Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Evan Mills is active.

Publication


Featured researches published by Evan Mills.


ACS Synthetic Biology | 2012

Engineering a Photoactivated Caspase-7 for Rapid Induction of Apoptosis

Evan Mills; Xi Chen; Elizabeth Pham; Stanley Wong; Kevin Truong

Apoptosis is a cell death program involved in the development of multicellular organisms, immunity, and pathologies ranging from cancer to HIV/AIDS. We present an engineered protein that causes rapid apoptosis of targeted cells in monolayer culture after stimulation with blue light. Cells transfected with the protein switch L57V, a tandem fusion of the light-sensing LOV2 domain and the apoptosis-executing domain from caspase-7, rapidly undergo apoptosis within 60 min after light stimulation. Constant illumination of under 5 min or oscillating with 1 min exposure had no effect, suggesting that cells have natural tolerance to a short duration of caspase-7 activity. Furthermore, the overexpression of Bcl-2 prevented L57V-mediated apoptosis, suggesting that although caspase-7 activation is sufficient to start apoptosis, it requires mitochondrial contribution to fully commit.


Chemistry & Biology | 2011

Ca2+-Mediated Synthetic Biosystems Offer Protein Design Versatility, Signal Specificity, and Pathway Rewiring

Evan Mills; Kevin Truong

Synthetic biosystems have been engineered that enable control of metazoan cell morphology, migration, and death. These systems possess signal specificity, but lack flexibility of input signal. To exploit the potential of Ca(2+) signaling, we designed RhoA chimeras for reversible, Ca(2+)-dependent control over RhoA morphology and migration. First, we inserted a calmodulin-binding peptide into a RhoA loop that activates or deactivates RhoA in response to Ca(2+) signals depending on the chosen peptide. Second, we localized the Ca(2+)-activated RhoA chimera to the plasma membrane, where it responded specifically to local Ca(2+) signals. Third, input control of RhoA morphology was rewired by coexpressing the Ca(2+)-activated RhoA chimera with Ca(2+)-transport proteins using acetylcholine, store-operated Ca(2+) entry, and blue light. Engineering synthetic biological systems with input versatility and tunable spatiotemporal responses motivates further application of Ca(2+) signaling in this field.


Cell Calcium | 2012

Split-intein mediated re-assembly of genetically encoded Ca2+ indicators

Stanley Wong; Ippei Kotera; Evan Mills; Hiroshi Suzuki; Kevin Truong

While genetically encoded Ca(2+) indicators (GECIs) allow Ca(2+) imaging in model organisms, the gene expression is often under the control of a single promoter that may drive expression beyond, the cell types of interest. To enable more cell-type specific targeting, GECIs can be brought under the, control of the intersecting expression from two promoters. Here, we present the splitting and, reassembly of two representative GECIs (TN-XL and GCaMP2) mediated by the split intein from Nostoc, punctiforme (NpuDnaE). While the split TN-XL biosensor offered ratiometric Ca(2+) imaging, it had a, diminished Ca(2+) response relative to the native TN-XL biosensor. In contrast, the split GCaMP2, biosensor retained similar Ca(2+) response to the native GCaMP2. The split GCaMP2 biosensor was, further targeted to the pharyngeal muscles of Caenorhabditis elegans where Ca(2+) signals from feeding C. elegans, were imaged. Thus, we envision that increased cell-type targetability of GECIs is feasible with two, complementary promoters.


Cell Calcium | 2010

Structure based design of a Ca2+-sensitive RhoA protein that controls cell morphology

Evan Mills; Elizabeth Pham; Kevin Truong

The Rho proteins are important regulators of cell morphology, and the prototypical protein RhoA is known to regulate contraction, blebbing and bleb retraction. We have identified and experimentally confirmed that RhoA has a binding site for calmodulin, a ubiquitous transducer of the Ca(2+) second messenger. Using structural modeling, a fusion protein was designed wherein RhoA activity was controlled by Ca(2+) via calmodulin. Living cells transfected with this synthetic protein underwent Ca(2+) sensitive and calmodulin-dependent bleb retraction within minutes. Further, the modularity of Ca(2+) signaling was exploited to induce bleb retraction in response to blue light (using channelrhodopsin-2) or exogenous chemicals (with acetylcholine receptor), showing input signal versatility. The widespread use of Ca(2+) signaling in nature suggests that fully exploring its signaling potential may allow powerful applications to other synthetic biological systems.


ACS Synthetic Biology | 2013

Programming membrane fusion and subsequent apoptosis into mammalian cells.

Seema Nagaraj; Evan Mills; Stanley Wong; Kevin Truong

By the delivery of specific natural or engineered proteins, mammalian cells can be programmed to perform increasingly sophisticated and useful functions. Here, we introduce a set of proteins that has potential value in cell-based therapies by programming a cell to target tumor cells. First, the delivery of VSV-G (vesicular stomatitis virus glycoprotein) allowed the cell to undergo membrane fusion with adjacent cells to form syncytia (i.e., a multinucleated cell) in conditions of low pH typically occurring at a tumor site. The formation of syncytia caused the clustering of nuclei along with an integration of the microtubule network and ER. Interestingly, the formation of syncytia between cells that are dynamically blebbing, a mode of migration preferred during tumor metastasis, resulted in the loss of these morphology changes. Lastly, the codelivery of VSV-G with L57R (an engineered photoactivated caspase-7) allowed cells to undergo low pH-dependent membrane fusion followed by blue light-dependent apoptosis. In cell-based therapies, the clearance of syncytia between tumor cells might further trigger an immune response against the tumor.


ACS Synthetic Biology | 2012

Engineered networks of synthetic and natural proteins to control cell migration.

Evan Mills; Elizabeth Pham; Seema Nagaraj; Kevin Truong

Mammalian cells reprogrammed with engineered transgenes have the potential to be useful therapeutic platforms because they can support large genetic networks, can be taken from a host or patient, and perform useful functions such as migration and secretion. Successful engineering of mammalian cells will require the development of modules that can perform well-defined, reliable functions, such as directed cell migration toward a chemical or physical signal. One inherently modular cellular pathway is the Ca(2+) signaling pathway: protein modules that mobilize and respond to Ca(2+) are combined across cell types to create complexity. We have designed a chimera of Rac1, a GTPase that controls cell morphology and migration, and calmodulin (CaM), a Ca(2+)-responsive protein, to control cell migration. The Rac1-CaM chimera (named RACer) controlled lamellipodia growth in response to Ca(2+). RACer was combined with LOVS1K (a previously engineered light-sensitive Ca(2+)-mobilizing module) and cytokine receptors to create protein networks where blue light and growth factors regulated cell morphology and, thereby, cell migration. To show the generalizability of our design, we created a Cdc42-CaM chimera that controls filopodia growth in response to Ca(2+). The insights that have been gained into Ca(2+) signaling and cell migration will allow future work to combine engineered protein systems to enable reprogrammed cell sensing of relevant therapeutic targets in vivo.


Cell Calcium | 2013

Analysis and regulation of amoeboid-like cell motility using synthetic Ca2+-sensitive proteins

Evan Mills; Kevin Truong

Several recent reports have demonstrated how engineered proteins can control cell motility, an important functional module for ultimately programming cells as therapeutics. We have reported two engineered proteins that regulate the blebbing cell morphology using chimeras of RhoA, a protein that regulates cytoskeletal tension. Here, we show that engineered switching of blebbing can be used to regulate cell motility. First, the analysis of morphology and motility characteristics showed that blebbing cells wobbled, or shifted, faster and less linearly than cells with a wild type morphology. Second, activating engineered protein switches that regulate cell morphology led to predictable changes in motility characteristics. Last, exogenous stimuli such as blue light, acetylcholine and VEGF-A were used to show that groups of proteins could cooperatively increase cell motility in vitro. This work demonstrates that control of RhoA can program the motility patterns of living cells and has implications in studying the relationship between cell morphology and motility.


Protein Engineering Design & Selection | 2013

Simultaneous assembly of two target proteins using split inteins for live cell imaging

Stanley Wong; Evan Mills; Kevin Truong

Inteins are protein elements that covalently reassemble proteins from two precursor fragments in a process known as protein splicing. They are commonly used to reassemble a single target protein by protein splicing, but a second target protein can potentially reassemble by intein dimerization. Here, we use the naturally occurring split DnaE intein from Nostoc punctiforme (NpuDnaE) to demonstrate the simultaneous assembly of two target proteins in several examples studied with live cell imaging: yellow fluorescent protein (YFP) with monomeric red fluorescent protein (mRFP), dominant positive mutant of RhoA GTPase with YFP and GCaMP2 Ca(2+) indicator with mRFP. These examples showed the versatility of the strategy along with some interesting attributes: first, the two target proteins are in equal stoichiometry; second, the extent of protein splicing can be reported by a fluorescent protein. In particular, the split GCaMP2 with mRFP could find applications in tissue-specific Ca(2+) imaging in transgenic organisms, where mRFP could control for motion-related intensity changes.


Cell Calcium | 2010

Engineering Ca2+/calmodulin-mediated modulation of protein translocation by overlapping binding and signaling peptide sequences

Evan Mills; Kevin Truong

Protein translocation is used by cells to regulate protein activity in time and space. Synthetic systems have studied the effect of second messengers and exogenous chemicals on translocation, and have used translocation-based sensors to monitor unrelated pathways such as caspase activity. We have created a synthetic Ca2+-inducible protein using calmodulin binding peptides that selectively reveal nuclear localization and export signals in low Ca2+ (0 microM) and high Ca2+ (10 microM) environments, respectively. Experiments in live cells showed that our construct translocates between the nucleolus and plasma membrane with time constants of approximately 2 h. Further, a single amino acid mutation (Cys20Ala) in our construct prevented translocation to the plasma membrane and instead targeted it the mitochondria as predicted by bioinformatic analysis. Lastly, we studied the effect of cell line, Ca2+ concentration, chemical inhibitors, and cell morphology on translocation and found these conditions affected the rate, extent and direction of translocation. Our work demonstrates the feasibility of engineering Ca2+/calmodulin-mediated modulation of protein translocation and suggests that more natural analogs may exist.


Protein Science | 2009

Rate and extent of protein localization is controlled by peptide‐binding domain association kinetics and morphology

Evan Mills; Kevin Truong

Protein localization is an important regulatory mechanism in many cell signaling pathways such as cytoskeletal organization and genetic regulation. The specific mechanism of protein localization determines the kinetics and morphological constraints of protein translocation, and thus affects the rate and extent of localization. To investigate the affect of localization kinetics and morphology on protein localization, we designed a protein localization system based on Ca2+‐calmodulin and Src homology 3 domain binding peptides that can translocate between specific localizations in response to a Ca2+ signal. We used a stochastic biomolecular simulator to predict that such a protein localization system will exhibit slower and less complete translocations when the association kinetics of a binding domain and peptide are reduced. As well, we predicted that increasing the diffusion resistance by manipulating the morphology of the system would similarly impair translocation speed and completeness. We then constructed a network of synthetic fusion proteins and showed that these predictions could be qualitatively confirmed in vitro. This work provides a basis for explaining the different characteristics (rate and extent) of protein transport and localization in cells as a consequence of the kinetics and morphology of the transport mechanism.

Collaboration


Dive into the Evan Mills's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Isaac T. S. Li

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Xi Chen

University of Toronto

View shared research outputs
Researchain Logo
Decentralizing Knowledge