1
|
Saldanha DJ, Rowat SJA, Stephenson H, Dorval Courchesne NM. Living Dual Heat- and pH-Responsive Textiles. ACS Synth Biol 2025; 14:564-574. [PMID: 39818762 DOI: 10.1021/acssynbio.4c00808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2025]
Abstract
Smart textiles that integrate multiple environmental sensing capabilities are an emerging frontier in wearable technology. In this study, we developed dual pH- and temperature-responsive textiles by combining engineered bacterial systems with bacterially derived proteins. For temperature sensing, we characterized the properties of a heat sensitive promoter, Phs, in Escherichia coli (E. coli) using enhanced green fluorescent protein as a reporter. Our findings demonstrate that the Phs promoter drives elevated gene expression at temperatures between 37 and 43 °C, maintaining sustained activity for several hours. Moreover, we found that short heat shocks can significantly boost expression levels of the Phs promoter. We successfully integrated E. coli expressing Phs-EGFP cells onto textiles and confirmed their ability to retain heat-responsive behavior after integration. To achieve pH responsiveness, we utilized curli fibers, genetically engineered to incorporate a pH-sensitive fluorescent protein, pHuji. pH-sensing curli fibers are bacterial proteins that have a proven track record of creating stable bioresponsive textile coatings. By embedding Phs-EGFP-expressing bacteria within curli fiber coatings, we created a dual-responsive textile capable of differentiating between acidic and alkaline environments while simultaneously responding to thermal stimuli. These multifunctional textiles exhibited dual environmental response and sensing capabilities. This work establishes a proof-of-concept for creating smart living textiles with modular functionalities, paving the way toward advanced bioresponsive materials.
Collapse
Affiliation(s)
- Dalia Jane Saldanha
- Department of Chemical Engineering, McGill University, Montreal, Quebec H3A 0C5, Canada
| | | | - Henry Stephenson
- Department of Chemical Engineering, McGill University, Montreal, Quebec H3A 0C5, Canada
| | | |
Collapse
|
2
|
Huang J, Fussenegger M. Programming mammalian cell behaviors by physical cues. Trends Biotechnol 2025; 43:16-42. [PMID: 39179464 DOI: 10.1016/j.tibtech.2024.07.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 07/24/2024] [Accepted: 07/26/2024] [Indexed: 08/26/2024]
Abstract
In recent decades, the field of synthetic biology has witnessed remarkable progress, driving advances in both research and practical applications. One pivotal area of development involves the design of transgene switches capable of precisely regulating specified outputs and controlling cell behaviors in response to physical cues, which encompass light, magnetic fields, temperature, mechanical forces, ultrasound, and electricity. In this review, we delve into the cutting-edge progress made in the field of physically controlled protein expression in engineered mammalian cells, exploring the diverse genetic tools and synthetic strategies available for engineering targeting cells to sense these physical cues and generate the desired outputs accordingly. We discuss the precision and efficiency limitations inherent in these tools, while also highlighting their immense potential for therapeutic applications.
Collapse
Affiliation(s)
- Jinbo Huang
- Department of Biosystems Science and Engineering, ETH Zurich, Klingelbergstrasse 48, CH-4056 Basel, Switzerland
| | - Martin Fussenegger
- Department of Biosystems Science and Engineering, ETH Zurich, Klingelbergstrasse 48, CH-4056 Basel, Switzerland; Faculty of Science, University of Basel, Klingelbergstrasse 48, CH-4056 Basel, Switzerland.
| |
Collapse
|
3
|
Sistemich L, Ebbinghaus S. Heat application in live cell imaging. FEBS Open Bio 2024; 14:1940-1954. [PMID: 39489617 PMCID: PMC11609584 DOI: 10.1002/2211-5463.13912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 08/29/2024] [Accepted: 10/01/2024] [Indexed: 11/05/2024] Open
Abstract
Thermal heating of biological samples allows to reversibly manipulate cellular processes with high temporal and spatial resolution. Manifold heating techniques in combination with live-cell imaging were developed, commonly tailored to customized applications. They include Peltier elements and microfluidics for homogenous sample heating as well as infrared lasers and radiation absorption by nanostructures for spot heating. A prerequisite of all techniques is that the induced temperature changes are measured precisely which can be the main challenge considering subcellular structures or multicellular organisms as target regions. This article discusses heating and temperature sensing techniques for live-cell imaging, leading to future applications in cell biology.
Collapse
Affiliation(s)
- Linda Sistemich
- Chair of Biophysical ChemistryRuhr‐University BochumGermany
- Research Center Chemical Sciences and Sustainability, Research Alliance RuhrBochumGermany
| | - Simon Ebbinghaus
- Chair of Biophysical ChemistryRuhr‐University BochumGermany
- Research Center Chemical Sciences and Sustainability, Research Alliance RuhrBochumGermany
| |
Collapse
|
4
|
Riquelme-Guzmán C, Stout AJ, Kaplan DL, Flack JE. Unlocking the potential of cultivated meat through cell line engineering. iScience 2024; 27:110877. [PMID: 39351194 PMCID: PMC11440241 DOI: 10.1016/j.isci.2024.110877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/04/2024] Open
Abstract
Cultivated meat has the potential to revolutionize food production, but its progress is hindered by fundamental shortcomings of mammalian cells with respect to industrial-scale bioprocesses. Here, we discuss the essential role of cell line engineering in overcoming these limitations, highlighting the balance between the benefits of enhanced cellular traits and the associated regulatory and consumer acceptance challenges. We believe that careful selection of cell engineering strategies, including both genetic and non-genetic modifications, can address this trade-off and is essential to advancing the field.
Collapse
Affiliation(s)
- Camilo Riquelme-Guzmán
- Biomedical Engineering Department, Tufts University Center for Cellular Agriculture, Tufts University, Medford, MA, USA
| | - Andrew J Stout
- Biomedical Engineering Department, Tufts University Center for Cellular Agriculture, Tufts University, Medford, MA, USA
- Deco Labs, Inc., Boston, MA, USA
| | - David L Kaplan
- Biomedical Engineering Department, Tufts University Center for Cellular Agriculture, Tufts University, Medford, MA, USA
| | - Joshua E Flack
- Department of Biotechnology, Delft University of Technology, Delft, the Netherlands
| |
Collapse
|
5
|
Siebart JC, Chan CS, Yao X, Su FY, Kwong GA. In vivo gene delivery to immune cells. Curr Opin Biotechnol 2024; 88:103169. [PMID: 38972172 PMCID: PMC11316639 DOI: 10.1016/j.copbio.2024.103169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 11/16/2023] [Accepted: 06/14/2024] [Indexed: 07/09/2024]
Abstract
Immune cell therapies are an emerging class of living drugs that rely on the delivery of therapeutic transgenes to enhance, modulate, or restore cell function, such as those that encode for tumor-targeting receptors or replacement proteins. However, many cellular immunotherapies are autologous treatments that are limited by high manufacturing costs, typical vein-to-vein time of 3-4 weeks, and severe immune-related adverse effects. To address these issues, different classes of gene delivery vehicles are being developed to target specific immune cell subsets in vivo to address the limitations of ex vivo manufacturing, modulate therapeutic responses in situ, and reduce on- and off-target toxicity. The success of in vivo gene delivery to immune cells - which is being tested at the preclinical and clinical stages of development for the treatment of cancer, infectious diseases, and autoimmunity - is paramount for the democratization of cellular immunotherapies.
Collapse
Affiliation(s)
- Jamison C Siebart
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA
| | - Ching S Chan
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA
| | - Xinyi Yao
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA
| | - Fang-Yi Su
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA
| | - Gabriel A Kwong
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA; Winship Cancer Institute of Emory University, Atlanta, GA 30322, USA; Institute for Electronics and Nanotechnology, Georgia Institute of Technology, Atlanta, GA 30332, USA; Parker H. Petit Institute of Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332, USA; Integrated Cancer Research Center, Georgia Institute of Technology, Atlanta, GA 30332, USA; Georgia ImmunoEngineering Consortium, Emory University and Georgia Institute of Technology, Atlanta, GA 30332, USA.
| |
Collapse
|
6
|
Teixeira AP, Fussenegger M. Synthetic Gene Circuits for Regulation of Next-Generation Cell-Based Therapeutics. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2309088. [PMID: 38126677 PMCID: PMC10885662 DOI: 10.1002/advs.202309088] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Indexed: 12/23/2023]
Abstract
Arming human cells with synthetic gene circuits enables to expand their capacity to execute superior sensing and response actions, offering tremendous potential for innovative cellular therapeutics. This can be achieved by assembling components from an ever-expanding molecular toolkit, incorporating switches based on transcriptional, translational, or post-translational control mechanisms. This review provides examples from the three classes of switches, and discusses their advantages and limitations to regulate the activity of therapeutic cells in vivo. Genetic switches designed to recognize internal disease-associated signals often encode intricate actuation programs that orchestrate a reduction in the sensed signal, establishing a closed-loop architecture. Conversely, switches engineered to detect external molecular or physical cues operate in an open-loop fashion, switching on or off upon signal exposure. The integration of such synthetic gene circuits into the next generation of chimeric antigen receptor T-cells is already enabling precise calibration of immune responses in terms of magnitude and timing, thereby improving the potency and safety of therapeutic cells. Furthermore, pre-clinical engineered cells targeting other chronic diseases are gathering increasing attention, and this review discusses the path forward for achieving clinical success. With synthetic biology at the forefront, cellular therapeutics holds great promise for groundbreaking treatments.
Collapse
Affiliation(s)
- Ana P. Teixeira
- Department of Biosystems Science and EngineeringETH ZurichKlingelbergstrasse 48BaselCH‐4056Switzerland
| | - Martin Fussenegger
- Department of Biosystems Science and EngineeringETH ZurichKlingelbergstrasse 48BaselCH‐4056Switzerland
- Faculty of ScienceUniversity of BaselKlingelbergstrasse 48BaselCH‐4056Switzerland
| |
Collapse
|
7
|
Cox JR, Fox A, Lenahan C, Pivnik L, Manion M, Blazeck J. Engineering CREB-activated promoters for adenosine-induced gene expression. Biotechnol J 2024; 19:e2300446. [PMID: 38403442 PMCID: PMC10901447 DOI: 10.1002/biot.202300446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 12/25/2023] [Accepted: 01/02/2024] [Indexed: 02/27/2024]
Abstract
Accumulation of the ribonucleoside, adenosine (ADO), triggers a cAMP response element binding protein (CREB)-mediated signaling pathway to suppress the function of immune cells in tumors. Here, we describe a collection of CREB-activated promoters that allow for strong and tunable ADO-induced gene expression in human cells. By optimizing number of CREB transcription factor binding sites and altering the core promoter region of CREB-based hybrid promoters, we created synthetic constructs that drive gene expression to higher levels than strong, endogenous mammalian promoters in the presence of ADO. These synthetic promoters are induced up to 47-fold by ADO, with minimal expression in their "off" state. We further determine that our CREB-based promoters are activated by other compounds that act as signaling analogs, and that combinatorial addition of ADO and these compounds has a synergistic impact on gene expression. Surprisingly, we also detail how background ADO degradation caused by the common cell culture media additive, fetal bovine serum (FBS), confounds experiments designed to determine ADO dose-responsiveness. We show that only after long-term heat deactivation of FBS can our synthetic promoters enable gene expression induction at physiologically relevant levels of ADO. Finally, we demonstrate that the strength of a CREB-based promoter is enhanced by incorporating other transcription factor binding sites.
Collapse
Affiliation(s)
- John Robert Cox
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, Georgia, USA
| | - Andrea Fox
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, Georgia, USA
| | - Conor Lenahan
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, Georgia, USA
| | - Liza Pivnik
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, Georgia, USA
| | - Matthew Manion
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, Georgia, USA
| | - John Blazeck
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, Georgia, USA
| |
Collapse
|
8
|
Giordano Attianese GMP, Ash S, Irving M. Coengineering specificity, safety, and function into T cells for cancer immunotherapy. Immunol Rev 2023; 320:166-198. [PMID: 37548063 DOI: 10.1111/imr.13252] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 07/03/2023] [Indexed: 08/08/2023]
Abstract
Adoptive T-cell transfer (ACT) therapies, including of tumor infiltrating lymphocytes (TILs) and T cells gene-modified to express either a T cell receptor (TCR) or a chimeric antigen receptor (CAR), have demonstrated clinical efficacy for a proportion of patients and cancer-types. The field of ACT has been driven forward by the clinical success of CD19-CAR therapy against various advanced B-cell malignancies, including curative responses for some leukemia patients. However, relapse remains problematic, in particular for lymphoma. Moreover, for a variety of reasons, relative limited efficacy has been demonstrated for ACT of non-hematological solid tumors. Indeed, in addition to pre-infusion challenges including lymphocyte collection and manufacturing, ACT failure can be attributed to several biological processes post-transfer including, (i) inefficient tumor trafficking, infiltration, expansion and retention, (ii) chronic antigen exposure coupled with insufficient costimulation resulting in T-cell exhaustion, (iii) a range of barriers in the tumor microenvironment (TME) mediated by both tumor cells and suppressive immune infiltrate, (iv) tumor antigen heterogeneity and loss, or down-regulation of antigen presentation machinery, (v) gain of tumor intrinsic mechanisms of resistance such as to apoptosis, and (vi) various forms of toxicity and other adverse events in patients. Affinity-optimized TCRs can improve T-cell function and innovative CAR designs as well as gene-modification strategies can be used to coengineer specificity, safety, and function into T cells. Coengineering strategies can be designed not only to directly support the transferred T cells, but also to block suppressive barriers in the TME and harness endogenous innate and adaptive immunity. Here, we review a selection of the remarkable T-cell coengineering strategies, including of tools, receptors, and gene-cargo, that have been developed in recent years to augment tumor control by ACT, more and more of which are advancing to the clinic.
Collapse
Affiliation(s)
- Greta Maria Paola Giordano Attianese
- Department of Oncology, Ludwig Institute for Cancer Research Lausanne, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Sarah Ash
- Department of Oncology, Ludwig Institute for Cancer Research Lausanne, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Melita Irving
- Department of Oncology, Ludwig Institute for Cancer Research Lausanne, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
9
|
Liu P, Foiret J, Situ Y, Zhang N, Kare AJ, Wu B, Raie MN, Ferrara KW, Qi LS. Sonogenetic control of multiplexed genome regulation and base editing. Nat Commun 2023; 14:6575. [PMID: 37852951 PMCID: PMC10584809 DOI: 10.1038/s41467-023-42249-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 10/05/2023] [Indexed: 10/20/2023] Open
Abstract
Manipulating gene expression in the host genome with high precision is crucial for controlling cellular function and behavior. Here, we present a precise, non-invasive, and tunable strategy for controlling the expression of multiple endogenous genes both in vitro and in vivo, utilizing ultrasound as the stimulus. By engineering a hyper-efficient dCas12a and effector under a heat shock promoter, we demonstrate a system that can be inducibly activated through thermal energy produced by ultrasound absorption. This system allows versatile thermal induction of gene activation or base editing across cell types, including primary T cells, and enables multiplexed gene activation using a single guide RNA array. In mouse models, localized temperature elevation guided by high-intensity focused ultrasound effectively triggers reporter gene expression in implanted cells. Our work underscores the potential of ultrasound as a clinically viable approach to enhance cell and gene-based therapies via precision genome and epigenome engineering.
Collapse
Affiliation(s)
- Pei Liu
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology, School of Medicine, Stanford University, Stanford, CA, USA
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Josquin Foiret
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology, School of Medicine, Stanford University, Stanford, CA, USA
| | - Yinglin Situ
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Nisi Zhang
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology, School of Medicine, Stanford University, Stanford, CA, USA
| | - Aris J Kare
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology, School of Medicine, Stanford University, Stanford, CA, USA
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Bo Wu
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology, School of Medicine, Stanford University, Stanford, CA, USA
| | - Marina N Raie
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology, School of Medicine, Stanford University, Stanford, CA, USA
| | - Katherine W Ferrara
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology, School of Medicine, Stanford University, Stanford, CA, USA.
| | - Lei S Qi
- Department of Bioengineering, Stanford University, Stanford, CA, USA.
- Sarafan ChEM-H, Stanford University, Stanford, CA, USA.
- Chan Zuckerberg Biohub - San Francisco, San Francisco, CA, USA.
| |
Collapse
|
10
|
Teixeira AP, Xue S, Huang J, Fussenegger M. Evolution of molecular switches for regulation of transgene expression by clinically licensed gluconate. Nucleic Acids Res 2023; 51:e85. [PMID: 37497781 PMCID: PMC10450161 DOI: 10.1093/nar/gkad600] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 06/22/2023] [Accepted: 07/16/2023] [Indexed: 07/28/2023] Open
Abstract
Synthetic biology holds great promise to improve the safety and efficacy of future gene and engineered cell therapies by providing new means of endogenous or exogenous control of the embedded therapeutic programs. Here, we focused on gluconate as a clinically licensed small-molecule inducer and engineered gluconate-sensitive molecular switches to regulate transgene expression in human cell cultures and in mice. Several switch designs were assembled based on the gluconate-responsive transcriptional repressor GntR from Escherichia coli. Initially we assembled OFF- and ON-type switches by rewiring the native gluconate-dependent binding of GntR to target DNA sequences in mammalian cells. Then, we utilized the ability of GntR to dimerize in the presence of gluconate to activate gene expression from a split transcriptional activator. By means of random mutagenesis of GntR combined with phenotypic screening, we identified variants that significantly enhanced the functionality of the genetic devices, enabling the construction of robust two-input logic gates. We also demonstrated the potential utility of the synthetic switch in two in vivo settings, one employing implantation of alginate-encapsulated engineered cells and the other involving modification of host cells by DNA delivery. Then, as proof-of-concept, the gluconate-actuated genetic switch was connected to insulin secretion, and the components encoding gluconate-induced insulin production were introduced into type-1 diabetic mice as naked DNA via hydrodynamic tail vein injection. Normoglycemia was restored, thereby showcasing the suitability of oral gluconate to regulate in situ production of a therapeutic protein.
Collapse
Affiliation(s)
- Ana Palma Teixeira
- Department of Biosystems Science and Engineering, ETH Zurich, Mattenstrasse 26, CH-4058Basel, Switzerland
| | - Shuai Xue
- Department of Biosystems Science and Engineering, ETH Zurich, Mattenstrasse 26, CH-4058Basel, Switzerland
| | - Jinbo Huang
- Department of Biosystems Science and Engineering, ETH Zurich, Mattenstrasse 26, CH-4058Basel, Switzerland
| | - Martin Fussenegger
- Department of Biosystems Science and Engineering, ETH Zurich, Mattenstrasse 26, CH-4058Basel, Switzerland
- Faculty of Science, University of Basel, Mattenstrasse 26, CH-4058Basel, Switzerland
| |
Collapse
|
11
|
Yan X, Liu X, Zhao C, Chen GQ. Applications of synthetic biology in medical and pharmaceutical fields. Signal Transduct Target Ther 2023; 8:199. [PMID: 37169742 PMCID: PMC10173249 DOI: 10.1038/s41392-023-01440-5] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Revised: 03/15/2023] [Accepted: 03/24/2023] [Indexed: 05/13/2023] Open
Abstract
Synthetic biology aims to design or assemble existing bioparts or bio-components for useful bioproperties. During the past decades, progresses have been made to build delicate biocircuits, standardized biological building blocks and to develop various genomic/metabolic engineering tools and approaches. Medical and pharmaceutical demands have also pushed the development of synthetic biology, including integration of heterologous pathways into designer cells to efficiently produce medical agents, enhanced yields of natural products in cell growth media to equal or higher than that of the extracts from plants or fungi, constructions of novel genetic circuits for tumor targeting, controllable releases of therapeutic agents in response to specific biomarkers to fight diseases such as diabetes and cancers. Besides, new strategies are developed to treat complex immune diseases, infectious diseases and metabolic disorders that are hard to cure via traditional approaches. In general, synthetic biology brings new capabilities to medical and pharmaceutical researches. This review summarizes the timeline of synthetic biology developments, the past and present of synthetic biology for microbial productions of pharmaceutics, engineered cells equipped with synthetic DNA circuits for diagnosis and therapies, live and auto-assemblied biomaterials for medical treatments, cell-free synthetic biology in medical and pharmaceutical fields, and DNA engineering approaches with potentials for biomedical applications.
Collapse
Affiliation(s)
- Xu Yan
- School of Life Sciences, Tsinghua University, 100084, Beijing, China
| | - Xu Liu
- PhaBuilder Biotech Co. Ltd., Shunyi District, Zhaoquan Ying, 101309, Beijing, China
| | - Cuihuan Zhao
- School of Life Sciences, Tsinghua University, 100084, Beijing, China
| | - Guo-Qiang Chen
- School of Life Sciences, Tsinghua University, 100084, Beijing, China.
- Center for Synthetic and Systems Biology, Tsinghua University, 100084, Beijing, China.
- MOE Key Lab for Industrial Biocatalysis, Dept Chemical Engineering, Tsinghua University, 100084, Beijing, China.
| |
Collapse
|
12
|
Escudero-Duch C, Muñoz-Moreno L, Martin-Saavedra F, Sanchez-Casanova S, Lerma-Juarez MA, Vilaboa N. Remote control of transgene expression using noninvasive near-infrared irradiation. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY. B, BIOLOGY 2023; 242:112697. [PMID: 36963296 DOI: 10.1016/j.jphotobiol.2023.112697] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Revised: 03/09/2023] [Accepted: 03/13/2023] [Indexed: 03/26/2023]
Abstract
This study investigated whether noninvasive near-infrared (NIR) energy could be transduced into heat in deep-seated organs in which adenovirus type-5 vectors tend to accumulate, thereby activating heat shock protein (HSP) promoter-mediated transgene expression, without local administration of photothermal agents. NIR irradiation of the subdiaphragmatic and left dorsocranial part of the abdominal cavity of adult immunocompetent C3H/HeNRj mice with an 808-nm laser effectively increased the temperature of the irradiated regions of the liver and spleen, respectively, resulting in the accumulation of the heat-inducible HSP70 protein. Spatial control of transgene expression was achieved in the NIR-irradiated regions of the mice administered an adenoviral vector carrying a firefly luciferase (fLuc) coding sequence controlled by a human HSP70B promoter, as assessed by bioluminescence and immunohistochemistry analyses. Levels of reporter gene expression were modulated by controlling NIR power density. Spatial control of transgene expression through NIR-focused activation of the HSP70B promoter, as well as temporal regulation by administering rapamycin was achieved in the spleens of mice inoculated with an adenoviral vector encoding a rapamycin-dependent transactivator driven by the HSP70B promoter and an adenoviral vector carrying a fLuc coding sequence controlled by the rapamycin-activated transactivator. Mice that were administered rapamycin and exposed to NIR light expressed fLuc activity in the splenic region, whereas no activity was detected in mice that were only administered rapamycin or vehicle or only NIR-irradiated. Thus, in the absence of any exogenously supplied photothermal material, remote control of heat-induced transgene expression can be achieved in the liver and spleen by means of noninvasive NIR irradiation.
Collapse
Affiliation(s)
- Clara Escudero-Duch
- CIBER de Bioingenieria, Biomateriales y Nanomedicina, CIBER-BBN, Madrid, Spain; Hospital Universitario La Paz-IdiPAZ, Paseo de la Castellana 261, 28046 Madrid, Spain
| | - Laura Muñoz-Moreno
- CIBER de Bioingenieria, Biomateriales y Nanomedicina, CIBER-BBN, Madrid, Spain; Hospital Universitario La Paz-IdiPAZ, Paseo de la Castellana 261, 28046 Madrid, Spain
| | - Francisco Martin-Saavedra
- CIBER de Bioingenieria, Biomateriales y Nanomedicina, CIBER-BBN, Madrid, Spain; Hospital Universitario La Paz-IdiPAZ, Paseo de la Castellana 261, 28046 Madrid, Spain
| | - Silvia Sanchez-Casanova
- CIBER de Bioingenieria, Biomateriales y Nanomedicina, CIBER-BBN, Madrid, Spain; Hospital Universitario La Paz-IdiPAZ, Paseo de la Castellana 261, 28046 Madrid, Spain
| | - Miguel Angel Lerma-Juarez
- CIBER de Bioingenieria, Biomateriales y Nanomedicina, CIBER-BBN, Madrid, Spain; Hospital Universitario La Paz-IdiPAZ, Paseo de la Castellana 261, 28046 Madrid, Spain
| | - Nuria Vilaboa
- CIBER de Bioingenieria, Biomateriales y Nanomedicina, CIBER-BBN, Madrid, Spain; Hospital Universitario La Paz-IdiPAZ, Paseo de la Castellana 261, 28046 Madrid, Spain.
| |
Collapse
|
13
|
Jiang Y, Fan M, Yang Z, Liu X, Xu Z, Liu S, Feng G, Tang S, Li Z, Zhang Y, Chen S, Yang C, Law WC, Dong B, Xu G, Yong KT. Recent advances in nanotechnology approaches for non-viral gene therapy. Biomater Sci 2022; 10:6862-6892. [PMID: 36222758 DOI: 10.1039/d2bm01001a] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Gene therapy has shown great potential in the treatment of many diseases by downregulating the expression of certain genes. The development of gene vectors as a vehicle for gene therapy has greatly facilitated the widespread clinical application of nucleic acid materials (DNA, mRNA, siRNA, and miRNA). Currently, both viral and non-viral vectors are used as delivery systems of nucleic acid materials for gene therapy. However, viral vector-based gene therapy has several limitations, including immunogenicity and carcinogenesis caused by the exogenous viral vectors. To address these issues, non-viral nanocarrier-based gene therapy has been explored for superior performance with enhanced gene stability, high treatment efficiency, improved tumor-targeting, and better biocompatibility. In this review, we discuss various non-viral vector-mediated gene therapy approaches using multifunctional biodegradable or non-biodegradable nanocarriers, including polymer-based nanoparticles, lipid-based nanoparticles, carbon nanotubes, gold nanoparticles (AuNPs), quantum dots (QDs), silica nanoparticles, metal-based nanoparticles and two-dimensional nanocarriers. Various strategies to construct non-viral nanocarriers based on their delivery efficiency of targeted genes will be introduced. Subsequently, we discuss the cellular uptake pathways of non-viral nanocarriers. In addition, multifunctional gene therapy based on non-viral nanocarriers is summarized, in which the gene therapy can be combined with other treatments, such as photothermal therapy (PTT), photodynamic therapy (PDT), immunotherapy and chemotherapy. We also provide a comprehensive discussion of the biological toxicity and safety of non-viral vector-based gene therapy. Finally, the present limitations and challenges of non-viral nanocarriers for gene therapy in future clinical research are discussed, to promote wider clinical applications of non-viral vector-based gene therapy.
Collapse
Affiliation(s)
- Yihang Jiang
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen, Guangdong 518055, China.
| | - Miaozhuang Fan
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen, Guangdong 518055, China.
| | - Zhenxu Yang
- School of Biomedical Engineering, The University of Sydney, Sydney, New South Wales 2006, Australia. .,The University of Sydney Nano Institute, The University of Sydney, Sydney, New South Wales 2006, Australia.,The Biophotonics and Mechanobioengineering Laboratory, The University of Sydney, Sydney, New South Wales 2006, Australia
| | - Xiaochen Liu
- School of Biomedical Engineering, The University of Sydney, Sydney, New South Wales 2006, Australia. .,The University of Sydney Nano Institute, The University of Sydney, Sydney, New South Wales 2006, Australia.,The Biophotonics and Mechanobioengineering Laboratory, The University of Sydney, Sydney, New South Wales 2006, Australia
| | - Zhourui Xu
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen, Guangdong 518055, China.
| | - Shikang Liu
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen, Guangdong 518055, China.
| | - Gang Feng
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen, Guangdong 518055, China.
| | - Shuo Tang
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen, Guangdong 518055, China.
| | - Zhengzheng Li
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen, Guangdong 518055, China.
| | - Yibin Zhang
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen, Guangdong 518055, China.
| | - Shilin Chen
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen, Guangdong 518055, China.
| | - Chengbin Yang
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen, Guangdong 518055, China.
| | - Wing-Cheung Law
- Department of Industrial and Systems Engineering, The Hong Kong Polytechnic University, Hung Hom, Kowloon 999077, Hong Kong, China
| | - Biqin Dong
- Guangdong Provincial Key Laboratory of Durability for Marine Civil Engineering, College of Civil and Transportation Engineering, Shenzhen University, Shenzhen 518060, China
| | - Gaixia Xu
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen, Guangdong 518055, China.
| | - Ken-Tye Yong
- School of Biomedical Engineering, The University of Sydney, Sydney, New South Wales 2006, Australia. .,The University of Sydney Nano Institute, The University of Sydney, Sydney, New South Wales 2006, Australia.,The Biophotonics and Mechanobioengineering Laboratory, The University of Sydney, Sydney, New South Wales 2006, Australia
| |
Collapse
|
14
|
Mansouri M, Ray PG, Franko N, Xue S, Fussenegger M. Design of programmable post-translational switch control platform for on-demand protein secretion in mammalian cells. Nucleic Acids Res 2022; 51:e1. [PMID: 36268868 PMCID: PMC9841418 DOI: 10.1093/nar/gkac916] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Revised: 09/11/2022] [Accepted: 10/20/2022] [Indexed: 01/29/2023] Open
Abstract
The development of novel strategies to program cellular behaviors is a central goal in synthetic biology, and post-translational control mediated by engineered protein circuits is a particularly attractive approach to achieve rapid protein secretion on demand. We have developed a programmable protease-mediated post-translational switch (POSH) control platform composed of a chimeric protein unit that consists of a protein of interest fused via a transmembrane domain to a cleavable ER-retention signal, together with two cytosolic inducer-sensitive split protease components. The protease components combine in the presence of the specific inducer to generate active protease, which cleaves the ER-retention signal, releasing the transmembrane-domain-linked protein for trafficking to the trans-Golgi region. A furin site placed downstream of the protein ensures cleavage and subsequent secretion of the desired protein. We show that stimuli ranging from plant-derived, clinically compatible chemicals to remotely controllable inducers such as light and electrostimulation can program protein secretion in various POSH-engineered designer mammalian cells. As proof-of-concept, an all-in-one POSH control plasmid encoding insulin and abscisic acid-activatable split protease units was hydrodynamically transfected into the liver of type-1 diabetic mice. Induction with abscisic acid attenuated glycemic excursions in glucose-tolerance tests. Increased blood levels of insulin were maintained for 12 days.
Collapse
Affiliation(s)
- Maysam Mansouri
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
| | - Preetam Guha Ray
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
| | - Nik Franko
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
| | - Shuai Xue
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
| | - Martin Fussenegger
- To whom correspondence should be addressed. Tel: +41 61 387 31 60; Fax: +41 61 387 39 88;
| |
Collapse
|
15
|
Stefanov BA, Mansouri M, Charpin-El Hamri G, Fussenegger M. Sunlight-Controllable Biopharmaceutical Production for Remote Emergency Supply of Directly Injectable Therapeutic Proteins. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2202566. [PMID: 36084222 DOI: 10.1002/smll.202202566] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 08/12/2022] [Indexed: 06/15/2023]
Abstract
Biopharmaceutical manufacturing requires specialized facilities and a long-range cold supply chain for the delivery of the therapeutics to patients. In order to produce biopharmaceuticals in locations lacking such infrastructure, a production process is designed that utilizes the trigger-inducible release of large quantities of a stored therapeutic protein from engineered endocrine cells within minutes to generate a directly injectable saline solution of the protein. To illustrate the versatility of this approach, it is shown that not only insulin, but also glucagon-like peptide 1 (GLP-1), nanoluciferase (NLuc), and the model biopharmaceutical erythropoietin (EPO) can be trigger-inducibly released, even when using biologically inactive insulin as a carrier. The facilitating beta cells are engineered with a controllable TRPV1-mediated Ca2+ influx that induces the fusion of cytoplasmic storage vesicles with the membrane, leading to the release of the stored protein. When required, the growth medium is exchanged for saline solution, and the system is stimulated with the small molecule capsaicin, with a hand-warming pack, or simply by using sunlight. Injection of insulin saline solution obtained in this way into a type-1 diabetes mouse model results in the regulation of blood glucose levels. It is believed that this system will be readily adaptable to deliver various biopharmaceutical proteins at remote locations.
Collapse
Affiliation(s)
- Bozhidar-Adrian Stefanov
- Department of Biosystems Science and Engineering, ETH Zürich, Mattenstrasse 26, Basel, 4058, Switzerland
| | - Maysam Mansouri
- Department of Biosystems Science and Engineering, ETH Zürich, Mattenstrasse 26, Basel, 4058, Switzerland
| | - Ghislaine Charpin-El Hamri
- Département Génie Biologique, Institut Universitaire de Technologie, Villeurbanne, Cedex F-69622, France
| | - Martin Fussenegger
- Department of Biosystems Science and Engineering, ETH Zürich, Mattenstrasse 26, Basel, 4058, Switzerland
- Faculty of Life Science, University of Basel, Basel, 4058, Switzerland
| |
Collapse
|
16
|
Mansouri M, Fussenegger M. Therapeutic cell engineering: designing programmable synthetic genetic circuits in mammalian cells. Protein Cell 2022; 13:476-489. [PMID: 34586617 PMCID: PMC9226217 DOI: 10.1007/s13238-021-00876-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Accepted: 08/02/2021] [Indexed: 12/01/2022] Open
Abstract
Cell therapy approaches that employ engineered mammalian cells for on-demand production of therapeutic agents in the patient's body are moving beyond proof-of-concept in translational medicine. The therapeutic cells can be customized to sense user-defined signals, process them, and respond in a programmable and predictable way. In this paper, we introduce the available tools and strategies employed to design therapeutic cells. Then, various approaches to control cell behaviors, including open-loop and closed-loop systems, are discussed. We also highlight therapeutic applications of engineered cells for early diagnosis and treatment of various diseases in the clinic and in experimental disease models. Finally, we consider emerging technologies such as digital devices and their potential for incorporation into future cell-based therapies.
Collapse
Affiliation(s)
- Maysam Mansouri
- Department of Biosystems Science and Engineering, ETH Zurich, Mattenstrasse 26, 4058, Basel, Switzerland
| | - Martin Fussenegger
- Department of Biosystems Science and Engineering, ETH Zurich, Mattenstrasse 26, 4058, Basel, Switzerland.
- Faculty of Science, University of Basel, Mattenstrasse 26, 4058, Basel, Switzerland.
| |
Collapse
|
17
|
Odda AH, Cheang TY, Alesary HF, Liu L, Qian X, Ullah N, Wang G, Pan Y, Xu AW. A multifunctional α-Fe 2O 3@PEDOT core-shell nanoplatform for gene and photothermal combination anticancer therapy. J Mater Chem B 2022; 10:1453-1462. [PMID: 35188170 DOI: 10.1039/d1tb02625a] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Exploration of versatile nanoplatforms within one single nanostructure for multidisciplinary treatment modalities, especially achieving a synergistic therapeutic efficacy of combinational gene/photothermal cancer therapy is still a great challenge in biomedicine and nanotechnology. In this study, a unique photothermal nanocarrier has successfully been designed and developed for a combination of gene therapy (GT) and photothermal therapy (PTT) of cancer cells. Surface-engineered iron oxides (α-Fe2O3) nanoparticles (NPs) with poly(3,4-ethylenedioxythiophene) (PEDOT) polymer coatings are synthesized using a one-pot in situ oxidative polymerization method. The results show that the as-prepared α-Fe2O3@PEDOT core-shell NPs with a uniform particle size exhibit positively charged surfaces, facilitating efficient siRNA Bcl-2 (B-cell lymphoma-2) uptake for delivery to breast cancer cells. More importantly, α-Fe2O3@PEDOT core-shell NPs not only display good biocompatibility and water dispersibility but also strong optical absorption enhancement in the Vis-NIR region as compared to α-Fe2O3 NPs. The obtained α-Fe2O3@PEDOT core-shell NPs show an efficient photothermal conversion efficacy (η = 54.3%) and photostability under NIR laser irradiation. As a result, both in vitro and in vivo biological studies on two types of breast cancer cells/tumors treated with α-Fe2O3@PEDOT-siRNA nanocomplexes demonstrate high cancer cell apoptosis and tumor inhibition induced by synergistic GT/PTT therapy under mild conditions compared to an individual GT or PTT alone. Taken together, this is the first example of the use of an α-Fe2O3@PEDOT core-shell nanoagent as a siRNA delivery nanocarrier for highly effective gene/photothermal combination anticancer therapy.
Collapse
Affiliation(s)
- Atheer Hameid Odda
- Department of Medical Oncology, The First Affiliated Hospital, Hefei National Laboratory for Physical Sciences at The Microscale, University of Science and Technology of China, Hefei 230026, P. R. China.
- Department of Biochemistry, College of Medicine, Department of Chemistry, College of Science, University of Kerbala, Kerbala 56001, Iraq
| | - Tuck-Yun Cheang
- Department of Breast Care Centre, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou 510080, P. R. China
| | - Hasan F Alesary
- Department of Biochemistry, College of Medicine, Department of Chemistry, College of Science, University of Kerbala, Kerbala 56001, Iraq
| | - Lirong Liu
- Department of Breast Care Centre, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou 510080, P. R. China
| | - Xiaojun Qian
- Department of Medical Oncology, The First Affiliated Hospital, Hefei National Laboratory for Physical Sciences at The Microscale, University of Science and Technology of China, Hefei 230026, P. R. China.
| | - Naseeb Ullah
- Department of Medical Oncology, The First Affiliated Hospital, Hefei National Laboratory for Physical Sciences at The Microscale, University of Science and Technology of China, Hefei 230026, P. R. China.
| | - Gang Wang
- Department of Medical Oncology, The First Affiliated Hospital, Hefei National Laboratory for Physical Sciences at The Microscale, University of Science and Technology of China, Hefei 230026, P. R. China.
| | - Yueyin Pan
- Department of Medical Oncology, The First Affiliated Hospital, Hefei National Laboratory for Physical Sciences at The Microscale, University of Science and Technology of China, Hefei 230026, P. R. China.
| | - An-Wu Xu
- Department of Medical Oncology, The First Affiliated Hospital, Hefei National Laboratory for Physical Sciences at The Microscale, University of Science and Technology of China, Hefei 230026, P. R. China.
| |
Collapse
|
18
|
Chee WKD, Yeoh JW, Dao VL, Poh CL. Highly Reversible Tunable Thermal-Repressible Split-T7 RNA Polymerases (Thermal-T7RNAPs) for Dynamic Gene Regulation. ACS Synth Biol 2022; 11:921-937. [PMID: 35089710 DOI: 10.1021/acssynbio.1c00545] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Temperature is a physical cue that is easy to apply, allowing cellular behaviors to be controlled in a contactless and dynamic manner via heat-inducible/repressible systems. However, existing heat-repressible systems are limited in number, rely on thermal sensitive mRNA or transcription factors that function at low temperatures, lack tunability, suffer delays, and are overly complex. To provide an alternative mode of thermal regulation, we developed a library of compact, reversible, and tunable thermal-repressible split-T7 RNA polymerase systems (Thermal-T7RNAPs), which fused temperature-sensitive domains of Tlpa protein with split-T7RNAP to enable direct thermal control of the T7RNAP activity between 30 and 42 °C. We generated a large mutant library with varying thermal performances via an automated screening framework to extend temperature tunability. Lastly, using the mutants, novel thermal logic circuitry was implemented to regulate cell growth and achieve active thermal control of the cell proportions within co-cultures. Overall, this technology expanded avenues for thermal control in biotechnology applications.
Collapse
Affiliation(s)
- Wai Kit David Chee
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, 4 Engineering Drive 3, 117583 Singapore
- NUS Synthetic Biology for Clinical and Technological Innovation (SynCTI), National University of Singapore, 28 Medical Drive, 117456 Singapore
| | - Jing Wui Yeoh
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, 4 Engineering Drive 3, 117583 Singapore
- NUS Synthetic Biology for Clinical and Technological Innovation (SynCTI), National University of Singapore, 28 Medical Drive, 117456 Singapore
| | - Viet Linh Dao
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, 4 Engineering Drive 3, 117583 Singapore
- NUS Synthetic Biology for Clinical and Technological Innovation (SynCTI), National University of Singapore, 28 Medical Drive, 117456 Singapore
| | - Chueh Loo Poh
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, 4 Engineering Drive 3, 117583 Singapore
- NUS Synthetic Biology for Clinical and Technological Innovation (SynCTI), National University of Singapore, 28 Medical Drive, 117456 Singapore
| |
Collapse
|
19
|
Jiao S, Bai C, Qi C, Wu H, Hu L, Li F, Yang K, Zhao C, Ouyang H, Pang D, Tang X, Xie Z. Identification and Functional Analysis of the Regulatory Elements in the pHSPA6 Promoter. Genes (Basel) 2022; 13:genes13020189. [PMID: 35205234 PMCID: PMC8872561 DOI: 10.3390/genes13020189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 01/14/2022] [Accepted: 01/19/2022] [Indexed: 12/10/2022] Open
Abstract
Functional and expressional research of heat shock protein A6 (HSPA6) suggests that the gene is of great value for neurodegenerative diseases, biosensors, cancer, etc. Based on the important value of pigs in agriculture and biomedicine and to advance knowledge of this little-studied HSPA member, the stress-sensitive sites in porcine HSPA6 (pHSPA6) were investigated following different stresses. Here, two heat shock elements (HSEs) and a conserved region (CR) were identified in the pHSPA6 promoter by a CRISPR/Cas9-mediated precise gene editing strategy. Gene expression data showed that sequence disruption of these regions could significantly reduce the expression of pHSPA6 under heat stress. Stimulation studies indicated that these regions responded not only to heat stress but also to copper sulfate, MG132, and curcumin. Further mechanism studies showed that downregulated pHSPA6 could significantly affect some important members of the HSP family that are involved in HSP40, HSP70, and HSP90. Overall, our results provide a new approach for investigating gene expression and regulation that may contribute to gene regulatory mechanisms, drug target selection, and breeding stock selection.
Collapse
Affiliation(s)
- Shuyu Jiao
- College of Animal Science, Jilin University, Changchun 130062, China; (S.J.); (C.B.); (C.Q.); (H.W.); (L.H.); (F.L.); (K.Y.); (C.Z.); (H.O.); (D.P.)
| | - Chunyan Bai
- College of Animal Science, Jilin University, Changchun 130062, China; (S.J.); (C.B.); (C.Q.); (H.W.); (L.H.); (F.L.); (K.Y.); (C.Z.); (H.O.); (D.P.)
| | - Chunyun Qi
- College of Animal Science, Jilin University, Changchun 130062, China; (S.J.); (C.B.); (C.Q.); (H.W.); (L.H.); (F.L.); (K.Y.); (C.Z.); (H.O.); (D.P.)
| | - Heyong Wu
- College of Animal Science, Jilin University, Changchun 130062, China; (S.J.); (C.B.); (C.Q.); (H.W.); (L.H.); (F.L.); (K.Y.); (C.Z.); (H.O.); (D.P.)
| | - Lanxin Hu
- College of Animal Science, Jilin University, Changchun 130062, China; (S.J.); (C.B.); (C.Q.); (H.W.); (L.H.); (F.L.); (K.Y.); (C.Z.); (H.O.); (D.P.)
| | - Feng Li
- College of Animal Science, Jilin University, Changchun 130062, China; (S.J.); (C.B.); (C.Q.); (H.W.); (L.H.); (F.L.); (K.Y.); (C.Z.); (H.O.); (D.P.)
| | - Kang Yang
- College of Animal Science, Jilin University, Changchun 130062, China; (S.J.); (C.B.); (C.Q.); (H.W.); (L.H.); (F.L.); (K.Y.); (C.Z.); (H.O.); (D.P.)
| | - Chuheng Zhao
- College of Animal Science, Jilin University, Changchun 130062, China; (S.J.); (C.B.); (C.Q.); (H.W.); (L.H.); (F.L.); (K.Y.); (C.Z.); (H.O.); (D.P.)
| | - Hongsheng Ouyang
- College of Animal Science, Jilin University, Changchun 130062, China; (S.J.); (C.B.); (C.Q.); (H.W.); (L.H.); (F.L.); (K.Y.); (C.Z.); (H.O.); (D.P.)
- Key Lab for Zoonoses Research, Ministry of Education, Animal Genome Editing Technology Innovation Center, Jilin University, Changchun 130062, China
- Chongqing Research Institute, Jilin University, Chongqing 401123, China
- Chongqing Jitang Biotechnology Research Institute Co., Ltd., Chongqing 401123, China
| | - Daxin Pang
- College of Animal Science, Jilin University, Changchun 130062, China; (S.J.); (C.B.); (C.Q.); (H.W.); (L.H.); (F.L.); (K.Y.); (C.Z.); (H.O.); (D.P.)
- Key Lab for Zoonoses Research, Ministry of Education, Animal Genome Editing Technology Innovation Center, Jilin University, Changchun 130062, China
- Chongqing Research Institute, Jilin University, Chongqing 401123, China
- Chongqing Jitang Biotechnology Research Institute Co., Ltd., Chongqing 401123, China
| | - Xiaochun Tang
- Key Lab for Zoonoses Research, Ministry of Education, Animal Genome Editing Technology Innovation Center, Jilin University, Changchun 130062, China
- Chongqing Research Institute, Jilin University, Chongqing 401123, China
- Chongqing Jitang Biotechnology Research Institute Co., Ltd., Chongqing 401123, China
- Correspondence: (X.T.); (Z.X.)
| | - Zicong Xie
- College of Animal Science, Jilin University, Changchun 130062, China; (S.J.); (C.B.); (C.Q.); (H.W.); (L.H.); (F.L.); (K.Y.); (C.Z.); (H.O.); (D.P.)
- Correspondence: (X.T.); (Z.X.)
| |
Collapse
|
20
|
Chee WKD, Yeoh JW, Dao VL, Poh CL. Thermogenetics: Applications come of age. Biotechnol Adv 2022; 55:107907. [PMID: 35041863 DOI: 10.1016/j.biotechadv.2022.107907] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 12/13/2021] [Accepted: 01/09/2022] [Indexed: 12/20/2022]
Abstract
Temperature is a ubiquitous physical cue that is non-invasive, penetrative and easy to apply. In the growing field of thermogenetics, through beneficial repurposing of natural thermosensing mechanisms, synthetic biology is bringing new opportunities to design and build robust temperature-sensitive (TS) sensors which forms a thermogenetic toolbox of well characterised biological parts. Recent advancements in technological platforms available have expedited the discovery of novel or de novo thermosensors which are increasingly deployed in many practical temperature-dependent biomedical, industrial and biosafety applications. In all, the review aims to convey both the exhilarating recent technological developments underlying the advancement of thermosensors and the exciting opportunities the nascent thermogenetic field holds for biomedical and biotechnology applications.
Collapse
Affiliation(s)
- Wai Kit David Chee
- Department of Biomedical Engineering, Faculty of Engineering, National University of Singapore, 4 Engineering Drive 3, Singapore 117583, Singapore; NUS Synthetic Biology for Clinical and Technological Innovation (SynCTI), Life Sciences Institute, National University of Singapore, 28 Medical Drive, Singapore 117456, Singapore
| | - Jing Wui Yeoh
- Department of Biomedical Engineering, Faculty of Engineering, National University of Singapore, 4 Engineering Drive 3, Singapore 117583, Singapore; NUS Synthetic Biology for Clinical and Technological Innovation (SynCTI), Life Sciences Institute, National University of Singapore, 28 Medical Drive, Singapore 117456, Singapore
| | - Viet Linh Dao
- Department of Biomedical Engineering, Faculty of Engineering, National University of Singapore, 4 Engineering Drive 3, Singapore 117583, Singapore; NUS Synthetic Biology for Clinical and Technological Innovation (SynCTI), Life Sciences Institute, National University of Singapore, 28 Medical Drive, Singapore 117456, Singapore
| | - Chueh Loo Poh
- Department of Biomedical Engineering, Faculty of Engineering, National University of Singapore, 4 Engineering Drive 3, Singapore 117583, Singapore; NUS Synthetic Biology for Clinical and Technological Innovation (SynCTI), Life Sciences Institute, National University of Singapore, 28 Medical Drive, Singapore 117456, Singapore.
| |
Collapse
|
21
|
Opto-thermal technologies for microscopic analysis of cellular temperature-sensing systems. Biophys Rev 2021; 14:41-54. [PMID: 35340595 PMCID: PMC8921355 DOI: 10.1007/s12551-021-00854-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 10/12/2021] [Indexed: 12/15/2022] Open
Abstract
AbstractCould enzymatic activities and their cooperative functions act as cellular temperature-sensing systems? This review introduces recent opto-thermal technologies for microscopic analyses of various types of cellular temperature-sensing system. Optical microheating technologies have been developed for local and rapid temperature manipulations at the cellular level. Advanced luminescent thermometers visualize the dynamics of cellular local temperature in space and time during microheating. An optical heater and thermometer can be combined into one smart nanomaterial that demonstrates hybrid function. These technologies have revealed a variety of cellular responses to spatial and temporal changes in temperature. Spatial temperature gradients cause asymmetric deformations during mitosis and neurite outgrowth. Rapid changes in temperature causes imbalance of intracellular Ca2+ homeostasis and membrane potential. Among those responses, heat-induced muscle contractions are highlighted. It is also demonstrated that the short-term heating hyperactivates molecular motors to exceed their maximal activities at optimal temperatures. We discuss future prospects for opto-thermal manipulation of cellular functions and contributions to obtain a deeper understanding of the mechanisms of cellular temperature-sensing systems.
Collapse
|
22
|
Control of the activity of CAR-T cells within tumours via focused ultrasound. Nat Biomed Eng 2021; 5:1336-1347. [PMID: 34385696 PMCID: PMC9015817 DOI: 10.1038/s41551-021-00779-w] [Citation(s) in RCA: 118] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 07/07/2021] [Indexed: 02/07/2023]
Abstract
Focused ultrasound can deliver energy safely and non-invasively into tissues at depths of centimetres. Here we show that the genetics and cellular functions of chimeric antigen receptor T cells (CAR-T cells) within tumours can be reversibly controlled by the heat generated by short pulses of focused ultrasound via a CAR cassette under the control of a promoter for the heat-shock protein. In mice with subcutaneous tumours, locally injected T cells with the inducible CAR and activated via focused ultrasound guided by magnetic resonance imaging mitigated on-target off-tumour activity and enhanced the suppression of tumour growth, compared with the performance of non-inducible CAR-T cells. Acoustogenetic control of the activation of engineered T cells may facilitate the design of safer cell therapies.
Collapse
|
23
|
Stefanov B, Teixeira AP, Mansouri M, Bertschi A, Krawczyk K, Hamri GC, Xue S, Fussenegger M. Genetically Encoded Protein Thermometer Enables Precise Electrothermal Control of Transgene Expression. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:e2101813. [PMID: 34496151 PMCID: PMC8564464 DOI: 10.1002/advs.202101813] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 07/05/2021] [Indexed: 05/25/2023]
Abstract
Body temperature is maintained at around 37 °C in humans, but may rise to 40 °C or more during high-grade fever, which occurs in most adults who are seriously ill. However, endogenous temperature sensors, such as ion channels and heat-shock promoters, are fully activated only at noxious temperatures above this range, making them unsuitable for medical applications. Here, a genetically encoded protein thermometer (human enhanced gene activation thermometer; HEAT) is designed that can trigger transgene expression in the range of 37-40 °C by linking a mutant coiled-coil temperature-responsive protein sensor to a synthetic transcription factor. To validate the construct, a HEAT-transgenic monoclonal human cell line, FeverSense, is generated and it is confirmed that it works as a fever sensor that can temperature- and exposure-time-dependently trigger reporter gene expression in vitro and in vivo. For translational proof of concept, microencapsulated designer cells stably expressing a HEAT-controlled insulin production cassette in a mouse model of type-1 diabetes are subcutaneously implanted and topical heating patches are used to apply heat corresponding to a warm sensation in humans. Insulin release is induced, restoring normoglycemia. Thus, HEAT appears to be suitable for practical electrothermal control of cell-based therapy, and may also have potential for next-generation treatment of fever-associated medical conditions.
Collapse
Affiliation(s)
| | - Ana P. Teixeira
- ETH ZürichDepartment of Biosystems Science and EngineeringMattenstrasse 26Basel4058Switzerland
| | - Maysam Mansouri
- ETH ZürichDepartment of Biosystems Science and EngineeringMattenstrasse 26Basel4058Switzerland
| | - Adrian Bertschi
- ETH ZürichDepartment of Biosystems Science and EngineeringMattenstrasse 26Basel4058Switzerland
| | - Krzysztof Krawczyk
- ETH ZürichDepartment of Biosystems Science and EngineeringMattenstrasse 26Basel4058Switzerland
| | | | - Shuai Xue
- ETH ZürichDepartment of Biosystems Science and EngineeringMattenstrasse 26Basel4058Switzerland
| | - Martin Fussenegger
- ETH ZürichDepartment of Biosystems Science and EngineeringMattenstrasse 26Basel4058Switzerland
- University of BaselFaculty of Life ScienceBasel4056Switzerland
| |
Collapse
|
24
|
Suciu M, Porav S, Radu T, Rosu MC, Lazar MD, Macavei S, Socaci C. Photodynamic effect of light emitting diodes on E. coli and human skin cells induced by a graphene-based ternary composite. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY B-BIOLOGY 2021; 223:112298. [PMID: 34474299 DOI: 10.1016/j.jphotobiol.2021.112298] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 08/17/2021] [Accepted: 08/18/2021] [Indexed: 01/10/2023]
Abstract
In this paper, the photodynamic effect of a ternary nanocomposite (TiO2-Ag/graphene) on Escherichia coli bacteria and two human cell lines: A375 (melanoma) and HaCaT (keratinocyte) after exposure to different wavelength domains (blue, green or red-Light Emitting Diode, LED) was analyzed. The results obtained through bioassays were correlated with the morphological, structural and spectral data obtained through FT-IR, XPS and UV-Vis spectroscopy, powder X-Ray diffractometry (XRD) and STEM/EDX techniques, leading to conclusions that showed different photodynamic activation mechanisms and effects on bacteria and human cells, depending on the wavelength. The nanocomposite proved a therapeutic potential for blue light-activated antibacterial treatment and revealed a keratinocyte cytotoxic effect under blue and green LEDs. The red light-nanocomposite duo gave a metabolic boost to normal keratinocytes and induced stasis to melanoma cells. The light and nanocomposite combination could be a potential therapy for bacterial keratosis or for skin tumors.
Collapse
Affiliation(s)
- Maria Suciu
- National Institute for Research and Development of Isotopic and Molecular Technologies, Donat 67-103 Str., RO-400293 Cluj-Napoca, Romania; Biology and Geology Faculty, Babes-Bolyai University, 5-7 Clinicilor Str, Cluj-Napoca, Romania
| | - Sebastian Porav
- National Institute for Research and Development of Isotopic and Molecular Technologies, Donat 67-103 Str., RO-400293 Cluj-Napoca, Romania
| | - Teodora Radu
- National Institute for Research and Development of Isotopic and Molecular Technologies, Donat 67-103 Str., RO-400293 Cluj-Napoca, Romania
| | - Marcela C Rosu
- National Institute for Research and Development of Isotopic and Molecular Technologies, Donat 67-103 Str., RO-400293 Cluj-Napoca, Romania
| | - Mihaela D Lazar
- National Institute for Research and Development of Isotopic and Molecular Technologies, Donat 67-103 Str., RO-400293 Cluj-Napoca, Romania
| | - Sergiu Macavei
- National Institute for Research and Development of Isotopic and Molecular Technologies, Donat 67-103 Str., RO-400293 Cluj-Napoca, Romania
| | - Crina Socaci
- National Institute for Research and Development of Isotopic and Molecular Technologies, Donat 67-103 Str., RO-400293 Cluj-Napoca, Romania.
| |
Collapse
|
25
|
Mansouri M, Xue S, Hussherr MD, Strittmatter T, Camenisch G, Fussenegger M. Smartphone-Flashlight-Mediated Remote Control of Rapid Insulin Secretion Restores Glucose Homeostasis in Experimental Type-1 Diabetes. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2021; 17:e2101939. [PMID: 34227232 DOI: 10.1002/smll.202101939] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 04/26/2021] [Indexed: 06/13/2023]
Abstract
Emerging digital assessment of biomarkers by linking health-related data obtained from wearable electronic devices and embedded health and fitness sensors in smartphones is opening up the possibility of creating a continuous remote-monitoring platform for disease management. It is considered that the built-in flashlight of smartphones may be utilized to remotely program genetically engineered designer cells for on-demand delivery of protein-based therapeutics. Here, the authors present smartphone-induced insulin release in β-cell line (iβ-cell) technology for traceless light-triggered rapid insulin secretion, employing the light-activatable receptor melanopsin to induce calcium influx and membrane depolarization upon illumination. This iβ-cell-based system enables repeated, reversible secretion of insulin within 15 min in response to light stimulation, with a high induction fold both in vitro and in vivo. It is shown that programmable percutaneous remote control of implanted microencapsulated iβ-cells with a smartphone's flashlight rapidly reverses hyperglycemia in a mouse model of type-1 diabetes.
Collapse
Affiliation(s)
- Maysam Mansouri
- Department of Biosystems Science and Engineering, ETH Zurich, Mattenstrasse 26, Basel, CH-4058, Switzerland
| | - Shuai Xue
- Department of Biosystems Science and Engineering, ETH Zurich, Mattenstrasse 26, Basel, CH-4058, Switzerland
| | - Marie-Didiée Hussherr
- Department of Biosystems Science and Engineering, ETH Zurich, Mattenstrasse 26, Basel, CH-4058, Switzerland
| | - Tobias Strittmatter
- Department of Biosystems Science and Engineering, ETH Zurich, Mattenstrasse 26, Basel, CH-4058, Switzerland
| | - Gieri Camenisch
- Department of Biosystems Science and Engineering, ETH Zurich, Mattenstrasse 26, Basel, CH-4058, Switzerland
| | - Martin Fussenegger
- Department of Biosystems Science and Engineering, ETH Zurich, Mattenstrasse 26, Basel, CH-4058, Switzerland
- Faculty of Science, University of Basel, Mattenstrasse 26, Basel, CH-4058, Switzerland
| |
Collapse
|
26
|
Miller IC, Zamat A, Sun LK, Phuengkham H, Harris AM, Gamboa L, Yang J, Murad JP, Priceman SJ, Kwong GA. Enhanced intratumoural activity of CAR T cells engineered to produce immunomodulators under photothermal control. Nat Biomed Eng 2021; 5:1348-1359. [PMID: 34385695 DOI: 10.1038/s41551-021-00781-2] [Citation(s) in RCA: 91] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Accepted: 07/11/2021] [Indexed: 12/17/2022]
Abstract
Treating solid malignancies with chimeric antigen receptor (CAR) T cells typically results in poor responses. Immunomodulatory biologics delivered systemically can augment the cells' activity, but off-target toxicity narrows the therapeutic window. Here we show that the activity of intratumoural CAR T cells can be controlled photothermally via synthetic gene switches that trigger the expression of transgenes in response to mild temperature elevations (to 40-42 °C). In vitro, heating engineered primary human T cells for 15-30 min led to over 60-fold-higher expression of a reporter transgene without affecting the cells' proliferation, migration and cytotoxicity. In mice, CAR T cells photothermally heated via gold nanorods produced a transgene only within the tumours. In mouse models of adoptive transfer, the systemic delivery of CAR T cells followed by intratumoural production, under photothermal control, of an interleukin-15 superagonist or a bispecific T cell engager bearing an NKG2D receptor redirecting T cells against NKG2D ligands enhanced antitumour activity and mitigated antigen escape. Localized photothermal control of the activity of engineered T cells may enhance their safety and efficacy.
Collapse
Affiliation(s)
- Ian C Miller
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, Atlanta, GA, USA
| | - Ali Zamat
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, Atlanta, GA, USA
| | - Lee-Kai Sun
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, Atlanta, GA, USA
| | - Hathaichanok Phuengkham
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, Atlanta, GA, USA
| | - Adrian M Harris
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, Atlanta, GA, USA
| | - Lena Gamboa
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, Atlanta, GA, USA
| | - Jason Yang
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope, Duarte, CA, USA
| | - John P Murad
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope, Duarte, CA, USA
| | - Saul J Priceman
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope, Duarte, CA, USA.,Department of Immuno-Oncology, Beckman Research Institute of City of Hope, Duarte, CA, USA
| | - Gabriel A Kwong
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, Atlanta, GA, USA. .,Institute for Electronics and Nanotechnology, Georgia Institute of Technology, Atlanta, GA, USA. .,Parker H. Petit Institute of Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA. .,Integrated Cancer Research Center, Georgia Institute of Technology, Atlanta, GA, USA. .,Georgia Immunoengineering Consortium, Emory University and Georgia Institute of Technology, Atlanta, GA, USA. .,Winship Cancer Institute, Emory University, Atlanta, GA, USA.
| |
Collapse
|
27
|
Cazier AP, Blazeck J. Advances in promoter engineering: novel applications and predefined transcriptional control. Biotechnol J 2021; 16:e2100239. [PMID: 34351706 DOI: 10.1002/biot.202100239] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 07/30/2021] [Accepted: 08/03/2021] [Indexed: 11/08/2022]
Abstract
Synthetic biology continues to progress by relying on more robust tools for transcriptional control, of which promoters are the most fundamental component. Numerous studies have sought to characterize promoter function, determine principles to guide their engineering, and create promoters with stronger expression or tailored inducible control. In this review, we will summarize promoter architecture and highlight recent advances in the field, focusing on the novel applications of inducible promoter design and engineering towards metabolic engineering and cellular therapeutic development. Additionally, we will highlight how the expansion of new, machine learning techniques for modeling and engineering promoter sequences are enabling more accurate prediction of promoter characteristics. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Andrew P Cazier
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, 311 Ferst St. NW, Atlanta, Georgia, 30332, USA
| | - John Blazeck
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, 311 Ferst St. NW, Atlanta, Georgia, 30332, USA
| |
Collapse
|
28
|
Su FY, Mac QD, Sivakumar A, Kwong GA. Interfacing Biomaterials with Synthetic T Cell Immunity. Adv Healthc Mater 2021; 10:e2100157. [PMID: 33887123 PMCID: PMC8349871 DOI: 10.1002/adhm.202100157] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 03/28/2021] [Indexed: 12/14/2022]
Abstract
The clinical success of cancer immunotherapy is providing exciting opportunities for the development of new methods to detect and treat cancer more effectively. A new generation of biomaterials is being developed to interface with molecular and cellular features of immunity and ultimately shape or control anti-tumor responses. Recent advances that are supporting the advancement of engineered T cells are focused here. This class of cancer therapy has the potential to cure disease in subsets of patients, yet there remain challenges such as the need to improve response rates and safety while lowering costs to expand their use. To provide a focused overview, recent strategies in three areas of biomaterials research are highlighted: low-cost cell manufacturing to broaden patient access, noninvasive diagnostics for predictive monitoring of immune responses, and strategies for in vivo control that enhance anti-tumor immunity. These research efforts shed light on some of the challenges associated with T cell immunotherapy and how engineered biomaterials that interface with synthetic immunity are gaining traction to solve these challenges.
Collapse
Affiliation(s)
- Fang-Yi Su
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, Atlanta, GA, 30332, USA
| | - Quoc D Mac
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, Atlanta, GA, 30332, USA
| | - Anirudh Sivakumar
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, Atlanta, GA, 30332, USA
| | - Gabriel A Kwong
- The Wallace H. Coulter Department of Biomedical Engineering, Institute for Electronics and Nanotechnology, Parker H. Petit Institute of Bioengineering and Bioscience, Integrated Cancer Research Center, Georgia Immunoengineering Consortium, Winship Cancer Institute, Emory University, Georgia Institute of Technology & Emory University, Atlanta, GA, 30332, USA
| |
Collapse
|
29
|
Madderson O, Teixeira AP, Fussenegger M. Emerging mammalian gene switches for controlling implantable cell therapies. Curr Opin Chem Biol 2021; 64:98-105. [PMID: 34216875 DOI: 10.1016/j.cbpa.2021.05.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 05/20/2021] [Accepted: 05/24/2021] [Indexed: 12/25/2022]
Abstract
Engineered cell-based therapies have emerged as a new paradigm in modern medicine, with several engineered T cell therapies currently approved to treat blood cancers and many more in clinical development. Tremendous progress in synthetic biology over the past two decades has allowed us to program cells with sophisticated sense-and-response modules that can effectively control therapeutic functions. In this review, we highlight recent advances in mammalian synthetic gene switches, focusing on devices designed for therapeutic applications. Although many gene switches responding to endogenous or exogenous molecular signals have been developed, the focus is shifting towards achieving remote-controlled production of therapeutic effectors by stimulating implanted engineered cells with traceless physical signals, such as light, electrical signals, magnetic fields, heat or ultrasound.
Collapse
Affiliation(s)
- Oliver Madderson
- ETH Zürich, Department of Biosystems Science and Engineering, Mattenstrasse 26, 4058, Basel, Switzerland
| | - Ana Palma Teixeira
- ETH Zürich, Department of Biosystems Science and Engineering, Mattenstrasse 26, 4058, Basel, Switzerland
| | - Martin Fussenegger
- ETH Zürich, Department of Biosystems Science and Engineering, Mattenstrasse 26, 4058, Basel, Switzerland; University of Basel, Faculty of Life Science, Basel, Switzerland.
| |
Collapse
|
30
|
Bretherton RC, DeForest CA. The Art of Engineering Biomimetic Cellular Microenvironments. ACS Biomater Sci Eng 2021; 7:3997-4008. [PMID: 33523625 DOI: 10.1021/acsbiomaterials.0c01549] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Cells and their surrounding microenvironment exist in dynamic reciprocity, where bidirectional feedback and feedforward crosstalk drives essential processes in development, homeostasis, and disease. With the ongoing explosion of customizable biomaterial innovation for dynamic cell culture, an ever-expanding suite of user-programmable scaffolds now exists to probe cell fate in response to spatiotemporally controlled biophysical and biochemical cues. Here, we highlight emerging trends in these efforts, emphasizing strategies that offer tunability over complex network mechanics, present biomolecular cues anisotropically, and harness cells as physiochemical actuators of the pericellular niche. Altogether, these material advances will lead to breakthroughs in our basic understanding of how cells interact with, integrate signals from, and influence their surrounding microenvironment.
Collapse
Affiliation(s)
- Ross C Bretherton
- Department of Bioengineering, University of Washington, Seattle, Washington 98105, United States.,Institute of Stem Cell & Regenerative Medicine, University of Washington, Seattle, Washington 98109, United States
| | - Cole A DeForest
- Department of Bioengineering, University of Washington, Seattle, Washington 98105, United States.,Institute of Stem Cell & Regenerative Medicine, University of Washington, Seattle, Washington 98109, United States.,Department of Chemical Engineering, University of Washington, Seattle, Washington 98195, United States.,Department of Chemistry, University of Washington, Seattle, Washington 98195, United States.,Molecular Engineering & Sciences Institute, University of Washington, Seattle, Washington 98195, United States
| |
Collapse
|
31
|
Corbett DC, Fabyan WB, Grigoryan B, O'Connor CE, Johansson F, Batalov I, Regier MC, DeForest CA, Miller JS, Stevens KR. Thermofluidic heat exchangers for actuation of transcription in artificial tissues. SCIENCE ADVANCES 2020; 6:6/40/eabb9062. [PMID: 32998880 PMCID: PMC7527231 DOI: 10.1126/sciadv.abb9062] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Accepted: 08/13/2020] [Indexed: 05/12/2023]
Abstract
Spatial patterns of gene expression in living organisms orchestrate cell decisions in development, homeostasis, and disease. However, most methods for reconstructing gene patterning in 3D cell culture and artificial tissues are restricted by patterning depth and scale. We introduce a depth- and scale-flexible method to direct volumetric gene expression patterning in 3D artificial tissues, which we call "heat exchangers for actuation of transcription" (HEAT). This approach leverages fluid-based heat transfer from printed networks in the tissues to activate heat-inducible transgenes expressed by embedded cells. We show that gene expression patterning can be tuned both spatially and dynamically by varying channel network architecture, fluid temperature, fluid flow direction, and stimulation timing in a user-defined manner and maintained in vivo. We apply this approach to activate the 3D positional expression of Wnt ligands and Wnt/β-catenin pathway regulators, which are major regulators of development, homeostasis, regeneration, and cancer throughout the animal kingdom.
Collapse
Affiliation(s)
- Daniel C Corbett
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA
- Institute for Stem Cell and Regenerative Medicine, Seattle, WA 98195, USA
| | - Wesley B Fabyan
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA
- Institute for Stem Cell and Regenerative Medicine, Seattle, WA 98195, USA
| | - Bagrat Grigoryan
- Department of Bioengineering, Rice University, Houston, TX 77005, USA
| | - Colleen E O'Connor
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA
- Institute for Stem Cell and Regenerative Medicine, Seattle, WA 98195, USA
| | - Fredrik Johansson
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA
- Institute for Stem Cell and Regenerative Medicine, Seattle, WA 98195, USA
| | - Ivan Batalov
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA
- Institute for Stem Cell and Regenerative Medicine, Seattle, WA 98195, USA
| | - Mary C Regier
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA
- Institute for Stem Cell and Regenerative Medicine, Seattle, WA 98195, USA
| | - Cole A DeForest
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA
- Institute for Stem Cell and Regenerative Medicine, Seattle, WA 98195, USA
- Department of Chemical Engineering, University of Washington, Seattle, WA 98195, USA
| | - Jordan S Miller
- Department of Bioengineering, Rice University, Houston, TX 77005, USA
| | - Kelly R Stevens
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA.
- Institute for Stem Cell and Regenerative Medicine, Seattle, WA 98195, USA
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA 98195, USA
- Brotman Baty Institute, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
32
|
Abstract
Genetically engineered T-cells are being developed to perform a variety of therapeutic functions. However, no robust mechanisms exist to externally control the activity of T-cells at specific locations within the body. Such spatiotemporal control could help mitigate potential off-target toxicity due to incomplete molecular specificity in applications such as T-cell immunotherapy against solid tumors. Temperature is a versatile external control signal that can be delivered to target tissues in vivo using techniques such as focused ultrasound and magnetic hyperthermia. Here, we test the ability of heat shock promoters to mediate thermal actuation of genetic circuits in primary human T-cells in the well-tolerated temperature range of 37-42 °C, and introduce genetic architectures enabling the tuning of the amplitude and duration of thermal activation. We demonstrate the use of these circuits to control the expression of chimeric antigen receptors and cytokines, and the killing of target tumor cells. This technology provides a critical tool to direct the activity of T-cells after they are deployed inside the body.
Collapse
|
33
|
Combes F, Meyer E, Sanders NN. Immune cells as tumor drug delivery vehicles. J Control Release 2020; 327:70-87. [PMID: 32735878 DOI: 10.1016/j.jconrel.2020.07.043] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 07/24/2020] [Accepted: 07/25/2020] [Indexed: 12/21/2022]
Abstract
This review article describes the use of immune cells as potential candidates to deliver anti-cancer drugs deep within the tumor microenvironment. First, the rationale of using drug carriers to target tumors and potentially decrease drug-related side effects is discussed. We further explain some of the current limitations when using nanoparticles for this purpose. Next, a comprehensive step-by-step description of the migration cascade of immune cells is provided as well as arguments on why immune cells can be used to address some of the limitations associated with nanoparticle-mediated drug delivery. We then describe the benefits and drawbacks of using red blood cells, platelets, granulocytes, monocytes, macrophages, myeloid-derived suppressor cells, T cells and NK cells for tumor-targeted drug delivery. An additional section discusses the versatility of nanoparticles to load anti-cancer drugs into immune cells. Lastly, we propose increasing the circulatory half-life and development of conditional release strategies as the two main future pillars to improve the efficacy of immune cell-mediated drug delivery to tumors.
Collapse
Affiliation(s)
- Francis Combes
- Laboratory of Gene Therapy, Department of Nutrition, Genetics and Ethology, Faculty of Veterinary Medicine, Ghent University, Heidestraat 19, 9820 Merelbeke, Belgium; Cancer Research Institute Ghent (CRIG), 9000 Ghent, Belgium
| | - Evelyne Meyer
- Cancer Research Institute Ghent (CRIG), 9000 Ghent, Belgium; Department of Pharmacology, Toxicology and Biochemistry, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, 9820 Merelbeke, Belgium
| | - Niek N Sanders
- Laboratory of Gene Therapy, Department of Nutrition, Genetics and Ethology, Faculty of Veterinary Medicine, Ghent University, Heidestraat 19, 9820 Merelbeke, Belgium; Cancer Research Institute Ghent (CRIG), 9000 Ghent, Belgium.
| |
Collapse
|
34
|
Gamboa L, Phung EV, Li H, Meyers JP, Hart AC, Miller IC, Kwong GA. Heat-Triggered Remote Control of CRISPR-dCas9 for Tunable Transcriptional Modulation. ACS Chem Biol 2020; 15:533-542. [PMID: 31904924 DOI: 10.1021/acschembio.9b01005] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
CRISPR-associated proteins (Cas) are enabling powerful new approaches to control mammalian cell functions, yet the lack of spatially defined, noninvasive modalities limits their use as biological tools. Here, we integrate thermal gene switches with dCas9 complexes to confer remote control of gene activation and suppression with short pulses of heat. Using a thermal switch constructed from the heat shock protein A6 (HSPA6) locus, we show that a single heat pulse 3-5 °C above basal temperature is sufficient to trigger expression of dCas9 complexes. We demonstrate that dCas9 fused to the transcriptional activator VP64 is functional after heat activation, and, depending on the number of heat pulses, drives transcription of endogenous genes GzmB and CCL21 to levels equivalent to that achieved by a constitutive viral promoter. Across a range of input temperatures, we find that downstream protein expression of GzmB closely correlates with transcript levels (R2 = 0.99). Using dCas9 fused with the transcriptional suppressor KRAB, we show that longitudinal suppression of the reporter d2GFP depends on key thermal input parameters including pulse magnitude, number of pulses, and dose fractionation. In living mice, we extend our study using photothermal heating to spatially target implanted cells to suppress d2GFP in vivo. Our study establishes a noninvasive and targeted approach to harness Cas-based proteins for modulation of gene expression to complement current methods for remote control of cell function.
Collapse
Affiliation(s)
- Lena Gamboa
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia 30332, United States
| | - Erick V. Phung
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia 30332, United States
| | - Haoxin Li
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia 30332, United States
| | - Jared P. Meyers
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia 30332, United States
| | - Anna C. Hart
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia 30332, United States
| | - Ian C. Miller
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia 30332, United States
| | - Gabriel A. Kwong
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia 30332, United States
- Institute for Electronics and Nanotechnology, Georgia Institute of Technology, Atlanta, Georgia 30332, United States
- Parker H. Petit Institute of Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia 30332, United States
- Integrated Cancer Research Center, Georgia Institute of Technology, Atlanta, Georgia 30332, United States
- Georgia Immunoengineering Consortium, Emory University and Georgia Institute of Technology, Atlanta, Georgia 30332, United States
| |
Collapse
|
35
|
Gamboa L, Zamat AH, Kwong GA. Synthetic immunity by remote control. Theranostics 2020; 10:3652-3667. [PMID: 32206114 PMCID: PMC7069089 DOI: 10.7150/thno.41305] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Accepted: 02/03/2020] [Indexed: 12/11/2022] Open
Abstract
Cell-based immunotherapies, such as T cells engineered with chimeric antigen receptors (CARs), have the potential to cure patients of disease otherwise refractory to conventional treatments. Early-on-treatment and long-term durability of patient responses depend critically on the ability to control the potency of adoptively transferred T cells, as overactivation can lead to complications like cytokine release syndrome, and immunosuppression can result in ineffective responses to therapy. Drugs or biologics (e.g., cytokines) that modulate immune activity are limited by mass transport barriers that reduce the local effective drug concentration, and lack site or target cell specificity that results in toxicity. Emerging technologies that enable site-targeted, remote control of key T cell functions - including proliferation, antigen-sensing, and target-cell killing - have the potential to increase treatment precision and safety profile. These technologies are broadly applicable to other immune cells to expand immune cell therapies across many cancers and diseases. In this review, we highlight the opportunities, challenges and the current state-of-the-art for remote control of synthetic immunity.
Collapse
Affiliation(s)
- Lena Gamboa
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, Atlanta, GA 30332, USA
| | - Ali H. Zamat
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, Atlanta, GA 30332, USA
| | - Gabriel A. Kwong
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, Atlanta, GA 30332, USA
- Institute for Electronics and Nanotechnology, Georgia Institute of Technology, Atlanta, GA 30332, USA
- Parker H. Petit Institute of Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332, USA
- Integrated Cancer Research Center, Georgia Institute of Technology, Atlanta, GA 30332, USA
- Georgia Immunoengineering Consortium, Emory University and Georgia Institute of Technology, Atlanta, GA 30332, USA
| |
Collapse
|
36
|
Kang P, Li X, Liu Y, Shiers SI, Xiong H, Giannotta M, Dejana E, Price TJ, Randrianalisoa J, Nielsen SO, Qin Z. Transient Photoinactivation of Cell Membrane Protein Activity without Genetic Modification by Molecular Hyperthermia. ACS NANO 2019; 13:12487-12499. [PMID: 31613606 PMCID: PMC7096286 DOI: 10.1021/acsnano.9b01993] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/02/2023]
Abstract
Precise manipulation of protein activity in living systems has broad applications in biomedical sciences. However, it is challenging to use light to manipulate protein activity in living systems without genetic modification. Here, we report a technique to optically switch off protein activity in living cells with high spatiotemporal resolution, referred to as molecular hyperthermia (MH). MH is based on the nanoscale-confined heating of plasmonic gold nanoparticles by short laser pulses to unfold and photoinactivate targeted proteins of interest. First, we show that protease-activated receptor 2 (PAR2), a G-protein-coupled receptor and an important pathway that leads to pain sensitization, can be photoinactivated in situ by MH without compromising cell proliferation. PAR2 activity can be switched off in laser-targeted cells without affecting surrounding cells. Furthermore, we demonstrate the molecular specificity of MH by inactivating PAR2 while leaving other receptors intact. Second, we demonstrate that the photoinactivation of a tight junction protein in brain endothelial monolayers leads to a reversible blood-brain barrier opening in vitro. Lastly, the protein inactivation by MH is below the nanobubble generation threshold and thus is predominantly due to the nanoscale heating. MH is distinct from traditional hyperthermia (that induces global tissue heating) in both its time and length scales: nanoseconds versus seconds, nanometers versus millimeters. Our results demonstrate that MH enables selective and remote manipulation of protein activity and cellular behavior without genetic modification.
Collapse
Affiliation(s)
- Peiyuan Kang
- Department of Mechanical Engineering, University of Texas at Dallas, 800 West Campbell Rd., Richardson, Texas 75080, USA
| | - Xiaoqing Li
- Department of Bioengineering, University of Texas at Dallas, 800 West Campbell Rd., Richardson, Texas 75080, USA
| | - Yaning Liu
- Department of Mechanical Engineering, University of Texas at Dallas, 800 West Campbell Rd., Richardson, Texas 75080, USA
| | - Stephanie I. Shiers
- School of Behavioral and Brain Sciences, University of Texas at Dallas, 800 West Campbell Rd., Richardson, Texas 75080, USA
| | - Hejian Xiong
- Department of Mechanical Engineering, University of Texas at Dallas, 800 West Campbell Rd., Richardson, Texas 75080, USA
| | - Monica Giannotta
- Vascular Biology Laboratory, The FIRC Institute of Molecular Oncology (IFOM), 20139 Milan, Italy
| | - Elisabetta Dejana
- Vascular Biology Laboratory, The FIRC Institute of Molecular Oncology (IFOM), 20139 Milan, Italy
- Department of Immunology, Genetics and Pathology, University of Uppsala, 751 05 Uppsala, Sweden
| | - Theodore John Price
- School of Behavioral and Brain Sciences, University of Texas at Dallas, 800 West Campbell Rd., Richardson, Texas 75080, USA
| | - Jaona Randrianalisoa
- Institut de Thermique, Mécanique, Matériaux (ITheMM EA 7548), University of Reims Champagne-Ardenne, Reims Cedex 2, 51687 France
| | - Steven O. Nielsen
- Department of Chemistry and Biochemistry, University of Texas at Dallas, 800 West Campbell Rd., Richardson, Texas 75080, USA
| | - Zhenpeng Qin
- Department of Mechanical Engineering, University of Texas at Dallas, 800 West Campbell Rd., Richardson, Texas 75080, USA
- Department of Bioengineering, University of Texas at Dallas, 800 West Campbell Rd., Richardson, Texas 75080, USA
- Department of Surgery, University of Texas at Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, Texas 75390, USA
- Corresponding Author: . Phone: (972)883-4440
| |
Collapse
|
37
|
Dahotre SN, Chang YM, Romanov AM, Kwong GA. DNA-Barcoded pMHC Tetramers for Detection of Single Antigen-Specific T Cells by Digital PCR. Anal Chem 2019; 91:2695-2700. [PMID: 30656939 PMCID: PMC6399736 DOI: 10.1021/acs.analchem.8b04153] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Antigen-specific T cells are found at low frequencies in circulation but carry important diagnostic information as liquid biomarkers in numerous biomedical settings, such as monitoring the efficacy of vaccines and cancer immunotherapies. To enable detection of antigen-specific T cells with high sensitivity, we develop peptide-MHC (pMHC) tetramers labeled with DNA barcodes to detect single T cells by droplet digital PCR (ddPCR). We show that site-specific conjugation of DNA via photocleavable linkers allows barcoded tetramers to stain T cells with similar avidity compared to conventional fluorescent tetramers and efficient recovery of barcodes by light with no loss in cell viability. We design an orthogonal panel of DNA-barcoded tetramers to simultaneously detect multiple antigen-specific T cell populations, including from a mouse model of viral infection, and discriminate single cancer-specific T cells with high diagnostic sensitivity and specificity. This approach of DNA-barcoding can be broadened to encompass additional rare cells for monitoring immunological health at the single cell level.
Collapse
Affiliation(s)
- Shreyas N. Dahotre
- Department of Biomedical Engineering, Wallace H. Coulter, Georgia Tech and Emory School of Medicine, Atlanta, GA 30332, United States
| | - Yun Min Chang
- Department of Biomedical Engineering, Wallace H. Coulter, Georgia Tech and Emory School of Medicine, Atlanta, GA 30332, United States
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA 30322, United States
| | - Anna M. Romanov
- Department of Biomedical Engineering, Wallace H. Coulter, Georgia Tech and Emory School of Medicine, Atlanta, GA 30332, United States
| | - Gabriel A. Kwong
- Department of Biomedical Engineering, Wallace H. Coulter, Georgia Tech and Emory School of Medicine, Atlanta, GA 30332, United States
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Tech, Atlanta, GA 30332, United States
- Institute for Electronics and Nanotechnology, Georgia Tech, Atlanta, GA, 30332, United States
- Integrated Cancer Research Center, Georgia Tech, Atlanta, GA, 30332, United States
- Georgia ImmunoEngineering Consortium, Georgia Tech and Emory University, Atlanta, GA 30332, United States
| |
Collapse
|