Network


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

Hotspot


Dive into the research topics where Audrey Lee-Gosselin is active.

Publication


Featured researches published by Audrey Lee-Gosselin.


Nature Chemical Biology | 2017

Tunable thermal bioswitches for in vivo control of microbial therapeutics

Dan I. Piraner; Mohamad Abedi; Brittany Moser; Audrey Lee-Gosselin; Mikhail G. Shapiro

Temperature is a unique input signal that could be used by engineered microbial therapeutics to sense and respond to host conditions or spatially targeted external triggers such as focused ultrasound. To enable these possibilities, we present two families of tunable, orthogonal, temperature-dependent transcriptional repressors providing switch-like control of bacterial gene expression at thresholds spanning the biomedically relevant range of 32-46 °C. We integrate these molecular bioswitches into thermal logic circuits and demonstrate their utility in three in vivo microbial therapy scenarios, including spatially precise activation using focused ultrasound, modulation of activity in response to a host fever, and self-destruction after fecal elimination to prevent environmental escape. This technology provides a critical capability for coupling endogenous or applied thermal signals to cellular function in basic research, biomedical and industrial applications.


Nature | 2018

Acoustic reporter genes for noninvasive imaging of microorganisms in mammalian hosts

Raymond W. Bourdeau; Audrey Lee-Gosselin; Anupama Lakshmanan; Arash Farhadi; Sripriya Ravindra Kumar; Suchita P. Nety; Mikhail G. Shapiro

The mammalian microbiome has many important roles in health and disease, and genetic engineering is enabling the development of microbial therapeutics and diagnostics. A key determinant of the activity of both natural and engineered microorganisms in vivo is their location within the host organism. However, existing methods for imaging cellular location and function, primarily based on optical reporter genes, have limited deep tissue performance owing to light scattering or require radioactive tracers. Here we introduce acoustic reporter genes, which are genetic constructs that allow bacterial gene expression to be visualized in vivo using ultrasound, a widely available inexpensive technique with deep tissue penetration and high spatial resolution. These constructs are based on gas vesicles, a unique class of gas-filled protein nanostructures that are expressed primarily in water-dwelling photosynthetic organisms as a means to regulate buoyancy. Heterologous expression of engineered gene clusters encoding gas vesicles allows Escherichia coli and Salmonella typhimurium to be imaged noninvasively at volumetric densities below 0.01% with a resolution of less than 100 μm. We demonstrate the imaging of engineered cells in vivo in proof-of-concept models of gastrointestinal and tumour localization, and develop acoustically distinct reporters that enable multiplexed imaging of cellular populations. This technology equips microbial cells with a means to be visualized deep inside mammalian hosts, facilitating the study of the mammalian microbiome and the development of diagnostic and therapeutic cellular agents.


Applied Physics Letters | 2017

Nonlinear ultrasound imaging of nanoscale acoustic biomolecules

David Maresca; Anupama Lakshmanan; Audrey Lee-Gosselin; Johan M. Melis; Yu-Li Ni; Raymond W. Bourdeau; Dennis M. Kochmann; Mikhail G. Shapiro

Ultrasound imaging is widely used to probe the mechanical structure of tissues and visualize blood flow. However, the ability of ultrasound to observe specific molecular and cellular signals is limited. Recently, a unique class of gas-filled protein nanostructures called gas vesicles (GVs) was introduced as nanoscale (∼250 nm) contrast agents for ultrasound, accompanied by the possibilities of genetic engineering, imaging of targets outside the vasculature and monitoring of cellular signals such as gene expression. These possibilities would be aided by methods to discriminate GV-generated ultrasound signals from anatomical background. Here, we show that the nonlinear response of engineered GVs to acoustic pressure enables selective imaging of these nanostructures using a tailored amplitude modulation strategy. Finite element modeling predicted a strongly nonlinear mechanical deformation and acoustic response to ultrasound in engineered GVs. This response was confirmed with ultrasound measurements in the range of 10 to 25 MHz. An amplitude modulation pulse sequence based on this nonlinear response allows engineered GVs to be distinguished from linear scatterers and other GV types with a contrast ratio greater than 11.5 dB. We demonstrate the effectiveness of this nonlinear imaging strategy in vitro, in cellulo, and in vivo.


Nature Protocols | 2017

Preparation of biogenic gas vesicle nanostructures for use as contrast agents for ultrasound and MRI

Anupama Lakshmanan; George J. Lu; Arash Farhadi; Suchita P. Nety; Martin Kunth; Audrey Lee-Gosselin; David Maresca; Raymond W. Bourdeau; Melissa Yin; Judy Yan; Christopher Witte; Dina Malounda; F. Stuart Foster; Leif Schröder; Mikhail G. Shapiro

Gas vesicles (GVs) are a unique class of gas-filled protein nanostructures that are detectable at subnanomolar concentrations and whose physical properties allow them to serve as highly sensitive imaging agents for ultrasound and MRI. Here we provide a protocol for isolating GVs from native and heterologous host organisms, functionalizing these nanostructures with moieties for targeting and fluorescence, characterizing their biophysical properties and imaging them using ultrasound and MRI. GVs can be isolated from natural cyanobacterial and haloarchaeal host organisms or from Escherichia coli expressing a heterologous GV gene cluster and purified using buoyancy-assisted techniques. They can then be modified by replacing surface-bound proteins with engineered, heterologously expressed variants or through chemical conjugation, resulting in altered mechanical, surface and targeting properties. Pressurized absorbance spectroscopy is used to characterize their mechanical properties, whereas dynamic light scattering (DLS)and transmission electron microscopy (TEM) are used to determine nanoparticle size and morphology, respectively. GVs can then be imaged with ultrasound in vitro and in vivo using pulse sequences optimized for their detection versus background. They can also be imaged with hyperpolarized xenon MRI using chemical exchange saturation transfer between GV-bound and dissolved xenon—a technique currently implemented in vitro. Taking 3–8 d to prepare, these genetically encodable nanostructures enable multimodal, noninvasive biological imaging with high sensitivity and potential for molecular targeting.


Nature Communications | 2018

Mapping the microscale origins of magnetic resonance image contrast with subcellular diamond magnetometry

Hunter C. Davis; Pradeep Ramesh; Aadyot Bhatnagar; Audrey Lee-Gosselin; John F. Barry; David R. Glenn; Ronald L. Walsworth; Mikhail G. Shapiro

Magnetic resonance imaging (MRI) is a widely used biomedical imaging modality that derives much of its contrast from microscale magnetic field patterns in tissues. However, the connection between these patterns and the appearance of macroscale MR images has not been the subject of direct experimental study due to a lack of methods to map microscopic fields in biological samples. Here, we optically probe magnetic fields in mammalian cells and tissues with submicron resolution and nanotesla sensitivity using nitrogen-vacancy diamond magnetometry, and combine these measurements with simulations of nuclear spin precession to predict the corresponding MRI contrast. We demonstrate the utility of this technology in an in vitro model of macrophage iron uptake and histological samples from a mouse model of hepatic iron overload. In addition, we follow magnetic particle endocytosis in live cells. This approach bridges a fundamental gap between an MRI voxel and its microscopic constituents.Magnetic resonance imaging derives its contrast from local magnetic fields, however the connection between these fields and macroscale contrast has not been established through direct experiments. Here, Davis et al. use diamond magnetometry to map local magnetic fields within mammalian cells with sub-micron resolution and predict macroscale contrast.


bioRxiv | 2018

Acoustically Targeted Chemogenetics for Noninvasive Control of Neural Circuits

Jerzy O. Szablowski; Brian Lue; Audrey Lee-Gosselin; Dina Malounda; Mikhail G. Shapiro

Neurological and psychiatric diseases often involve the dysfunction of specific neural circuits in particular regions of the brain. Existing treatments, including drugs and implantable brain stimulators, aim to modulate the activity of these circuits, but are typically not cell type-specific, lack spatial targeting or require invasive procedures. Here, we introduce an approach to modulating neural circuits noninvasively with spatial, cell-type and temporal specificity. This approach, called acoustically targeted chemogenetics, or ATAC, uses transient ultrasonic opening of the blood brain barrier to transduce neurons at specific locations in the brain with virally-encoded engineered G-protein-coupled receptors, which subsequently respond to systemically administered bio-inert compounds to activate or inhibit the activity of these neurons. We demonstrate this concept in mice by using ATAC to noninvasively modify and subsequently activate or inhibit excitatory neurons within the hippocampus, showing that this enables pharmacological control of memory formation. This technology allows a brief, noninvasive procedure to make one or more specific brain regions capable of being selectively modulated using orally bioavailable compounds, thereby overcoming some of the key limitations of conventional brain therapies.


Nature Materials | 2018

Acoustically modulated magnetic resonance imaging of gas-filled protein nanostructures

George J. Lu; Arash Farhadi; Jerzy O. Szablowski; Audrey Lee-Gosselin; Samuel Barnes; Anupama Lakshmanan; Raymond W. Bourdeau; Mikhail G. Shapiro

Non-invasive biological imaging requires materials capable of interacting with deeply penetrant forms of energy such as magnetic fields and sound waves. Here, we show that gas vesicles (GVs), a unique class of gas-filled protein nanostructures with differential magnetic susceptibility relative to water, can produce robust contrast in magnetic resonance imaging (MRI) at sub-nanomolar concentrations, and that this contrast can be inactivated with ultrasound in situ to enable background-free imaging. We demonstrate this capability in vitro, in cells expressing these nanostructures as genetically encoded reporters, and in three model in vivo scenarios. Genetic variants of GVs, differing in their magnetic or mechanical phenotypes, allow multiplexed imaging using parametric MRI and differential acoustic sensitivity. Additionally, clustering-induced changes in MRI contrast enable the design of dynamic molecular sensors. By coupling the complementary physics of MRI and ultrasound, this nanomaterial gives rise to a distinct modality for molecular imaging with unique advantages and capabilities.Gas-filled vesicles derived from photosynthetic microbes are shown to elicit magnetic resonance imaging contrast in vitro and in vivo with the potential for acoustically modulated multiplexing and molecular sensing.


Nature Biomedical Engineering | 2018

Acoustically targeted chemogenetics for the non-invasive control of neural circuits

Jerzy O. Szablowski; Audrey Lee-Gosselin; Brian Lue; Dina Malounda; Mikhail G. Shapiro

Neurological and psychiatric disorders are often characterized by dysfunctional neural circuits in specific regions of the brain. Existing treatment strategies, including the use of drugs and implantable brain stimulators, aim to modulate the activity of these circuits. However, they are not cell-type-specific, lack spatial targeting or require invasive procedures. Here, we report a cell-type-specific and non-invasive approach based on acoustically targeted chemogenetics that enables the modulation of neural circuits with spatiotemporal specificity. The approach uses ultrasound waves to transiently open the blood–brain barrier and transduce neurons at specific locations in the brain with virally encoded engineered G-protein-coupled receptors. The engineered neurons subsequently respond to systemically administered designer compounds to activate or inhibit their activity. In a mouse model of memory formation, the approach can modify and subsequently activate or inhibit excitatory neurons within the hippocampus, with selective control over individual brain regions. This technology overcomes some of the key limitations associated with conventional brain therapies.The combination of focused ultrasound and virally encoded receptors engineered to be activated by a designer drug enables, on intravenous administration of the drug, the non-invasive activation or inhibition of brain regions in mice, with cell-type and spatiotemporal specificity.


Nature Biomedical Engineering | 2017

Localization of Microscale Devices In Vivo using Addressable Transmitters Operated as Magnetic Spins

Manuel Monge; Audrey Lee-Gosselin; Mikhail G. Shapiro; Azita Emami


arXiv: Medical Physics | 2016

Mapping the Microscale Origins of MRI Contrast with Subcellular NV Diamond Magnetometry

Hunter C. Davis; Pradeep Ramesh; Aadyot Bhatnagar; Audrey Lee-Gosselin; John F. Barry; David R. Glenn; Ronald L. Walsworth; Mikhail G. Shapiro

Collaboration


Dive into the Audrey Lee-Gosselin's collaboration.

Top Co-Authors

Avatar

Mikhail G. Shapiro

California Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

Anupama Lakshmanan

California Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

Raymond W. Bourdeau

California Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

Arash Farhadi

California Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

Dina Malounda

California Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

Hunter C. Davis

California Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

Jerzy O. Szablowski

California Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

Pradeep Ramesh

California Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

Aadyot Bhatnagar

California Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

Brian Lue

California Institute of Technology

View shared research outputs
Researchain Logo
Decentralizing Knowledge