FLAG-Co-RHD into Capsaspora cells
results in nuclear localization. Capsaspora cells were transfected with FLAG-tagged
vectors for full-length Co-NF-κB, mutant Co-RHD, and mutant Co-Cterm. The cells were
stained using anti-FLAG antiserum (left panels) and HOECHST (middle panels), and
then MERGED (right panels). Scale bars are 2 µM.
35
bioRxiv preprint doi: https://doi.org/10.1101/2021.03.15.435342; this version posted November 14, 2021. The copyright holder for this preprint
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
available under aCC-BY 4.0 International license.
Fig. 5. NF-κB is differentially expressed during the different life stages of
Capsaspora and its proposed roles in development and immunity. (A) Top: The
FPKM values from Sebé- Pedrós et al. (9) of NF-κB at each life stage, done in triplicate.
Agg, Aggregative (yellow), Filo, Filopodic (green), Cys, Cystic (blue). Error bars are
standard deviation. Bottom: Images taken with a light microscope of each life stage
(Agg, Filo, and Cys from left to right). Yellow scale bar is 20 µM. (B) Capsaspora wholecell extracts were created from each life stage (see Methods). 70 µg of each extract was
then used in an electromobility shift assay (EMSA). Lane 1 contains only free probe.
36
bioRxiv preprint doi: https://doi.org/10.1101/2021.03.15.435342; this version posted November 14, 2021. The copyright holder for this preprint
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
available under aCC-BY 4.0 International license.
Lanes 2-4 contain lysates from Agg, Filo, and Cys life stages incubated with a
radioactive κB-site probe. Lanes 5 and 9 are empty. Lanes 6-8, and lanes 10-12 contain
lysates from Agg, Filo, and Cys life stages as indicated, and were incubated with an
excess (10X and 25X, respectively) of unlabeled κB-site probe. Lane 13 contains the
Cys lysate incubated with 25X unlabeled IRF-site probe. NF-κB complexes and Free
Probe are indicated with arrows. The dashed lines indicate where the gel was cut to
remove excess lanes. (C) NF-κB expression may influence the expression of genes that
are involved in developmental and immune system processes. The expression profiles
of these genes correlate with NF-κB mRNA expression in each life stage (Agg, low; Filo,
medium; Cys, high), and were identified via Biological Processes GO analysis. Two of
the genes in this list (SYK and SRC) also contain κB sites in the 500 bp upstream of
their TSS.
37
bioRxiv preprint doi: https://doi.org/10.1101/2021.03.15.435342; this version posted November 14, 2021. The copyright holder for this preprint
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
available under aCC-BY 4.0 International license.
Fig. 6. Characterization of cellular and molecular properties of three
choanoflagellate NF-κBs. (A) FLAG-tagged NF-κB proteins used in these experiments.
38
bioRxiv preprint doi: https://doi.org/10.1101/2021.03.15.435342; this version posted November 14, 2021. The copyright holder for this preprint
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
available under aCC-BY 4.0 International license.
From top to bottom the drawings depict the three NF-κB-like proteins from the
transcriptome of A. spectabilis (5). RHDs are in green (Rel Homology Domain). (B) AntiFLAG Western blot of lysates from HEK 293T cells transfected with the indicated
expression vectors. (C and D) Indirect immunofluorescence of DF-1 chicken cells (C) or
Capsaspora cells (D) transfected with the indicated expression vectors. Cells were then
stained with anti-FLAG antiserum (left panels) and HOESCHT (middle panels), and then
MERGED in the right panels. Yellow scale bar in Capsaspora panels is 2 µM. (E) A κBsite electromobility shift assay (EMSA) using each of the indicated lysates from (B). The
NF-κB complexes and free probe are indicated by arrows. (F) A κB-site luciferase
reporter gene assay was performed with the indicated proteins in HEK 293 cells.
Luciferase activity is relative (Rel.) to that seen with the empty vector control (1.0), and
values are averages of three assays performed with triplicate samples with standard
error. Values are shown on a log scale. (G) A GAL4-site LacZ reporter gene assay was
performed with GAL4-fusion proteins in yeast Y190 cells. Values are average units of
two assays performed with four samples with standard error. (H) Coimmunoprecipitation
(IP) assays of MYC-tagged As-NF-κB2 and As-NF-κB3. In each IP assay, MYC-As-NFκBs were co-transfected with pcDNA FLAG, FLAG-As-NF-κB1, 2 or 3 as indicated. An
IP using anti-FLAG beads was performed. Anti-FLAG (top) and anti-MYC (middle)
Western blotting was then performed. An anti-MYC Western blot of the whole-cell (WC)
lysates was also performed (bottom).
39