Download as pdf or txt
Download as pdf or txt
You are on page 1of 12

Featured

Communication

Optogenetic Control of the Canonical


Wnt Signaling Pathway During Xenopus
laevis Embryonic Development

Vishnu V. Krishnamurthy a, Hyojeong Hwang b, Jia Fu b, Jing Yang b⇑ and


Kai Zhang a,c,d,e,f⇑

a - Department of Biochemistry, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA


b - Department of Comparative Biosciences, University of Illinois at Urbana-Champaign, Urbana, IL 61802, USA
c - Neuroscience Program, University of Illinois at Urbana–Champaign, Urbana, IL 61801, USA
d - Center for Biophysics and Quantitative Biology, University of Illinois at Urbana–Champaign, Urbana, IL 61801, USA
e - Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana–Champaign, Urbana,
IL 61801, USA
f - Cancer Center at Illinois, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA

Correspondence to Jing Yang and Kai Zhang:


2001 S Lincoln Ave, VMBSB3411, Urbana, IL 61802,
USA (J. Yang). 600 South Mathews Avenue,
314 B Roger Adams Laboratory, Urbana, IL 61801,
USA. Fax: 1 217 244 5858. (K. Zhang).
yangj@illinois.edu (J. Yang), kaizkaiz@illinois.edu
(K. Zhang) Vishnu V. Krishnamurthy Hyojeong Hwang Jia Fu

https://doi.org/10.1016/j.jmb.2021.167050
Edited by M. Yaniv

Abstract
Optogenetics uses light-inducible protein-protein
interactions to precisely control the timing, local- Jing Yang Kai Zhang

ization, and intensity of signaling activity. The pre-


cise spatial and temporal resolution of this
emerging technology has proven extremely attrac-
tive to the study of embryonic development, a pro-
gram faithfully replicated to form the same
organism from a single cell. We have previously
performed a comparative study for optogenetic
activation of receptor tyrosine kinases, where we
found that the cytoplasm-to-membrane
translocation-based optogenetic systems outper-
Legend: The low-density lipoprotein receptor protein 6, or LRP6,
form the membrane-anchored dimerization sys- mediates the canonical Wnt signaling pathway and plays an essential
tems in activating the receptor tyrosine kinase role during development and disease. The authors demonstrated
signaling in live Xenopus embryos. Here, we deter- optogenetic activation of the Wnt pathway by light-inducible cytoplasm-
to-membrane translocation of the LRP6 cytosolic domain, which induces
mine if this engineering strategy can be general- body axis duplication in developing frog embryos. The cover refers to the
ized to other signaling pathways involving article by Krishnamurthy et al. “Optogenetic control of the canonical Wnt
membrane-bound receptors. As a proof of con- signaling pathway during Xenopus laevis embryonic development ” in
JMB Volume 433, Issue 18, Pages 1–12. Ella Maru Studio designed the
cept, we demonstrate that the cytoplasm-to- illustration.
membrane translocation of the low-density lipopro-
tein receptor-related protein-6 (LRP6), a
membrane-bound coreceptor for the canonical

0022-2836/Ó 2021 Elsevier Ltd. All rights reserved. Journal of Molecular Biology 433 (2021) 167050
V.V. Krishnamurthy, H. Hwang, J. Fu, et al. Journal of Molecular Biology 433 (2021) 167050

Wnt pathway, triggers Wnt activity. Optogenetic activation of LRP6 leads to axis duplication in developing
Xenopus embryos, indicating that the cytoplasm-to-membrane translocation of the membrane-bound
receptor could be a generalizable strategy for the construction of optogenetic systems.
Ó 2021 Elsevier Ltd. All rights reserved.

Introduction membrane. This cytoplasm-to-membrane


translocation-(CMT)-based strategy has been suc-
Membrane-bound receptors serve as antennas cessfully utilized to construct multiple optogenetic
for cells and integrate signals from the receptor tyrosine kinases (RTK) such as CMT-
extracellular environment. These signals are optoTrkA, TrkB, TrkC, and FGFR.8,9 We speculated
then transmitted to the cell interior and regulate that cytoplasmic expression of the RTKs’ intracellu-
the cellular responses to environmental stimuli. lar domain (ICD) effectively lowers the avidity of the
Under physiological conditions, ligand binding otherwise membrane-bound receptor ICDs, and
induces a conformational change to activate the therefore reduces the basal activity of the optoge-
receptor. Because one type of ligand could bind netic RTK system.
to different types of receptors or different In this work, we aim to determine if a similar
isoforms of the same receptor, understanding engineering strategy could be generalized to other
the mechanism by which cells achieve signaling signaling pathways that involve membrane-bound
specificity remains challenging. For instance, in receptors. We focus on the canonical Wnt
the context of neurotrophic signaling pathways, signaling pathway, which mediates essential
nerve growth factors could bind to both TrkA functions in embryogenesis, organogenesis, and
and p75NTR, each of which elicits distinct stem cell self-renewal in adult tissues.10,11 Promi-
cellular outcomes.1 nent roles of the canonical Wnt pathway during ver-
To delineate the signaling outcomes from tebrate early embryonic development include
individual receptor tyrosine kinases (RTKs), we patterning of the dorsal/ventral and anterior-
and others have developed optogenetic RTKs that posterior body axes,12 regulation of somitogene-
use light-induced protein-protein interaction to sis,13,14 cell fate specification in the skeletal15 and
either dimerize the receptor or translocate the nervous systems,16 and development and mainte-
intracellular domain of the RTK to the plasma nance of zonation in the adult liver,17,18 to name a
membrane.2–7 Mounting evidence suggests that few. Activation of the canonical Wnt pathway is
these optogenetic systems activate downstream mediated by ligand binding to the membrane-
signaling in a ligand-independent way, thereby bound coreceptor, LRP6. Here, we show that
enabling intricate perturbation of the temporal pro- light-mediated cytoplasm-to-membrane transloca-
files in RTK signaling. More importantly, the precise tion of the cytosolic domain of LRP6 (LRP6c) effec-
spatial resolution allows for light-guided cellular tively activates the canonical Wnt pathway.
growth and neuronal repair in multicellular organ- Signaling outcome was confirmed in both culture
isms such as Drosophila39 and zebrafish.40 cells and live Xenopus laevis embryos. Notably,
In our previous work, we performed a we successfully demonstrate that optogenetic con-
comparative study of optogenetic approaches to trol of the Wnt signaling results in duplication of the
activate receptor tyrosine kinases in cultured body axis in developing Xenopus embryos, which
mammalian cell monolayers and Xenopus involves the formation of ectopic Spemann Orga-
embryos.8 We found that optimal optogenetic nizer during gastrulation.
design requires a sufficient dynamic range (i.e., light
versus dark outcome) and a relatively low basal
activity. For instance, although optogenetically-
induced dimerization of membrane-bound TrkA pro- Results and discussion
duced an acceptable dynamic range in the signal
readout, its high basal activity in the dark reduced Membrane-anchoring of LRP6c enhances its
its effectiveness in live embryos. Adjusting the signaling activity
expression level of optogenetic TrkA could amelio- We previously showed that the cytoplasm-to-
rate the basal activity to an extent but requires membrane translocation-based optogenetic
time-consuming empirical testing to achieve systems enable precise activation of the RTK
desired activity levels. A better approach to mini- signaling, raising the possibility that this
mize basal activity involves expressing the recep- engineering strategy may be generalized for the
tor’s intracellular domain in the cytosolic development of novel optogenetic tools to
compartment where homodimerization is less likely, modulate signaling pathways involving membrane-
followed by light-mediated translocation to the bound receptors.8 To test this hypothesis, we focus
dimerization-conducive environment of the plasma on the Wnt pathway, which plays numerous roles
2
V.V. Krishnamurthy, H. Hwang, J. Fu, et al. Journal of Molecular Biology 433 (2021) 167050

during embryonic development, adult tissue home- activated, is responsible for triggering the down-
ostasis, and tumorigenesis. stream intracellular signaling cascade. Truncations
To determine if the cytoplasm-to-membrane of the ECD or the ICD produce a constitutively
translocation-based optogenetic strategy works for active or a dominant-negative version of the protein,
the Wnt pathway, we chose LRP6, the coreceptor respectively.21 Interestingly, LRP6c alone cannot
of the Wnt ligand.19 It has been reported that the activate Wnt-independent b-catenin signaling
full-length LRP6 exists in an inactive state by default unless expressed at very high levels, whereas the
and is ineffective in manifesting the Wnt phenotype TM-containing LRP6c can potently activate sig-
in Xenopus embryos in the absence of the ligand.20 nalling.22 In agreement with this report, we found
LRP6 contains an extracellular ligand-binding GFP-tagged LRP6c elicited no detectable activity
domain (ECD), a transmembrane domain (TMD), in the widely used Wnt-responsive TopFlash repor-
and a cytosolic domain. The ligand-responsive ter assay. In contrast, the membrane-anchored
ECD serves to regulate the protein’s activity, form showed significantly higher activity (Figure 1
whereas the C-terminal end of the molecule, once (a)), which suggests that LRP6c may be an ideal

3
V.V. Krishnamurthy, H. Hwang, J. Fu, et al. Journal of Molecular Biology 433 (2021) 167050

candidate for developing an optogenetic tool to con- increase of activity was only a modest two-fold over
trol the Wnt pathway via the cytoplasm-to- the basal level in the dark (Figure 1(f), Figure 2(a)
membrane translocation strategy. system 2).

Light-inducible cytoplasm-to-membrane Improving the dynamic range of OptoLRP6 by


translocation of LRP6c activates Wnt signaling structural optimization
The cytoplasm-to-membrane translocation- We sought to optimize the construct to achieve a
based optogenetic systems we had previously better light/dark dynamic range to match the levels
engineered relies on blue light-induced achievable with a canonical Wnt ligand (~25–30
heterodimerization of the photolyase-homologous fold).25 As a first step, we eliminated mCherry,
region (PHR) domain from Arabidopsis bringing LRP6c closer to the plasma membrane
cryptochrome2 (CRY2) protein and its when recruited. This construct significantly
heterodimerizing partner cryptochrome interacting enhanced the fold change produced by OptoLRP6,
basic helix-loop-helix N-terminal domain (CIBN).23 reaching a 12-fold light/dark ratio (Figure 2(a), sys-
We adapted the system by replacing Raf1 with tem 3). When we supplemented the membrane-
human LRP6c to create an OptoLRP6 (Figure 1 targeting sites by co-transfecting a membrane-
(b)). When expressed in cells, this single polypro- anchored CIBN plasmid (CIBN-CaaX) alongside
tein CRY2PHR-mCherry-LRP6c-P2A-CIBN  2-G OptoLRP6 lacking mCherry, we observed remark-
FP-CaaX splits into a cytosolic CRY2PHR- ably improved TopFlash reporter activity, producing
mCherry-LRP6c and a membrane-anchored a 46-fold light-over-dark ratio. (Figure 2(a), system
CIBN  2-GFP-CaaX due to the ribosomal skipping 4). These results suggested that LRP6c proximity
activity of the P2A peptide. Since blue light has to the plasma membrane and increased binding
been shown to promote CRY2-CIBN interaction avidity are more conducive to LRP6c activation,
and, to a lesser extent CRY2 homo-association,24 drawing similarities to our earlier experiences with
we reasoned that LRP6c fused to CRY2PHR Raf1.23 Because the phosphorylation of LRP6c,
should experience both a translocation to the especially at its five “PPPSPxS” motifs A-E,20 is cru-
membrane-located CIBN and a clustering effect cial to its recruitment of Axin, we sought to improve
from CRY2 oligomerization, both of which should the activation state of LRP6c by facilitating its phos-
enhance the LRP6c activity. We then confirmed that phorylation. The transmembrane protein TMEM198
blue light could induce the translocation of the has been shown to enhance LRP6 function by pro-
LRP6c to the plasma membrane via live-cell imag- moting its aggregation and CK1c-induced phospho-
ing (Figure 1(c) and (d)). The membrane transloca- rylation.26 Fusing the cytosolic domain of TMEM198
tion reached saturation levels with a few short to LRP6c resulted in more robust LRP6c phospho-
pulses (Figure 1(e), t1/2 ~ 4 sec). In HEK293T cells rylation by CK1c.26 We speculated that including
overexpressing OptoLRP6, blue light also this short tail region of TMEM (TMEMc) will
increased TopFlash luciferase levels compared to enhance the functionality of our optogenetic con-
the dark controls. However, this light-induced struct. We fused TMEMc to the membrane-

3
Figure 1. Construction and validation of the OptoLRP6 system. (a) TopFlash luciferase assay for HEK293T
cells in 48-well plates transiently transfected with the indicated mass of EGFP-labeled LRP6c or LRP6c-CaaX
constructs. Twenty-four hours following transfection, the cells were lysed, and luciferase assay was performed.
Values represent mean ± s.d. of three biological replicates (n = 3). Representative images of BHK21 cells transiently
transfected with EGFP-labeled LRP6c or LRP6c-CaaX shown in the inset. (b) Schematic of the OptoLRP6 construct
and its proposed mechanism of action. Cytosolic CRY2-mCherry-LRP6c is reversibly recruited to plasma membrane-
anchored CIBN with blue-light and subsequently homo-associates via the double CIBN. (c) Epifluorescence
microscopic images of OptoLRP6 in transfected HEK293T cells. Cells plated on glass coverslips were transiently
transfected and imaged after 24 h. Images from the EGFP and mCherry channels were recorded before and after
administration of ten short (100 ms) blue light (488 nm) pulses using the microscope 100  objective. (d) Intensity plot
profiles of lines 1 and 2 represented in (c). Pre-blue light profiles (black) and the post-blue light profiles (blue) are
indistinguishable for GFP, whereas mCherry shows marked breaking of symmetry due to its redistribution from the
cytosol to the plasma membrane. (e) Kinetics of plasma membrane translocation of cytosolic mCh-LRP6c in
HEK293T transfected with OptoLRP6. Cells were imaged under the 100  objective while being pulsed with 100 ms
pulses of blue light every 5 s. Each point represents the average membrane/cytoplasmic fluorescence ratio of
arbitrarily selected regions of interest from ten cells (n = 10) after background normalization (f) TopFlash luciferase
assay for HEK293T cells grown in 48-well plates and transiently transfected with 50 ng OptoLRP6 along with two
luciferase reporter constructs, one of which serves as a transfection normalization control. Sixteen hours following
transfection, the cells were illuminated for 24 h with 450 mW/cm2 blue light (460 nm), and luciferase assay was
performed. The readings were normalized to non-illuminated OptoLRP6-transfected cells. Values represent
mean ± s.d. of three biological replicates (n = 3).
4
V.V. Krishnamurthy, H. Hwang, J. Fu, et al. Journal of Molecular Biology 433 (2021) 167050

anchored CIBN, reasoning that CRY2-LRP6c plasma membrane. We also obtained significant
recruited to the CIBN-TMEMc can be phosphory- gains in signaling activity when we substituted
lated more strongly. We observed that this fusion CIBN with its truncated form CIB81,27 which fur-
indeed further activated the TopFlash reporter more ther shortens the distance between LRP6c and
than 18-fold upon blue light illumination (Figure 2 the membrane (Figure 2(a), system 6). Substitu-
(a), system 5), a nine-fold improvement over the tion of 2  CIBN with 4  CIB81 resulted in a
original OptoLRP6 (Figure 2(a), system 2). TopFlash activity close to 50-fold, thereby sur-
Because the activity of LRP6 is strongly passing our desired activity level within a single
correlated with both its clustering ability and its construct (Figure 2(a), system 8). To confirm the
membrane proximity, we sought to increase the observed enhancement of luciferase activity
local concentration of CRY2-LRP6c at the resulted from LRP6c activity, we truncated the last

5
V.V. Krishnamurthy, H. Hwang, J. Fu, et al. Journal of Molecular Biology 433 (2021) 167050

three phosphorylation repeats (C, D, and E) of hours. Cells were categorized into different
LRP6c (LRP6cABDCDE). As expected, groups, including non-transfected (orange), b-
LRP6cABDCDE failed to show any degree of catenin-mRuby2 singly transfected (red),
activity in the reporter assay (Figure 2(a), system OptoLRP6opt (or CIBN-EGFP-CaaX) singly
7), suggesting that the luciferase signal is induced transfected (green), or doubly-transfected (pink)
by LRP6c activity. We thus referred to this opti- (Figure S1). Blue light-treatment led to an
mized 4  CIB81-containing construct (Figure 2 exposure time-dependent increase in the
(a), system 8, and Figure 2(b)) as OptoLRP6opt. emergence of mRuby2-positive cells in the
Comparable to a mock-transfected control, OptoLRP6opt-transfected population, but not in
OptoLRP6opt exhibited low basal activity in the the control group consisting of b-catenin-mRuby2
dark and showed evident dose-dependent activity co-transfected with CIBN-GFP-CaaX, indicating
between 10 ng and 100 ng of transfected DNA per the stabilization of b-catenin-mRuby2 upon
well in a standard 48-well tissue culture plate OptoLRP6opt activation (Figure 2(d)). Such
(1.1 cm2 per well) (Figure 2(c)). stabilization peaked at 6 hours and slightly
decreased at 8 hours, likely due to intrinsic b-
catenin-mRuby2 degradation. We also noted that
OptoLRP6opt activation causes b-catenin
the basal level of double-positive cells is higher in
stabilization in HEK293T cells
OptoLRP6opt transfected cells (45%) compared
To confirm that OptoLPR6opt activates the with CIBN-EGFP-CaaX-transfected cells (15%),
canonical Wnt signaling pathway, we probed the likely a result of the slight stabilization effect from
level of stabilized b-catenin, the primary OptoLRP6opt overexpression. Because b-catenin-
downstream effector of LRP6. In the absence of mRuby2 is constitutively unstable, we imposed a
the Wnt ligand, the destruction complex, a tertiary conservative gate for our flow experiment so that
complex containing Axin, adenomatous polyposis even cells with faint b-catenin-mRuby2 signal
coli (APC), the Ser/Thr kinases GSK-3 and CK1a, could be identified. Indeed, the basal-level b-
protein phosphatase 2A (PP2A), and the E3- catenin-mRuby2 stabilization in the dark (45%)
ubiquitin ligase b-TrCP, phosphorylates b-catenin, does not cause significant Wnt activation, as
which subsequently undergoes proteasomal shown in the following embryonic assays.
degradation.28 Wnt ligand-binding causes dephos- Besides causing an increase in the percentage of
phorylation and stabilization of b-catenin, allowing double-positive cells, OptoLRP6opt activation also
its association with T-cell factor in the nucleus to led to a significant increase of normalized b-
regulate transcription. catenin-mRuby2 intensity (Figure 2(e)),
To demonstrate that light-activated OptoLRP6opt suggesting an accumulation of stabilized b-
enhances b-catenin levels and to follow the kinetics catenin. The normalized b-catenin-mRuby2
of its stabilization, we carried out two-color flow intensity peaked at 2 hours after light stimulation
cytometry experiments in HEK293T cells co- and reached a plateau afterward, suggesting
transfected with b-catenin-mRuby2 and establishing a new balance between b-catenin-
OptoLRP6opt (labeled with EGFP) stimulated with mRuby2 stabilization and degradation. In contrast,
different duration of blue light ranging from 0 to 8 in cells co-transfected with b-catenin-mRuby2 and

3
Figure 2. Optimization of the OptoLRP6 system. (a) TopFlash luciferase assays for the modified OptoLRP6
constructs bearing various improvements described in the main text. Cell plating, transfection, and illumination were
done as described in Figure 1(f). For cells co-transfected with CIBN-CaaX, 150 ng of the construct was used. A
plasmid containing the mRuby2 fluorophore was used as a filler to keep the total transfected DNA constant across all
experimental conditions. The control sample is CIBN-CaaX. The values are fold-changes of light- over dark-incubated
samples. The fold-changes are indicated at the right of each column. The numbers left to the columns serve as
system identifiers. The five phosphorylation “PPPSPxS” motifs A-E are indicated as red circles on LRP6c. The mutant
in (a) lacks three of these motifs. RLU is relative luciferase units. TMEMc is marked as a blue “T” in the scheme. The
data were obtained from three biological replicates (n = 3). (b) Schematic of the optimized OptoLRP6opt system. (c)
Dose-resistance of non-illuminated HEK293T cells (black bars) in 48-well plates transfected with the indicated doses
of OptoLRP6opt. Cells illuminated with 450 mW/cm2 blue light show luciferase signals scaling nearly linearly with dose
(grey bars). The control sample is CIBN-CaaX. (n = 3) (d) Flow-cytometry analysis of double-positive percentage in
HEK293T cells co-transfected with b-catenin-mRuby2 and OptoLRP6opt (blue) or CIBN-EGFP-CaaX (black) in
response to blue light stimulation from 0 to 8 hours. (e) Flow-cytometry analysis of normalized b-catenin-mRuby2
fluorescence intensity in HEK293T cells co-transfected with b-catenin-mRuby2 and OptoLRP6opt (blue) or CIBN-
EGFP-CaaX (black) in response to blue light stimulation from 0 to 8 hours. Cell counts for each conditions (double
positive/total EGFP expression) are 0 h: 2957/6575, 2 h: 2261/4847, 4 h: 13265/22962, 6 h: 9551/14474, 8 h: 2947/
4664 for OptoLRP6opt and 0 h: 4114/29200, 2 h: 4656/31127, 4 h: 5230/31127, 6 h: 3351/21929, 8 h: 4339/27836 for
CIBN-EGFP-CaaX.
6
V.V. Krishnamurthy, H. Hwang, J. Fu, et al. Journal of Molecular Biology 433 (2021) 167050

CIBN-EGFP-CaaX, blue light stimulation did not in Xenopus laevis embryos (Figure 3(a)). Normal
change the b-catenin-mRuby2 levels (Figure 2(e)). Xenopus embryos contain a single Spemann
Organizer, which induces dorsal tissues during
development by secreting several bone
OptoLRP6opt activation elicits duplication of morphogenetic protein (BMP) inhibitors, including
the body axis in Xenopus embryos Chordin, Noggin, and Follistatin.29 To confirm that
To test whether OptoLRP6opt functions in whole OptoLRP6opt elicits the canonical Wnt signaling,
organisms, we carried out optogenetic activation we used an animal cap assay to determine the opto-

7
V.V. Krishnamurthy, H. Hwang, J. Fu, et al. Journal of Molecular Biology 433 (2021) 167050

genetic transcriptional induction of xnr3 and si- embryos exposed to blue light showed severe
amois, both of which contain multiple TCF/LEF truncation of the head (Figure 3(f) and 3(h)).
binding sites in their promoter and are direct targets These results demonstrate the functionality of
of Wnt signaling. We injected 10 pg and 50 pg of OptoLRP6opt in embryos. To our knowledge,
OptoLRP6opt mRNA into the animal pole of fertil- OptoLRP6opt represents the first successful
ized eggs. Half of the embryos were exposed to optogenetic tool capable of activating the
blue light from stage 6 to stage 8.5, while other canonical Wnt signaling in vertebrate embryos.
embryos were cultured in the dark. Animal caps Notably, Bugaj et al. demonstrated that clustering
were dissected at stage 9 and harvested at stage LRP6c in the cytoplasm using CRY2 is sufficient to
10 for RT-PCR. As expected, OptoLRP6opt activa- induce Wnt responses in cultured cells.31 Since the
tion increased the mRNA level of xnr3 and siamois application of blue-light optogenetics in tissues or
in a dose-dependent manner (Figure 3(b)), demon- organisms is more challenging due to limited light
strating activation of the canonical Wnt signaling penetration, and CRY2 clustering has a higher
pathway in animal caps. Since ectopic activation power requirement when compared with CRY2-
of Wnt signaling in Xenopus embryos induces a CIBN hetero-association,32 we compared the light
secondary Organizer,30 we asked if OptoLRP6opt responsivity of these two systems at different blue
can induce axis duplication. OptoLRP6opt tran- light intensities. We observed higher light respon-
scripts were microinjected into a ventral blastomere siveness with our system as predicted (Figure S2
at the 4-cell stage. The embryos were illuminated (a)). We then compared the activities of OptoLR-
with blue light from the 32-cell stage to the mid- P6opt and CRY2-mCh-LRP6c in the Xenopus axis
blastula transition (MBT) and fixed at stage 10 for duplication assay. Unlike the successful induction
Chordin in situ hybridization (ISH). OptoLRP6opt- of axis duplication by OptoLRP6opt (Figure S2(b)
injected embryos subject to illumination showed and S2(d)), the same doses of cytosolic CRY2-
two chordin expression domains, demonstrating LRP6 system were insufficient to induce the sec-
the formation of the ectopic Spemann Organizer ondary axis under blue light stimulation (Figure S2
(Figure 3(c)). Furthermore, the secondary Orga- (c) and S2(e)), consistent with the result from the
nizer was fully functional as evidenced by the cultured cells. Thus, the OptoLRP6opt could be
appearance of a secondary body axis (Figure 3 applied to multicellular tissue or organisms with
(d), 3(e), and 3(g)) in a dose-dependent manner, higher light sensitivity.
independent of and indistinguishable from the pri-
mary axis, a phenotypic trait of Wnt overexpression. Conclusion
Whereas ventral activation of Wnt signaling leads
to axis duplication, dorsal activation of the Wnt In summary, we report the generation of a CRY2-
pathway causes posteriorization during CIBN-based optogenetic tool that activates
development. To determine if OptoLRP6opt could canonical Wnt signaling at the organismal level
induce such a phenotype, we injected mRNA into using LRP6. Our design reiterates the suitability of
both dorsal blastomeres at the four-cell stage, employing plasma membrane proximity as a
cultured embryos in light and dark, and compared switch to activate intracellular signaling pathways.
the embryonic development at the tailbud stage. This and other recent reports9,23 indicate that the
Whereas embryos in the dark developed normally, cytoplasm-to-plasma membrane translocation-

3
Figure 3. Activation of OptoLRP6opt in Xenopus laevis embryos. (a) Experimental scheme for OptoLRP6opt
activation. Fertilized eggs were uninjected or microinjected with in vitro-transcribed OptoLRP6opt mRNA and fixed at
the gastrula stage after illumination with 5 mW/cm2 blue light lasting from the 32-cell stage till the MBT (stage 8.5). (b)
Expression of xnr3 and siamois was analyzed by RT-PCR with different doses (10 pg and 50 pg) of OptoLRP6opt
mRNA upon blue light stimulation with the conditions indicated in (a). (c) The expression of Chordin was analyzed by
in situ hybridization. Light-activated embryos show two expression domains of Chordin (blue arrows), concordant with
the induction of a secondary Organizer by blue light. (d) Blue light-activated OptoLRP6opt-injected embryos
examined at the neurula stage show two body axes (arrows). (e) Dorsal and ventral views of an OptoLRP6opt-
injected, light-activated embryo in the tailbud stage, showing further development of body axes and a full complement
of ectopic anterior structures including 2 heads and 4 eyes. (f) Side views of tailbud stage uninjected embryos (upper
panel) and embryos that were dorsally-injected with OptoLRP6opt (middle and lower panel). Compared with
uninjected embryos and injected ones kept in the dark, light activation of OptoLRP6opt causes truncation of head
(lower panel). (g) Quantitative analysis of percentage of embryos with duplicated axis shown in (d). The number of
embryos for each condition (dark, light) is n = 18,17 (0 pg), 18, 20 (10 pg), 19, 21 (25 pg), 17, 14 (50 pg), and 15, 19
(100 pg). (h) Quantitative analysis of percentage of embryos with normal head and no head phenotypes upon
dorsally-injected OptoLRP6opt activated by blue light shown in (f). The number of embryos for each condition is
n = 18 (0 pg), 6 (50 pg, dark), and 17 (50 pg, light).
8
V.V. Krishnamurthy, H. Hwang, J. Fu, et al. Journal of Molecular Biology 433 (2021) 167050

based optogenetic systems can be generalized to and sodium pyruvate (Corning #10-013-CV),
signaling pathways involving membrane-bound supplemented with 10% fetal bovine serum
receptors. (Sigma-Aldrich) and 1  penicillin-streptomycin
We expect that our system will be applicable (Corning #30-002-CI). Cells were maintained in a
across different vertebrate cells and tissues, standard incubator at 37 °C with 5% CO2.
especially since the Wnt pathway and its Transfections were performed using Turbofect
components, such as LRP6, are well conserved. reagent (ThermoFisher Scientific #R0534)
Moreover, the same unmodified construct following manufacturer instructions. For Luciferase
OptoLRP6opt was potent in both human kidney assays, the dishes were transfected at 50%
cells and Xenopus embryos. Whether this tool can confluence.
be feasibly applied in the interior tissues of bigger
animal models such as mice or rat remains to be Live-cell imaging for protein localization
tested, largely due to the limited penetration depth
of the blue light. Nonetheless, technological For light-mediated membrane translocation of
innovations such as upconversion nanoparticles33 proteins, HEK293T cells were transfected with
and fiber-optic light guides34,35 compensate for the CMT-OptoLRP6 and recovered overnight before
limitation in blue light tissue penetrance. In addition, fluorescence imaging. Cells were incubated with
red-light or infra-red light-driven optogenetic tools light for 24 h before live-cell imaging with the
could be more favourable.6,36 Since our construct EGFP fluorescence. An epi-illumination inverted
is modular, we expect the mechanistic principles fluorescence microscope (Leica DMI8) equipped
and motif elements to be transferrable to such with a 10  objective and a light-emitting diode
new designs. Efforts along that direction are already illuminator (SOLA SE II 365) was used to visualize
underway. transfected cells. Green fluorescence was
detected using the EGFP filter cube (Leica,
excitation filter 472/30, dichroic mirror 495, and
Materials and methods emission filter 520/35). The exposure time for both
fluorescence channels was 200 ms. Confocal
Plasmids
imaging was performed using a Zeiss LSM 700
CRY2PHR-mCherry-LRP6c-P2A-CIBN  2- microscope. Excitation beams were focused via a
GFP-CaaX (OptoLRP6) was generated by 60  dry objective.
linearization of the 2-CIBN-containing optoRaf123
with KpnI and Kpn2I, which removes Raf1, followed
Luciferase assays
by insertion of human LRP6 intracellular domain
(NCBI NP_002327.2; residues 1394–1613) using Cells grown in the appropriate dishes were
In-Fusion cloning. CRY2PHR-LRP6c-P2A-CIBN transiently transfected with the luciferase and
 2-GFP-CaaX was generated by removing control plasmids as explained in the figures and
CRY2-mCh using NheI and Kpn2I and ligating lysed after the indicated times by incubating with
CRY2. Human TMEM198 cytosolic domain (NCBI 1  passive lysis buffer and gently shaking at
NP_001290027.1; residues 235–360) was ampli- room temperature for 30 min. The lysate was
fied from HEK293T cDNA and inserted into the centrifuged at maximum speed on a tabletop
BamHI site following the P2A peptide in the previ- centrifuge (17000  g; 10 min), and the cleared
ous construct. The ABDCDE mutant of LRP6c supernatant was used for luciferase
was generated by removing residues 1575–1613 measurements in a Promega GloMax
of LRP6c by PCR and cloned as described earlier. Luminometer (Promega Cat. #E5311) as per
OptoLRP6opt was generated by removing the manufacturer recommendations. The Dual-
2  CIBN module by inverted PCR and infusion of luciferase reporter assay system and the control
a gene-block encoding 4  CIB81 module. The pRL-TK vector for luciferase assays were
entire open reading frame for OptoLRP6 was ampli- procured from Promega (Cat. #E1980, #E2241).
fied using high-fidelity PCR and inserted into the
XbaI site of pCS2+. M50 Super 8  TopFlash was Flow cytometry and analysis
a gift from Randall Moon (Addgene plasmid #
12456; RRID: Addgene_12456). HEK293T cells grown in 12-well plates were
transfected with 500 ng of total DNA per well
consisting of 200 ng of b-catenin-mRuby2 and
Cell cultures and transfection
300 ng of OptoLRP6opt or CIBN-EGFP-CaaX
BHK21 cells were used for epifluorescence plasmids. Individual wells were subjected to
microscopy. HEK293T cells were used for continuous 750 lW/cm2 blue light (465 nm, lab-
luciferase assays and epifluorescence built LED array) 16 h after transfection from 0 to 8
microscopy. BHK21 and HEK293T cells were hours. Following the illumination, the cells were
cultured in Dulbecco’s modified Eagle’s Medium immediately trypsinized, pelleted in individual
(DMEM) containing 4.5 g/L glucose, L-glutamine, 1.5 mL tubes and fixed for 15 min in 4%
9
V.V. Krishnamurthy, H. Hwang, J. Fu, et al. Journal of Molecular Biology 433 (2021) 167050

Formaldehyde-containing PBS (ThermoFisher curation, Formal analysis, Validation. Jia Fu: Data
Scientific #FB002). The fixed cells were next curation, Formal analysis. Jing Yang:
centrifuged and resuspended in cold PBS Conceptualization, Funding acquisition, Project
containing 2.5 mM EDTA and 25 mM HEPES, administration, Supervision, Writing - review &
followed by flow cytometry (BD FACS ARIA II editing. Kai Zhang: Conceptualization, Funding
sorter) for analysis. acquisition, Project administration, Supervision,
The percentage of EGFP, mRuby2 double- Writing - original draft.
positive cells in Figure 2(d) was calculated as:
%Double-positive cells
#Double-positive cells
¼  100 Acknowledgements
#Double-positive cells þ #GFP single positive cells
Normalized b-catenin fluorescence intensity in The research reported in this publication was
Figure 2(e) was calculated as follows: First, the supported by the School of Molecular and Cellular
OptoLRP6opt (or CIBN-EGFP-CaaX) and b- Biology at UIUC, the National Institute of General
catenin-mRuby2 expression levels in the double- Medical Sciences of the National Institutes of
positive population were benchmarked to the Health and the National Institute of Environmental
respective average intensity of all single-positive Health Sciences under Award Numbers
cells in the eGFP + and mRuby + channels R01GM132438 and R01MH124827 (both to K.Z.),
(Figure S1) to obtain IGFP(t) and ImRuby(t). The b- and R35GM131810 (J.Y.).
catenin-mRuby2 intensity was then calculated as
the ratio ImRuby2(t)/IGFP(t) at each time point. Finally, Declaration of Competing Interest
the normalized b-catenin fluorescence intensity
was calculated by dividing all ratios by the value of The authors declare that they have no known
dark sample (i.e., 0 h) for OptoLRP6opt (or CIBN- competing financial interests or personal
EGFP-CaaX). relationships that could have appeared to
influence the work reported in this paper.
Xenopus embryos harvest and manipulations
Xenopus embryos were obtained and
Appendix A. Supplementary material
microinjections were performed as described.37 Supplementary data to this article can be found
When performing microinjection, embryos were online at https://doi.org/10.1016/j.jmb.2021.
placed in 0.5  Marc’s Modified Ringer’s with 3% 167050.
Ficoll. RNA was injected at the 4-cell stage using
a Narishige IM300 microinjector. The dosage of
Received 3 February 2021;
RNA for microinjection is indicated in the text or fig-
Accepted 16 April 2021;
ures. After injection, embryos were cultured in 0.2
Available online 19 May 2021
 Marc’s Modified Ringer’s and illuminated with
blue light from the 32-cell stage to the MBT. Keywords:
Embryos were harvested at various stages for optogenetics;
imaging or in situ hybridization. In situ hybridization Wnt signaling;
was performed according to the standard proto- axis duplication;
col.38 Embryos were imaged with a Leica M165 Xenopus laevis
FC dissecting scope connected to a Leica
DFC450 C digital camera using Leica Application
Suite software v4.0. The animal cap assay and
RT-PCR primers were performed as previously References
described.38 All Xenopus procedures were
approved by the University of Illinois at Urbana- 1. Chao, M.V., (2003). Neurotrophins and their receptors: a
Champaign Institutional Animal Care and Use Com- convergence point for many signalling pathways. Nat. Rev.
mittee (IACUC) under animal protocol #20125, and Neurosci., 4, 299–309.
performed in accordance with the recommenda- 2. Chang, K.Y., Woo, D., Jung, H., Lee, S., Kim, S., Won, J.,
tions of the Guide for the Care and Use of Labora- et al., (2014). Light-inducible receptor tyrosine kinases that
tory Animals of the National Institutes of Health. regulate neurotrophin signalling. Nat. Commun., 5, 4057.
3. Grusch, M., Schelch, K., Riedler, R., Reichhart, E., Differ,
C., Berger, W., et al., (2014). Spatio-temporally precise
CRediT authorship contribution activation of engineered receptor tyrosine kinases by light.
statement EMBO J., 33, 1713–1726.
4. Khamo, J.S., Krishnamurthy, V.V., Chen, Q., Diao, J.,
Vishnu V. Krishnamurthy: Conceptualization, Zhang, K., (2019). Optogenetic delineation of receptor
Data curation, Formal analysis, Methodology, tyrosine kinase subcircuits in PC12 cell differentiation. Cell
Writing - original draft. Hyojeong Hwang: Data Chem. Biol., 26, 400–410.
10
V.V. Krishnamurthy, H. Hwang, J. Fu, et al. Journal of Molecular Biology 433 (2021) 167050

5. Kim, N., Kim, J.M., Lee, M., Kim, C.Y., Chang, K.Y., Heo, 23. Krishnamurthy, V.V., Khamo, J.S., Mei, W., Turgeon, A.J.,
W.D., (2014). Spatiotemporal control of fibroblast growth Ashraf, H.M., Mondal, P., et al., (2016). Reversible
factor receptor signals by blue light. Chem. Biol., 21, 903– optogenetic control of kinase activity during differentiation
912. and embryonic development. Development., 143, 4085–
6. Leopold, A.V., Chernov, K.G., Shemetov, A.A., Verkhusha, 4094.
V.V., (2019). Neurotrophin receptor tyrosine kinases 24. Che, D.L., Duan, L., Zhang, K., Cui, B., (2015). The dual
regulated with near-infrared light. Nat. Commun., 10, 1129. characteristics of light-induced cryptochrome 2, homo-
7. Csanaky, K., Hess, M.W., Klimaschewski, L., (2019). oligomerization and heterodimerization, for optogenetic
Membrane-associated, not cytoplasmic or nuclear, manipulation in mammalian cells. ACS Synth. Biol., 4,
FGFR1 induces neuronal differentiation. Cells, 8, 243. 1124–1135.
8. Krishnamurthy, V.V., Fu, J., Oh, T.J., Khamo, J., Yang, J., 25. Shah, K.V., Chien, A.J., Yee, C., Moon, R.T., (2008).
Zhang, K., (2020). A generalizable optogenetic strategy to CTLA-4 is a direct target of Wnt/beta-catenin signaling and
regulate receptor tyrosine kinases during vertebrate is expressed in human melanoma tumors. J. Invest.
embryonic development. J. Mol. Biol., 432, 3149–3158. Dermatol., 128, 2870–2879.
9. Duan, L., Hope, J.M., Guo, S., Ong, Q., Francois, A., 26. Liang, J., Fu, Y., Cruciat, C.M., Jia, S., Wang, Y., Tong, Z.,
Kaplan, L., et al., (2018). Optical activation of TrkA et al., (2011). Transmembrane protein 198 promotes LRP6
signaling. ACS Synth. Biol., 7, 1685–1693. phosphorylation and Wnt signaling activation. Mol. Cell.
10. Masuda, T., Ishitani, T., (2017). Context-dependent Biol., 31, 2577–2590.
regulation of the beta-catenin transcriptional complex 27. Taslimi, A., Zoltowski, B., Miranda, J.G., Pathak, G.P.,
supports diverse functions of Wnt/beta-catenin signaling. Hughes, R.M., Tucker, C.L., (2016). Optimized second-
J. Biochem., 161, 9–17. generation CRY2-CIB dimerizers and photoactivatable Cre
11. Clevers, H., (2006). Wnt/beta-catenin signaling in recombinase. Nat. Chem. Biol., 12, 425–430.
development and disease. Cell, 127, 469–480. 28. Stamos, J.L., Weis, W.I., (2013). The beta-catenin
12. Hikasa, H., Sokol, S.Y., (2013). Wnt signaling in vertebrate destruction complex. Cold Spring Harb. Perspect. Biol., 5
axis specification. Cold Spring Harb. Perspect. Biol., 5 29. Plouhinec, J.L., Zakin, L., Moriyama, Y., De Robertis, E.M.,
13. W.C. Dunty, Jr., K.K. Biris, R.B. Chalamalasetty, M.M. (2013). Chordin forms a self-organizing morphogen
Taketo, M. Lewandoski, T.P. Yamaguchi, Wnt3a/beta- gradient in the extracellular space between ectoderm and
catenin signaling controls posterior body development by mesoderm in the Xenopus embryo. Proc. Natl. Acad. Sci.
coordinating mesoderm formation and segmentation, U. S. A., 110, 20372–20379.
Development 2008;135:85-94. 30. Funayama, N., Fagotto, F., McCrea, P., Gumbiner, B.M.,
14. Linker, C., Lesbros, C., Gros, J., Burrus, L.W., Rawls, A., (1995). Embryonic axis induction by the armadillo repeat
Marcelle, C., (2005). beta-Catenin-dependent Wnt domain of beta-catenin: evidence for intracellular signaling.
signalling controls the epithelial organisation of somites J. Cell Biol., 128, 959–968.
through the activation of paraxis. Development., 132, 31. Bugaj, L.J., Choksi, A.T., Mesuda, C.K., Kane, R.S.,
3895–3905. Schaffer, D.V., (2013). Optogenetic protein clustering and
15. Day, T.F., Guo, X., Garrett-Beal, L., Yang, Y., (2005). signaling activation in mammalian cells. Nat. Methods, 10,
Wnt/beta-catenin signaling in mesenchymal progenitors 249–252.
controls osteoblast and chondrocyte differentiation during 32. Duan, L., Hope, J., Ong, Q., Lou, H.Y., Kim, N., McCarthy, C.,
vertebrate skeletogenesis. Dev. Cell, 8, 739–750. et al., (2017). Understanding CRY2 interactions for optical
16. Machon, O., Backman, M., Machonova, O., Kozmik, Z., control of intracellular signaling. Nat. Commun., 8, 547.
Vacik, T., Andersen, L., et al., (2007). A dynamic gradient 33. Bergeron, D., (2018). Nanoparticle-mediated upconversion
of Wnt signaling controls initiation of neurogenesis in the of near-infrared light: a step closer to optogenetic
mammalian cortex and cellular specification in the neuromodulation in humans. Stereotact. Funct.
hippocampus. Dev. Biol., 311, 223–237. Neurosurg., 96, 270–271.
17. Birchmeier, W., (2016). Orchestrating Wnt signalling for 34. Mohanty, S.K., Lakshminarayananan, V., (2015). Optical
metabolic liver zonation. Nat. Cell Biol., 18, 463–465. techniques in optogenetics. J. Mod. Opt., 62, 949–970.
18. Burke, Z.D., Reed, K.R., Yeh, S.W., Meniel, V., Sansom, 35. Montagni, E., Resta, F., Mascaro, A.L.A., Pavone, F.S.,
O.J., Clarke, A.R., et al., (2018). Spatiotemporal regulation (2019). Optogenetics in brain research: from a strategy to
of liver development by the Wnt/beta-catenin pathway. Sci. investigate physiological function to a therapeutic tool.
Rep., 8, 2735. Photonics, 6, 92.
19. Li, Y., Lu, W., King, T.D., Liu, C.C., Bijur, G.N., Bu, G., 36. Leopold, A.V., Pletnev, S., Verkhusha, V.V., (2020).
(2010). Dkk1 stabilizes Wnt co-receptor LRP6: Bacterial phytochrome as a scaffold for engineering of
implication for Wnt ligand-induced LRP6 down-regulation. receptor tyrosine kinases controlled with near-infrared light.
PLoS ONE, 5 J. Mol. Biol., 432, 3749–3760.
20. Tamai, K., Zeng, X., Liu, C., Zhang, X., Harada, Y., Chang, 37. Sive, H.L., Grainger, R.M., Harland, R.M., (2000). Early
Z., et al., (2004). A mechanism for Wnt coreceptor Development of Xenopus laevis: A Laboratory Manual.
activation. Mol. Cell, 13, 149–156. Cold Spring Harbor Laboratory Press.
21. MacDonald, B.T., He, X., (2012). Frizzled and LRP5/6 38. Rorick, A.M., Mei, W.Y., Liette, N.L., Phiel, C., El-Hodiri, H.
receptors for Wnt/beta-catenin signaling. Cold Spring Harb. M., Yang, J., (2007). PP2A : B56 epsilon is required for eye
Perspect. Biol., 4 induction and eye field separation. Dev. Biol., 302, 477–
22. Metcalfe, C., Mendoza-Topaz, C., Mieszczanek, J., Bienz, 493.
M., (2010). Stability elements in the LRP6 cytoplasmic tail 39. Wang, Q., Fan, H., Li, F., Skeeters, S., Krishnamurthy, V.,
confer efficient signalling upon DIX-dependent Song, Y., Zhang, K., (2020). Optical control of ERK and
polymerization. J. Cell Sci., 123, 1588–1599. AKT signaling promotes axon regeneration and functional

11
V.V. Krishnamurthy, H. Hwang, J. Fu, et al. Journal of Molecular Biology 433 (2021) 167050

recovery of PNS and CNS in Drosophila. eLife, 9, e57395. Range Optogenetic Control of Axon Guidance Overcomes
https://doi.org/10.7554/eLife.57395. Developmental Boundaries and Defects. Developmental
40. Harris, J., Wang, A., Boulanger-Weill, J., Santoriello, C., Cell, 53 (5), 577–588. https://doi.org/10.1016/j.
Foianini, S., Lichtman, J., Zon, L., Arlotta, P., (2020). Long- devcel.2020.05.009.

12

You might also like