Processing
Synthetic signaling circuits promise to revolutionize cell therapies by enabling engineered cells to sense, compute, and respond with high precision. Yet, achieving orthogonal and reversible control remains a major challenge. Existing systems often depend on irreversible safety switches (e.g. iCasp9) or provide only slow, incomplete reversibility through antigen-gated or pharmacologic inhibition, limiting dynamic regulation. To overcome these constraints, we developed PHOENICS: a synthetic phosphorylation circuit that enables rapid and reversible signal switching at the post-translational level. In PHOENICS, a synthetic kinase-phosphatase pair dynamically controls the phosphorylation state of a substrate, which determines binding to a phosphobinder (SH2) and thereby regulates downstream responses. We demonstrated that substrate phosphorylation is specific and orthogonal, occurring exclusively in the presence of the synthetic kinase. The SH2-substrate interaction proved fully reversible and concentration-dependent as increasing kinase levels strengthened complex formation, while higher phosphatase expression progressively diminished it. These core processing units lay the foundation for programmable and dynamically regulated signaling in future cell therapies.
Aim
Engineering complex, interaction-dependent fusion proteins presents a significant challenge. Adopting a bottom-up strategy, we characterized each functional core component in a simplified environment to enable rapid Design-Build-Test-Learn cycles, facilitating thorough part optimization and improved downstream engineering. Because effective (de-)phosphorylation and subsequent (de-)dimerization underpin our system's processing layer, understanding their kinetics and regulatory behavior is critical to achieve the desired circuit response. Prior to integration, we developed assays providing a modular framework for validating individual circuit components. After first validating phosphorylation with western blot analysis, we investigated phosphorylation-dependent protein-protein interactions using splitFAST, which provides dimerization-specific fluorescence at the single-cell level by flow cytometry.
Cellular signaling networks coordinate how cells perceive and respond to their environment through cascades of post-translational modifications and regulated protein interactions. Among these, phosphorylation and dephosphorylation are central regulators of protein activity, localization, and molecular interactions. Such signaling mechanisms control key processes including cell proliferation (MAPK/ERK), metabolism (PI3K-Akt), immune activation (JAK-STAT), and cell cycle regulation (CDK pathways) (Cohen, 2000). Within these networks, modular adaptor domains translate phosphorylation states into specific molecular assemblies and downstream outputs (Pawson & Scott, 2005).
Building on these natural systems, synthetic phosphorylation circuits have been developed to reconstruct or reprogram signal transduction using defined molecular components. By coupling engineered kinases and phosphatases to modular substrates and interaction domains, these systems allow precise and tunable control of signaling dynamics while remaining orthogonal to endogenous pathways (Lim & Pawson, 2010).
Based on previous work by Yang et al. (2025), who established a modular framework for synthetic phosphorylation networks, we implemented a synthetic phosphorylation circuit composed of four core processing components: an engineered ABL kinase serving as the phosphorylation module, a PTPN1 phosphatase providing reversible control, a CD3ζ substrate carrying immunoreceptor tyrosine-based activation motifs (ITAMs), and an SH2 binder that recognizes phosphorylated sites to couple signal state to downstream function.
The ABL kinase was chosen for its well-characterized activity toward tyrosine-rich motifs, including
ITAM
sequences (Moresco & Koleske, 2003). While native ABL is not inherently orthogonal to cellular
substrates,
we employed a truncated version (residues 218-511; UniProt-ID: P00519) lacking regulatory and
localization
domains.
Kinase-substrate specificity is achieved through engineered coiled-coil recruitment using
bZipEE and bZipRR motifs to spatially confine phosphorylation to the synthetic substrate (Acharya
et
al., 2002). The resulting ABL-bZipEE fusion constitutes the SynKinase module.
As a
counteracting
enzyme, we used a truncated variant of PTPN1 phosphatase (residues 3-321; UniProt-ID: P18031)
comprising only the
catalytic domain. Removal of regulatory and targeting regions eliminates ER localization and reduces
interactions with endogenous substrates (Tonks, 2006). The enzyme was likewise fused to bZipEE,
forming
the SynPhosphatase, which enables reversible dephosphorylation within the synthetic network.
The
synthetic substrate (SynSubstrate) consists of three tandem repeats of the CD3ζ C-terminal ITAM motif
(residues 131-164; UniProt-ID: P20963). In natural T-cell signaling, these motifs mediate receptor
activation through site-specific phosphorylation and SH2-domain binding (Pawson,
2004). In our design,
repeating the motif three times enhances the dynamic range and sensitivity of
phosphorylation-dependent responses. To
detect
and translate substrate phosphorylation, we used two SH2 domains from ZAP-70 (residues 2-259;
UnirProt-ID:
P43403), which specifically recognize phosphorylated CD3ζ ITAMs (Hatada
et al., 1995). This
module, termed the SynPhosphobinder, acts as a phosphorylation-dependent interaction unit that
couples
the
modified substrate to downstream synthetic effectors while remaining insulated from endogenous
signaling.
Phosphorylation of CD3ζ
Validating functional phosphorylation of our SynSubstrate by our SynKinase is essential to couple accuracy of dependent read-out methods. Western blotting against the phosphorylated residue in the absence of the SynKinase also gives an insight into background noise facilitated by endogenous enzyme activity. We verified that coiled coil mediated proximity between our SynKinase, a truncated ABL kinase, and the SynSubstrate leads to efficient phosphorylation.
To verify the orthogonality of SynKinase-mediated phosphorylation, we performed Western blot analysis using anti-pY antibodies specific for phosphorylated tyrosine residues within the ITAM motifs. Substrate presence in each sample was confirmed by Myc-tag fusion and subsequent anti-Myc staining (Fig. 1).
Figure 1: Schematic of SynKinase and SynSubstrate constructs used for phosphorylation validation. The cytosolic SynKinase consists of a truncated ABL kinase fused to a bZipEE dimerization domain. The SynSubstrate contains three tandem CD3ζ ITAM motifs (3x CD3ζ) fused to the complementary bZipRR domain and a C-terminal triple Myc tag for detection.
Western blot analysis confirmed that SynSubstrate phosphorylation occurs exclusively in the presence of SynKinase. The anti-pY antibody detected no phosphorylation signal in the SynSubstrate-only condition (Fig. 2, lane 1, top panel), whereas a strong signal appeared upon SynSubstrate + SynKinase cotransfection (lane 2, top panel). Myc-tag staining verified substrate expression in both conditions, confirming that the absence of phosphorylation in the control was not due to missing substrate. The α-Myc signal cannot be quantitatively compared between samples because residual anti-pY binding persisted after antibody reuse without stripping. Nevertheless, equal SynSubstrate plasmid amounts were transfected in all conditions, supporting the conclusion that SynKinase specifically and orthogonally phosphorylates SynSubstrate.
Figure 2: Western blot verification of SynSubstrate phosphorylation by SynKinase. HEK293T cells were transfected with equal amounts of SynSubstrate either alone or together with SynKinase. Phosphorylation was detected using a phospho-tyrosine-specific (anti-pY) antibody. A phosphorylation signal was observed exclusively in the presence of SynKinase, confirming kinase-dependent modification of the SynSubstrate.
Dimerization
Efficient dimerization between phosphorylated SynSub and SH2, coupled with their dissociation upon dephosphorylation, is essential for mediating phosphorylation-dependent signaling output. This phosphorylation-dependent dimerization was confirmed using the split fluorescent protein system SplitFAST. SynKinase titration demonstrated a dose-dependent increase of dimerization corresponding to phosphorylation levels, which was effectively attenuated in a dose-dependent manner by SynPhosphatase.
To develop and test the basic circuit functionality, we established a split-fluorescent protein system enabling us to directly observe dimerization between CD3ζ and SH2. This strategy allowed us to investigate interaction dynamics before further engineering a dimerization dependent output. SplitFAST is a split version of RspA-FAST, a protein that fluorescences in the presence of the small molecule HMBR (Rakotoarison et al., 2024)(Fig. 3). Exhibiting low self-complementation and a high dynamic range it allows for effective studying of protein protein interactions.
Figure 3: Schematic of the CD3ζ-SH2 dimerization dependent splitFAST reporter read-out Upon coiled-coil interaction between RspA(N)-CD3ζ-bZipRR and the SynKinase, tyrosine residues within the CD3ζ are phosphorylated. This increases affinity to the SH2, allowing for phosphorylation-specific dimerization. Dimerization between RspA(N)-CD3ζ-bZipRR and RspA(C)-SH2 brings the splitFAST halves into proximity giving a fluorescent readout upon HMBR addition.
Figure 4: Validating phosphorylation dependent dimerization using splitFAST. HEK293T cells were transfected 24 hours after seeding. Y-axis description corresponds to the ng amounts of SynKinase, 100 ng of both RspA(N)-CD3𝜁-bZipRR and RspA(C)-SH2 were co-transfected. Dimerization dependent SplitFAST fluorescence was quantified by flow cytometry 48 hours after transfection, employing excitation lasers at 488 nm and 561 nm. Emission was recorded at 525 nm for splitFAST (Ligand: TFLime) and at 610 nm for constitutively expressed mCherry, serving as a transfection control. Data is depicted as the geometric mean of fluorescence +/- geometric SEM with ~30000 single cells
Reversibility of phosphorylation-dependent dimerization is crucial for dynamic signal processing and logic gate computation. To test this, we assessed whether SynPhosphatase could access and dephosphorylate CD3ζ, thereby disrupting CD3ζ-SH2 binding. Increasing SynPhosphatase expression progressively reduced splitFAST fluorescence, indicating dissociation (Fig. 5). The dose-dependent decline in signal, reaching background levels at high SynPhosphatase concentrations, confirms effective dephosphorylation and circuit reversibility.
Figure 5: Validating dephosphorylation dependent dissociation using splitFAST. HEK293T cells were transfected 24 hours after seeding. Y-axis description corresponds to the ng amounts of SynPhosphatase, 100 ng of RspA(N)-CD3𝜁-bZipRR, RspA(C)-SH2 and SynKinase were co-transfected. Dimerization dependent SplitFAST fluorescence was quantified by flow cytometry 48 h post transfection, employing excitation lasers at 488 nm and 561 nm. Emission was recorded at 525 nm for splitFAST (Ligand: TFLime) and at 610 nm for constitutively expressed mCherry, serving as a transfection control. Data is depicted as the geometric mean of fluorescence +/- geometric SEM with ~30000 single cells
Discussion
Using a bottom-up approach, we systematically characterized the functionality and
interaction dynamics of the core PHOENICS components before advancing to higher circuit integration.
Computational modeling with SPARC complemented these experiments by providing quantitative insights into the
parameter space underlying phosphorylation-dependent behavior. By establishing assays to directly validate
phosphorylation and to monitor dimerization of CD3ζ and SH2, we created a robust framework for iterative
circuit optimization.
Western blot analysis confirmed functional phosphorylation and dephosphorylation catalyzed by the SynKinase
and SynPhosphatase. SplitFAST assays further demonstrated that SynKinase activity drives CD3ζ-SH2
association, whereas SynPhosphatase expression induces dissociation, confirming the reversibility of the
interaction.
Moving forward, tuning the interaction dynamics between these components will enable fine control over
circuit performance and response kinetics. Testing coiled-coil variants of different affinities will help
determine how binding strength influences temporal behavior and the potential for switch-like responses. In
parallel, modeling these variants will guide experimental design by prioritizing configurations with desired
kinetic or ultrasensitive properties. Additional exploration of alternative tyrosine kinases, phosphatases,
and ITAM motifs may further expand the circuit's dynamic range and functional versatility.
Responding
In cell therapies, precise temporal control of activation and deactivation is critical. Therapeutic
cells must rapidly respond to disease cues and deactivate once the signal subsides. Building on our core
processing modules, we coupled phosphorylation-dependent signaling to two effector systems, enabling
engineered cells to translate post-translational states into reversible functional responses.
First, we established a transcriptional output layer by linking substrate phosphorylation directly to
gene expression. Transcriptional activators and DNA-binding domains were fused to SH2 and CD3ζ, allowing
phosphorylation-dependent dimerization to initiate transcription.
Second, to achieve faster and dynamically reversible control, we implemented a phosphorylation-inducible
protease system in which phosphorylation activates a protease that releases pre-synthesized proteins,
enabling rapid and tunable secretion.
Together, these modules demonstrate how PHOENICS connects post-translational signaling to gene
expression and secretion, forming a reversible effector layer for precise and adaptive control of
therapeutic cell activity.
Aim
After validating kinase-dependent phosphorylation and dimerization of CD3ζ and SH2, the next step was to
couple the phosphorylation signal to a meaningful effector function. For this, we planned to establish two
parallel output systems, each with distinct use cases.
In our expression-based system, the SynSubstrate was fused to a Gal4 DNA-Binding Domain (GAL4DBD), while
the
SH2-domain was linked to a VP64 transcriptional transactivator. Once the substrate is phosphorylated, it
is
engaged by SH2, which results in transcription factor reconstitution and subsequent gene
expression.
However, in a therapeutic
context, precision demands speed, because cellular composition can shift from malignant to
healthy within the same tissue. To create an effector platform that matches the rapid dynamics of our
phosphorylation circuit, we implemented the RELEASE system (Vlahos et al.,
2022). For this, we engineered a
phosphorylation-inducible Tobacco Etch Virus protease (PhosphoTEV) to control protein secretion on a
post-translational level.
The split Gal4DBD-VP64 Transcription System
The Gal4 DNA-binding domain (Gal4DBD) originates from the yeast transcription factor Gal4, which
regulates
galactose metabolism genes by binding specific Upstream Activating Sequences (UAS). Its zinc cluster
motif
coordinates two zinc ions and stabilizes a fold that enables precise recognition of palindromic
CGG(N11)CCG repeats. Two Gal4DBD monomers dimerize on DNA to achieve strong and specific
promoter
binding (Hellauer et al., 1996).
The VP64 activation domain is a
synthetic
construct derived from the herpes simplex virus protein VP16. It consists of four random repeats of
the minimal
VP16 activation domain (Beerli et al., 1998) and enhances
transcription by
recruiting mediator proteins and histone acetyltransferases, which promote RNA polymerase II assembly
(Mittler et al., 2003).
In synthetic systems, Gal4DBD and VP64
are often used
together to form a modular
transcriptional activator, in which Gal4DBD provides DNA-binding specificity while VP64 drives
promoter activation
(Hu et et al., 2014).
Protein Secretion in Mammalian Cells
The secretory pathway processes over 25% of mammalian proteins. Nascent proteins are translated in the rough endoplasmic reticulum (ER) and enter the ER lumen via co-translational translocation (Benavides-López et al., 2025). Inside the ER, proteins fold with help from molecular chaperones. Misfolded proteins undergo ER-associated degradation (ERAD), while correctly folded proteins proceed forward (Benavides-López et al., 2025). COPII-coated vesicles mediate anterograde transport from ER to the Golgi complex while COPI-coated vesicles handle retrograde transport from Golgi back to the ER, recycling essential components (Benavides-López et al., 2025).
ER Retention and Protease-Controlled Secretion
In mammalian cells, proteins destined to remain in the endoplasmic reticulum (ER) are retained
through C-terminal
dilysine motifs, which are recognized by the COPI vesicle machinery and coatomer complex responsible
for
retrograde transport from the Golgi back to the ER (Barlowe & Helenius, 2016;
Hoffman et
al., 2003).
The Retained Endoplasmic Cleavable Secretion (RELEASE) system exploits
this mechanism
by tethering a protein of interest (POI) to the ER membrane with a dilysine or diarginine motif
downstream of a
TEV cleavage site (Fig. 6). Under normal conditions, the POI is retained in the ER
through
COPI-mediated
transport. Upon TEV protease cleavage, the retention signal is removed and the POI enters the
secretion pathway. A
furin cleavage site between the transmembrane domain and POI ensures proper processing in the
trans-Golgi for
final secretion (Vlahos et al., 2022).
Figure 6: Visualization of the RELEASE system. The POI (yellow circle) is expressed and tethered to the ER membrane with a transmembrane domain (orange). As long as the TEV protease (pink pacman) does not cleave off the ER retention signal at its cleavage site (pink circle), it is retained in the ER. Upon cleavage, it undergoes the secretory pathway through the Golgi complex and is secreted into the extracellular space.
Expression
Using the split composite transcription factor of Gal4DBD and VP64, we established a robust and sensitive output that translates phosphorylation signal into protein expression. The system is sensitive towards (de-)phosphorylation over a wide range of both kinase and phosphatase amounts.
To build on the previously validated phosphorylation and dimerization events, a transcriptional effector circuit was implemented that translates enzymatic activity into a measurable output. Therefore, Gal4DBD is fused to the synthetic substrate, while VP64 is attached to the SH2 domain. SynKinase, recruited via complementary coiled-coils (bZipEE/bZipRR), phosphorylates CD3ζ and enables SH2-VP64 binding. This interaction reconstitutes a functional Gal4DBD-VP64 transcription factor that activates NanoLuciferase expression. Conversely, SynPhosphatase dephosphorylates CD3ζ, preventing SH2-VP64 recruitment and thereby reporter activation (Fig. 7).
Figure 7: Schematic representation of the phosphorylation circuit and Nanoluciferase reporter assay. SynKinase (ABL-bZipEE) phosphorylates the SynSubstrate (Gal4DBD-CD3ζ-bZipRR) in a proximity-dependent manner, while SynPhosphatase (PTPN1-bZipEE) counteracts this modification through dephosphorylation. Phosphorylated SynSubstrate recruits the SH2-VP64 binder, which, together with Gal4DBD, activates transcription from a minimal promoter and induces NanoLuciferase expression. For clarity, CD3ζ (three ITAM repeats) and SH2 (two domains) are illustrated as single units.
A kinase titration revealed a dose-dependent increase in reporter activity that plateaued above 40 ng ABL-bZipEE, with no significant differences between 40, 50, and 60 ng, indicating saturation of substrate phosphorylation. Transcriptional activation was therefore limited by SynSubstrate availability rather than kinase abundance (Fig. 8).
Figure 8: Dose-response of SynKinase. HEK293T cells were transfected with SynSubstrate (5 ng) and SH2-VP64 (25 ng) in a 1:5 ratio together with increasing amounts of cytosolic SynKinase. The Dual-Luciferase assay was performed 48 hours post transfection, with NanoLuciferase normalized to Firefly. Data are presented as the mean ± SEM with n = 3 technical replicates. Statistical significance was determined with ordinary one-way ANOVA and Tukey's multiple comparisons test (single pooled variance).
Figure 9: Titration of SynPhosphatase. HEK293T cells were transfected with 20 ng SynKinase, 25 ng SH2-VP64, and 5 ng Gal4DBD-CD3ζ-bZipRR (SynSubstrate), while SynPhosphatase (PTPN1-bZipEE) was titrated from 1 to 20 ng as indicated. Reporter activity was measured 48 h post transfection using a Dual Luciferase assay, with NanoLuciferase normalized to constitutively expressed Firefly luciferase. Data are shown as mean ± SEM (n = 3 technical replicates). Statistical significance was determined with ordinary one-way ANOVA with Tukey's multiple comparison test (single pooled variance).
Together, the kinase and phosphatase titration experiments demonstrate that the phosphorylation circuit enables precise, reversible, and tunable control of transcriptional activation. Reporter expression increased proportionally with SynKinase input until phosphorylation sites on the SynSubstrate became saturated, while rising SynPhosphatase levels progressively reduced the signal back to baseline. These findings confirm that circuit output directly reflects dynamic balance between kinase- and phosphatase-mediated modification states, establishing a controllable molecular switch that links enzymatic activity to gene expression.
Secretion
To allow the implementation of the RELEASE system into our phosphorylation circuits, we engineered and characterized two different designs of a phosphorylation-inducible TEV protease. This module allows for rapid protein secretion without the delay of gene expression.
To engineer a phosphorylation-inducible TEV protease, we developed two different designs. In the first one we used a split TEV system where the N-terminal half was linked to an SH2-domain, while the C-terminal half was linked to a SynSubstrate and bZipRR (Fig. 10A). In a second iteration, the two constructs were joined into one by linking them through a semi-rigid linker between the SH2 and bZipRR domains (Fig. 10B). In both versions, the association of the bZip coiled-coils causes phosphorylation of the SynSubstrate so that the SH2 domain binds to it, resulting in the reconstitution of the TEV protease. The activated protease in turn causes the secretion of a luminescent NanoLuc luciferase (NanoLuc) reporter, which could be measured in the supernatant.
Figure 10: Different PhosphoTEV designs. Reconstitution of split TEV halves depends on the phosphorylation-inducible binding of SH2 to CD3ζ. (A) In the split PhosphoTEV design, the interacting domains are located on different constructs. (B) In the linked PhosphoTEV, SH2 and bZipRR are connected through a variable hinge.
Figure 11: SynKinase activates PhosphoTEV and induces secretion. HEK293T cells were co-transfected with split and linked PhosphoTEV variants without (orange) and with (purple) SynKinase, and the NanLuc-RELEASE plasmid. Secreted NanoLuc in supernatant was measured 48h post-transfection. Positive and negative controls (yellow) with a full TEV and no TEV are shown for comparison. Data is depicted as the mean +/- SEM with n = 3 technical replicates. Statistical significance was calculated with an ordinary One-way ANOVA with Tukey's multiple comparisons test. **P<0.01, ****P<0.0001; ns, not significant.
A key advantage of phosphorylation-based signaling circuits lies in the reversibility dependent on kinase and phosphatase activity. To assess whether this behaviour applies to the RELEASE system, different amounts of PhosphoTEV were co-transfected with SynKinase alone or in combination with SynPhosphatase. After 48 hours, NanoLuc luminescence in the supernatant was measured (Fig. 12). As shown before, the background of PhosphoTEV without kinase or phosphatase did not differ significantly from the RELEASE background without any TEV. With only SynKinase, there was a 2.6-fold induction in the luminescence signal. This induction could be quenched significantly with the addition of SynPhosphatase, reducing the increase by about 50 %, showing that the activation of PhosphoTEV is reversible and sensitive to SynPhosphatase. With the successful (de-)activation of PhosphoTEV using SynKinase and -Phosphatase, we could establish the fully post-translational RELEASE response module, able to be integrated into more sophisticated circuits.
Figure 12: SynPhosphatase deactivates PhosphoTEV and reduces secretion. HEK293T cells were co-transfected with the linked PhosphoTEV and either no enzyme (yellow), only SynKinase (orange), or in combination with SynPhosphatase (purple), and the NanoLuc-RELEASE plasmid. Secreted NanoLuc in supernatant was measured 48h post-transfection. Data is depicted as the mean +/- SEM with n = 3 technical replicates. Statistical significance was calculated with an ordinary Two-way ANOVA with Tukey's multiple comparison's test. ***P<0.001, ****P<0.0001; Negative and positive controls were excluded from the test and only shown for reference.
Discussion
These phosphorylation‑responsive systems show that intracellular kinase activity can be routed through
SH2-CD3ζ recognition to drive either a transcriptional or a proteolytic output, yielding a modular
signal-response architecture with dynamic, tunable control across molecular layers. Kinase input increases
activation until substrate or promoter occupancy saturates, while phosphatase input rapidly quenches
activity by removing the phosphorylation off ITAMs required for SH2 engagement, underscoring phosphorylation
as a universal
signaling language that integrates diverse upstream cues with programmable downstream functions.
The Gal4‑VP64 output module amplifies phosphorylation signals into gene expression by reconstituting a
composite activator only upon SH2 binding to phosphorylated CD3ζ fused to Gal4DBD at UAS sites. The observed
large dynamic range, validated by kinase and phosphatase titrations, was strengthened by stoichiometric and
localization
optimizations that increase effective
nuclear molarity. The natural rates of transcription and translation in biological circuits tend to favor
stable,
long-lasting responses rather than rapid changes. Future improvements - such as adding destabilization
domains,
degrons that target proteins for degradation, or RNA-based controls like responsive 3′ UTR decay elements
and
ribozyme switches - could speed up signal repression and enhance the temporal precision of circuit
responses,
while maintaining robust output levels.
The PhosphoTEV-RELEASE output module provides immediate, post‑translational actuation by directly coupling
phosphorylation state to ER exit and secretion. ER retention ensures low basal output by preventing
premature
reporter release. Upon phosphorylation-dependent SH2 engagement, TEV protease activity is reconstituted,
resulting in removal of the ER retention tag, permitting anterograde trafficking and thus rapid reporter
secretion.
The linked PhosphoTEV architecture improves the on‑state activtiy,
likely through higher effective molarity and favorable geometry, while ER retention buffers off‑state
leakage. The remaining background is consistent with spontaneous fragment association or partial retention
escape. Future efforts will focus on further reducing this leakage, on one hand through the optimization of
the RELEASE construct by exchanging the transmembrane domain to reduce retention escape and increase
expression levels. On the other hand, the PhosphoTEV construct will be further improved through linker
optimization, which may greatly impact background, as shown in the context of allosterically controlled
proteins.
Treating gene expression and secretion as two swappable output modules enables circuits to be adapted to
application demands: the expression arm delivers strong, programmable amplification with slower onset, while
the secretion arm delivers rapid, transient output with minimal background leakage. Layering these outputs
can
yield hierarchical, staged dynamics that better approximate natural cascades - fast post‑translational
responses leading or gating slower transcriptional programs - while maintaining orthogonality and reversible
control suitable for therapeutic contexts. Coupling both modules to ligand‑responsive kinases further
supports pharmacological control, reinforcing the practicality of phosphorylation‑gated logic in living
cells.
Sensing
Precise recognition of extracellular cues is crucial for the safe and adaptable development of cell therapies. Existing synthetic receptors are often limited by slow, transcription-based signaling and poor modularity. To overcome these constraints, we developed a phosphorylation-based sensing layer that links ligand detection to rapid, post-translational computation within the PHOENICS framework. This system integrates synthetic GPCR and Modular Extracellular Sensor Architecture (MESA) receptors in a plug-and-play fashion to translate ligand binding into phosphorylation or dephosphorylation of the synthetic substrate CD3ζ, thereby regulating SH2-mediated transcriptional reconstitution. Both receptor scaffolds allow for complex combinatorial signal processing, facilitating both activation and repression, and were shown to functionally sense tumor associated proteins including VEGF and TNF-α. The GPCR design enables particularly fast, binary switch-like responses with minimal background and a large dynamic range while MESA offers enhanced modularity coupled with high output strength. Using our in silico platform SPARC, we designed and validated de novo protein binders against cancer-relevant targets such as GDF-15, thereby extending the plug-and-play retargeting of both receptor classes. The combination of these features establishes a rapid, reversible, and modular input layer that bridges extracellular sensing with post-translational computation - providing a foundation for personalized cancer therapies capable of responding precisely and dynamically to complex tumor environments.
Aim
To establish sensing capabilities with our intracellular phosphorylation logic, we wanted to engineer two
modular receptor platforms with distinct use cases. In order to develop a receptor scaffold able to
respond with all-or-nothing-behaviour, we planned to engineer the synthetic κ-opioid receptor DREADD
(KORD) (Vardy et al., 2015) and additionally adapt the PAGER platform
(Kalogriopoulos
et al., 2025). By
replacing the intracellular signaling machinery with our SynPhosphatase and -Kinase, receptor
activation
can facilitate direct (de-)phosphorylation of CD3𝜁, our SynSubstrate.
In parallel, we wanted to develop MESA receptors similarly fused to our SynKinase and SynPhosphatase. MESA
offers the distinct benefit of a stronger total activation and quenching, albeit with the tradeoff of a
diminished dynamic range.
To expand accessible tumor targets, we established a CF2H-based validation pipeline for computationally
designed de novo binders, demonstrating this approach with GDF-15-targeting constructs. Together, these
experiments aimed to deliver a modular, bidirectional sensing layer capable of processing complex tumor
microenvironment cues.
Cells sense environmental cues such as small molecules, proteins, and surface signals through diverse
receptors, including G protein-coupled receptors, receptor tyrosine kinases, and ion channels. Many of
these receptors act on pathways involving phosphorylation, a fast, reversible and tunable
post-translational process. Kinases and phosphatases rapidly and reversibly set the
phosphorylation state of chosen substrates, turning extracellular cues into graded intracellular
responses on second-to-minute timescales (Farahani et al., 2023; Zhang
et al.,
2005). This
architectural logic - co-recruitment of catalytic domains to defined substrates, tight spatial
control, and rapid off-kinetics - makes phosphorylation an attractive approach for
engineering synthetic sense-and-respond systems that are both fast and precise.
Current synthetic receptors such as CARs and SynNotch receptors established that extracellular signals
can be coupled to customized transcriptional responses, but their activation physics (clustering and
force for CARs; proteolytic release for SynNotch) constrain antigen scope and largely commit
computation to the gene-expression layer, with inherent delays and potential basal leakiness.
Recent work has integrated phosphorylation control with the Modular Extracellular Sensing Architecture
(MESA) to engineer modular, orthogonal phosphorylation cycles in human cells thus integrating upstream
receptor activation with downstream gene expression. MESA addressed a central limitation by
introducing a self-contained, orthogonal receptor that dimerizes upon ligand and
triggers engineered outputs through modular intracellular cassettes; importantly, MESA's
ectodomains
and transmembrane/linker elements can be swapped or tuned to retarget specificity and performance
(Daringer et al., 2014). Improving the original MESA architecture
by
using receptor-coupled
kinases
demonstrated that phospho-circuits can be predictively tuned and interconnected while remaining
largely insulated from endogenous phosphorylation networks (Yang
et al., 2025). Yet, even in
these
systems, inhibitory control typically resides within the internal cycle rather than at the
receptor
itself.
Synthetic GPCRs have recently emerged as alternative modular scaffolds for
programmable sensing. The PAGER framework fuses a nanobody with an auto-inhibitory peptide to
the
𝜅-Opioid Receptor DREADD so that antigen binding relieves inhibition and permits small molecule
activation, enabling modular antigen-gated control over transgene expression, endogenous
G-protein
signaling, or real-time readouts (Kalogriopoulos et al., 2025).
PAGER highlights two
properties
essential for therapeutic translation - retargetability via simple binder
exchange
and drug addressability - yet still primarily decodes through transcriptional
modules
or native GPCR pathways. A phosphorylation-native decoder placed immediately downstream of
such GPCRs
would preserve modular antigen gating while delivering the speed and reversibility of
protein-protein
signaling.
Despite these advances, a specific capability has been missing at the sensing layer to allow the
implementation of advanced receptor construct into cellular therapies: a bidirectional,
ligand-programmable phosphorylation interface that is both (i) reversible
- providing receptor-level ON control via a kinase and OFF control via a phosphatase on a
post-translational level - and (ii) plug-and-play retargetable to the soluble
cues
that characterize the tumor microenvironment (TME). Prior orthogonal phospho-systems
established
reversible cycles and model-guided tuning, and they showed coupling to receptors, but to our
knowledge
they did not provide a general phosphatase-fused, ligand-gated receptor to
facilitate
extracellularly mediated inhibition on a post-translational level. Filling this gap would
enable fast
N-IMPLY safety logic and drug-tunable dormancy directly at the receptor-circuit
boundary-capabilities
that are particularly valuable for oncological applications, where multi-ligand contexts and
patient-specific profiles demand flexible decoding and stringent OFF control (Norton et al.,
2015;
Yang et al., 2025).
GPCR
As one of our modular sensing systems, we engineered multiple synthetic GPCR scaffolds offering different levels of sensing capabilities and modularity in HEK293T cells. This architecture allows for modular sensing of varied peptide and small molecule ligands and is able to enact a binary, switch-like response.
To effectively drive gene expression in an inducible, ligand-specific manner, a receptor should be
host-orthogonal, exhibit low background, and be readily inducible. In this context, we chose the
𝜅-Opioid receptor DREADD (KORD), an engineered GPCR inducible by the small-molecule Salvinorin B
(SalB), as a receptor scaffold for our phosphorylation circuit (Fig. 13). By fusing
its intracellular C-terminal tail with our SynKinase, a truncated ABL kinase able to induce specific,
proximity-based phosphorylation, we created a novel receptor construct: KORD-ABL. Together with an
engineered version of 𝛽-Arrestin2, an endogenous regulator of GPCR activity, it builds a system that
is able to specifically recruit and phosphorylate our SynSubstrate upon receptor activation.
SalB binding triggers a conformational change that enables GRK-mediated phosphorylation of residues in
the receptor's C-terminal tail (Fig. 13). This recruits β-Arrestin2-bZipEE that
can bind
Gal4DBD-Cd3ζ-bZipRR via a coiled-coil interaction. The resulting positioning of the CD3ζ near
the ABL kinase fused to the GPCR leads to its phosphorylation. The phosphorylated Gal4DBD-CD3ζ-bZipRR
subsequently dimerizes with SH2-VP64, forming a transcriptional activator that binds the 5xUAS motif
and
drives transgene expression.
Figure 13: Schematic overview of KORD-ABL coupled to the phosphorylation circuit. In the absence of SalB, 𝛽-Arrestin2-bZipEE cannot bind KORD-ABL. Receptor activation through SalB presence leads to a conformational change, allowing GRK binding and subsequent phosphorylation of the C-terminal tail of KORD. This allows for the binding of 𝛽-Arrestin2-bZipEE. Connection to the intracellular PHOENICS circuit is facilitated by the fused bZipEE domain on 𝛽-Arrestin2 and its spatial organization in the proximity of the ABL kinase upon KORD binding. Bound 𝛽-Arrestin2-bZipEE recruits Gal4DBD-CD3𝜁-bZipRR, which is subsequently phosphorylated by ABL. Dissociation from the KORD due to the transient coiled-coil interactions allows dimerization between the phosphorylated CD3𝜁 and SH2, reconstituting the split transcription factor Gal4DBD-VP64. Shuttling to the nucleus then facilitates transgene expression under control of the 5xUAS-minCMV promoter.
To evaluate the synthetic KORD-ABL receptor, we investigated its ability to drive NanoLuciferase expression upon induction by its small molecule ligand SalB. We co-transfected HEK293T cells with KORD-ABL, 𝛽-Arrestin2-bZipEE, as well as our phosphorylation circuit and constitutively expressed Firefly luciferase to normalize to cell viability. Cells were induced with 500 nM SalB 24 hours after transfection. Nano- and Firefly Luciferase activity was determined 48 hours after transfection using the Nano-Glo Dual-Luciferase Reporter Assay System. As controls, we first used the entire phosphorylation circuit without KORD-ABL, as well as separately without 𝛽-Arrestin2-bZipEE. In three biological replicates, induction of KORD-ABL significantly increases NanoLuciferase expression by 28-fold compared to its uninduced condition (p < 0.0001), exhibiting a switch-like behavior to drive gene expression (Fig. 14). Moreover, KORD-ABL does not exhibit significant leakiness or background noise compared to control conditions without KORD-ABL or 𝛽-Arrestin2-bZipEE (p > 0.99). This demonstrates the functionality of a novel receptor architecture that can drive transgene expression in a switch-like manner, without exhibiting significant background or interference in native signaling pathways. This data was further corroborated by measuring mCherry expression upon KORD-ABL induction on a single-cell basis using flow cytometry.
Figure 14: KORD-ABL represents a novel receptor platform that senses and responds to extracellular cues in a switch-like manner. HEK293T cells were transfected 24 hours after seeding. NanoLuc and Firefly Luciferase expression was measured 48 hours after transfection using the Dual Luciferase assay. Induction occurred 24 hours transfection using DMSO or SalB [500 nM]. Negative 1 corresponds to KORD-ABL and the full phosphorylation-dependent expression circuit without 𝛽-Arrestin2-bZipEE. Negative 2 corresponds to 𝛽-Arrestin2-bZipEE and the full phosphorylation-dependent expression circuit without KORD-ABL. 30/5 ng refers to the condition containing the full receptor construct, with 30 ng of KORD-ABL, 5 ng of 𝛽-Arrestin2-bZipEE, and the full phosphorylation-dependent expression circuit. Data is depicted as the mean +/- SEM with n = 3 biological replicates. Two-way ANOVA with Sidak's multiple comparison test. ****P < 0.0001; NS, not significant.
A critical variable in synthetic systems is the ability to respond to extracellular cues in a fast and steady manner. To evaluate this property, we characterized the response kinetics of our combined sensing, processing, and output system. For this purpose, SalB induction was performed in cells co-transfected with KORD-ABL, β-Arrestin2-bZipEE, the phosphorylation circuit, and a normalization plasmid. Induction occurred at different time points before measurement to capture dynamic changes in system activity. NanoLuciferase expression increases significantly by 10-fold compared to its uninduced condition 5 hours after induction (p = 0.0049). Expression also correlates linearly with time (R2 = 0.97), reaching a total fold change of 151-fold compared to its uninduced condition measured 32 h after induction (Fig. 15). This highlights how KORD-ABL, in conjunction with our phosphorylation circuit, drives rapid gene expression after sensing a small molecule and processing the signal at the post-translational level.
Figure 15: KORD-ABL drives fast gene expression upon ligand sensing . Line plot showing an increase in expression over time. HEK293T cells were transfected 24 hours after seeding. NanoLuc and Firefly Luciferase expression was measured 48 hours after transfection using the Dual Luciferase assay. Induction occurred at the indicated time points before measurement with either DMSO or SalB [2 µM]. Each condition was transfected with 50 ng of KORD-ABL and 5 ng of 𝛽-Arrestin2-bZipEE, Overlaid with a linear regression curve (R2 = 0.97). Data is depicted as the mean +/- SEM with n = 3 technical replicates. Due to technical errors during sample induction, one value is missing for all induced conditions. Two-way ANOVA with Sidak's multiple comparison test. **P < 0.01; ****P < 0.0001.
To expand the existing receptor repertoire to go beyond gene-expression mediated repression, we set out to establish a receptor construct that is able to directly interact with, and quench our phosphorylation circuit on a post-translational level. Fusing our SynPhosphatase, a truncated version of the PTPN1 phosphatase, to the C-terminal tail of the KORD, we aimed to enable ligand-inducible dephosphorylation and subsequent quenching of expression (Fig. 16)
Figure 16: Schematic overview of KORD-PTPN1 coupled to the phosphorylation circuit. In the absence of SalB, 𝛽-Arrestin2-bZipEE is unable to bind KORD-PTPN1. Receptor activation through SalB presence leads to a conformational change, allowing GRK binding and subsequent phosphorylation of the C-terminal tail of KORD. This allows for the binding of β-Arrestin2-bZipEE. Connection to the intracellular PHOENICS circuit is facilitated by the fused bZipEE domain on β-Arrestin2 and its spatial organization in the proximity of the PTPN1 phosphatase upon KORD binding. Bound 𝛽-Arrestin2-bZipEE recruits Gal4DBD-CD3ζ-bZipRR, which is subsequently dephosphorylated by PTPN1. Dephosphorylation of CD3ζ leads to dissociation of Gal4DBD-CD3ζ-bZipRR and Sh2-VP64, thereby splitting the transcription factor and quenching gene expression.
To evaluate the ability of KORD-PTPN1 to interact with and facilitate dephosphorylation of CD3𝜁, as well as to interrogate full circuit compatibility, we co-transfected KORD-ABL, KORD-PTPN1, 𝛽-Arrestin2-bZipEE, and our phosphorylation circuit with a 5x UAS NanoLuciferase expression cassette as well as a normalization plasmid. Cells were induced with 500 nM SalB 24 hours after transfection. Expressed NanoLuciferase was measured 48 hours after transfection using a plate reader. Titrating increasing amounts of KORD-PTPN1 against KORD-ABL led to a significant decrease in phosphorylation-mediated expression, indicating a dose-dependent ability to dephosphorylate and quench expression starting at a 2:3 ratio (KORD-PTPN1:KORD-ABL; p = 0.004) (Fig. 17). Increasing KORD-PTPN1 amounts relative to KORD-ABL further resulted in a 4.3-fold reduction in the total expressed NanoLuciferase (p < 0.0001). Overall, this indicates that KORD-PTPN1 can facilitate a dose-dependent, linear suppression of total output, confirming its inhibitory role within the circuit.
Figure 17: Titration of KORD-PTPN1 against KORD-ABL. HEK293T cells were transfected 24 hours after seeding. NanoLuc and Firefly Luciferase expression was measured using the Dual Luciferase assay 48 hours after transfection. Induction occurred 24 h after transfection, using either DMSO or SalB [500 nM]. Each condition was transfected with 30 ng of KORD-ABL and 5 ng of 𝛽-Arrestin2-bZipEE, as well as the amount of KORD-PTPN1 indicated on the X-axis. Data is depicted as the mean +/- SEM with n = 3 technical replicates. Two-way ANOVA with Sidak's multiple comparison test. ***P<0.001, ****P<0.0001.
To further evaluate the total quenching ability of KORD-PTPN1 on a single cell level, we co-transfected KORD-ABL, KORD-PTPN1, 𝛽-Arrestin2-bZipEE, and our phosphorylation circuit with a 5x UAS mCherry expression cassette as well as a normalization plasmid. Cells were induced with 500 nM SalB 24 hours after transfection. Expressed mCherry fluorescence was determined 48 hours after transfection using Flow cytometry. Cells were gated based on GFP fluorescence from a transfected normalization plasmid with constitutive expression. As a control, 180 ng of KORD-ABL was transfected together with the complete phosphorylation circuit and measured under both uninduced (DMSO) and induced (SalB) conditions. For simplicity, only induced conditions of KORD-ABL + KORD-PTPN1 are shown. mCherry fluorescence of DMSO control conditions was equivalent to that of the only KORD-ABL condition, confirming minimal basal activation. Co-transfection of equal amounts of KORD-ABL and KORD-PTPN1 with the expression circuit quenched expression by 6.8-fold. At high KORD-PTPN1 levels (2:1 ratio of KORD-PTPN1 to KORD-ABL), fluorescence approached background intensity (1.4-fold above full quenching achieved by the cytosolic SynPhosphatase), indicating that KORD-PTPN1 efficiently quenches KORD-ABL–mediated signaling (Fig. 18).
Figure 18: KORD-PTPN1 quenches expression on a post-translational level. HEK293T cells were transfected with the complete expression-coupled PHOENICS circuit 24 hours after seeding, with varying amounts of KORD-ABL and KORD-PTPN1 as indicated. Cells were induced 24 hours after transfection with DMSO or SalB [500 nM]. mCherry fluorescence was quantified with flow cytometry 48 hours after transfection. 180/0+ DMSO: Uninduced control exhibiting only background fluorescence in the absence of SalB. 180/0+ SalB: Induced positive control showing the signal achievable without quenching by KORD-PTPN1. Test conditions show the plasmid amounts between KORD-ABL and KORD-PTPN1 (180/180: 180 ng KORD-ABL and 180 ng KORD-PTPN1; intracellular components remain constant ). Fluorescence profiles of viable single cell populations (>13000 per condition) are displayed in the ridgeline plot on the left, geometric mean fluorescence (RFU) ± geometric SEM are shown in the barplot to the right.
Building on an extension of the KORD architecture, the Programmable Antigen-Gated Engineered G Protein-coupled Receptor (PAGER) (Kalogriopoulos et al., 2025), we engineered a receptor that detects both a small molecule and a soluble or surface-bound ligand. Targeting the tumor-relevant antigen vascular epithelial growth factor (VEGF), we show the ability to sense, process, and respond to physiologically relevant cues using the PAGER-ABL construct in conjunction with the PHOENICS circuit. This approach enables the PAGER architecture to overcome the existing limitations of synthetic receptors by introducing dual modularity: the extracellular nanobody domain can be readily exchanged to detect diverse soluble or membrane-associated ligands, while the intracellular GPCR backbone can be replaced with β-arrestin-recruiting receptors, ensuring signaling orthogonality.
Furthermore, this design enables flexible reprogramming of input specificity and allows combinatorial processing of signals, while supporting adaptable rewiring of downstream outputs, allowing for seamless integration into the PHOENICS circuit with fully reversible signaling dynamics.
The PAGER-ABL construct consists of the KORD-ABL backbone and intracellular domains. In addition, it contains an extracellular nanobody N-terminally fused with Arodyn, a peptide antagonist of KORD, facilitating constant auto-inhibition. Ligand binding by the nanobody displaces Arodyn from the orthosteric site of KORD, allowing SalB to bind, triggering a conformational change that enables GRK-mediated phosphorylation of residues in the receptor's C-terminal tail (Fig. 19). This recruits β-Arrestin2-bZipEE that can bind Gal4DBD-Cd3ζ-bZipRR via a coiled-coil interaction. The resulting positioning of the CD3ζ near the ABL kinase fused to the GPCR leads to its phosphorylation. The phosphorylated Gal4DBD-CD3ζ-bZipRR subsequently dimerizes with SH2-VP64, forming a transcriptional activator that binds the 5xUAS motif and drives transgene expression.
Figure 19: Schematic overview of VEGF sensing PAGER-ABL coupled to the phosphorylation circuit. In the absence of SalB or VEGF, 𝛽-Arrestin2-bZipEE is unable to bind KORD-ABL. Presence of VEGF results in nanobody binding, displacing Arodyn from the orthosteric pocket of PAGER-ABL. This sensitizes the PAGER to SalB, allowing SalB-dependent activation. Receptor activation leads to a conformational change, allowing GRK binding and subsequent phosphorylation of the C-terminal tail of KORD. This allows for the binding of β-Arrestin2-bZipEE. Connection to the intracellular PHOENICS circuit is facilitated by the fused bZipEE domain on β-Arrestin2 and its spatial organization in the proximity of the ABL kinase upon KORD binding. Bound β-Arrestin2-bZipEE recruits Gal4DBD-CD3ζ-bZipRR, which ABL subsequently phosphorylates. Dissociation from the KORD due to the transient coiled-coil interactions allows dimerization between the phosphorylated CD3𝜁 and SH2, reconstituting the split transcription factor Gal4DBD-VP64. Shuttling to the nucleus then facilitates transgene expression under control of the 5xUAS-minCMV promoter.
To test whether the VEGF-sensing PAGER-ABL receptor could act as an AND gate and drive expression through the PHOENICS circuit, we co-transfected cells with VEGF-PAGER-ABL, the phosphorylation circuit, a NanoLuciferase reporter, and a constitutive control for normalization. HEK293T cells were stimulated 24 hours after transfection with SalB (100 nM, 250 nM, or 500 nM) and VEGF165 (500 nM). Nano- and Firefly Luciferase activity was determined 48 hours after transfection using the Nano-Glo Dual-Luciferase Reporter Assay System. Induction of PAGER-ABL with either DMSO, SalB, or VEGF did not induce a significant increase in NanoLuciferase expression (p > 0.05). Induction with SalB + VEGF resulted in significant, dose-dependent increases of NanoLuciferase expression. Compared to SalB only, SalB + VEGF induced a 44.2-fold, 48.8-fold, and 28-fold change in expression (Fig. 20). This shows the functionality of a novel receptor architecture that can drive transgene expression in an AND logic gate, binary switch-like manner, without exhibiting significant background or interference in native signaling pathways. Furthermore, the receptor architecture can sense tumor-relevant antigens and induce expression in a dose-dependent manner.
Figure 20: VEGF sensing PAGER-ABL represents a novel receptor platform, sensing and responding to a surface-bound or soluble ligand and a small molecule extracellular cues in an AND logic, switch-like manner . Barplot showing NanoLuciferase expression in the presence of different ligands and at different SalB concentrations. HEK293T cells were transfected 24 hours after seeding. NanoLuc and Firefly Luciferase expression was measured 48 hours after transfection using the Dual Luciferase assay. Induction occurred 24 hours after transfection with DMSO, DMSO + VEGF [500 nM], SalB or VEGF [500 nM] + SalB. Each condition was transfected with 60 ng of KORD-ABL and 5 ng of 𝛽-Arrestin2-bZipEE. Data is depicted as the mean +/- SEM with n = 3 technical replicates. Two-way ANOVA with Tukey's multiple comparisons test was performed. ****P < 0.0001; NS, not significant.
MESA
To expand the sensing layer of our system, we engineered and optimized rapalog-inducible two-chain Modular Extracellular Sensing Architecture (MESA) receptor system in HEK293T cells. With this we aimed to establish an additional receptor architecture as part of our PHOENICS toolbox, to enable straight forward retargeting towards additional cancer-relevant ligands.
Our MESA receptor design was inspired by a recent publication (Yang et al., 2025) and consists of a rapalog binding domain-fused ABL kinase chain (FRB-ABL) and a rapalog binding domain-attached bZipEE coiled coil chain (FKBP-bZipEE), which heterodimerize upon addition of the rapalog (AP21967). Proximity between receptor-fused kinase and substrate-recruiting coiled coil results in the phosphorylation of our regulatory substrate CD3ζ. This activates our previously established expression platform: an SH2-Gal4DBD reader protein and a bZipRR-VP64 activator, which is fused to CD3ζ. SH2 selective binding to phosphorylated CD3ζ enables the reconstitution of a functional Gal4DBD-VP64 transcription factor, driving NanoLuciferase reporter expression (Fig. 21).
Figure 21: NanoLuciferase expression coupled PHOENICS circuit with rapalog induction via MESA kinase receptors. Adapted MESA-receptors consist of a bZipEE coiled coil-carrying and a ABL kinase fused chain. Receptors bind rapalog via extracellular FRB/FKBP domains, mediating receptor dimerization and inducing proximity between receptors fused ABL kinase and coiled coil bZipEE. bZipEE recruits the substrate-construct via a complementary coiled coil bZipRR. Proximity-induced phosphorylation of CD3ζ results in binding of SH2. Gal4DBD and VP64 reconstitute. This drives NanoLuciferase reporter expression upon nuclear shuttling of the reconstituted transcription factor complex, producing a measurable luminescence signal.
To verify that ligand-dependent receptor-dimerization leads to substrate-recruitment and phosphorylation, cells were co-transfected with equal amounts of FRB-ABL and FKBP-bZipEE receptor constructs in combination with a bZipRR-fused CD3ζ-substrate. To assess ligand-dependent substrate-phosphorylation, western blot with phosphospecific and anti-Myc Antibody-staining was performed for rapalog-induced and uninduced conditions (Fig. 22A). To confirm ligand-inducible expression via our Nanoluciferase platform, cells were co-transfected with optimal ratios of FRB-ABL and FKBP-bZipEE, alongside the phosphorylation-responsive protein-circuit components and NanoLuciferase reporter construct. NanoLuciferase activity was quantified by luminometry and normalized by Firefly luciferase, which was co-transfected at equimolar amounts across all conditions, accounting for possible variations in cell viability and transfection efficiency (Fig. 22B).
Figure 22: Verification of rapalog-induced phosphorylation and SH2-mediated reporter activation. (A) Western blot of myc-tagged substrate using anti-phosphotyrosine (pY) and anti-myc antibodies. The strong myc signal in the positive control results from reusing the same secondary antibody without stripping after pY detection, leading to signal accumulation. (B) Quantification of SH2-mediated reporter activation using the Nano-Glo assay . Cells were transfected with the complete expression-coupled PHOENICS circuit, varying only the receptor configuration as indicated. Receptor conditions contained a 1:9 ratio of ABL to bZipEE halves (10 ng total DNA). Pos. ctrl: cytosolic kinase fused to a coiled coil; neg. ctrl: FRB-ABL receptor half lacking the coiled coil half. NanoLuciferase luminescence values (AU) are normalized by Firefly luminescence (AU) and display mean ± SEM. Normalized luminescence was analyzed by two-way ANOVA. P values: ns > 0.05; * ≤ 0.05; ** ≤ 0.01; *** ≤ 0.001; **** ≤ 0.0001.
In the Western Blot, cytosolic ABL in combination with the coiled coil-fused substrate serves as a positive control for robust phosphorylation. As a negative control, we used a receptor construct without coiled coil, disrupting substrate-recruitment to the receptor fused ABL kinase. Anti-Myc-staining shows clear bands for both rapalog-induced and uninduced receptors at the length of our CD3ζ substrate, confirming successful protein loading. Anti-phosphorylation-staining results in a clear band for the induced condition, while uninduced receptors did not exhibit detectable phosphorylation.
In luminometry, cytosolic ABL in combination with intracellular circuit components again serves as a positive control. As a negative control, the FRB-ABL receptor chain and circuit components were transfected without the FKBP partner to assess background phosphorylation. Rapalog induction produced a 3.4-fold increase in NanoLuciferase expression compared to uninduced conditions, demonstrating robust ligand-dependent transcriptional activation through our synthetic phosphorylation receptor platform.
This demonstrates ligand-dependent phosphorylation of our CD3ζ substrate using our receptor designs and verifies ligand-inducible expression via our MESA-receptor-coupled phosphorylation circuit.
After establishing ligand-dependent activation of gene expression, we engineered what is, to our knowledge, an entirely novel platform for inhibitory ligand sensing and integrated it into our circuit. For this, we replaced the truncated ABL kinase with the truncated phosphatase PTPN1 on the FRB receptor chain. This MESA-receptor now dephosphorylates our regulatory substrate upon ligand-induced dimerization, which prevents SH2-mediated transcription factor reconstitution and inhibits expression (Fig. 23).
Figure 23: Rapalog-mediated suppression of NanoLuciferase expression via the PHOENICS circuit coupled to MESA phosphatase receptors. Ligand-dependent control of proximity-based dephosphorylation was achieved by replacing the intracellular kinase domain of one MESA receptor with a phosphatase, while retaining the bZipEE motif on the other receptor half. Co-transfection with a cytosolic kinase ensures constitutive phosphorylation of the substrate, enabling detection of ligand-induced dephosphorylation events. Upon rapalog binding, MESA receptor dimerization brings the phosphatase into proximity with the substrate, promoting dephosphorylation. Loss of substrate phosphorylation disrupts SH2 binding and prevents reconstitution of the Gal4DBD and VP64 transcriptional activator, thereby suppressing NanoLuciferase reporter expression and reducing luminescence output.
To demonstrate ligand-dependent inhibition of expression, we co-transfected optimal ratios of our inhibitory receptor constructs alongside cytosolic ABL to maintain basal substrate phosphorylation, the intracellular circuit components, and the NanoLuciferase reporter construct. NanoLuciferase activity was again quantified by luminometry and Firefly Luciferase normalization (Fig. 24).
Figure 24: Rapalog-induced reporter expression quenching by phosphatase receptor. Quantification of SH2-mediated reporter activation using the Nano-Glo assay. Cells were transfected with the complete expression-coupled PHOENICS circuit, varying only the receptor configuration as indicated. Neg. ctrl: FKBP-bZipEE receptor half lacking the phosphatase-fused construct. Receptor conditions contained a 1:4 ratio of PTPN1 to bZipEE halves (10 ng total DNA). NanoLuciferase luminescence values (AU) are normalized by Firefly luminescence (AU) and display mean ± SEM. Luminescence was analyzed by two-way ANOVA. P values: ns > 0.05; * ≤ 0.05; ** ≤ 0.01; *** ≤ 0.001; **** ≤ 0.0001.
As a negative control for background gene expression, we transfected intracellular circuit components and our FRBP-bZipEE construct. Rapalog treatment of functional receptor conditions resulted in a 2.8-fold reduction in normalized NanoLuciferase expression compared to uninduced states, verifying reliable, ligand-dependent inhibition of gene expression.
This demonstrates functionality of our phosphatase-fused receptors and enables intracellular processing of two opposing ligand signals with our MESA-architectures coupled to our phosphorylation circuit.
After integrating stimulatory and inhibitory ligand sensing into our circuit, we set out to verify target flexibility of our synthetic receptor architecture. By exchanging the extracellular domains of our receptor constructs with anti-TNFα scFvs, we established soluble protein-dependent activation of gene expression (Fig. 25).
Figure 25: NanoLuciferase expression coupled PHOENICS circuit with TNFα induciton via MESA kinase receptor. Adaptation of the initial reporter assay to assess TNFα-mediated reporter expression via anti-TNFα-scFv-fused MESA-receptors. MESA-receptors bind TNFα via extracellular anti-TNFα-scFvs. TNFα-binding mediates receptor dimerization, inducing proximity between receptors fused ABL kinase and coiled coil bZipEE. bZipEE recruits the substrate-construct via a complementary coiled coil bZipRR. Proximity-induced phosphorylation of CD3ζ results in binding of SH2. Gal4DBD and VP64 reconstitute. This drives NanoLuciferase reporter expression upon nuclear shuttling of the reconstituted transcription factor complex, producing a measurable luminescence signal.
To show induction of gene expression by TNFα-sensing, we co-transfected our adapted receptor constructs, intracellular circuit components, and the NanoLuciferase reporter construct. NanoLuciferase activity was quantified by luminometry and Firefly Luciferase normalization (Fig. 26).
Figure 26: TNF-induced reporter activation by MESA kinase receptor. Quantification of SH2-mediated reporter activation using the Nano-Glo assay. Cells were transfected with the complete expression-coupled PHOENICS circuit, varying only the receptor configuration as indicated. Neg. ctrl: RAPA-ABL receptor half lacking the bZipEE half. Receptor conditions contained a 1:4 ratio of ABL to bZipEE halves (10 ng total DNA). NanoLuciferase luminescence values (AU) are normalized by Firefly luminescence (AU) and display mean ± SEM. Luminescence was analyzed by two-way ANOVA. P values: ns > 0.05; * ≤ 0.05; ** ≤ 0.01; *** ≤ 0.001; **** ≤ 0.0001.
As a positive control, cytosolic ABL kinase was added to facilitate strong substrate phosphorylation. Conditions including only the intracellular circuit components alone as well as the ABL receptor monomer served as negative controls of background gene expression and background phosphorylation byFRB-ABL without its FKBP-bZipEE-partner. For functional receptor conditions, treatment with TNFα resulted in a 1.8-fold increase of NanoLuciferase expression, confirming soluble-protein-dependent activation of our circuit, albeit needing optimization to reduce background expression.
To refine expression control for minimized background activity, we utilized our dry lab model SPARC and optimized receptor transmembrane domains (TMDs) with coarse-grained molecular dynamics. To reduce spontaneous dimerization we exchanged CD28 and CD28-M3 TMDs with FGFR1 TMDs. Additionally we fine tuned receptor construct ratios for precise TNFα-inducible phosphorylation of our regulatory substrate. To assess phosphorylation efficiency of different designs at single-cell resolution, we adapted our circuit for a flow cytometry readout. We replaced Gal4DBD and VP64 with halves of the splitFast fluorophore system to quantify SH2-mediated assembly of the intracellular circuit components. Upon reconstitution of circuit components and addition of HMBR, SplitFast becomes fluorescently active (Fig. 27).
Figure 27: TNFα-inducted split fluorophore reconstitution in PHOENICS circuit via MESA kinase receptor. MESA-receptors bind TNFα via extracellular anti-TNFα-scFvs. TNFα-binding mediates receptor dimerization, inducing proximity between receptors fused ABL kinase and coiled coil bZipEE. bZipEE recruits the substrate-construct via a complementary coiled coil bZipRR. Proximity-induced phosphorylation of CD3ζ results in binding of SH2. Split fluorophore constructs RspA(C) and RspA(N) reconstitute, yielding fluorescence upon addition of HMBR, producing a measurable reporter for CD3ζ-SH2 association.
To demonstrate reduced background activity of our improved TNFα-MESA, we compared the previous design with FGFR1 TMD constructs, which was transfected at optimal ratios of ABL-chain and bZipEE-chain, as predicted by SPARC, and quantified CD3ζ phosphorylation with flow cytometry (Fig. 28).
Figure 28: TNFα-induced CD3ζ-SH2 constitution via MESA-like kinase receptor. Quantification of TNFα-induced phosphorylation-mediated CD3ζ-SH2 constitution. Cells were transfected with the complete expression-coupled PHOENICS circuit, varying only the receptor configuration as indicated. Neg. ctrl: No receptor. Receptor conditions contained specified ratios of ABL to bZipEE halves (170 ng total DNA in 12-well plate wells). SplitFast fluorescence was quantified with flow cytometry. Dimerization dependent SplitFAST fluorescence was quantified by flow cytometry 48 hours after transfection, employing excitation lasers at 488 nm and 561 nm. Emission was recorded at 525 nm for splitFAST (Ligand: TFLime) and at 610 nm for constitutively expressed mCherry, serving as a transfection control. (A) Fluorescence profiles of viable single cell populations are displayed in ridgeline plot, (B) geometric mean fluorescence (RFU) ± geometric SEM with ~9000 single cells are shown in barplot.
As a negative control, we transfected our SplitFast-fused intracellular components without receptor constructs to display background fluorescence without substrate-phosphorylation. For functional receptor conditions, FGFR1-transmembrane domains and an optimized ratio of receptor constructs for reduced background activity exhibit a clear shift of the cell population towards higher fluorescence upon TNFα treatment. With these changes in place we move from a 1.5-fold increase of geometric mean of fluorescence between uninduced and induced states for the old ratios and transmembrane domains to a 3.7-fold increase. Optimized receptor construct ratios and transmembrane domains result in significantly lower background phosphorylation in the uninduced state, likely due to minimized spontaneous dimerization. We expect to transfer these improvements in dynamic range to our expression system, which is mediated by the constitution of the intracellular circuit components measured in this assay, in the near future.
Overall, this serves as a proof-of-concept for processing soluble-protein ligands in our phosphorylation circuit via our MESA-receptor platform. We showcase its target flexibility by readily exchangeable extracellular binding domains, opening up various retargeting options to tumor-microenvironment associated soluble proteins.
After establishing both, small-molecule and soluble-protein gated synthetic MESA-receptors as a sensing layer, we proceeded to validate simultaneous processing of multiple ligand-signals in our phosphorylation circuit. Guided by SPARCs mathematical modeling predicting optimal receptor ratios, we integrated both our kinase-carrying TNFα-receptors and our ralapog-gated phosphatase-receptors within one cell, forming a cellular processing unit: Gene expression is only triggered when the stimulatory TNFα-signal outweighs the inhibitory rapalog input, providing an additional safety layer and enabling precise environmental control of cellular outputs (Fig. 29).
Figure 29: NanoLuciferase expression coupled PHOENICS circuit with TNFα induction and rapalog inhibition via MESA receptors. Adaptation of the initial reporter assays to assess TNFα-induced reporter expression via anti-TNFα-scFv-fused MESA-receptors and rapalog-controlled quenching. ABL kinase carrying MESA-receptors bind TNFα via extracellular anti-TNFα-scFvs. TNFα-binding mediates receptor dimerization, inducing proximity between ABL and coiled coil bZipEE. bZipEE recruits the substrate-construct via a complementary coiled coil bZipRR, resulting in proximity-induced phosphorylation of CD3ζ. SH2 binds phosphorylated CD3ζ, with subsequent Gal4DBD-VP64 transcription factor constitution and NanoLuciferase reporter expression. Rapalog-binding of PTPN1 phosphatase-carrying receptor induced dimerization, resulting in substrate dephosphorylation, quenching reporter expression. NanoLuciferase expression can be quantified by luminometry.
To showcase effective expression regulation in a two-ligand-setting, we co-transfected stimulatory and inhibitory receptors alongside our intracellular circuit components and the NanoLuciferase reporter construct. NanoLuciferase activity was quantified by luminometry and normalized by Firefly Luciferase (Fig. 30).
Figure 30: Dual-receptor logic for processing dual-ligand-specific reporter activation. Quantification of SH2-mediated reporter activation using the Nano-Glo assay. Cells were transfected with the complete expression-coupled PHOENICS circuit, varying only the receptor configuration as indicated. Neg. ctrl: No receptor. Receptors were co-transfected with 10 ng total DNA using a 1:4 ratio of TNF to rapalog receptors. For the rapalog receptor, a 1:3 ratio of phosphatase to bZipEE halves was used; for the TNF receptor, a 1:5 ratio of TNF-ABL to bZipEE halves. NanoLuciferase luminescence values (AU) are normalized by Firefly luminescence (AU) and display mean ± SEM. Luminescence was analyzed by two-way ANOVA. P values: ns > 0.05; * ≤ 0.05; ** ≤ 0.01; *** ≤ 0.001; **** ≤ 0.0001.
Circuit components without receptor constructs serve as a negative control displaying only background expression. For conditions including both, stimulatory TNFα-receptor and inhibitory rapalog-receptor, TNFα treatment produced a 2.0-fold increase relative to the uninduced state. Simultaneous TNFα and rapalog exposure fully suppressed activation, returning NanoLuciferase to baseline expression of the uninduced system. This demonstrates functional ligand-dependent AND-NOT gating of expression. Future experiments will focus on the reduction of background activity by optimizing transmembrane domains and calibrating receptor stoichiometry, as shown above. By adjusting the relative abundance of stimulatory versus inhibitory receptors, the switching threshold of stimulatory versus inhibitory ligand concentrations can be positioned at defined ligand ratios. This maintains dormancy in healthy tissues and robust expression of therapeutically relevant effectors when tumor microenvironment specific ligand profiles are encountered.
Overall, these results serve as a proof-of-concept for the computation of opposing signals with our phosphorylation circuit, which can be tailored to produce precise cellular outputs. By exchanging extracellular binding domains on our MESA-receptors, we have showcased straightforward retargetability of our receptors to cancer-relevant soluble-proteins. This unlocks various potential targets that shape and are indicative of the tumor microenvironment. We have further expanded the accessible ligand space with our dry lab efforts by successfully generated several de novo protein binders for cancer-relevant ligands with experimental validation demonstrating our pipeline's reliability. This enables personalized applications of the PHOENICS system, which can be re-tuned to programmed ligand combinations, allowing therapeutic effector functions to be gated by ligand ratios characteristic of specific tumor microenvironments.
Binder Validation
To enable modular retargeting of our receptor platforms toward cancer-relevant ligands, we established a streamlined pipeline integrating computational de novo binder design with functional validation using cell-free two-hybrid (CF2H) screening. Screening five binders against GDF-15, a protein tumor related protein, we identified a promising binder, validating this approach for expanding the sensing capabilities of our PHOENICS toolbox.
Having designed protein binders for retargeting our sensory receptor platforms by exchange of extracellular binding domains on MESAs or nanobody-replacement on our GPCRs, we set out to achieve experimental validation of our binder designs. To rapidly assess computationally designed binders against GDF-15, a protein upregulated in cancers as shown by (Zhao et al., 2020). We implemented the Cell-Free Two-Hybrid (CF2H) system as described by (Capin et al., 2025). This approach enables protein-protein interaction screening without requiring cloning, protein purification, or specialized equipment. This method utilizes a cell-free expression system to selectively express reporter sfGFP by binder-mediated reconstitution of split transcription factors. Biotinylated GDF-15-Fc was complexed with streptavidin to induce multimerization and then spiked into cell-free reactions containing linear DNA encoding CIopt-fused binder constructs and a pRM-sfGFP reporter plasmid. After expression, binder-target interaction induces CIopt dimerization, which activates pRM promoter-driven sfGFP expression, providing a fluorescent readout to functionally assess binding capabilities of the binder design to its designated target (Fig. 31)
Figure 31: Cell-Free Two-Hybrid (CF2H) assay for functional binder validation. Functional validation of protein-binders with sfGFP expression readout. Biotinylated target proteins complex with the streptavidin tetramer. This complex is added to a cell-free expression system in combination with linearized DNA encoding for a CIoptDBD-fused binder and a reporter plasmid, encoding sfGFP under pRM-promotor control. CIoptDBD-binder expression results in binding to target protein with subsequent CIopt-dimerization. Two dimerized CIopt domains bind to Operon 1 and Operon 2 of the pRM-promoter inducing expression and subsequently yielding a measurable fluorescence signal.
We observed some basal promoter leakage for the negative control, indicating moderate residual pRM activity in the absence of CIopt activation. This was however significantly lower compared to our positive control, confirming the viability of this assay. Binder B3 demonstrated highest sfGFP fluorescence upon ligand induction out of all tested candidates, while in the absence of biotinylated GDF-15 it produced fluorescence not higher than the autofluorescence of the cell-free extract (Fig. 32). This confirms that sfGFP was specifically expressed due to binder-target interaction rather than non-specific aggregation. Binder-3 exhibited higher fluorescence than our positive control, however direct comparison of binding affinities is limited by varying protein concentrations as binders were expressed in situ by the cell-free system. Additionally the molar amount of GDF-15 binder DNA slightly exceeded the amount of the positive control binder.
Fig 32: Functional validation of de novo binders with the CF2H assay. Functional assessment of in silico generated de novo binders against GDF-15 using the CF2H assay. Neg. ctrl: No DBD-binder encoding DNA. Pos. ctrl: DNA encoding for NbALFA nanobody paired cognate ALFA-tag epitope. sfGFP fluorescence [AU] was measured for induction with GDF-15 and compared to fluorescence without target protein, displaying mean ± SEM. Fluorescence was analyzed by two-way ANOVA. P values: ns > 0.05; * ≤ 0.05; ** ≤ 0.01; *** ≤ 0.001; **** ≤ 0.0001.
While quantitative comparison is thus limited, these results qualitatively confirm the binding capabilities of our de novo designed binder to GDF-15. Binders B2 and B7 also showed detectable binding activity, albeit without or reduced significant difference compared to background sfGFP expression. B1 and B5 did not produce detectable sfGFP fluorescence over the autofluorescence of the extract measured in Neg. CT2. Overall, these results demonstrate that our de novo binder actively targets cancer-relevant GDF-15. This indicates the potency of our dry lab model SPARC in designing functional binders for select target proteins and highlights the utility of CF2H as an accessible and rapid validation platform for de novo designed protein binders. Going forward, this established pipeline linking in silico design and experimental validation enables us to engineer custom binding moieties for retargeting our receptor platforms.
Discussion
In this year's iGEM cycle, we established two distinct receptor platforms to connect extracellular ligand detection to our phosphorylation circuit: a small-molecule or protein ligand-gated MESA architecture and a synthetic GPCR-based receptor design sensing either small molecules alone or in combination with protein cues. Both systems rely on the same intracellular principle of ligand-induced recruitment of the regulatory CD3ζ substrate, inducing proximity to receptor-fused kinases for stimulatory ligands or phosphatases for inhibitory signaling. Resulting phosphorylation or dephosphorylation of CD3ζ regulates gene expression or secretion via SH2-mediated transcription factor reconstitution. Despite this shared design logic, the two receptor classes exhibited strikingly different behaviors in terms of background activity, dynamic range, and modularity, offering complementary advantages and limitations.
We created our synthetic GPCR sensing platform by fusing a truncated version of the ABL kinase or PTPN1 phosphatase to the κ-opioid receptor DREADD (KORD) and recruiting the CD3ζ substrate to β-Arrestin2 via complementary coiled coils. When the GPCR is activated by ligand binding, it undergoes a conformational change that mediates 𝛽-Arrestin2-bZipEE binding, thus ensuring that proximity-induced (de–)phosphorylation events occur exclusively after ligand binding.
This architecture exhibited highly accurate ligand-inducible control over reporter expression with robust fold changes in both luminometry and flow cytometry. By integrating either SynKinase or SynPhosphatase fusions, we could readily switch between ligand-inducible activation (KORD-ABL) and inhibition (KORD-PTPN1), enabling processing of both stimulatory and inhibitory signals. Motivated by these results, we further extended this GPCR platform by implementing PAGER (Programmable Antigen-gated G-Protein coupled Engineered Receptors) designs, where extracellular nanobodies carrying an arodyn-blocking peptide enabled dual-input AND-gating of small-molecule and protein ligands. After achieving proof-of-concept for the PAGER design as a sensory component in our phosphorylation circuit, we retargeted PAGER to VEGF, successfully establishing cancer-relevant soluble protein sensing coupled to an expression output. Collectively, these results establish GPCR-based (de-)phosphorylation receptors as a highly versatile, tight platform for precise environmental control of cellular activity in the PHOENICS circuit.
Our MESA designs were engineered by fusing ABL kinase or PTPN1 phosphatase to receptors carrying rapalog-binding domains and recruiting CD3ζ via a coiled coil. Ligand binding induces receptor dimerization and brings the substrate into proximity to the kinase or phosphatase, yielding ligand-inducible transcriptional activation. Furthermore, by exchanging extracellular domains (ECDs) for scFvs, we showed that MESAs are readily retargetable to soluble proteins such as TNFα, confirming their modularity as a sensing layer. Finally, by integrating kinase- and phosphatase-fused MESA receptors in one cell, we demonstrated PHOENICS' ability to process opposing extracellular cues, resulting in flexible switching between active and dormant states.
Our current MESA designs exhibit moderate spontaneous background dimerization, indicated by elevated reporter expression compared to baseline levels in the absence of an inducing ligand. Despite optimization cycles guided by our SPARC modeling, including transmembrane domain engineering and construct ratio tuning, leaky expression in the OFF-state could only be partially suppressed. This resulted in smaller fold changes compared to our synthetic GPCRs and limits the dynamic range of our MESA receptors. While our MESA platform provides a versatile and modular architecture, we plan to continue optimization efforts, which will likely prove advantageous for applications requiring highly precise control of cellular outputs.
Together, our results establish two complementary receptor platforms for phosphorylation-based signal processing. MESAs excel in their modularity and straightforward retargetability, offering a rapid path to new ligand inputs. GPCRs provide a novel receptor class with tight OFF states and the ability to gate multiple ligand modalities via a single construct. Our receptor constructs have been engineered as plug-and-play components of our PHOENICS circuit and can effectively enable the processing of multiple opposing signals coupled to therapeutic gene expression or secretion. This paves the way for customized applications of the PHOENICS system, which can be retuned to programmed ligand combinations targeting TME-specific characteristics.
Outlook
Throughout our engineering cycles, we identified stoichiometry and ligand concentration as critical design parameters across both platforms. For GPCRs, optimal ratios of receptor to β-Arrestin2 modules and appropriate SalB concentrations were necessary to achieve maximal induction while minimizing background. For MESAs, tuning transmembrane domains and receptor monomer stoichiometry was essential to mitigate spontaneous dimerization. These optimization steps were closely guided by our computational model SPARC and highlight the importance of iterative Design-Build-Test-Learn cycles in developing synthetic receptor systems with predictable input-output behaviors. Building on these insights, our next steps focus on further suppressing MESA background activity through alternative transmembrane scaffolds and dimerization interfaces.
In parallel, we aim to expand our sensory platform by leveraging SPARC to design MESA-ECDs and arodyn-fused nanobodies for our GPCR architecture against additional tumor-relevant targets. Additionally, we plan to implement multi-input logic by combining multiple receptor modules to enable more sophisticated tumor microenvironment discrimination. SPARC will be instrumental in guiding these expansions, enabling rational design of custom binding domains, optimal circuit compositions and predictive tuning of receptor stoichiometry for maximum therapeutic efficacy.
Circuit Assembly
With all system components validated and characterized, it was finally time to put it all together and assemble the PHOENICS circuit. To demonstrate the modularity and applicability of our system, we implemented two different circuits, applying our sense-process-respond logic with both a tumor-relevant ligand and a therapeutically applicable output protein.
Using the previously established VEGF-PAGER-ABL, our optimized PhosphoTEV and the NanoLuc-RELEASE reporter, we implemented a circuit to make RELEASE VEGF-inducible (Fig. 33). To simulate an application in which SalB is present systematically and circuit assembly should occur only in the presence of VEGF, NanoLuc secretion was compared between SalB alone and with VEGF. Co-stimulation 24 hours post-transfection with SalB led to a significant increase in secretion (p=0.03), corresponding to a modest 1.5-fold change. To estimate the basal activity of SalB alone, a separate no-kinase RELEASE condition was included for reference. In this comparison, SalB alone appeared to generate elevated background relative to the no-kinase baseline, suggesting partial leakiness of PAGER gating in the absence of VEGF.
Figure 33: VEGF-inducible NanoLuc secretion. VEGF-inducible NanoLuc secretion. HEK293T cells were transfected with VEGF-PAGER-ABL, PhosphoTEV, and NanoLuc-RELEASE plasmids 24 hours post-seeding. 24 hours post-transfection, they were induced with either SalB [500 nM] or SalB [500 nM] + VEGF [500 nM]. The data is shown as the mean +/- SEM with n = 3 technical replicates. A 1-tailed Student's t-test was performed to test for an increase with VEGF induction. Positive and negative controls were excluded from testing and are shown as a reference. *P<0.05.
To prove the modularity of our secretion module, we exchanged the NanoLuc-RELEASE reporter with Interleukin-12 (IL-12). IL-12 is a potent cytokine that activates natural killer and cytotoxic T cells and exhibits anti-angiogenic effects, inhibiting tumor growth and counteracting the immune-suppressive tumor microenvironment. We validated it on a simple circuit using two different amounts of the KORD-ABL construct. The cells were induced with SalB after 24 hours and secreted IL-12 was measured in the supernatant another 24 hours later using a sandwich ELISA with a chemiluminescent readout (Fig. 34). For both KORD-ABL amounts, a significant increase in secreted IL-12 could be observed. However, there was a substantial increase without SalB induction compared to the negative control without any protease, indicating leakage of the system. Nevertheless, this proves the system working as planned in principle.
Figure 34: SalB induced IL-12 secretion using PhosphoTEV-RELEASE and KORD-ABL. HEK293T cells were co-transfected with β-Arrestin2-bZipEE, IL-12-RELEASE, PhosphoTEV and KORD-ABL plasmids. Cells were induced with 3 µM SalB 24h post-transfection. IL-12 in 100 µL of supernatant was measured by ELISA with a chemiluminescence readout 48h post-transfection. Data is depicted as the mean +/- SEM with n = 3 technical replicates. Statistical significance was calculated with a Two-way ANOVA with Sidak's multiple comparisons test. *P<0.05, **P<0.01.
Discussion
These results demonstrate a proof of concept for fully post-translational, receptor-driven protein secretion, mediated by phosphorylation. We successfully validated all system components individually, thoroughly characterizing them and showed them to work in conjunction. Thus, we established the PHOENICS toolbox as a modular framework that allows reprogramming to different input signals, as well as effector functions. The proven reversibility of our signal transduction through inducible dephosphorylation allows our system to integrate multiple signals and thus adapt to different therapeutic contexts. We present a set of modular receptors, as well as two output systems with different advantages, with either a superior specificity or rapid response.
Additional Modules
Membrane Localization using Temperature (Melt)
To enable sensing of additional environmental cues characteristic of the tumor microenvironment, such as elevated temperature, hypoxia, and pH, we set out to engineer complementary sensory modules beyond small-molecule and protein ligands. While experiments for hypoxia and pH sensing are ongoing, we have achieved promising results for temperature-controlled gene expression.
Figure 35: Mechanism of temperature-gated transcription with Melt40. Schematic of the working model. At cooler temperatures, Melt40 (purple) oligomerizes and is recruited to the plasma membrane via the STIM polybasic segment, sequestering the fused Gal4DBD-VP64 away from DNA. Upon heating (red arrow), Melt disengages from the membrane and the SV40 NLS drives nuclear import of Gal4DBD-VP64. In the nucleus, Gal4DBD binds UAS sites and VP64 activates transcription of the mCherry reporter, which accumulates in the cytosol (red). Cooling reverses the process (blue arrow), returning the fusion to the membrane and reducing transcriptional output.
Melt is a temperature-inducible membrane translocator consisting of a mutated LOV domain that undergoes conformational change around 40°C (Benman et al., 2025). We designed a temperature-sensing system by fusing it with a Gal4 DNA-binding domain (Gal4DBD) and VP64 transcriptional activator. This construct facilitates nuclear shuttling via a nuclear localization sequence (NLS). Our Melt-construct was combined with an mCherry reporter controlled by a Gal4-UAS promoter (Fig. 35). To measure temperature-induced membrane-dissociation, we incubated transfected cells at 37°C and 40°C and measured reporter expression with flow cytometry (Fig. 36).
Figure 36: Temperature-induced reporter expression via Melt. Quantification of temperature-induced mCherry reporter expression via membrane-dissociating transcription factor-fused Melt. Cells were transfected with mCherry reporter construct under UAS-promotor control. Neg. ctrl: whiteout Melt. Melt conditions contained specified construct version, with Gal4DBD-VP64 fused C-terminally. Transfected cells were incubated for 12-16h at 37°C or 40°C before measurement. mCherry fluorescence was quantified with flow cytometry. Fluorescence profiles of viable single cell populations are displayed in the ridgeline plot on the left, geometric mean fluorescence (RFU) ± geometric SEM are shown in the barplot to the right.
As a negative control we transfected cells solely with the mCherry reporter construct, which exhibited minimal basal expression for both incubation temperatures. When incubated at 37°C, our Melt construct exhibited moderate background expression, as indicated by a detectable shift of the cell population towards higher fluorescence and an elevated geometric mean of fluorescence. However, induction at 40°C produced a significantly higher population of fluorescent cells and a 6.6-fold increase of measured mean mCherry fluorescence. Notably, when comparing mCherry expression through a fused Gal4DBD-VP64 construct at 37°C and 40°C, only a minor (1.1-fold) increase of reporter expression was detected (Fig. 37). Temperature differences alone thus do not account for the observed effect in this assay.
Figure 37: Minimal temperature-induced variation of reporter expression without Melt. Quantification of mCherry reporter expression via Gal4DBD-VP64 transcription factor. Cells were transfected with mCherry reporter construct under UAS-promotor control and Gal4DBD-VP64 transcription factor. Transfected cells were incubated for 12-16h at 37°C or 40°C before measurement. mCherry fluorescence was quantified with flow cytometry. Fluorescence profiles of viable single cell populations are displayed in ridgeline plot on the left, geometric mean fluorescence (RFU) ± geometric SEM are shown in barplot to the right.
Overall, these results demonstrate robust temperature control of expression and suggest a promising candidate for a temperature-sensing module in our circuit, albeit needing some optimization to minimize background membrane dissociation. Our next steps aim to implement Melt as another sensing platform in PHOENICS, through SH2 domain fusion. In this configuration, temperature elevation would induce SH2 release from the membrane, freeing it to participate in transcription factor reconstitution within our established phosphorylation-based circuit (Fig. 38). We also plan to tune LOV domain variants to reduce background activity at physiological temperature (37°C) while achieving selective induction at pathologically relevant elevated temperatures. Critically, this temperature-sensing capability could serve as an externally controllable safety mechanism. It could locally confine cell effector functions to tumor sites that frequently exhibit elevated temperature levels (Yahara et al., 2003). Moreover, therapeutic activation could be spatiotemporally triggered by elevating tumor temperature through focused ultrasound.
Figure 38: Minimal temperature-induced variation of reporter expression without MELT. Quantification of mCherry reporter expression via Gal4DBD-VP64 transcription factor. Cells were transfected with an mCherry reporter construct under UAS-promoter control and the Gal4DBD-VP64 transcription factor. Transfected cells were incubated for 16h at 37°C or 40°C before measurement. mCherry fluorescence was quantified with flow cytometry. Fluorescence profiles of viable single cell populations are displayed in a ridgeline plot on the left, geometric mean fluorescence (RFU) ± geometric SEM are shown in a barplot to the right.
Two-Component System
We implemented the bacterial two-component system EnvZ/OmpR as an additional signaling layer. Using histidine-aspartate phosphorylation, a mechanism absent in mammalian cells, it is fully orthogonal and can function in parallel with the PHOENICS system. We successfully achieved OmpR activation through substrate phosphorylation, demonstrating modular signal control.
Bacteria employ two component systems (TCSs) to sense and adapt to environmental changes. Each system consists of a sensor histidine kinase (HK) and a response regulator (RR). The HK, often membrane-bound, detects external cues such as changes in osmolarity, pH, or nutrient availability (Stock et al., 2000). Most HKs form homodimers in which each subunit phosphorylates the other in a trans-autophosphorylation reaction. Upon stimulation, it undergoes ATP-dependent autophosphorylation on a conserved histidine residue and transfers the phosphoryl group to an aspartate residue in the RR. Phosphorylation induces conformational changes in the RR that modulates its activity, often altering protein-protein or protein-DNA interactions to regulate downstream processes. Many HKs also act as phosphatases, dephosphorylating their cognate RR to reset signaling, enabling dynamic and reversible control of cellular adaptation (Gao & Stock, 2009; Papon & Stock, 2019).
The EnvZ/OmpR system is a well characterized TCS. In Escherichia coli, it regulates outer membrane porins (OmpF, OmpC) that respond to osmotic changes. EnvZ, a membrane-bound HK, senses osmolarity through its periplasmic domain and phosphorylates the RR OmpR. Phosphorylated OmpR binds DNA to control transcription of OmpF and OmpC, activating OmpF under low osmolarity and OmpC under high osmolarity. This graded response finetunes membrane permeability to maintain cellular homeostasis (Kenney & Anand, 2020).
In synthetic biology, TCSs are increasingly used as modular tools to engineer programmable signaling pathways. Their simple, reversible phosphotransfer mechanism allows precise control of signal processing and dynamic regulation of cellular responses. A key advantage is their complete orthogonality to mammalian signaling, as TCS rely on histidine-aspartate phosphorylation, a chemistry absent in eukaryotic cells. This prevents cross-reactivity with endogenous pathways and enables the design of insulated, tunable circuits for synthetic sensing and gene control application (Scheller et al., 2020; Schmidl et al., 2019).
Building on these benefits, we adapted the synthetic signaling architecture described by (Jones et al., 2022).The naturally bifunctional HK EnvZ was truncated to remove its transmembrane domain and further mutated to yield monomeric cytosolic variants with isolated enzymatic activities, one acting as a kinase and the other as a phosphatase. The kinase variant (EnvZK) comprises two Dimerization and Histidine Phosphotransfer domains (DHp; "A") and one Catalytic and ATP-binding domain (CA; "B"). In contrast, the phosphate (EnvZP) variant is formed by two monomers that are held together via a GCN4 coiled-coil, forcing the DHp domains into a fixed rotational state (Fig. 39)
Figure 39: Schematic of the engineered EnvZ/OmpR two-component system. The bacterial EnvZ/OmpR signaling pair was adapted for mammalian cells by creating separate kinase and phosphatase variants of EnvZ. The kinase (EnvZK) consists of two DHp and one CA domain (AAB variant), enabling phosphorylation of the response regulator OmpR. The phosphatase (EnvZP) is formed by two AB monomers, which are held together by GCN4 coiled-coil linkers (shown as grey bars) to maintain the active conformation. For clarity, DHp and CA domains are not explicitly depicted. When OmpR becomes phosphorylated, it binds to the OmpR-binding site (OmpR-bs) upstream of the reporter and induces Firefly luciferase expression in a phosphorylation-dependent manner.
Reporter activity was measured under varying kinase and phosphatase conditions using 30 ng of OmpR. In the absence of both enzymes, a basal signal was detected, indicating background activity. Expression of the EnvZ kinase progressively increased reporter output, with a significant elevation already observed at 20 ng compared to the enzyme-free condition, and 40 ng resulting in maximal activation. Co-expression of 20 ng kinase with 20 ng of the phosphatase variant barely reduced reporter activity compared to the 20 ng kinase condition, suggesting that the phosphatase exhibits limited dephosphorylating efficiency under these conditions. EnvZP alone caused only a minor, non-significant decrease relative to the enzyme-free control (Fig. 40). Together, these results demonstrate that the cytosolic kinase efficiently phosphorylates and activates OmpR, while the phosphatase counteracts this effect only weakly and does not substantially affect basal reporter levels.
Figure 40: Quantification of OmpR-dependent reporter activation by cytosolic EnvZ variants. HEK293T cells were transfected with 30 ng of OmpR together with the indicated amounts of kinase (EnvZK) and phosphatase (EnvZP) variants. The reporter plasmid contains an OmpR-binding site upstream of a Firefly luciferase gene, enabling transcriptional activation upon OmpR phosphorylation. Reporter activity was measured 48 hours post transfection using a Dual Luciferase assay, with Firefly luminescence normalized to constitutively expressed Renilla luciferase. Data are shown as mean ± SEM (n = 3 technical replicates). Statistical significance was determined using one-way ANOVA with Tukey's multiple comparison test (***p < 0.001, ****p < 0.0001, ns = not significant).
This background likely originates from unphosphorylated OmpR binding to the promoter, as has been reported for the native EnvZ/OmpR system. Although phosphorylation enhances OmpR dimerization and DNA affinity, several studies have shown that unphosphorylated OmpR can still associate with target promoters and weakly activate transcription (Kenney & Anand, 2020). This agrees with observations from Dr. Jones, who confirmed that OmpR can bind DNA even in its inactive form during one of our interviews with him.
Interestingly, co-expression EnvZP with OmpR resulted in a slightly reduced signal compared to OmpR alone. One possible explanation is that the phosphatase interacts with OmpR to form a non-productive complex, thereby sequestering OmpR away from the promoter. EnvZ and OmpR are known to form stable cytoplasmic complexes independently of OmpR phosphorylation (Qin et al., 2001), and such interactions could transiently limit promoter occupancy.
As demonstrated previously for ABL-bZipEE and PTPN1-bZipEE, a logical next step would be to couple
the
OmpR/EnvZ system to synthetic receptor platforms such as MESA or PAGER architectures, enabling
extracellular
control over kinase and phosphatase activity. Further optimization will be required to minimize
background
activity, improve dephosphorylation efficiency, and explore alternative coiled-coil pairs to fine-tune
enzyme interaction and signaling strength.These improvements will be particularly important for
potential
applications in cell therapy, where unintended basal activity or incomplete dephosphorylation could
lead to
undesired cellular responses.
A major advantage of this system is its full orthogonality to
mammalian
signaling pathways, allowing implementation in a broad range of human cell types, including immune
cells,
where SH2/CD3ζ-based systems may trigger unwanted cross-reactivity. Moreover, this module could be
combined
with the PHOENICS platform to create parallel, non-interfering signaling layers that operate on
distinct
substrates, providing greater flexibility and modularity in synthetic signaling circuit design.
- Acharya, A., Ruvinov, S. B., Gal, J., Moll, J. R., & Vinson, C. (2002). A heterodimerizing leucine zipper coiled coil system for examining the specificity of a position interactions: Amino acids I, V, L, N, A, and K. Biochemistry, 41(48), 14122-14131. https://doi.org/10.1021/bi020486r
- Barlowe, C., & Helenius, A. (2016). Cargo Capture and Bulk Flow in the Early Secretory Pathway. Annual Review of Cell and Developmental Biology, 32, 197–222. https://doi.org/10.1146/annurev-cellbio-111315-125016
- Beerli, R. R., Segal, D. J., Dreier, B., & Barbas, C. F. (1998). Toward controlling gene expression at will: Specific regulation of the erbB-2/HER-2 promoter by using polydactyl zinc finger proteins constructed from modular building blocks. Proceedings of the National Academy of Sciences of the United States of America, 95(25), 14628–14633. https://doi.org/10.1073/pnas.95.25.14628
- Benavides-López, S., Muñoz-Echeverri, L. M., Trujillo-Roldán, M. A., & Valdez-Cruz, N. A. (2025). The flow of proteins through the mammalian secretory pathway described by a phenomenological-based semi-physical model. Biochemical Engineering Journal, 220, 109759. https://doi.org/10.1016/j.bej.2025.109759
- Benman, W., Huang, Z., Iyengar, P., Wilde, D., Mumford, T. R., & Bugaj, L. J. (2025). A temperature-inducible protein module for control of mammalian cell fate. Nature Methods, 22(3), 539–549. https://doi.org/10.1038/s41592-024-02572-4
- Capin, J., Mayonove, P., DeVisch, A., Becher, A., Ngo, G., Courbet, A., Ragotte, R. J., Cohen Gonsaud, M., Espeut, J., & Bonnet, J. (2025). CF2H: a Cell-Free Two-Hybrid platform for rapid protein binder screening. bioRxiv. https://doi.org/10.1101/2025.07.16.665152
- Chassin, H., Müller, M., Tigges, M., Scheller, L., Lang, M., & Fussenegger, M. (2019). A modular degron library for synthetic circuits in mammalian cells. Nature Communications, 10(1), 2013. https://doi.org/10.1038/s41467-019-09974-5
- Chen, H., Ge, X., Li, C., Zeng, J., & Wang, X. (2024). Structure and assembly of the human IL-12 signaling complex. Structure, 32(10), 1640–1651.e5. https://doi.org/10.1016/j.str.2024.07.010
- Cohen, P. (2000). The regulation of protein function by multisite phosphorylation – a 25 year update. Trends in Biochemical Sciences, 25(12), 596–601. https://doi.org/10.1016/S0968-0004(00)01712-6
- Daringer, N. M., Dudek, R. M., Schwarz, K. A., & Leonard, J. N. (2014). Modular Extracellular Sensor Architecture for Engineering Mammalian Cell-based Devices. ACS Synthetic Biology, 3(12), 892–902. https://doi.org/10.1021/sb400128g
- Dolberg, T. B., Meger, A. T., Boucher, J. D., Corcoran, W. K., Schauer, E. E., Prybutok, A. N., Raman, S., & Leonard, J. N. (2021). Computation-guided optimization of split protein systems. Nature Chemical Biology, 17(5), 531–539. https://doi.org/10.1038/s41589-020-00729-8
- Engstrom, M. D., & Pfleger, B. F. (2017). Transcription control engineering and applications in synthetic biology. Synthetic and Systems Biotechnology, 2(3), 176–191. https://doi.org/10.1016/j.synbio.2017.09.003
- Farahani, P. E., Yang, X., Mesev, E. V., Fomby, K. A., Brumbaugh-Reed, E. H., Bashor, C. J., Nelson, C. M., & Toettcher, J. E. (2023). pYtags enable spatiotemporal measurements of receptor tyrosine kinase signaling in living cells. eLife, 12, e82863. https://doi.org/10.7554/eLife.82863
- Gao, R., & Stock, A. M. (2009). Biological insights from structures of two-component proteins. Annual Review of Microbiology, 63, 133–154. https://doi.org/10.1146/annurev.micro.091208.073214
- Hatada, M. H., Lu, X., Laird, E. R., Green, J., Morgenstern, J. P., Lou, M., Marr, C. S., Phillips, T. B., Ram, M. K., & Theriault, K. (1995). Molecular basis for interaction of the protein tyrosine kinase ZAP-70 with the T-cell receptor. Nature, 377(6544), 32–38. https://doi.org/10.1038/377032a0
- Hellauer, K., Rochon, M. H., & Turcotte, B. (1996). A novel DNA binding motif for yeast zinc cluster proteins: The Leu3p and Pdr3p transcriptional activators recognize everted repeats. Molecular and Cellular Biology, 16(11), 6096. https://doi.org/10.1128/MCB.16.11.6096
- Hoffman, G. R., Rahl, P. B., Collins, R. N., & Cerione, R. A. (2003). Conserved Structural Motifs in Intracellular Trafficking Pathways: Structure of the γCOP Appendage Domain. Molecular Cell, 12(3), 615–625. https://doi.org/10.1016/j.molcel.2003.08.002
- Hu, J., Lei, Y., Wong, W.-K., Liu, S., Lee, K.-C., He, X., You, W., Zhou, R., Guo, J.-T., Chen, X., Peng, X., Sun, H., Huang, H., Zhao, H., & Feng, B. (2014). Direct activation of human and mouse Oct4 genes using engineered TALE and Cas9 transcription factors. Nucleic Acids Research, 42(7), 4375–4390. https://doi.org/10.1093/nar/gku109
- Jones, R. D., Qian, Y., Ilia, K., Wang, B., Laub, M. T., Del Vecchio, D., & Weiss, R. (2022). Robust and tunable signal processing in mammalian cells via engineered covalent modification cycles. Nature Communications, 13(1), 1720. https://doi.org/10.1038/s41467-022-29338-w
- Kalogriopoulos, N. A., Tei, R., Yan, Y., Klein, P. M., Ravalin, M., Cai, B., Soltesz, I., Li, Y., & Ting, A. Y. (2025). Synthetic GPCRs for programmable sensing and control of cell behaviour. Nature, 637(8044), 230–239. https://doi.org/10.1038/s41586-024-08282-3
- Kasembeli, M. M., Rodas, J., & Tweardy, D. J. (2025). Update on Structure and Function of SH2 Domains: Mechanisms and Emerging Targeting Strategies. International Journal of Molecular Sciences, 26(18), 9060. https://doi.org/10.3390/ijms26189060
- Kenney, L. J., & Anand, G. S. (2020). EnvZ/OmpR Two-Component Signaling: An Archetype System That Can Function Noncanonically. EcoSal Plus, 9(1), ESP-0001–2019. https://doi.org/10.1128/ecosalplus.ESP-0001-2019
- Lim, W. A., & Pawson, T. (2010). Phosphotyrosine signaling: Evolving a new cellular communication system. Cell, 142(5), 661–667. https://doi.org/10.1016/j.cell.2010.08.023
- Mittler, G., Stühler, T., Santolin, L., Uhlmann, T., Kremmer, E., Lottspeich, F., Berti, L., & Meisterernst, M. (2003). A novel docking site on Mediator is critical for activation by VP16 in mammalian cells. The EMBO Journal, 22(24), 6494–6504. https://doi.org/10.1093/emboj/cdg619
- Moresco, E. M., & Koleske, A. J. (2003). Regulation of neuronal morphogenesis and synaptic function by Abl family kinases. Current Opinion in Neurobiology, 13(5), 535–544. https://doi.org/10.1016/j.conb.2003.08.002
- Norton, K.-A., Popel, A. S., & Pandey, N. B. (2015). Heterogeneity of chemokine cell-surface receptor expression in triple-negative breast cancer. American Journal of Cancer Research, 5(4), 1295–1307.
- Papon, N., & Stock, A. M. (2019). Two-component systems. Current Biology, 29(15), R724–R725. https://doi.org/10.1016/j.cub.2019.06.010
- Pawson, T. (2004). Specificity in signal transduction: From phosphotyrosine–SH2 domain interactions to complex cellular systems. Cell, 116(2), 191–203. https://doi.org/10.1016/S0092-8674(03)01077-8
- Pawson, T., & Scott, J. D. (2005). Protein phosphorylation in signaling – 50 years and counting. Trends in Biochemical Sciences, 30(6), 286–290. https://doi.org/10.1016/j.tibs.2005.04.013
- Qin, L., Yoshida, T., & Inouye, M. (2001). The critical role of DNA in the equilibrium between OmpR and phosphorylated OmpR mediated by EnvZ in Escherichia coli. Proceedings of the National Academy of Sciences of the United States of America, 98(3), 908–913. https://doi.org/10.1073/pnas.031383098
- Renna, P., Ripoli, C., Dagliyan, O., Pastore, F., Rinaudo, M., Re, A., Paciello, F., & Grassi, C. (2022). Engineering a switchable single-chain TEV protease to control protein maturation in living neurons. Bioengineering & Translational Medicine, 7(2), e10292. https://doi.org/10.1002/btm2.10292
- Román, E., Coman, I., Prieto, D., Alonso-Monge, R., & Pla, J. (2019). Implementation of a CRISPR-based system for gene regulation in Candida albicans. mSphere, 4(1), e00001-19. https://doi.org/10.1128/mSphere.00001-19
- Scheller, L., Schmollack, M., Bertschi, A., Mansouri, M., Saxena, P., & Fussenegger, M. (2020). Phosphoregulated orthogonal signal transduction in mammalian cells. Nature Communications, 11(1), 3085. https://doi.org/10.1038/s41467-020-16895-1
- Schmidl, S. R., Ekness, F., Sofjan, K., Daeffler, K. N.-M., Brink, K. R., Landry, B. P., Gerhardt, K. P., Dyulgyarov, N., Sheth, R. U., & Tabor, J. J. (2019). Rewiring bacterial two-component systems by modular DNA-binding domain swapping. Nature Chemical Biology, 15(7), 690–698. https://doi.org/10.1038/s41589-019-0286-6
- Shah, K., Al-Haidari, A., Sun, J., & Kazi, J. U. (2021). T cell receptor (TCR) signaling in health and disease. Signal Transduction and Targeted Therapy, 6(1), 412. https://doi.org/10.1038/s41392-021-00823-w
- Stock, A. M., Robinson, V. L., & Goudreau, P. N. (2000). Two-component signal transduction. Annual Review of Biochemistry, 69, 183–215. https://doi.org/10.1146/annurev.biochem.69.1.183
- Szczesna-Skorupa, E., & Kemper, B. (2011). The signal-anchor sequence of CYP2C1 inserts into the membrane as a hairpin structure. Biochemical and Biophysical Research Communications, 415(2), 405–409. https://doi.org/10.1016/j.bbrc.2011.10.082
- Tonks, N. K. (2006). Protein tyrosine phosphatases: From genes, to function, to disease. Nature Reviews Molecular Cell Biology, 7(11), 833–846. https://doi.org/10.1038/nrm2039
- Vardy, E., Robinson, J. E., Li, C., Olsen, R. H. J., DiBerto, J. F., Giguere, P. M., Sassano, F. M., Huang, X.-P., Zhu, H., Urban, D. J., White, K. L., Rittiner, J. E., Crowley, N. A., Pleil, K. E., Mazzone, C. M., Mosier, P. D., Song, J., Kash, T. L., Malanga, C. J., … Roth, B. L. (2015). A new DREADD facilitates the multiplexed chemogenetic interrogation of behavior. Neuron, 86(4), 936–946. https://doi.org/10.1016/j.neuron.2015.03.065
- Vlahos, A. E., Kang, J., Aldrete, C. A., Zhu, R., Chong, L. S., Elowitz, M. B., & Gao, X. J. (2022). Protease-controlled secretion and display of intercellular signals. Nature Communications, 13(1), 912. https://doi.org/10.1038/s41467-022-28623-y
- Wolf, B., Shehu, P., Brenker, L., Bachmann, A. von, Kroell, A.-S., Southern, N., Holderbach, S., Eigenmann, J., Aschenbrenner, S., Mathony, J., & Niopek, D. (2024). Rational engineering of allosteric protein switches by in silico prediction of domain insertion sites. bioRxiv, 2024.12.04.626757. https://doi.org/10.1101/2024.12.04.626757
- Yahara, T., Koga, T., Yoshida, S., Nakagawa, S., Deguchi, H., & Shirouzu, K. (2003). Relationship between microvessel density and thermographic hot areas in breast cancer. Surgery Today, 33(4), 243–248. https://doi.org/10.1007/s005950300055
- Yang, X., Rocks, J. W., Jiang, K., Walters, A. J., Rai, K., Liu, J., Nguyen, J., Olson, S. D., Mehta, P., Collins, J. J., Daringer, N. M., & Bashor, C. J. (2025). Engineering synthetic phosphorylation signaling networks in human cells. Science, 387(6729), 74–81. https://doi.org/10.1126/science.adm8485
- Zhang, Y., Wolf-Yadlin, A., Ross, P. L., Pappin, D. J., Rush, J., Lauffenburger, D. A., & White, F. M. (2005). Time-resolved mass spectrometry of tyrosine phosphorylation sites in the epidermal growth factor receptor signaling network reveals dynamic modules. Molecular & Cellular Proteomics, 4(9), 1240–1250. https://doi.org/10.1074/mcp.M500089-MCP200
- Zhao, Z., Zhang, J., Yin, L., Yang, J., Zheng, Y., Zhang, M., Ni, B., & Wang, H. (2020). Upregulated GDF-15 expression facilitates pancreatic ductal adenocarcinoma progression through orphan receptor GFRAL. Aging, 12(22), 22564–22581. https://doi.org/10.18632/aging.103830