1
|
Unda SR, Pomeranz LE, Marongiu R, Yu X, Kelly L, Hassanzadeh G, Molina H, Vaisey G, Wang P, Dyke JP, Fung EK, Grosenick L, Zirkel R, Antoniazzi AM, Norman S, Liston CM, Schaffer C, Nishimura N, Stanley SA, Friedman JM, Kaplitt MG. Bidirectional regulation of motor circuits using magnetogenetic gene therapy. SCIENCE ADVANCES 2024; 10:eadp9150. [PMID: 39383230 PMCID: PMC11463271 DOI: 10.1126/sciadv.adp9150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Accepted: 09/05/2024] [Indexed: 10/11/2024]
Abstract
Here, we report a magnetogenetic system, based on a single anti-ferritin nanobody-TRPV1 receptor fusion protein, which regulated neuronal activity when exposed to magnetic fields. Adeno-associated virus (AAV)-mediated delivery of a floxed nanobody-TRPV1 into the striatum of adenosine-2a receptor-Cre drivers resulted in motor freezing when placed in a magnetic resonance imaging machine or adjacent to a transcranial magnetic stimulation device. Functional imaging and fiber photometry confirmed activation in response to magnetic fields. Expression of the same construct in the striatum of wild-type mice along with a second injection of an AAVretro expressing Cre into the globus pallidus led to similar circuit specificity and motor responses. Last, a mutation was generated to gate chloride and inhibit neuronal activity. Expression of this variant in the subthalamic nucleus in PitX2-Cre parkinsonian mice resulted in reduced c-fos expression and motor rotational behavior. These data demonstrate that magnetogenetic constructs can bidirectionally regulate activity of specific neuronal circuits noninvasively in vivo using clinically available devices.
Collapse
Affiliation(s)
- Santiago R. Unda
- Laboratory of Molecular Neurosurgery, Department of Neurological Surgery, Weill Cornell Medical College, Cornell University, New York, NY 10065, USA
| | - Lisa E. Pomeranz
- Laboratory of Molecular Genetics, Rockefeller University, New York, NY 10065, USA
| | - Roberta Marongiu
- Laboratory of Molecular Neurosurgery, Department of Neurological Surgery, Weill Cornell Medical College, Cornell University, New York, NY 10065, USA
| | - Xiaofei Yu
- School of Life Sciences, Fudan University, Shanghai 200433, China
| | - Leah Kelly
- Laboratory of Molecular Genetics, Rockefeller University, New York, NY 10065, USA
| | | | - Henrik Molina
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10019, USA
| | - George Vaisey
- Laboratory of Molecular Neurobiology and Biophysics, Rockefeller University, New York, NY 10065, USA
| | - Putianqi Wang
- Laboratory of Molecular Genetics, Rockefeller University, New York, NY 10065, USA
| | - Jonathan P. Dyke
- Citigroup Bioimaging Center, Weill Cornell Medical College, Cornell University, New York, NY 10065, USA
| | - Edward K. Fung
- Citigroup Bioimaging Center, Weill Cornell Medical College, Cornell University, New York, NY 10065, USA
| | - Logan Grosenick
- Department of Psychiatry, Weill Cornell Medical College, Cornell University, New York, NY 10065, USA
| | - Rick Zirkel
- Meining School of Biomedical Engineering, Cornell University, Ithaca, NY 14850, USA
| | - Aldana M. Antoniazzi
- Laboratory of Molecular Neurosurgery, Department of Neurological Surgery, Weill Cornell Medical College, Cornell University, New York, NY 10065, USA
| | - Sofya Norman
- Laboratory of Molecular Neurosurgery, Department of Neurological Surgery, Weill Cornell Medical College, Cornell University, New York, NY 10065, USA
| | - Conor M. Liston
- Department of Psychiatry, Weill Cornell Medical College, Cornell University, New York, NY 10065, USA
| | - Chris Schaffer
- Meining School of Biomedical Engineering, Cornell University, Ithaca, NY 14850, USA
| | - Nozomi Nishimura
- Meining School of Biomedical Engineering, Cornell University, Ithaca, NY 14850, USA
| | - Sarah A. Stanley
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10019, USA
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10019, USA
| | - Jeffrey M. Friedman
- Laboratory of Molecular Genetics, Rockefeller University, New York, NY 10065, USA
- Howard Hughes Medical Institute, The Rockefeller University, New York, NY 10065, USA
| | - Michael G. Kaplitt
- Laboratory of Molecular Neurosurgery, Department of Neurological Surgery, Weill Cornell Medical College, Cornell University, New York, NY 10065, USA
| |
Collapse
|
2
|
Chuang KH, Qian C, Gilad AA, Pelled G. Magnetogenetic stimulation inside MRI induces spontaneous and evoked changes in neural circuits activity in rats. Front Neurosci 2024; 18:1459120. [PMID: 39411150 PMCID: PMC11473493 DOI: 10.3389/fnins.2024.1459120] [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] [Received: 07/03/2024] [Accepted: 09/16/2024] [Indexed: 10/19/2024] Open
Abstract
The ability to modulate specific neural circuits and simultaneously visualize and measure brain activity with MRI would greatly impact our understanding of brain function in health and disease. The combination of neurostimulation methods and functional MRI in animal models have already shown promise in elucidating fundamental mechanisms associated with brain activity. We developed an innovative magnetogenetics neurostimulation technology that can trigger neural activity through magnetic fields. Similar to other genetic-based neuromodulation methods, magnetogenetics offers cell-, area-, and temporal-specific control of neural activity. The magnetogenetic protein-Electromagnetic Perceptive Gene (EPG)-is activated by non-invasive magnetic fields, providing a unique way to target neural circuits by the MRI static and gradient fields while simultaneously measuring their effect on brain activity. EPG was expressed in rat's visual cortex and the amplitude of low-frequency fluctuation, resting-state functional connectivity (FC), and sensory activation was measured using a 7T MRI. The results demonstrate that EPG-expressing rats had significantly higher signal fluctuations in the visual areas and stronger FC in sensory areas consistent with known anatomical visuosensory and visuomotor connections. This new technology complements the existing neurostimulation toolbox and provides a means to study brain function in a minimally-invasive way which was not possible previously.
Collapse
Affiliation(s)
- Kai-Hsiang Chuang
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD, Australia
- Australian Research Council Training Centre for Innovation in Biomedical Imaging Technology, Brisbane, QLD, Australia
| | - Chunqi Qian
- Department of Radiology, Michigan State University, East Lansing, MI, United States
| | - Assaf A. Gilad
- Department of Radiology, Michigan State University, East Lansing, MI, United States
- Department of Chemical Engineering and Materials Science, Michigan State University, East Lansing, MI, United States
| | - Galit Pelled
- Department of Radiology, Michigan State University, East Lansing, MI, United States
- Department of Mechanical Engineering, Michigan State University, East Lansing, MI, United States
| |
Collapse
|
3
|
Ricci C, Abbandonato G, Giannangeli M, Matthews L, Almásy L, Sartori B, Podestà A, Caselli A, Boffi A, Thiel G, Del Favero E, Moroni A. Ferritin at different iron loading: From biological to nanotechnological applications. Int J Biol Macromol 2024; 276:133812. [PMID: 39032902 DOI: 10.1016/j.ijbiomac.2024.133812] [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: 05/21/2024] [Revised: 07/08/2024] [Accepted: 07/09/2024] [Indexed: 07/23/2024]
Abstract
The characterization of the structure of ferritin in solution and the arrangement of iron stored in its cavity are intriguing subjects for both cell biology and applied science, since the protein structure, stability, and easiness of production make it an ideal tool for biomedical applications. We characterized the ferritin structure over a wide range of iron loadings by visible light, X-ray, and neutron scattering techniques. We found that the arrangement of iron ions inside the protein cage resulted in a more disposable arrangement at lower loading factors and then in a crystalline structure. At very high iron content the inner core is composed of magnetite more than ferrihydrite, and the shell of the protein is elastically deformed by the iron crystal growth in an ellipsoidal arrangement. The application of an external radiofrequency (RF) magnetic field affected ferritins at low iron loading factors. Notably the RF modified the iron disposition towards a more dispersed arrangement. The structural characterization of the ferritin at different LFs and in presence of magnetic fields provides useful insights into their physiological behaviour and can help in the design and fine-tuning of ferritin-based nanosystems for biotechnological applications.
Collapse
Affiliation(s)
| | | | | | - Lauren Matthews
- ESRF, The European Synchrotron, 71 avenue des Martyrs, 38043 Grenoble, France
| | - László Almásy
- HUN-REN Centre for Energy Research, POB 49, Budapest 1525, Hungary
| | - Barbara Sartori
- Institute of Inorganic Chemistry, Graz University of Technology, Stremayrgasse 9/4, Graz, Austria
| | - Alessandro Podestà
- Department of Physics "Aldo Pontremoli", University of Milan, Milan, Italy
| | | | - Alberto Boffi
- Department of Biochemical Sciences, Sapienza University of Rome, Italy
| | - Gerhard Thiel
- Department of Biosciences, University of Milan, Milan, Italy
| | | | - Anna Moroni
- Department of Biosciences, University of Milan, Milan, Italy
| |
Collapse
|
4
|
Huang J, Fussenegger M. Programming mammalian cell behaviors by physical cues. Trends Biotechnol 2024:S0167-7799(24)00208-7. [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] [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
|
5
|
Ding Q, Liu L. Reprogramming cellular metabolism to increase the efficiency of microbial cell factories. Crit Rev Biotechnol 2024; 44:892-909. [PMID: 37380349 DOI: 10.1080/07388551.2023.2208286] [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: 11/17/2022] [Accepted: 04/11/2023] [Indexed: 06/30/2023]
Abstract
Recent studies are increasingly focusing on advanced biotechnological tools, self-adjusting smart microorganisms, and artificial intelligent networks, to engineer microorganisms with various functions. Microbial cell factories are a vital platform for improving the bioproduction of medicines, biofuels, and biomaterials from renewable carbon sources. However, these processes are significantly affected by cellular metabolism, and boosting the efficiency of microbial cell factories remains a challenge. In this review, we present a strategy for reprogramming cellular metabolism to enhance the efficiency of microbial cell factories for chemical biosynthesis, which improves our understanding of microbial physiology and metabolic control. Current methods are mainly focused on synthetic pathways, metabolic resources, and cell performance. This review highlights the potential biotechnological strategy to reprogram cellular metabolism and provide novel guidance for designing more intelligent industrial microbes with broader applications in this growing field.
Collapse
Affiliation(s)
- Qiang Ding
- School of Life Sciences, Anhui University, Hefei, China
- Key Laboratory of Human Microenvironment and Precision Medicine of Anhui Higher Education Institutes, Anhui University, Hefei, Anhui, China
- Anhui Key Laboratory of Modern Biomanufacturing, Hefei, Anhui, China
| | - Liming Liu
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China
| |
Collapse
|
6
|
Hernández-Morales M, Morales-Weil K, Han SM, Han V, Tran T, Benner EJ, Pegram K, Meanor J, Miller EW, Kramer RH, Liu C. Electrophysiological Mechanisms and Validation of Ferritin-Based Magnetogenetics for Remote Control of Neurons. J Neurosci 2024; 44:e1717232024. [PMID: 38777598 PMCID: PMC11270515 DOI: 10.1523/jneurosci.1717-23.2024] [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: 09/07/2023] [Revised: 05/07/2024] [Accepted: 05/10/2024] [Indexed: 05/25/2024] Open
Abstract
Magnetogenetics was developed to remotely control genetically targeted neurons. A variant of magnetogenetics uses magnetic fields to activate transient receptor potential vanilloid (TRPV) channels when coupled with ferritin. Stimulation with static or RF magnetic fields of neurons expressing these channels induces Ca2+ transients and modulates behavior. However, the validity of ferritin-based magnetogenetics has been questioned due to controversies surrounding the underlying mechanisms and deficits in reproducibility. Here, we validated the magnetogenetic approach Ferritin-iron Redistribution to Ion Channels (FeRIC) using electrophysiological (Ephys) and imaging techniques. Previously, interference from RF stimulation rendered patch-clamp recordings inaccessible for magnetogenetics. We solved this limitation for FeRIC, and we studied the bioelectrical properties of neurons expressing TRPV4 (nonselective cation channel) and transmembrane member 16A (TMEM16A; chloride-permeable channel) coupled to ferritin (FeRIC channels) under RF stimulation. We used cultured neurons obtained from the rat hippocampus of either sex. We show that RF decreases the membrane resistance (Rm) and depolarizes the membrane potential in neurons expressing TRPV4FeRIC RF does not directly trigger action potential firing but increases the neuronal basal spiking frequency. In neurons expressing TMEM16AFeRIC, RF decreases the Rm, hyperpolarizes the membrane potential, and decreases the spiking frequency. Additionally, we corroborated the previously described biochemical mechanism responsible for RF-induced activation of ferritin-coupled ion channels. We solved an enduring problem for ferritin-based magnetogenetics, obtaining direct Ephys evidence of RF-induced activation of ferritin-coupled ion channels. We found that RF does not yield instantaneous changes in neuronal membrane potentials. Instead, RF produces responses that are long-lasting and moderate, but effective in controlling the bioelectrical properties of neurons.
Collapse
Affiliation(s)
- Miriam Hernández-Morales
- Department of Electrical Engineering and Computer Sciences, University of California, Berkeley, California 94720
- Helen Wills Neuroscience Institute, University of California, Berkeley, California 94720
| | - Koyam Morales-Weil
- Department of Electrical Engineering and Computer Sciences, University of California, Berkeley, California 94720
- Helen Wills Neuroscience Institute, University of California, Berkeley, California 94720
| | - Sang Min Han
- Department of Electrical Engineering and Computer Sciences, University of California, Berkeley, California 94720
- Helen Wills Neuroscience Institute, University of California, Berkeley, California 94720
| | - Victor Han
- Department of Electrical Engineering and Computer Sciences, University of California, Berkeley, California 94720
- Helen Wills Neuroscience Institute, University of California, Berkeley, California 94720
| | - Tiffany Tran
- Helen Wills Neuroscience Institute, University of California, Berkeley, California 94720
| | - Eric J Benner
- Division of Neonatology, Department of Pediatrics, Duke University Medical Center, Jean and George Brumley, Jr. Neonatal-Perinatal Institute, Durham, North Carolina 27710
| | - Kelly Pegram
- Division of Neonatology, Department of Pediatrics, Duke University Medical Center, Jean and George Brumley, Jr. Neonatal-Perinatal Institute, Durham, North Carolina 27710
| | - Jenna Meanor
- Division of Neonatology, Department of Pediatrics, Duke University Medical Center, Jean and George Brumley, Jr. Neonatal-Perinatal Institute, Durham, North Carolina 27710
| | - Evan W Miller
- Helen Wills Neuroscience Institute, University of California, Berkeley, California 94720
- Department of Chemistry, University of California, Berkeley, California 94720
- Department of Molecular and Cell Biology, University of California, Berkeley, California 94720
| | - Richard H Kramer
- Department of Molecular and Cell Biology, University of California, Berkeley, California 94720
| | - Chunlei Liu
- Department of Electrical Engineering and Computer Sciences, University of California, Berkeley, California 94720
- Helen Wills Neuroscience Institute, University of California, Berkeley, California 94720
| |
Collapse
|
7
|
Vyas A, Petrášek Z, Nidetzky B. Limits of Non-invasive Enzymatic Activation by Local Temperature Control. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2312220. [PMID: 38344893 DOI: 10.1002/smll.202312220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Indexed: 07/26/2024]
Abstract
Enzymatic activity depends on and can therefore be regulated by temperature. Selective modulation of the activity of different enzymes in one reaction pot would require temperature control local to each type of enzyme. It has been suggested previously that immobilization of enzyme on magnetic nanoparticles and exposing them to alternating magnetic field can enhance the reaction rate. This enhancement has been explained as being mediated by temperature increase caused by dissipation of the absorbed field energy in the form of heat. However, the possibility of spatially limiting this temperature increase on the microscale has been questioned. Here, it is investigated whether an activity enhancement of the enzyme sucrose phosphorylase immobilized on magnetic beads can be achieved, how this effect is related to the increase in temperature, and whether temperature differences within one reaction pot could be generated in this way. It is found that alternating magnetic field stimulation leads to increased enzymatic activity fully attributable to the increase of bulk temperature. Both theoretical analysis and experimental data indicate that no local heating near the particle surface takes place. It is further concluded that relevant increase of surface temperature can be obtained only with macroscopic, millimeter-sized, magnetic particles.
Collapse
Affiliation(s)
- Anisha Vyas
- Institute of Biotechnology and Biochemical Engineering, Graz University of Technology, Petersgasse 12, Graz, A-8010, Austria
| | - Zdeněk Petrášek
- Institute of Biotechnology and Biochemical Engineering, Graz University of Technology, Petersgasse 12, Graz, A-8010, Austria
| | - Bernd Nidetzky
- Institute of Biotechnology and Biochemical Engineering, Graz University of Technology, Petersgasse 12, Graz, A-8010, Austria
- Austrian Centre of Industrial Biotechnology, Krenngasse 37, Graz, A-8010, Austria
| |
Collapse
|
8
|
Latypova AA, Yaremenko AV, Pechnikova NA, Minin AS, Zubarev IV. Magnetogenetics as a promising tool for controlling cellular signaling pathways. J Nanobiotechnology 2024; 22:327. [PMID: 38858689 PMCID: PMC11163773 DOI: 10.1186/s12951-024-02616-z] [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: 03/28/2024] [Accepted: 06/04/2024] [Indexed: 06/12/2024] Open
Abstract
Magnetogenetics emerges as a transformative approach for modulating cellular signaling pathways through the strategic application of magnetic fields and nanoparticles. This technique leverages the unique properties of magnetic nanoparticles (MNPs) to induce mechanical or thermal stimuli within cells, facilitating the activation of mechano- and thermosensitive proteins without the need for traditional ligand-receptor interactions. Unlike traditional modalities that often require invasive interventions and lack precision in targeting specific cellular functions, magnetogenetics offers a non-invasive alternative with the capacity for deep tissue penetration and the potential for targeting a broad spectrum of cellular processes. This review underscores magnetogenetics' broad applicability, from steering stem cell differentiation to manipulating neuronal activity and immune responses, highlighting its potential in regenerative medicine, neuroscience, and cancer therapy. Furthermore, the review explores the challenges and future directions of magnetogenetics, including the development of genetically programmed magnetic nanoparticles and the integration of magnetic field-sensitive cells for in vivo applications. Magnetogenetics stands at the forefront of cellular manipulation technologies, offering novel insights into cellular signaling and opening new avenues for therapeutic interventions.
Collapse
Affiliation(s)
- Anastasiia A Latypova
- Institute of Future Biophysics, Dolgoprudny, 141701, Russia
- Moscow Center for Advanced Studies, Moscow, 123592, Russia
| | - Alexey V Yaremenko
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA.
- Aristotle University of Thessaloniki, Thessaloniki, 54124, Greece.
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, 117997, Russia.
| | - Nadezhda A Pechnikova
- Aristotle University of Thessaloniki, Thessaloniki, 54124, Greece
- Saint Petersburg Pasteur Institute, Saint Petersburg, 197101, Russia
| | - Artem S Minin
- M.N. Mikheev Institute of Metal Physics of the Ural Branch of the Russian Academy of Sciences, Yekaterinburg, 620108, Russia
| | - Ilya V Zubarev
- Institute of Future Biophysics, Dolgoprudny, 141701, Russia.
| |
Collapse
|
9
|
Chen C, Chen H, Wang P, Wang X, Wang X, Chen C, Pan W. Reactive Oxygen Species Activate a Ferritin-Linked TRPV4 Channel under a Static Magnetic Field. ACS Chem Biol 2024; 19:1151-1160. [PMID: 38648729 DOI: 10.1021/acschembio.4c00090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/25/2024]
Abstract
Magnetogenetics has shown great potential for cell function and neuromodulation using heat or force effects under different magnetic fields; however, there is still a contradiction between experimental effects and underlying mechanisms by theoretical computation. In this study, we aimed to investigate the role of reactive oxygen species (ROS) in mechanical force-dependent regulation from a physicochemical perspective. The transient receptor potential vanilloid 4 (TRPV4) cation channels fused to ferritin (T4F) were overexpressed in HEK293T cells and exposed to static magnetic fields (sMF, 1.4-5.0 mT; gradient: 1.62 mT/cm). An elevation of ROS levels was found under sMF in T4F-overexpressing cells, which could lead to lipid oxidation. Compared with the overexpression of TRPV4, ferritin in T4F promoted the generation of ROS under the stimulation of sMF, probably related to the release of iron ions from ferritin. Then, the resulting ROS regulated the opening of the TRPV4 channel, which was attenuated by the direct addition of ROS inhibitors or an iron ion chelator, highlighting a close relationship among iron release, ROS production, and TRPV4 channel activation. Taken together, these findings indicate that the produced ROS under sMF act on the TRPV4 channel, regulating the influx of calcium ions. The study would provide a scientific basis for the application of magnetic regulation in cellular or neural regulation and disease treatment and contribute to the development of the more sensitive regulatory technology.
Collapse
Affiliation(s)
- Changyou Chen
- Beijing Key Laboratory of Bioelectromagnetism, Institute of Electrical Engineering, Chinese Academy of Sciences, Beijing 100190, China
- France-China International Laboratory of Evolution and Development of Magnetotactic Multicellular Organisms, Beijing 100190, China
| | - Haitao Chen
- Beijing Key Laboratory of Bioelectromagnetism, Institute of Electrical Engineering, Chinese Academy of Sciences, Beijing 100190, China
- France-China International Laboratory of Evolution and Development of Magnetotactic Multicellular Organisms, Beijing 100190, China
| | - Pingping Wang
- Beijing Key Laboratory of Bioelectromagnetism, Institute of Electrical Engineering, Chinese Academy of Sciences, Beijing 100190, China
- France-China International Laboratory of Evolution and Development of Magnetotactic Multicellular Organisms, Beijing 100190, China
| | - Xue Wang
- Beijing Key Laboratory of Bioelectromagnetism, Institute of Electrical Engineering, Chinese Academy of Sciences, Beijing 100190, China
- France-China International Laboratory of Evolution and Development of Magnetotactic Multicellular Organisms, Beijing 100190, China
- University of the Chinese Academy of Sciences, Beijing 100049, China
| | - Xuting Wang
- Beijing Key Laboratory of Bioelectromagnetism, Institute of Electrical Engineering, Chinese Academy of Sciences, Beijing 100190, China
- France-China International Laboratory of Evolution and Development of Magnetotactic Multicellular Organisms, Beijing 100190, China
- University of the Chinese Academy of Sciences, Beijing 100049, China
| | - Chuanfang Chen
- Beijing Key Laboratory of Bioelectromagnetism, Institute of Electrical Engineering, Chinese Academy of Sciences, Beijing 100190, China
- France-China International Laboratory of Evolution and Development of Magnetotactic Multicellular Organisms, Beijing 100190, China
| | - Weidong Pan
- Beijing Key Laboratory of Bioelectromagnetism, Institute of Electrical Engineering, Chinese Academy of Sciences, Beijing 100190, China
- France-China International Laboratory of Evolution and Development of Magnetotactic Multicellular Organisms, Beijing 100190, China
| |
Collapse
|
10
|
Chen C, Chen H, Wang P, Wang X, Wang X, Chen C. Ca 2+ Overload Decreased Cellular Viability in Magnetic Hyperthermia without a Macroscopic Temperature Rise. ACS Biomater Sci Eng 2024; 10:2995-3005. [PMID: 38654432 DOI: 10.1021/acsbiomaterials.3c01875] [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: 04/26/2024]
Abstract
Magnetic hyperthermia is a crucial medical engineering technique for treating diseases, which usually uses alternating magnetic fields (AMF) to interplay with magnetic substances to generate heat. Recently, it has been found that in some cases, there is no detectable temperature increment after applying an AMF, which caused corresponding effects surprisingly. The mechanisms involved in this phenomenon are not yet fully understood. In this study, we aimed to explore the role of Ca2+ overload in the magnetic hyperthermia effect without a perceptible temperature rise. A cellular system expressing the fusion proteins TRPV1 and ferritin was prepared. The application of an AMF (518 kHz, 16 kA/m) could induce the fusion protein to release a large amount of iron ions, which then participates in the production of massive reactive oxygen radicals (ROS). Both ROS and its induced lipid oxidation enticed the opening of ion channels, causing intracellular Ca2+ overload, which further led to decreased cellular viability. Taken together, Ca2+ overload triggered by elevated ROS and the induced oxidation of lipids contributes to the magnetic hyperthermia effect without a perceptible temperature rise. These findings would be beneficial for expanding the application of temperature-free magnetic hyperthermia, such as in cellular and neural regulation, design of new cancer treatment methods.
Collapse
Affiliation(s)
- Changyou Chen
- Beijing Key Laboratory of Bioelectromagnetism, Institute of Electrical Engineering, Chinese Academy of Sciences, Beijing 100190, China
- France-China International Laboratory of Evolution and Development of Magnetotactic Multicellular Organisms, Beijing 100190, China
| | - Haitao Chen
- Beijing Key Laboratory of Bioelectromagnetism, Institute of Electrical Engineering, Chinese Academy of Sciences, Beijing 100190, China
- France-China International Laboratory of Evolution and Development of Magnetotactic Multicellular Organisms, Beijing 100190, China
| | - Pingping Wang
- Beijing Key Laboratory of Bioelectromagnetism, Institute of Electrical Engineering, Chinese Academy of Sciences, Beijing 100190, China
- France-China International Laboratory of Evolution and Development of Magnetotactic Multicellular Organisms, Beijing 100190, China
| | - Xue Wang
- Beijing Key Laboratory of Bioelectromagnetism, Institute of Electrical Engineering, Chinese Academy of Sciences, Beijing 100190, China
- France-China International Laboratory of Evolution and Development of Magnetotactic Multicellular Organisms, Beijing 100190, China
- University of the Chinese Academy of Sciences, Beijing 100049, China
| | - Xuting Wang
- Beijing Key Laboratory of Bioelectromagnetism, Institute of Electrical Engineering, Chinese Academy of Sciences, Beijing 100190, China
- France-China International Laboratory of Evolution and Development of Magnetotactic Multicellular Organisms, Beijing 100190, China
- University of the Chinese Academy of Sciences, Beijing 100049, China
| | - Chuanfang Chen
- Beijing Key Laboratory of Bioelectromagnetism, Institute of Electrical Engineering, Chinese Academy of Sciences, Beijing 100190, China
- France-China International Laboratory of Evolution and Development of Magnetotactic Multicellular Organisms, Beijing 100190, China
| |
Collapse
|
11
|
Unda SR, Pomeranz LE, Marongiu R, Yu X, Kelly L, Hassanzadeh G, Molina H, Vaisey G, Wang P, Dyke JP, Fung EK, Grosenick L, Zirkel R, Antoniazzi AM, Norman S, Liston CM, Schaffer C, Nishimura N, Stanley SA, Friedman JM, Kaplitt MG. Bidirectional Regulation of Motor Circuits Using Magnetogenetic Gene Therapy Short: Magnetogenetic Regulation of Motor Circuits. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.07.13.548699. [PMID: 37503198 PMCID: PMC10369996 DOI: 10.1101/2023.07.13.548699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
Here we report a novel suite of magnetogenetic tools, based on a single anti-ferritin nanobody-TRPV1 receptor fusion protein, which regulated neuronal activity when exposed to magnetic fields. AAV-mediated delivery of a floxed nanobody-TRPV1 into the striatum of adenosine 2a receptor-cre driver mice resulted in motor freezing when placed in an MRI or adjacent to a transcranial magnetic stimulation (TMS) device. Functional imaging and fiber photometry both confirmed activation of the target region in response to the magnetic fields. Expression of the same construct in the striatum of wild-type mice along with a second injection of an AAVretro expressing cre into the globus pallidus led to similar circuit specificity and motor responses. Finally, a mutation was generated to gate chloride and inhibit neuronal activity. Expression of this variant in subthalamic nucleus in PitX2-cre parkinsonian mice resulted in reduced local c-fos expression and motor rotational behavior. These data demonstrate that magnetogenetic constructs can bidirectionally regulate activity of specific neuronal circuits non-invasively in-vivo using clinically available devices.
Collapse
Affiliation(s)
- Santiago R. Unda
- Laboratory of Molecular Neurosurgery, Department of Neurological Surgery, Weill Cornell Medical College, Cornell University; New York, NY, USA
| | - Lisa E. Pomeranz
- Laboratory of Molecular Genetics, Rockefeller University; New York, NY, USA
| | - Roberta Marongiu
- Laboratory of Molecular Neurosurgery, Department of Neurological Surgery, Weill Cornell Medical College, Cornell University; New York, NY, USA
| | - Xiaofei Yu
- School of Life Sciences, Fudan University, Shanghai, 200433
| | - Leah Kelly
- Laboratory of Molecular Genetics, Rockefeller University; New York, NY, USA
| | | | - Henrik Molina
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - George Vaisey
- Laboratory of Molecular Neurobiology and Biophysics, Rockefeller University, New York, NY 10065, USA
| | - Putianqi Wang
- Laboratory of Molecular Genetics, Rockefeller University; New York, NY, USA
| | - Jonathan P. Dyke
- Citigroup Bioimaging Center, Weill Cornell Medical College, Cornell University; New York, NY, USA
| | - Edward K. Fung
- Citigroup Bioimaging Center, Weill Cornell Medical College, Cornell University; New York, NY, USA
| | - Logan Grosenick
- Department of Psychiatry, Weill Cornell Medical College, Cornell University; New York, NY, USA
| | - Rick Zirkel
- Meining School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | - Aldana M. Antoniazzi
- Laboratory of Molecular Neurosurgery, Department of Neurological Surgery, Weill Cornell Medical College, Cornell University; New York, NY, USA
| | - Sofya Norman
- Laboratory of Molecular Neurosurgery, Department of Neurological Surgery, Weill Cornell Medical College, Cornell University; New York, NY, USA
| | - Conor M. Liston
- Department of Psychiatry, Weill Cornell Medical College, Cornell University; New York, NY, USA
| | - Chris Schaffer
- Meining School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | - Nozomi Nishimura
- Meining School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | - Sarah A. Stanley
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Michael G. Kaplitt
- Laboratory of Molecular Neurosurgery, Department of Neurological Surgery, Weill Cornell Medical College, Cornell University; New York, NY, USA
| |
Collapse
|
12
|
Pomeranz L, Li R, Yu X, Kelly L, Hassanzadeh G, Molina H, Gross D, Brier M, Vaisey G, Wang P, Jimenez-Gonzalez M, Garcia-Ocana A, Dordick J, Friedman J, Stanley S. Magnetogenetic cell activation using endogenous ferritin. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.06.20.545120. [PMID: 37786709 PMCID: PMC10541561 DOI: 10.1101/2023.06.20.545120] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/04/2023]
Abstract
The ability to precisely control the activity of defined cell populations enables studies of their physiological roles and may provide therapeutic applications. While prior studies have shown that magnetic activation of ferritin-tagged ion channels allows cell-specific modulation of cellular activity, the large size of the constructs made the use of adeno-associated virus, AAV, the vector of choice for gene therapy, impractical. In addition, simple means for generating magnetic fields of sufficient strength have been lacking. Toward these ends, we first generated a novel anti-ferritin nanobody that when fused to transient receptor potential cation channel subfamily V member 1, TRPV1, enables direct binding of the channel to endogenous ferritin in mouse and human cells. This smaller construct can be delivered in a single AAV and we validated that it robustly enables magnetically induced cell activation in vitro. In parallel, we developed a simple benchtop electromagnet capable of gating the nanobody-tagged channel in vivo. Finally, we showed that delivering these new constructs by AAV to pancreatic beta cells in combination with the benchtop magnetic field delivery stimulates glucose-stimulated insulin release to improve glucose tolerance in mice in vivo. Together, the novel anti-ferritin nanobody, nanobody-TRPV1 construct and new hardware advance the utility of magnetogenetics in animals and potentially humans.
Collapse
Affiliation(s)
- Lisa Pomeranz
- Laboratory of Molecular Genetics, Rockefeller University, New York, NY 10065, USA
| | - Rosemary Li
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Xiaofei Yu
- School of Life Sciences, Fudan University, Shanghai, 200433
| | - Leah Kelly
- Laboratory of Molecular Genetics, Rockefeller University, New York, NY 10065, USA
| | | | - Henrik Molina
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Daniel Gross
- Current address, Dept. of Radiology, Weill Cornell Medicine, 1300 York Avenue New York, NY 10065
| | - Matthew Brier
- Department of Chemical and Biological Engineering, Rensselaer Polytechnic Institute, Troy, NY 12180
| | - George Vaisey
- Laboratory of Molecular Neurobiology and Biophysics, Rockefeller University, New York, NY 10065, USA
| | - Putianqi Wang
- Laboratory of Molecular Genetics, Rockefeller University, New York, NY 10065, USA
| | - Maria Jimenez-Gonzalez
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Adolfo Garcia-Ocana
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Molecular and Cellular Endocrinology, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute, City of Hope, Duarte, CA, 91010
| | - Jonathan Dordick
- Department of Chemical and Biological Engineering, Rensselaer Polytechnic Institute, Troy, NY 12180
| | - Jeffrey Friedman
- Laboratory of Molecular Genetics, Rockefeller University, New York, NY 10065, USA
| | - Sarah Stanley
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| |
Collapse
|
13
|
Grady CJ, Castellanos Franco EA, Schossau J, Ashbaugh RC, Pelled G, Gilad AA. A putative design for the electromagnetic activation of split proteins for molecular and cellular manipulation. Front Bioeng Biotechnol 2024; 12:1355915. [PMID: 38605993 PMCID: PMC11007078 DOI: 10.3389/fbioe.2024.1355915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 03/05/2024] [Indexed: 04/13/2024] Open
Abstract
The ability to manipulate cellular function using an external stimulus is a powerful strategy for studying complex biological phenomena. One approach to modulate the function of the cellular environment is split proteins. In this method, a biologically active protein or an enzyme is fragmented so that it reassembles only upon a specific stimulus. Although many tools are available to induce these systems, nature has provided other mechanisms to expand the split protein toolbox. Here, we show a novel method for reconstituting split proteins using magnetic stimulation. We found that the electromagnetic perceptive gene (EPG) changes conformation due to magnetic field stimulation. By fusing split fragments of a certain protein to both termini of the EPG, the fragments can be reassembled into a functional protein under magnetic stimulation due to conformational change. We show this effect with three separate split proteins: NanoLuc, APEX2, and herpes simplex virus type-1 thymidine kinase. Our results show, for the first time, that reconstitution of split proteins can be achieved only with magnetic fields. We anticipate that this study will be a starting point for future magnetically inducible split protein designs for cellular perturbation and manipulation. With this technology, we can help expand the toolbox of the split protein platform and allow better elucidation of complex biological systems.
Collapse
Affiliation(s)
- Connor J. Grady
- Department of Biomedical Engineering, Michigan State University, East Lansing, MI, United States
| | | | - Jory Schossau
- Department of Chemical Engineering and Materials Science, Michigan State University, East Lansing, MI, United States
- Department of Computer Science and Engineering, Michigan State University, East Lansing, MI, United States
| | - Ryan C. Ashbaugh
- Department of Electrical and Computer Engineering, Michigan State University, East Lansing, MI, United States
| | - Galit Pelled
- Department of Mechanical Engineering, Michigan State University, East Lansing, MI, United States
- Department of Radiology, Michigan State University, East Lansing, MI, United States
| | - Assaf A. Gilad
- Department of Chemical Engineering and Materials Science, Michigan State University, East Lansing, MI, United States
- Department of Radiology, Michigan State University, East Lansing, MI, United States
| |
Collapse
|
14
|
Hahmann J, Ishaqat A, Lammers T, Herrmann A. Sonogenetics for Monitoring and Modulating Biomolecular Function by Ultrasound. Angew Chem Int Ed Engl 2024; 63:e202317112. [PMID: 38197549 DOI: 10.1002/anie.202317112] [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: 11/10/2023] [Revised: 01/01/2024] [Accepted: 01/08/2024] [Indexed: 01/11/2024]
Abstract
Ultrasound technology, synergistically harnessed with genetic engineering and chemistry concepts, has started to open the gateway to the remarkable realm of sonogenetics-a pioneering paradigm for remotely orchestrating cellular functions at the molecular level. This fusion not only enables precisely targeted imaging and therapeutic interventions, but also advances our comprehension of mechanobiology to unparalleled depths. Sonogenetic tools harness mechanical force within small tissue volumes while preserving the integrity of the surrounding physiological environment, reaching depths of up to tens of centimeters with high spatiotemporal precision. These capabilities circumvent the inherent physical limitations of alternative in vivo control methods such as optogenetics and magnetogenetics. In this review, we first discuss mechanosensitive ion channels, the most commonly utilized sonogenetic mediators, in both mammalian and non-mammalian systems. Subsequently, we provide a comprehensive overview of state-of-the-art sonogenetic approaches that leverage thermal or mechanical features of ultrasonic waves. Additionally, we explore strategies centered around the design of mechanochemically reactive macromolecular systems. Furthermore, we delve into the realm of ultrasound imaging of biomolecular function, encompassing the utilization of gas vesicles and acoustic reporter genes. Finally, we shed light on limitations and challenges of sonogenetics and present a perspective on the future of this promising technology.
Collapse
Affiliation(s)
- Johannes Hahmann
- Institute of Technical and Macromolecular Chemistry, RWTH Aachen University, Worringerweg 1, 52074, Aachen, Germany
- DWI - Leibniz Institute for Interactive Materials, Forckenbeckstr. 50, 52074, Aachen, Germany
- Max Planck School Matter to Life, Jahnstr. 29, 69120, Heidelberg, Germany
| | - Aman Ishaqat
- Institute of Technical and Macromolecular Chemistry, RWTH Aachen University, Worringerweg 1, 52074, Aachen, Germany
- DWI - Leibniz Institute for Interactive Materials, Forckenbeckstr. 50, 52074, Aachen, Germany
| | - Twan Lammers
- Institute for Experimental Molecular Imaging (ExMI), Center for Biohybrid Medical Systems (CBMS), RWTH Aachen University Clinic, Forckenbeckstr. 55, 52074, Aachen, Germany
| | - Andreas Herrmann
- Institute of Technical and Macromolecular Chemistry, RWTH Aachen University, Worringerweg 1, 52074, Aachen, Germany
- DWI - Leibniz Institute for Interactive Materials, Forckenbeckstr. 50, 52074, Aachen, Germany
- Max Planck School Matter to Life, Jahnstr. 29, 69120, Heidelberg, Germany
| |
Collapse
|
15
|
Mundell JW, Brier MI, Orloff E, Stanley SA, Dordick JS. Alternating magnetic fields drive stimulation of gene expression via generation of reactive oxygen species. iScience 2024; 27:109186. [PMID: 38420587 PMCID: PMC10901079 DOI: 10.1016/j.isci.2024.109186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 12/23/2023] [Accepted: 02/06/2024] [Indexed: 03/02/2024] Open
Abstract
Magnetogenetics represents a method for remote control of cellular function. Previous work suggests that generation of reactive oxygen species (ROS) initiates downstream signaling. Herein, a chemical biology approach was used to elucidate further the mechanism of radio frequency-alternating magnetic field (RF-AMF) stimulation of a TRPV1-ferritin magnetogenetics platform that leads to Ca2+ flux. RF-AMF stimulation of HEK293T cells expressing TRPV1-ferritin resulted in ∼30% and ∼140% increase in intra- and extracellular ROS levels, respectively. Mutations to specific cysteine residues in TRPV1 responsible for ROS sensitivity eliminated RF-AMF driven Ca2+-dependent transcription of secreted embryonic alkaline phosphatase (SEAP). Using a non-tethered (to TRPV1) ferritin also eliminated RF-AMF driven SEAP production, and using specific inhibitors, ROS-activated TRPV1 signaling involves protein kinase C, NADPH oxidase, and the endoplasmic reticulum. These results suggest ferritin-dependent ROS activation of TRPV1 plays a key role in the initiation of magnetogenetics, and provides relevance for potential applications in medicine and biotechnology.
Collapse
Affiliation(s)
- Jordan W. Mundell
- Department of Chemical and Biological Engineering, Rensselaer Polytechnic Institute, Troy, NY 12180, USA
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY 12180, USA
| | - Matthew I. Brier
- Department of Chemical and Biological Engineering, Rensselaer Polytechnic Institute, Troy, NY 12180, USA
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY 12180, USA
| | - Everest Orloff
- Department of Chemical and Biological Engineering, Rensselaer Polytechnic Institute, Troy, NY 12180, USA
| | - Sarah A. Stanley
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Jonathan S. Dordick
- Department of Chemical and Biological Engineering, Rensselaer Polytechnic Institute, Troy, NY 12180, USA
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY 12180, USA
- Departments of Biomedical Engineering and Biological Sciences, Rensselaer Polytechnic Institute, Troy, NY 12180, USA
| |
Collapse
|
16
|
Lu Y, Gu F, Ma Y, Li R, Luo Y, Da X, Jiang L, Li X, Liu Y. Simultaneous Delivery of Doxorubicin and EZH2-Targeting siRNA by Vortex Magnetic Nanorods Synergistically Improved Anti-Tumor Efficacy in Triple-Negative Breast Cancer. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2301307. [PMID: 37376877 DOI: 10.1002/smll.202301307] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 05/19/2023] [Indexed: 06/29/2023]
Abstract
Triple-negative breast cancer (TNBC), one of the most aggressive types of breast cancer, currently lacks a targeted therapy and has a high clinical recurrence rate. The present study reports an engineered magnetic nanodrug based on Fe3 O4 vortex nanorods coated with a macrophage membrane loaded with doxorubicin (DOX) and Enhancer of zeste 2 polycomb repressive complex 2 subunit (EZH2) siRNA. This novel nanodrug displays excellent tissue penetration and preferential tumor accumulation. More importantly, it significantly increases tumor suppression compared to chemotherapy, suggesting the synergistic activity of the combination of doxorubicin and EZH2-inhibition. Importantly, owing to tumor-targeted delivery, nanomedicine shows an excellent safety profile after systemic delivery, unlike conventional chemotherapy. In summary, chemotherapy and gene therapy are combined into a novel magnetic nanodrug carrying doxorubicin and EZH2 siRNA, which shows promising clinical application potential in TNBC therapy.
Collapse
Affiliation(s)
- Yunshu Lu
- Department of Breast Surgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, P. R. China
| | - Fenfen Gu
- Department of Clinical Pharmacy, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, P. R. China
| | - Yuwei Ma
- Department of Breast Surgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, P. R. China
| | - Ruonan Li
- Department of Clinical Pharmacy, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, P. R. China
- School of Materials and Chemistry, University of Shanghai for Science and Technology, Shanghai, 200093, P. R. China
| | - Yi Luo
- Biotheus Inc., Guangdong Province, Zhuhai, 519080, P. R. China
- Clinical Pharmacy Innovation Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, P. R. China
| | - Xianhong Da
- Department of Clinical Pharmacy, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, P. R. China
- School of Materials and Chemistry, University of Shanghai for Science and Technology, Shanghai, 200093, P. R. China
| | - Lan Jiang
- Department of Clinical Pharmacy, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, P. R. China
- School of Materials and Chemistry, University of Shanghai for Science and Technology, Shanghai, 200093, P. R. China
| | - Xiang Li
- School of Materials and Chemistry, University of Shanghai for Science and Technology, Shanghai, 200093, P. R. China
| | - Yan Liu
- Department of Clinical Pharmacy, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, P. R. China
- Clinical Pharmacy Innovation Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, P. R. China
| |
Collapse
|
17
|
Huang J, Xue S, Buchmann P, Teixeira AP, Fussenegger M. An electrogenetic interface to program mammalian gene expression by direct current. Nat Metab 2023; 5:1395-1407. [PMID: 37524785 PMCID: PMC10447240 DOI: 10.1038/s42255-023-00850-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 06/23/2023] [Indexed: 08/02/2023]
Abstract
Wearable electronic devices are playing a rapidly expanding role in the acquisition of individuals' health data for personalized medical interventions; however, wearables cannot yet directly program gene-based therapies because of the lack of a direct electrogenetic interface. Here we provide the missing link by developing an electrogenetic interface that we call direct current (DC)-actuated regulation technology (DART), which enables electrode-mediated, time- and voltage-dependent transgene expression in human cells using DC from batteries. DART utilizes a DC supply to generate non-toxic levels of reactive oxygen species that act via a biosensor to reversibly fine-tune synthetic promoters. In a proof-of-concept study in a type 1 diabetic male mouse model, a once-daily transdermal stimulation of subcutaneously implanted microencapsulated engineered human cells by energized acupuncture needles (4.5 V DC for 10 s) stimulated insulin release and restored normoglycemia. We believe this technology will enable wearable electronic devices to directly program metabolic interventions.
Collapse
Affiliation(s)
- Jinbo Huang
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
| | - Shuai Xue
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
| | - Peter Buchmann
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
| | - Ana Palma Teixeira
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
| | - Martin Fussenegger
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland.
- Faculty of Science, University of Basel, Basel, Switzerland.
| |
Collapse
|
18
|
Seo JY, Park SB, Kim SY, Seo GJ, Jang HK, Lee TJ. Acoustic and Magnetic Stimuli-Based Three-Dimensional Cell Culture Platform for Tissue Engineering. Tissue Eng Regen Med 2023; 20:563-580. [PMID: 37052782 PMCID: PMC10313605 DOI: 10.1007/s13770-023-00539-8] [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: 12/22/2022] [Revised: 02/16/2023] [Accepted: 03/15/2023] [Indexed: 04/14/2023] Open
Abstract
In a conventional two-dimensional (2D) culture method, cells are attached to the bottom of the culture dish and grow into a monolayer. These 2D culture methods are easy to handle, cost-effective, reproducible, and adaptable to growing many different types of cells. However, monolayer 2D cell culture conditions are far from those of natural tissue, indicating the need for a three-dimensional (3D) culture system. Various methods, such as hanging drop, scaffolds, hydrogels, microfluid systems, and bioreactor systems, have been utilized for 3D cell culture. Recently, external physical stimulation-based 3D cell culture platforms, such as acoustic and magnetic forces, were introduced. Acoustic waves can establish acoustic radiation force, which can induce suspended objects to gather in the pressure node region and aggregate to form clusters. Magnetic targeting consists of two components, a magnetically responsive carrier and a magnetic field gradient source. In a magnetic-based 3D cell culture platform, cells are aggregated by changing the magnetic force. Magnetic fields can manipulate cells through two different methods: positive magnetophoresis and negative magnetophoresis. Positive magnetophoresis is a way of imparting magnetic properties to cells by labeling them with magnetic nanoparticles. Negative magnetophoresis is a label-free principle-based method. 3D cell structures, such as spheroids, 3D network structures, and cell sheets, have been successfully fabricated using this acoustic and magnetic stimuli-based 3D cell culture platform. Additionally, fabricated 3D cell structures showed enhanced cell behavior, such as differentiation potential and tissue regeneration. Therefore, physical stimuli-based 3D cell culture platforms could be promising tools for tissue engineering.
Collapse
Affiliation(s)
- Ju Yeon Seo
- Division of Biomedical Convergence, Department of Medical Biotechnology, College of Biomedical Science, Kangwon National University, Chuncheon-si, Gangwon-do, 24341, Republic of Korea
- Department of Biomedical Science, Kangwon National University, Chuncheon-si, Gangwon-do, 24341, Republic of Korea
| | - Song Bin Park
- Department of Bio-Health Technology, College of Biomedical Science, Kangwon National University, Chuncheon-si, Gangwon-do, 24341, Republic of Korea
| | - Seo Yeon Kim
- Division of Biomedical Convergence, Department of Medical Biotechnology, College of Biomedical Science, Kangwon National University, Chuncheon-si, Gangwon-do, 24341, Republic of Korea
| | - Gyeong Jin Seo
- Division of Biomedical Convergence, Department of Medical Biotechnology, College of Biomedical Science, Kangwon National University, Chuncheon-si, Gangwon-do, 24341, Republic of Korea
| | - Hyeon-Ki Jang
- Division of Chemical Engineering and Bioengineering, College of Art Culture and Engineering, Kangwon National University, Chuncheon-si, Gangwon-do, 24341, Republic of Korea
| | - Tae-Jin Lee
- Division of Biomedical Convergence, Department of Medical Biotechnology, College of Biomedical Science, Kangwon National University, Chuncheon-si, Gangwon-do, 24341, Republic of Korea.
- Department of Bio-Health Convergence, Kangwon National University, Chuncheon-si, Gangwon-do, 24341, Republic of Korea.
| |
Collapse
|
19
|
Lee S, Jiao M, Zhang Z, Yu Y. Nanoparticles for Interrogation of Cell Signaling. ANNUAL REVIEW OF ANALYTICAL CHEMISTRY (PALO ALTO, CALIF.) 2023; 16:333-351. [PMID: 37314874 PMCID: PMC10627408 DOI: 10.1146/annurev-anchem-092822-085852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Cell functions rely on signal transduction-the cascades of molecular interactions and biochemical reactions that relay extracellular signals to the cell interior. Dissecting principles governing the signal transduction process is critical for the fundamental understanding of cell physiology and the development of biomedical interventions. The complexity of cell signaling is, however, beyond what is accessible by conventional biochemistry assays. Thanks to their unique physical and chemical properties, nanoparticles (NPs) have been increasingly used for the quantitative measurement and manipulation of cell signaling. Even though research in this area is still in its infancy, it has the potential to yield new, paradigm-shifting knowledge of cell biology and lead to biomedical innovations. To highlight this importance, we summarize in this review studies that pioneered the development and application of NPs for cell signaling, from quantitative measurements of signaling molecules to spatiotemporal manipulation of cell signal transduction.
Collapse
Affiliation(s)
- Seonik Lee
- Department of Chemistry, Indiana University, Bloomington, Indiana, USA;
| | - Mengchi Jiao
- Department of Chemistry, Indiana University, Bloomington, Indiana, USA;
| | - Zihan Zhang
- Department of Chemistry, Indiana University, Bloomington, Indiana, USA;
| | - Yan Yu
- Department of Chemistry, Indiana University, Bloomington, Indiana, USA;
| |
Collapse
|
20
|
Karatum O, Han M, Erdogan ET, Karamursel S, Nizamoglu S. Physical mechanisms of emerging neuromodulation modalities. J Neural Eng 2023; 20:031001. [PMID: 37224804 DOI: 10.1088/1741-2552/acd870] [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: 11/16/2022] [Accepted: 05/24/2023] [Indexed: 05/26/2023]
Abstract
One of the ultimate goals of neurostimulation field is to design materials, devices and systems that can simultaneously achieve safe, effective and tether-free operation. For that, understanding the working mechanisms and potential applicability of neurostimulation techniques is important to develop noninvasive, enhanced, and multi-modal control of neural activity. Here, we review direct and transduction-based neurostimulation techniques by discussing their interaction mechanisms with neurons via electrical, mechanical, and thermal means. We show how each technique targets modulation of specific ion channels (e.g. voltage-gated, mechanosensitive, heat-sensitive) by exploiting fundamental wave properties (e.g. interference) or engineering nanomaterial-based systems for efficient energy transduction. Overall, our review provides a detailed mechanistic understanding of neurostimulation techniques together with their applications toin vitro, in vivo, and translational studies to guide the researchers toward developing more advanced systems in terms of noninvasiveness, spatiotemporal resolution, and clinical applicability.
Collapse
Affiliation(s)
- Onuralp Karatum
- Department of Electrical and Electronics Engineering, Koc University, Istanbul 34450, Turkey
| | - Mertcan Han
- Department of Electrical and Electronics Engineering, Koc University, Istanbul 34450, Turkey
| | - Ezgi Tuna Erdogan
- Department of Physiology, Koc University School of Medicine, Istanbul 34450, Turkey
| | - Sacit Karamursel
- Department of Physiology, Koc University School of Medicine, Istanbul 34450, Turkey
| | - Sedat Nizamoglu
- Department of Electrical and Electronics Engineering, Koc University, Istanbul 34450, Turkey
- Department of Biomedical Science and Engineering, Koc University, Istanbul 34450, Turkey
| |
Collapse
|
21
|
Ma T, Ding Q, Liu C, Wu H. Electromagnetic fields regulate calcium-mediated cell fate of stem cells: osteogenesis, chondrogenesis and apoptosis. Stem Cell Res Ther 2023; 14:133. [PMID: 37194107 DOI: 10.1186/s13287-023-03303-w] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 03/28/2023] [Indexed: 05/18/2023] Open
Abstract
Electromagnetic fields (EMF) are increasing in popularity as a safe and non-invasive therapy. On the one hand, it is widely acknowledged that EMF can regulate the proliferation and differentiation of stem cells, promoting the undifferentiated cells capable of osteogenesis, angiogenesis, and chondroblast differentiation to achieve bone repair purpose. On the other hand, EMF can inhibit tumor stem cells proliferation and promote apoptosis to suppress tumor growth. As an essential second messenger, intracellular calcium plays a role in regulating cell cycle, such as proliferation, differentiation and apoptosis. There is increasing evidence that the modulation of intracellular calcium ion by EMF leads to differential outcomes in different stem cells. This review summarizes the regulation of channels, transporters, and ion pumps by EMF-induced calcium oscillations. It furtherly discusses the role of molecules and pathways activated by EMF-dependent calcium oscillations in promoting bone and cartilage repair and inhibiting tumor stem cells growth.
Collapse
Affiliation(s)
- Tian Ma
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Qing Ding
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Chaoxu Liu
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China.
| | - Hua Wu
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China.
| |
Collapse
|
22
|
Quintana D, Bounds HA, Brown J, Wang M, Bhatla N, Wiegert JS, Adesnik H. Dissociating instructive from permissive roles of brain circuits with reversible neural activity manipulations. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.11.540397. [PMID: 37214966 PMCID: PMC10197619 DOI: 10.1101/2023.05.11.540397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Neuroscientists rely on targeted perturbations and lesions to causally map functions in the brain1. Yet, since the brain is highly interconnected, manipulation of one area can impact behavior through indirect effects on many other brain regions, complicating the interpretation of such results2,3. On the other hand, the often-observed recovery of behavior performance after lesion can cast doubt on whether the lesioned area was ever directly involved4,5. Recent studies have highlighted how the results of acute and irreversible inactivation can directly conflict4-6, making it unclear whether a brain area is instructive or merely permissive in a specific brain function. To overcome this challenge, we developed a three-stage optogenetic approach which leverages the ability to precisely control the temporal period of regional inactivation with either brief or sustained illumination. Using a visual detection task, we found that acute optogenetic inactivation of the primary visual cortex (V1) suppressed task performance if cortical inactivation was intermittent across trials within each behavioral session. However, when we inactivated V1 for entire behavioral sessions, animals quickly recovered performance in just one to two days. Most importantly, after returning these recovered animals to intermittent cortical inactivation, they quickly reverted to failing on optogenetic inactivation trials. These data support a revised model where the cortex is the default circuit that instructs perceptual performance in basic sensory tasks. More generally, this novel, temporally controllable optogenetic perturbation paradigm can be broadly applied to brain circuits and specific cell types to assess whether they are instructive or merely permissive in a brain function or behavior.
Collapse
Affiliation(s)
- Daniel Quintana
- Department of Molecular and Cell Biology, University of California, Berkeley
| | - Hayley A Bounds
- Department of Molecular and Cell Biology, University of California, Berkeley
- The Helen Wills Neuroscience Institute
| | - Jennifer Brown
- Department of Molecular and Cell Biology, University of California, Berkeley
| | - May Wang
- Department of Molecular and Cell Biology, University of California, Berkeley
| | - Nikhil Bhatla
- Department of Molecular and Cell Biology, University of California, Berkeley
| | - J Simon Wiegert
- University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Department of Neurophysiology, MCTN, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Hillel Adesnik
- Department of Molecular and Cell Biology, University of California, Berkeley
- The Helen Wills Neuroscience Institute
| |
Collapse
|
23
|
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: 18] [Impact Index Per Article: 18.0] [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
|
24
|
Maity D, Guha Ray P, Buchmann P, Mansouri M, Fussenegger M. Blood-Glucose-Powered Metabolic Fuel Cell for Self-Sufficient Bioelectronics. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2300890. [PMID: 36893359 DOI: 10.1002/adma.202300890] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 02/28/2023] [Indexed: 05/26/2023]
Abstract
Currently available bioelectronic devices consume too much power to be continuously operated on rechargeable batteries, and are often powered wirelessly, with attendant issues regarding reliability, convenience, and mobility. Thus, the availability of a robust, self-sufficient, implantable electrical power generator that works under physiological conditions would be transformative for many applications, from driving bioelectronic implants and prostheses to programing cellular behavior and patients' metabolism. Here, capitalizing on a new copper-containing, conductively tuned 3D carbon nanotube composite, an implantable blood-glucose-powered metabolic fuel cell is designed that continuously monitors blood-glucose levels, converts excess glucose into electrical power during hyperglycemia, and produces sufficient energy (0.7 mW cm-2 , 0.9 V, 50 mm glucose) to drive opto- and electro-genetic regulation of vesicular insulin release from engineered beta cells. It is shown that this integration of blood-glucose monitoring with elimination of excessive blood glucose by combined electro-metabolic conversion and insulin-release-mediated cellular consumption enables the metabolic fuel cell to restore blood-glucose homeostasis in an automatic, self-sufficient, and closed-loop manner in an experimental model of type-1 diabetes.
Collapse
Affiliation(s)
- Debasis Maity
- Department of Biosystems Science and Engineering, ETH Zurich, Mattenstrasse 26, Basel, CH-4058, Switzerland
| | - Preetam Guha Ray
- Department of Biosystems Science and Engineering, ETH Zurich, Mattenstrasse 26, Basel, CH-4058, Switzerland
| | - Peter Buchmann
- Department of Biosystems Science and Engineering, ETH Zurich, Mattenstrasse 26, Basel, CH-4058, Switzerland
| | - Maysam Mansouri
- 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
|
25
|
Central Nervous System Nanotechnology. Nanomedicine (Lond) 2023. [DOI: 10.1007/978-981-16-8984-0_29] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
|
26
|
Martinez-Sanchez N, Sweeney O, Sidarta-Oliveira D, Caron A, Stanley SA, Domingos AI. The sympathetic nervous system in the 21st century: Neuroimmune interactions in metabolic homeostasis and obesity. Neuron 2022; 110:3597-3626. [PMID: 36327900 PMCID: PMC9986959 DOI: 10.1016/j.neuron.2022.10.017] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 08/23/2022] [Accepted: 10/10/2022] [Indexed: 11/06/2022]
Abstract
The sympathetic nervous system maintains metabolic homeostasis by orchestrating the activity of organs such as the pancreas, liver, and white and brown adipose tissues. From the first renderings by Thomas Willis to contemporary techniques for visualization, tracing, and functional probing of axonal arborizations within organs, our understanding of the sympathetic nervous system has started to grow beyond classical models. In the present review, we outline the evolution of these findings and provide updated neuroanatomical maps of sympathetic innervation. We offer an autonomic framework for the neuroendocrine loop of leptin action, and we discuss the role of immune cells in regulating sympathetic terminals and metabolism. We highlight potential anti-obesity therapeutic approaches that emerge from the modern appreciation of SNS as a neural network vis a vis the historical fear of sympathomimetic pharmacology, while shifting focus from post- to pre-synaptic targeting. Finally, we critically appraise the field and where it needs to go.
Collapse
Affiliation(s)
| | - Owen Sweeney
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3PT, UK
| | - Davi Sidarta-Oliveira
- Physician-Scientist Graduate Program, Obesity and Comorbidities Research Center, School of Medical Sciences, University of Campinas, Campinas, Brazil
| | - Alexandre Caron
- Faculty of Pharmacy, Université Laval, Québec City, QC G1V 0A6, Canada
| | - Sarah A Stanley
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Ana I Domingos
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3PT, UK.
| |
Collapse
|
27
|
Shin W, Jeong S, Lee JU, Jeong SY, Shin J, Kim HH, Cheon J, Lee JH. Magnetogenetics with Piezo1 Mechanosensitive Ion Channel for CRISPR Gene Editing. NANO LETTERS 2022; 22:7415-7422. [PMID: 36069378 DOI: 10.1021/acs.nanolett.2c02314] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Regulation of genetic activity in single cells and tissues is pivotal to determine key cellular functions in current biomedicine, yet the conventional biochemical activators lack spatiotemporal precision due to the diffusion-mediated slow kinetics and nonselectivity. Here, we describe a magnetogenetic method for target-specific activation of a clustered regularly interspaced short palindromic repeats (CRISPR) system for the regulation of intracellular proteins. We used magnetomechanical force generated by the magnetic nanostructure to activate pre-encoded Piezo1, the mechanosensitive ion channel, on the target cell. The activated Piezo1 further triggers the intracellular Ca2+ signaling pathway, inducing the pre-encoded genes to express genes of interest (GOIs), which is Cas9 protein for the CRISPR regulation of the target proteins. We demonstrated that this magnetogenetic CRISPR system successfully edits the target genome for both in vitro and pseudo-in vivo environments, providing a versatile magnetic platform for remote gene editing of animals with various size scales.
Collapse
Affiliation(s)
- Wookjin Shin
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul 03722, Republic of Korea
| | - Sumin Jeong
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul 03722, Republic of Korea
- Department of Chemistry, Yonsei University, Seoul 03722, Republic of Korea
| | - Jung-Uk Lee
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul 03722, Republic of Korea
- Department of Chemistry, Yonsei University, Seoul 03722, Republic of Korea
| | - Soo Yeun Jeong
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul 03722, Republic of Korea
| | - Jeonghong Shin
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul 03722, Republic of Korea
- Department of Pharmacology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
- Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Hyongbum Henry Kim
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul 03722, Republic of Korea
- Department of Pharmacology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
- Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Jinwoo Cheon
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul 03722, Republic of Korea
- Graduate Program of Nano Biomedical Engineering (NanoBME), Advanced Science Institute, Yonsei University, Seoul 03722, Republic of Korea
- Department of Chemistry, Yonsei University, Seoul 03722, Republic of Korea
| | - Jae-Hyun Lee
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul 03722, Republic of Korea
- Graduate Program of Nano Biomedical Engineering (NanoBME), Advanced Science Institute, Yonsei University, Seoul 03722, Republic of Korea
| |
Collapse
|
28
|
Mao Z, Lin X, Wang P, Yan H. Iron oxide nanoparticles for biomedical applications: an updated patent review (2015-2021). Expert Opin Ther Pat 2022; 32:939-952. [PMID: 35929879 DOI: 10.1080/13543776.2022.2109413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Iron oxide nanoparticles (IONPs) hold the edges of great magnetic properties and fine nanoparticle characteristics, making them an attractive therapeutic agent. In the past seven years, more in-depth investigations were devoted to the intrinsic structure, magnetic properties, and biological effects of IONPs, expanding the range of their therapeutic application scenes. AREAS COVERED This review focuses on the development of IONPs for biomedical applications from the angle of the patent literature reported during the period 2015-2021. EXPERT OPINION While the magnetic properties of IONPs have been extensively explored, the precise control of IONP behavior through external magnetic fields remains a challenge. Further digging into the biological effects of IONPs will facilitate the development of IONP-based immune therapies. Long-term reliable safety evaluations are of necessity and significance to promote the process of clinical translation.
Collapse
Affiliation(s)
- Zeyuan Mao
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing, People's Republic of China
| | - Xin Lin
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing, People's Republic of China
| | - Pengfei Wang
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing, People's Republic of China
| | - Hailong Yan
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing, People's Republic of China
| |
Collapse
|
29
|
Sebesta C, Torres Hinojosa D, Wang B, Asfouri J, Li Z, Duret G, Jiang K, Xiao Z, Zhang L, Zhang Q, Colvin VL, Goetz SM, Peterchev AV, Dierick HA, Bao G, Robinson JT. Subsecond multichannel magnetic control of select neural circuits in freely moving flies. NATURE MATERIALS 2022; 21:951-958. [PMID: 35761060 PMCID: PMC10965118 DOI: 10.1038/s41563-022-01281-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Accepted: 05/09/2022] [Indexed: 06/15/2023]
Abstract
Precisely timed activation of genetically targeted cells is a powerful tool for the study of neural circuits and control of cell-based therapies. Magnetic control of cell activity, or 'magnetogenetics', using magnetic nanoparticle heating of temperature-sensitive ion channels enables remote, non-invasive activation of neurons for deep-tissue applications and freely behaving animal studies. However, the in vivo response time of thermal magnetogenetics is currently tens of seconds, which prevents precise temporal modulation of neural activity. Moreover, magnetogenetics has yet to achieve in vivo multiplexed stimulation of different groups of neurons. Here we produce subsecond behavioural responses in Drosophila melanogaster by combining magnetic nanoparticles with a rate-sensitive thermoreceptor (TRPA1-A). Furthermore, by tuning magnetic nanoparticles to respond to different magnetic field strengths and frequencies, we achieve subsecond, multichannel stimulation. These results bring magnetogenetics closer to the temporal resolution and multiplexed stimulation possible with optogenetics while maintaining the minimal invasiveness and deep-tissue stimulation possible only by magnetic control.
Collapse
Affiliation(s)
- Charles Sebesta
- Department of Bioengineering, Rice University, Houston, TX, USA
| | | | - Boshuo Wang
- Department of Psychiatry & Behavioral Sciences, School of Medicine, Duke University, Durham, NC, USA
| | - Joseph Asfouri
- Department of Electrical and Computer Engineering, Rice University, Houston, TX, USA
| | - Zhongxi Li
- Department of Electrical and Computer Engineering, School of Engineering, Duke University, Durham, NC, USA
| | - Guillaume Duret
- Department of Electrical and Computer Engineering, Rice University, Houston, TX, USA
| | - Kaiyi Jiang
- Department of Bioengineering, Rice University, Houston, TX, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Zhen Xiao
- Department of Chemistry, Brown University, Providence, RI, USA
| | - Linlin Zhang
- Department of Bioengineering, Rice University, Houston, TX, USA
| | - Qingbo Zhang
- Department of Bioengineering, Rice University, Houston, TX, USA
| | - Vicki L Colvin
- Department of Chemistry, Brown University, Providence, RI, USA
| | - Stefan M Goetz
- Department of Psychiatry & Behavioral Sciences, School of Medicine, Duke University, Durham, NC, USA
- Department of Electrical and Computer Engineering, School of Engineering, Duke University, Durham, NC, USA
- Department of Neurosurgery, School of Medicine, Duke University, Durham, NC, USA
- Institute of Brain Sciences, Duke University, Durham, NC, USA
- Department of Engineering, School of Technology, University of Cambridge, Cambridge, UK
| | - Angel V Peterchev
- Department of Psychiatry & Behavioral Sciences, School of Medicine, Duke University, Durham, NC, USA
- Department of Electrical and Computer Engineering, School of Engineering, Duke University, Durham, NC, USA
- Department of Neurosurgery, School of Medicine, Duke University, Durham, NC, USA
- Department of Biomedical Engineering, School of Engineering, Duke University, Durham, NC, USA
| | - Herman A Dierick
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
| | - Gang Bao
- Department of Bioengineering, Rice University, Houston, TX, USA
| | - Jacob T Robinson
- Department of Bioengineering, Rice University, Houston, TX, USA.
- Department of Electrical and Computer Engineering, Rice University, Houston, TX, USA.
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
30
|
Zhao H, Xue S, Hussherr MD, Teixeira AP, Fussenegger M. Autonomous push button-controlled rapid insulin release from a piezoelectrically activated subcutaneous cell implant. SCIENCE ADVANCES 2022; 8:eabm4389. [PMID: 35704573 PMCID: PMC9200281 DOI: 10.1126/sciadv.abm4389] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Accepted: 04/28/2022] [Indexed: 05/25/2023]
Abstract
Traceless physical cues are desirable for remote control of the in situ production and real-time dosing of biopharmaceuticals in cell-based therapies. However, current optogenetic, magnetogenetic, or electrogenetic devices require sophisticated electronics, complex artificial intelligence-assisted software, and external energy supplies for power and control. Here, we describe a self-sufficient subcutaneous push button-controlled cellular implant powered simply by repeated gentle finger pressure exerted on the overlying skin. Pushing the button causes transient percutaneous deformation of the implant's embedded piezoelectric membrane, which produces sufficient low-voltage energy inside a semi-permeable platinum-coated cell chamber to mediate rapid release of a biopharmaceutical from engineered electro-sensitive human cells. Release is fine-tuned by varying the frequency and duration of finger-pressing stimulation. As proof of concept, we show that finger-pressure activation of the subcutaneous implant can restore normoglycemia in a mouse model of type 1 diabetes. Self-sufficient push-button devices may provide a new level of convenience for patients to control their cell-based therapies.
Collapse
Affiliation(s)
- Haijie Zhao
- Department of Biosystems Science and Engineering, ETH Zurich, Mattenstrasse 26, CH-4058 Basel, Switzerland
| | - Shuai Xue
- Department of Biosystems Science and Engineering, ETH Zurich, Mattenstrasse 26, CH-4058 Basel, Switzerland
| | - Marie-Didiée Hussherr
- Department of Biosystems Science and Engineering, ETH Zurich, Mattenstrasse 26, CH-4058 Basel, Switzerland
| | - Ana Palma Teixeira
- Department of Biosystems Science and Engineering, ETH Zurich, Mattenstrasse 26, CH-4058 Basel, Switzerland
| | - Martin Fussenegger
- Department of Biosystems Science and Engineering, ETH Zurich, Mattenstrasse 26, CH-4058 Basel, Switzerland
- Faculty of Science, University of Basel, Mattenstrasse 26, CH-4058 Basel, Switzerland
| |
Collapse
|
31
|
Improvement of synaptic plasticity by nanoparticles and the related mechanisms: Applications and prospects. J Control Release 2022; 347:143-163. [PMID: 35513209 DOI: 10.1016/j.jconrel.2022.04.049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 04/27/2022] [Accepted: 04/28/2022] [Indexed: 11/20/2022]
Abstract
Synaptic plasticity is an important basis of learning and memory and participates in brain network remodelling after different types of brain injury (such as that caused by neurodegenerative diseases, cerebral ischaemic injury, posttraumatic stress disorder (PTSD), and psychiatric disorders). Therefore, improving synaptic plasticity is particularly important for the treatment of nervous system-related diseases. With the rapid development of nanotechnology, increasing evidence has shown that nanoparticles (NPs) can cross the blood-brain barrier (BBB) in different ways, directly or indirectly act on nerve cells, regulate synaptic plasticity, and ultimately improve nerve function. Therefore, to better elucidate the effect of NPs on synaptic plasticity, we review evidence showing that NPs can improve synaptic plasticity by regulating different influencing factors, such as neurotransmitters, receptors, presynaptic membrane proteins and postsynaptic membrane proteins, and further discuss the possible mechanism by which NPs improve synaptic plasticity. We conclude that NPs can improve synaptic plasticity and restore the function of damaged nerves by inhibiting neuroinflammation and oxidative stress, inducing autophagy, and regulating ion channels on the cell membrane. By reviewing the mechanism by which NPs regulate synaptic plasticity and the applications of NPs for the treatment of neurological diseases, we also propose directions for future research in this field and provide an important reference for follow-up research.
Collapse
|
32
|
Xie C, Kang P, Cazals J, Castelán OM, Randrianalisoa J, Qin Z. Single pulse heating of a nanoparticle array for biological applications. NANOSCALE ADVANCES 2022; 4:2090-2097. [PMID: 35530423 PMCID: PMC9063739 DOI: 10.1039/d1na00766a] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/24/2021] [Accepted: 02/07/2022] [Indexed: 06/14/2023]
Abstract
With the ability to convert external excitation into heat, nanomaterials play an essential role in many biomedical applications. Two modes of nanoparticle (NP) array heating, nanoscale-confined heating (NCH) and macroscale-collective heating (MCH), have been found and extensively studied. Despite this, the resulting biological response at the protein level remains elusive. In this study, we developed a computational model to systematically investigate the single-pulsed heating of the NP array and corresponding protein denaturation/activation. We found that NCH may lead to targeted protein denaturation, however, nanoparticle heating does not lead to nanoscale selective TRPV1 channel activation. The excitation duration and NP concentration are primary factors that determine a window for targeted protein denaturation, and together with heating power, we defined quantified boundaries for targeted protein denaturation. Our results boost our understandings of the NCH and MCH under realistic physical constraints and provide robust guidance to customize biomedical platforms with desired NP heating.
Collapse
Affiliation(s)
- Chen Xie
- Department of Mechanical Engineering, University of Texas at Dallas800 West Campbell Road EW31RichardsonTexas 75080USA
| | - Peiyuan Kang
- Department of Mechanical Engineering, University of Texas at Dallas800 West Campbell Road EW31RichardsonTexas 75080USA
| | - Johan Cazals
- Department of Mechanical Engineering, University of Texas at Dallas800 West Campbell Road EW31RichardsonTexas 75080USA
| | - Omar Morales Castelán
- Department of Mechanical Engineering, University of Texas at Dallas800 West Campbell Road EW31RichardsonTexas 75080USA
| | - Jaona Randrianalisoa
- Institut de Thermique, Mécanique, Matériaux (ITheMM EA 7548), University of Reims Champagne-ArdenneReimsCedex 251687France
| | - Zhenpeng Qin
- Department of Mechanical Engineering, University of Texas at Dallas800 West Campbell Road EW31RichardsonTexas 75080USA
- Department of Bioengineering, Center for Advanced Pain Studies, University of Texas at Dallas800 West Campbell RoadRichardsonTexas 75080USA
- Department of Surgery, University of Texas at Southwestern Medical Center5323 Harry Hines BoulevardDallasTexas 75390USA
| |
Collapse
|
33
|
Hydrothermal Synthesis of β-Nb 2ZnO 6 Nanoparticles for Photocatalytic Degradation of Methyl Orange and Cytotoxicity Study. Int J Mol Sci 2022; 23:ijms23094777. [PMID: 35563173 PMCID: PMC9105501 DOI: 10.3390/ijms23094777] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Revised: 04/05/2022] [Accepted: 04/24/2022] [Indexed: 02/01/2023] Open
Abstract
β-Nb2ZnO6 nanoparticles were synthesized by a hydrothermal process and calcined at two temperatures, 500 °C and 700 °C, and assigned as A and B, respectively. X-ray diffraction, together with transmission electron microscopy, revealed that the β-Nb2ZnO6 nanoparticles calcined at 700 °C (B) were more crystalline than the β-Nb2ZnO6 calcined at 500 °C (A) with both types of nanoparticles having an average size of approximately 100 nm. The physiochemical, photocatalytic, and cytotoxic activities of both types of β-Nb2ZnO6 nanoparticles (A and B) were examined. Interestingly, the photodegradation of methyl orange, used as a standard for environmental pollutants, was faster in the presence of the β-Nb2ZnO6 nanoparticles calcined at 500 °C (A) than in the presence of those calcined at 700 °C (B). Moreover, the cytotoxicity was evaluated against different types of cancer cells and the results indicated that both types of β-Nb2ZnO6 nanoparticles (A and B) exhibited high cytotoxicity against MCF-7 and HCT116 cells but low cytotoxicity against HeLa cells after 24 and 48 h of treatment. Overall, both products expressed similar EC50 values on tested cell lines and high cytotoxicity after 72 h of treatment. As a photocatalyst, β-Nb2ZnO6 nanoparticles (A) could be utilized in different applications including the purification of the environment and water from specific pollutants. Further biological studies are required to determine the other potential impacts of utilizing β-Nb2ZnO6 nanoparticles in the biomedical application field.
Collapse
|
34
|
Zou L, Xu K, Tian H, Fang Y. Remote neural regulation mediated by nanomaterials. NANOTECHNOLOGY 2022; 33:272002. [PMID: 35442216 DOI: 10.1088/1361-6528/ac62b1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 03/29/2022] [Indexed: 06/14/2023]
Abstract
Neural regulation techniques play an essential role in the functional dissection of neural circuits and also the treatment of neurological diseases. Recently, a series of nanomaterials, including upconversion nanoparticles (UCNPs), magnetic nanoparticles (MNPs), and silicon nanomaterials (SNMs) that are responsive to remote optical or magnetic stimulation, have been applied as transducers to facilitate localized control of neural activities. In this review, we summarize the latest advances in nanomaterial-mediated neural regulation, especially in a remote and minimally invasive manner. We first give an overview of existing neural stimulation techniques, including electrical stimulation, transcranial magnetic stimulation, chemogenetics, and optogenetics, with an emphasis on their current limitations. Then we focus on recent developments in nanomaterial-mediated neural regulation, including UCNP-mediated fiberless optogenetics, MNP-mediated magnetic neural regulation, and SNM-mediated non-genetic neural regulation. Finally, we discuss the possibilities and challenges for nanomaterial-mediated neural regulation.
Collapse
Affiliation(s)
- Liang Zou
- CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, People's Republic of China
- CAS Center for Excellence in Brain Science and Intelligence Technology, Institute of Neuroscience, Chinese Academy of Sciences, Shanghai 200031, People's Republic of China
- University of Chinese Academy of Sciences, Beijing 100049, People's Republic of China
| | - Ke Xu
- CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, People's Republic of China
- CAS Center for Excellence in Brain Science and Intelligence Technology, Institute of Neuroscience, Chinese Academy of Sciences, Shanghai 200031, People's Republic of China
- University of Chinese Academy of Sciences, Beijing 100049, People's Republic of China
| | - Huihui Tian
- CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, People's Republic of China
| | - Ying Fang
- CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, People's Republic of China
- CAS Center for Excellence in Brain Science and Intelligence Technology, Institute of Neuroscience, Chinese Academy of Sciences, Shanghai 200031, People's Republic of China
- University of Chinese Academy of Sciences, Beijing 100049, People's Republic of China
| |
Collapse
|
35
|
Filippi M, Garello F, Yasa O, Kasamkattil J, Scherberich A, Katzschmann RK. Engineered Magnetic Nanocomposites to Modulate Cellular Function. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2104079. [PMID: 34741417 DOI: 10.1002/smll.202104079] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 09/13/2021] [Indexed: 06/13/2023]
Abstract
Magnetic nanoparticles (MNPs) have various applications in biomedicine, including imaging, drug delivery and release, genetic modification, cell guidance, and patterning. By combining MNPs with polymers, magnetic nanocomposites (MNCs) with diverse morphologies (core-shell particles, matrix-dispersed particles, microspheres, etc.) can be generated. These MNCs retain the ability of MNPs to be controlled remotely using external magnetic fields. While the effects of these biomaterials on the cell biology are still poorly understood, such information can help the biophysical modulation of various cellular functions, including proliferation, adhesion, and differentiation. After recalling the basic properties of MNPs and polymers, and describing their coassembly into nanocomposites, this review focuses on how polymeric MNCs can be used in several ways to affect cell behavior. A special emphasis is given to 3D cell culture models and transplantable grafts, which are used for regenerative medicine, underlining the impact of MNCs in regulating stem cell differentiation and engineering living tissues. Recent advances in the use of MNCs for tissue regeneration are critically discussed, particularly with regard to their prospective involvement in human therapy and in the construction of advanced functional materials such as magnetically operated biomedical robots.
Collapse
Affiliation(s)
- Miriam Filippi
- Soft Robotics Laboratory, ETH Zurich, Tannenstrasse 3, Zurich, 8092, Switzerland
| | - Francesca Garello
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Via Nizza 52, Torino, 10126, Italy
| | - Oncay Yasa
- Soft Robotics Laboratory, ETH Zurich, Tannenstrasse 3, Zurich, 8092, Switzerland
| | - Jesil Kasamkattil
- Department of Biomedicine, University Hospital Basel, Hebelstrasse 20, Basel, 4031, Switzerland
| | - Arnaud Scherberich
- Department of Biomedicine, University Hospital Basel, Hebelstrasse 20, Basel, 4031, Switzerland
- Department of Biomedical Engineering, University of Basel, Gewerbestrasse 14, Allschwil, 4123, Switzerland
| | - Robert K Katzschmann
- Soft Robotics Laboratory, ETH Zurich, Tannenstrasse 3, Zurich, 8092, Switzerland
| |
Collapse
|
36
|
Chen WH, Onoe T, Kamimura M. Noninvasive near-infrared light triggers the remote activation of thermo-responsive TRPV1 channels in neurons based on biodegradable/photothermal polymer micelles. NANOSCALE 2022; 14:2210-2220. [PMID: 35084002 DOI: 10.1039/d1nr07242k] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
In this study, we developed a novel biodegradable/photothermal polymer micelle-based remote-activation method for a temperature-sensitive ion channel, namely transient receptor potential cation channel subfamily V member 1 (TRPV1). Biodegradable/photothermal polymer micelles containing indocyanine green (ICG-micelles) were prepared using a simple one-pod mixing method. The obtained ICG-micelles showed biocompatibility and biodegradability. Furthermore, under tissue-penetrable near-infrared (NIR) laser irradiation, the ICG-micelles exhibited excellent photothermal effects and NIR emission. Moreover, NIR light-induced remote activation of neurons was successfully performed. ICG-micelles loaded with anti-TRPV1 antibodies effectively bound TRPV1 on cell membranes, and accelerated Ca2+ ion influx into neuronal cells was induced under NIR irradiation. Based on these findings, it is anticipated that the ICG-micelles can serve as a novel noninvasive remote-activation tool for neuronal cells.
Collapse
Affiliation(s)
- Wei-Hsu Chen
- Department of Materials Science and Technology, Faculty of Advanced Engineering, Tokyo University of Science, 6-3-1 Niijuku, Katsushika, Tokyo 125-8585, Japan.
| | - Taiki Onoe
- Department of Materials Science and Technology, Faculty of Advanced Engineering, Tokyo University of Science, 6-3-1 Niijuku, Katsushika, Tokyo 125-8585, Japan.
| | - Masao Kamimura
- Department of Materials Science and Technology, Faculty of Advanced Engineering, Tokyo University of Science, 6-3-1 Niijuku, Katsushika, Tokyo 125-8585, Japan.
| |
Collapse
|
37
|
Del Sol-Fernández S, Martínez-Vicente P, Gomollón-Zueco P, Castro-Hinojosa C, Gutiérrez L, Fratila RM, Moros M. Magnetogenetics: remote activation of cellular functions triggered by magnetic switches. NANOSCALE 2022; 14:2091-2118. [PMID: 35103278 PMCID: PMC8830762 DOI: 10.1039/d1nr06303k] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Accepted: 11/13/2021] [Indexed: 05/03/2023]
Abstract
During the last decade, the possibility to remotely control intracellular pathways using physical tools has opened the way to novel and exciting applications, both in basic research and clinical applications. Indeed, the use of physical and non-invasive stimuli such as light, electricity or magnetic fields offers the possibility of manipulating biological processes with spatial and temporal resolution in a remote fashion. The use of magnetic fields is especially appealing for in vivo applications because they can penetrate deep into tissues, as opposed to light. In combination with magnetic actuators they are emerging as a new instrument to precisely manipulate biological functions. This approach, coined as magnetogenetics, provides an exclusive tool to study how cells transform mechanical stimuli into biochemical signalling and offers the possibility of activating intracellular pathways connected to temperature-sensitive proteins. In this review we provide a critical overview of the recent developments in the field of magnetogenetics. We discuss general topics regarding the three main components for magnetic field-based actuation: the magnetic fields, the magnetic actuators and the cellular targets. We first introduce the main approaches in which the magnetic field can be used to manipulate the magnetic actuators, together with the most commonly used magnetic field configurations and the physicochemical parameters that can critically influence the magnetic properties of the actuators. Thereafter, we discuss relevant examples of magneto-mechanical and magneto-thermal stimulation, used to control stem cell fate, to activate neuronal functions, or to stimulate apoptotic pathways, among others. Finally, although magnetogenetics has raised high expectations from the research community, to date there are still many obstacles to be overcome in order for it to become a real alternative to optogenetics for instance. We discuss some controversial aspects related to the insufficient elucidation of the mechanisms of action of some magnetogenetics constructs and approaches, providing our opinion on important challenges in the field and possible directions for the upcoming years.
Collapse
Affiliation(s)
- Susel Del Sol-Fernández
- Instituto de Nanociencia y Materiales de Aragón (INMA), CSIC-Universidad de Zaragoza, Zaragoza 50009, Spain.
| | - Pablo Martínez-Vicente
- Instituto de Nanociencia y Materiales de Aragón (INMA), CSIC-Universidad de Zaragoza, Zaragoza 50009, Spain.
| | - Pilar Gomollón-Zueco
- Instituto de Nanociencia y Materiales de Aragón (INMA), CSIC-Universidad de Zaragoza, Zaragoza 50009, Spain.
| | - Christian Castro-Hinojosa
- Instituto de Nanociencia y Materiales de Aragón (INMA), CSIC-Universidad de Zaragoza, Zaragoza 50009, Spain.
| | - Lucía Gutiérrez
- Instituto de Nanociencia y Materiales de Aragón (INMA), CSIC-Universidad de Zaragoza, Zaragoza 50009, Spain.
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Spain
- Departamento de Química Analítica, Universidad de Zaragoza, Zaragoza 50009, Spain
| | - Raluca M Fratila
- Instituto de Nanociencia y Materiales de Aragón (INMA), CSIC-Universidad de Zaragoza, Zaragoza 50009, Spain.
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Spain
- Departamento de Química Orgánica, Universidad de Zaragoza, C/Pedro Cerbuna 12, Zaragoza 50009, Spain
| | - María Moros
- Instituto de Nanociencia y Materiales de Aragón (INMA), CSIC-Universidad de Zaragoza, Zaragoza 50009, Spain.
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Spain
| |
Collapse
|
38
|
Yu Y, Payne C, Marina N, Korsak A, Southern P, García‐Prieto A, Christie IN, Baker RR, Fisher EMC, Wells JA, Kalber TL, Pankhurst QA, Gourine AV, Lythgoe MF. Remote and Selective Control of Astrocytes by Magnetomechanical Stimulation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2104194. [PMID: 34927381 PMCID: PMC8867145 DOI: 10.1002/advs.202104194] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 11/15/2021] [Indexed: 05/06/2023]
Abstract
Astrocytes play crucial and diverse roles in brain health and disease. The ability to selectively control astrocytes provides a valuable tool for understanding their function and has the therapeutic potential to correct dysfunction. Existing technologies such as optogenetics and chemogenetics require the introduction of foreign proteins, which adds a layer of complication and hinders their clinical translation. A novel technique, magnetomechanical stimulation (MMS), that enables remote and selective control of astrocytes without genetic modification is described here. MMS exploits the mechanosensitivity of astrocytes and triggers mechanogated Ca2+ and adenosine triphosphate (ATP) signaling by applying a magnetic field to antibody-functionalized magnetic particles that are targeted to astrocytes. Using purpose-built magnetic devices, the mechanosensory threshold of astrocytes is determined, a sub-micrometer particle for effective MMS is identified, the in vivo fate of the particles is established, and cardiovascular responses are induced in rats after particles are delivered to specific brainstem astrocytes. By eliminating the need for device implantation and genetic modification, MMS is a method for controlling astroglial activity with an improved prospect for clinical application than existing technologies.
Collapse
Affiliation(s)
- Yichao Yu
- Centre for Advanced Biomedical ImagingDivision of MedicineUniversity College London72 Huntley StreetLondonWC1E 6DDUK
| | - Christopher Payne
- Centre for Advanced Biomedical ImagingDivision of MedicineUniversity College London72 Huntley StreetLondonWC1E 6DDUK
| | - Nephtali Marina
- Centre for Cardiovascular and Metabolic NeuroscienceResearch Department of Neuroscience, Physiology and PharmacologyUniversity College LondonGower StreetLondonWC1E 6BTUK
| | - Alla Korsak
- Centre for Cardiovascular and Metabolic NeuroscienceResearch Department of Neuroscience, Physiology and PharmacologyUniversity College LondonGower StreetLondonWC1E 6BTUK
| | - Paul Southern
- Healthcare Biomagnetics LaboratoryUniversity College London21 Albemarle StreetLondonW1S 4BSUK
| | - Ana García‐Prieto
- Healthcare Biomagnetics LaboratoryUniversity College London21 Albemarle StreetLondonW1S 4BSUK
- Departamento Física Aplicada IUniversidad del País VascoBilbao48013Spain
| | - Isabel N. Christie
- Centre for Cardiovascular and Metabolic NeuroscienceResearch Department of Neuroscience, Physiology and PharmacologyUniversity College LondonGower StreetLondonWC1E 6BTUK
| | - Rebecca R. Baker
- Centre for Advanced Biomedical ImagingDivision of MedicineUniversity College London72 Huntley StreetLondonWC1E 6DDUK
| | - Elizabeth M. C. Fisher
- Department of Neuromuscular DiseasesQueen Square Institute of NeurologyUniversity College LondonQueen SquareLondonWC1N 3BGUK
| | - Jack A. Wells
- Centre for Advanced Biomedical ImagingDivision of MedicineUniversity College London72 Huntley StreetLondonWC1E 6DDUK
| | - Tammy L. Kalber
- Centre for Advanced Biomedical ImagingDivision of MedicineUniversity College London72 Huntley StreetLondonWC1E 6DDUK
| | - Quentin A. Pankhurst
- Healthcare Biomagnetics LaboratoryUniversity College London21 Albemarle StreetLondonW1S 4BSUK
| | - Alexander V. Gourine
- Centre for Cardiovascular and Metabolic NeuroscienceResearch Department of Neuroscience, Physiology and PharmacologyUniversity College LondonGower StreetLondonWC1E 6BTUK
| | - Mark F. Lythgoe
- Centre for Advanced Biomedical ImagingDivision of MedicineUniversity College London72 Huntley StreetLondonWC1E 6DDUK
| |
Collapse
|
39
|
Wang S, Xu J, Li W, Sun S, Gao S, Hou Y. Magnetic Nanostructures: Rational Design and Fabrication Strategies toward Diverse Applications. Chem Rev 2022; 122:5411-5475. [PMID: 35014799 DOI: 10.1021/acs.chemrev.1c00370] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
In recent years, the continuous development of magnetic nanostructures (MNSs) has tremendously promoted both fundamental scientific research and technological applications. Different from the bulk magnet, the systematic engineering on MNSs has brought a great breakthrough in some emerging fields such as the construction of MNSs, the magnetism exploration of multidimensional MNSs, and their potential translational applications. In this review, we give a detailed description of the synthetic strategies of MNSs based on the fundamental features and application potential of MNSs and discuss the recent progress of MNSs in the fields of nanomedicines, advanced nanobiotechnology, catalysis, and electromagnetic wave adsorption (EMWA), aiming to provide guidance for fabrication strategies of MNSs toward diverse applications.
Collapse
Affiliation(s)
- Shuren Wang
- Beijing Key Laboratory of Magnetoelectric Materials and Devices, School of Materials Science and Engineering, Beijing Innovation Centre for Engineering Science and Advanced Technology, Peking University, Beijing 100871, China
| | - Junjie Xu
- Beijing Key Laboratory of Magnetoelectric Materials and Devices, School of Materials Science and Engineering, Beijing Innovation Centre for Engineering Science and Advanced Technology, Peking University, Beijing 100871, China
| | - Wei Li
- Beijing Key Laboratory of Magnetoelectric Materials and Devices, School of Materials Science and Engineering, Beijing Innovation Centre for Engineering Science and Advanced Technology, Peking University, Beijing 100871, China
| | - Shengnan Sun
- Beijing Key Laboratory of Magnetoelectric Materials and Devices, School of Materials Science and Engineering, Beijing Innovation Centre for Engineering Science and Advanced Technology, Peking University, Beijing 100871, China
| | - Song Gao
- Beijing Key Laboratory of Magnetoelectric Materials and Devices, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China.,Institute of Spin-X Science and Technology, South China University of Technology, Guangzhou 511442, China
| | - Yanglong Hou
- Beijing Key Laboratory of Magnetoelectric Materials and Devices, School of Materials Science and Engineering, Beijing Innovation Centre for Engineering Science and Advanced Technology, Peking University, Beijing 100871, China
| |
Collapse
|
40
|
Fan H. Central Nervous System Nanotechnology. Nanomedicine (Lond) 2022. [DOI: 10.1007/978-981-13-9374-7_29-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
|
41
|
Gaburjáková M, Gaburjáková J, Krejčíová E, Kosnáč D, Kosnáčová H, Nagy Š, Polák Š, Sabo M, Trnka M, Kopáni M. Blocking effect of ferritin on the ryanodine receptor-isoform 2. Arch Biochem Biophys 2021; 712:109031. [PMID: 34534540 DOI: 10.1016/j.abb.2021.109031] [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: 05/11/2021] [Revised: 09/08/2021] [Accepted: 09/08/2021] [Indexed: 10/20/2022]
Abstract
Iron, an essential element for most living organism, participates in a wide variety of physiological processes. Disturbance in iron homeostasis has been associated with numerous pathologies, particularly in the heart and brain, which are the most susceptible organs. Under iron-overload conditions, the generation of reactive oxygen species leads to impairment in Ca2+ signaling, fundamentally implicated in cardiac and neuronal physiology. Since iron excess is accompanied by increased expression of iron-storage protein, ferritin, we examined whether ferritin has an effect on the ryanodine receptor - isoform 2 (RYR2), which is one of the major components of Ca2+ signaling. Using the method of planar lipid membranes, we show that ferritin induced an abrupt, permanent blockage of the RYR2 channel. The ferritin effect was strongly voltage dependent and competitively antagonized by cytosolic TEA+, an impermeant RYR2 blocker. Our results collectively indicate that monomeric ferritin highly likely blocks the RYR2 channel by a direct electrostatic interaction within the wider region of the channel permeation pathway.
Collapse
Affiliation(s)
- Marta Gaburjáková
- Centre of Biosciences, Institute of Molecular Physiology and Genetics, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Jana Gaburjáková
- Centre of Biosciences, Institute of Molecular Physiology and Genetics, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Eva Krejčíová
- Centre of Biosciences, Institute of Molecular Physiology and Genetics, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Daniel Kosnáč
- Institute of Medical Physics, Biophysics, Informatics and Telemedicine, Faculty of Medicine, Comenius University, Bratislava, Slovakia; Department of Simulation and Virtual Medical Education, Faculty of Medicine, Comenius University, Bratislava, Slovakia
| | - Helena Kosnáčová
- Department of Simulation and Virtual Medical Education, Faculty of Medicine, Comenius University, Bratislava, Slovakia; Slovak Academy of Sciences, Department of Genetics, Cancer Research Institute, Biomedical Research Center, Bratislava, Slovakia
| | - Štefan Nagy
- Institute of Materials and Machine Mechanics, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Štefan Polák
- Institute of Histology and Embryology, Faculty of Medicine, Comenius University, Bratislava, Slovakia
| | - Michal Sabo
- Institute of Medical Physics, Biophysics, Informatics and Telemedicine, Faculty of Medicine, Comenius University, Bratislava, Slovakia
| | - Michal Trnka
- Institute of Medical Physics, Biophysics, Informatics and Telemedicine, Faculty of Medicine, Comenius University, Bratislava, Slovakia
| | - Martin Kopáni
- Institute of Medical Physics, Biophysics, Informatics and Telemedicine, Faculty of Medicine, Comenius University, Bratislava, Slovakia.
| |
Collapse
|
42
|
Evaluating methods and protocols of ferritin-based magnetogenetics. iScience 2021; 24:103094. [PMID: 34622149 PMCID: PMC8479696 DOI: 10.1016/j.isci.2021.103094] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 07/15/2021] [Accepted: 09/03/2021] [Indexed: 11/22/2022] Open
Abstract
FeRIC (Ferritin iron Redistribution to Ion Channels) is a magnetogenetic technique that uses radiofrequency (RF) alternating magnetic fields to activate the transient receptor potential channels, TRPV1 and TRPV4, coupled to cellular ferritins. In cells expressing ferritin-tagged TRPV, RF stimulation increases the cytosolic Ca2+ levels via a biochemical pathway. The interaction between RF and ferritin increases the free cytosolic iron levels that, in turn, trigger chemical reactions producing reactive oxygen species and oxidized lipids that activate the ferritin-tagged TRPV. In this pathway, it is expected that experimental factors that disturb the ferritin expression, the ferritin iron load, the TRPV functional expression, or the cellular redox state will impact the efficiency of RF in activating ferritin-tagged TRPV. Here, we examined several experimental factors that either enhance or abolish the RF control of ferritin-tagged TRPV. The findings may help optimize and establish reproducible magnetogenetic protocols.
Collapse
|
43
|
Liu X, Zhang Y, Guo Y, Jiao W, Gao X, Lee WSV, Wang Y, Deng X, He Y, Jiao J, Zhang C, Hu G, Liang X, Fan H. Electromagnetic Field-Programmed Magnetic Vortex Nanodelivery System for Efficacious Cancer Therapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:e2100950. [PMID: 34279055 PMCID: PMC8456207 DOI: 10.1002/advs.202100950] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 05/06/2021] [Indexed: 05/05/2023]
Abstract
Effective delivery of anticancer drugs into the nucleus for pharmacological action is impeded by a series of intratumoral transport barriers. Despite the significant potential of magnetic nanovehicles in electromagnetic field (EF)-activated drug delivery, modularizing a tandem magnetoresponsive activity in a one-nanoparticle system to meet different requirements at both tissue and cellular levels remain highly challenging. Herein, a strategy is described by employing sequential EF frequencies in inducing a succession of magnetoresponses in the magnetic nanovehicles that aims to realize cascaded tissue penetration and nuclear accumulation. This nanovehicle features ferrimagnetic vortex-domain iron oxide nanorings coated with a thermo-responsive polyethylenimine copolymer (PI/FVIOs). It is shown that the programmed cascading of low frequency (Lf)-EF-induced magnetophoresis and medium frequency (Mf)-EF-stimulated magneto-thermia can steer the Doxorubicin (DOX)-PI/FVIOs to the deep tissue and subsequently trigger intracellular burst release of DOX for successful nuclear entry. By programming the order of different EF frequencies, it is demonstrated that first-stage Lf-EF and subsequent Mf-EF operation enables DOX-PI/FVIOs to effectively deliver 86.2% drug into the nucleus in vivo. This nanodelivery system empowers potent antitumoral activity in various models of intractable tumors, including DOX-resistant MCF-7 breast cancer cells, triple-negative MDA-MB-231 breast cancer cells, and BxPC-3 pancreatic cancer cells with poor permeability.
Collapse
Affiliation(s)
- Xiaoli Liu
- Key Laboratory of Resource Biology and Biotechnology in Western ChinaMinistry of EducationSchool of MedicineNorthwest UniversityXi'anShaanxi710069China
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and NanosafetyCAS Center for Excellence in NanoscienceNational Center for Nanoscience and Technology of ChinaNo. 11, First North Road, ZhongguancunBeijing100190China
| | - Yifan Zhang
- Key Laboratory of Synthetic and Natural Functional Molecule Chemistry of the Ministry of EducationCollege of Chemistry and Materials ScienceNorthwest UniversityXi'an710127China
| | - Yu Guo
- Department of Engineering MechanicsZhejiang UniversityHangzhou310027China
| | - Wangbo Jiao
- Key Laboratory of Synthetic and Natural Functional Molecule Chemistry of the Ministry of EducationCollege of Chemistry and Materials ScienceNorthwest UniversityXi'an710127China
| | - Xiao Gao
- Key Laboratory of Resource Biology and Biotechnology in Western ChinaMinistry of EducationSchool of MedicineNorthwest UniversityXi'anShaanxi710069China
| | - Wee Siang Vincent Lee
- Department of Materials Science and EngineeringNational University of SingaporeSingapore117573
| | - Yanyun Wang
- Key Laboratory of Synthetic and Natural Functional Molecule Chemistry of the Ministry of EducationCollege of Chemistry and Materials ScienceNorthwest UniversityXi'an710127China
| | - Xia Deng
- School of Life Sciences and Electron Microscopy Center of Lanzhou UniversityLanzhou UniversityLanzhou730000China
| | - Yuan He
- Key Laboratory of Synthetic and Natural Functional Molecule Chemistry of the Ministry of EducationCollege of Chemistry and Materials ScienceNorthwest UniversityXi'an710127China
| | - Ju Jiao
- Department of Nuclear MedicineThe Third Affiliated Hospital of Sun Yat‐sen University600 Tianhe RoadGuangzhouGuangdong510630China
| | - Ce Zhang
- State Key Laboratory of Cultivation Base for Photoelectric Technology and Functional MaterialsLaboratory of Optoelectronic Technology of Shaanxi ProvinceNational Center for International Research of Photoelectric Technology & Nanofunctional Materials and ApplicationInstitute of Photonics and Photon‐TechnologyNorthwest UniversityXuefu Street No. 1Xi'an710127China
| | - Guoqing Hu
- Department of Engineering MechanicsZhejiang UniversityHangzhou310027China
| | - Xing‐Jie Liang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and NanosafetyCAS Center for Excellence in NanoscienceNational Center for Nanoscience and Technology of ChinaNo. 11, First North Road, ZhongguancunBeijing100190China
- University of Chinese Academy of SciencesNo.19(A) Yuquan Road, Shijingshan DistrictBeijing100049China
| | - Haiming Fan
- Key Laboratory of Resource Biology and Biotechnology in Western ChinaMinistry of EducationSchool of MedicineNorthwest UniversityXi'anShaanxi710069China
- Key Laboratory of Synthetic and Natural Functional Molecule Chemistry of the Ministry of EducationCollege of Chemistry and Materials ScienceNorthwest UniversityXi'an710127China
| |
Collapse
|
44
|
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: 3.0] [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
|
45
|
Stephens K, Zakaria FR, VanArsdale E, Payne GF, Bentley WE. Electronic signals are electrogenetically relayed to control cell growth and co-culture composition. Metab Eng Commun 2021; 13:e00176. [PMID: 34194997 PMCID: PMC8233222 DOI: 10.1016/j.mec.2021.e00176] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 05/20/2021] [Accepted: 05/31/2021] [Indexed: 01/17/2023] Open
Abstract
There is much to be gained by enabling electronic interrogation and control of biological function. While the benefits of bioelectronics that rely on potential-driven ionic flows are well known (electrocardiograms, defibrillators, neural prostheses, etc) there are relatively few advances targeting nonionic molecular networks, including genetic circuits. Redox activities combine connectivity to electronics with the potential for specific genetic control in cells. Here, electrode-generated hydrogen peroxide is used to actuate an electrogenetic "relay" cell population, which interprets the redox cue and synthesizes a bacterial signaling molecule (quorum sensing autoinducer AI-1) that, in turn, signals increased growth rate in a second population. The dramatically increased growth rate of the second population is enabled by expression of a phosphotransferase system protein, HPr, which is important for glucose transport. The potential to electronically modulate cell growth via direct genetic control will enable new opportunities in the treatment of disease and manufacture of biological therapeutics and other molecules.
Collapse
Affiliation(s)
- Kristina Stephens
- Fischell Department of Bioengineering, University of Maryland, College Park, USA.,Institute for Bioscience and Biotechnology Research, University of Maryland, College Park, USA.,Robert E. Fischell Institute for Biomedical Devices, University of Maryland, College Park, USA
| | - Fauziah Rahma Zakaria
- Fischell Department of Bioengineering, University of Maryland, College Park, USA.,Institute for Bioscience and Biotechnology Research, University of Maryland, College Park, USA.,Robert E. Fischell Institute for Biomedical Devices, University of Maryland, College Park, USA
| | - Eric VanArsdale
- Fischell Department of Bioengineering, University of Maryland, College Park, USA.,Institute for Bioscience and Biotechnology Research, University of Maryland, College Park, USA.,Robert E. Fischell Institute for Biomedical Devices, University of Maryland, College Park, USA
| | - Gregory F Payne
- Institute for Bioscience and Biotechnology Research, University of Maryland, College Park, USA.,Robert E. Fischell Institute for Biomedical Devices, University of Maryland, College Park, USA
| | - William E Bentley
- Fischell Department of Bioengineering, University of Maryland, College Park, USA.,Institute for Bioscience and Biotechnology Research, University of Maryland, College Park, USA.,Robert E. Fischell Institute for Biomedical Devices, University of Maryland, College Park, USA
| |
Collapse
|
46
|
Bhave G, Chen JC, Singer A, Sharma A, Robinson JT. Distributed sensor and actuator networks for closed-loop bioelectronic medicine. MATERIALS TODAY (KIDLINGTON, ENGLAND) 2021; 46:125-135. [PMID: 34366697 PMCID: PMC8336425 DOI: 10.1016/j.mattod.2020.12.020] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
Designing implantable bioelectronic systems that continuously monitor physiological functions and simultaneously provide personalized therapeutic solutions for patients remains a persistent challenge across many applications ranging from neural systems to bioelectronic organs. Closed-loop systems typically consist of three functional blocks, namely, sensors, signal processors and actuators. An effective system, that can provide the necessary therapeutics, tailored to individual physiological factors requires a distributed network of sensors and actuators. While significant progress has been made, closed-loop systems still face many challenges before they can truly be considered as long-term solutions for many diseases. In this review, we consider three important criteria where materials play a critical role to enable implantable closed-loop systems: Specificity, Biocompatibility and Connectivity. We look at the progress made in each of these fields with respect to a specific application and outline the challenges in creating bioelectronic technologies for the future.
Collapse
|
47
|
Huang Q, Garrett A, Bose S, Blocker S, Rios AC, Clevers H, Shen X. The frontier of live tissue imaging across space and time. Cell Stem Cell 2021; 28:603-622. [PMID: 33798422 PMCID: PMC8034393 DOI: 10.1016/j.stem.2021.02.010] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
What you see is what you get-imaging techniques have long been essential for visualization and understanding of tissue development, homeostasis, and regeneration, which are driven by stem cell self-renewal and differentiation. Advances in molecular and tissue modeling techniques in the last decade are providing new imaging modalities to explore tissue heterogeneity and plasticity. Here we describe current state-of-the-art imaging modalities for tissue research at multiple scales, with a focus on explaining key tradeoffs such as spatial resolution, penetration depth, capture time/frequency, and moieties. We explore emerging tissue modeling and molecular tools that improve resolution, specificity, and throughput.
Collapse
Affiliation(s)
- Qiang Huang
- Department of Pediatric Surgery, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004 Shaanxi, China; Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC 27708, USA
| | - Aliesha Garrett
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC 27708, USA
| | - Shree Bose
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC 27708, USA
| | - Stephanie Blocker
- Center for In Vitro Microscopy, Duke University, Durham, NC 27708, USA
| | - Anne C Rios
- Princess Máxima Center for Pediatric Oncology, Utrecht 3584, the Netherlands; Department of Cancer Research, Oncode Institute, Hubrecht Institute-KNAW Utrecht, Utrecht 3584, the Netherlands
| | - Hans Clevers
- Princess Máxima Center for Pediatric Oncology, Utrecht 3584, the Netherlands; Department of Cancer Research, Oncode Institute, Hubrecht Institute-KNAW Utrecht, Utrecht 3584, the Netherlands; Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Center (UMC) Utrecht, Utrecht 3584, the Netherlands
| | - Xiling Shen
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC 27708, USA.
| |
Collapse
|
48
|
Lipid Oxidation Induced by RF Waves and Mediated by Ferritin Iron Causes Activation of Ferritin-Tagged Ion Channels. Cell Rep 2021; 30:3250-3260.e7. [PMID: 32160534 DOI: 10.1016/j.celrep.2020.02.070] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Revised: 12/19/2019] [Accepted: 02/18/2020] [Indexed: 12/26/2022] Open
Abstract
One approach to magnetogenetics uses radiofrequency (RF) waves to activate transient receptor potential channels (TRPV1 and TRPV4) that are coupled to cellular ferritins. The mechanisms underlying this effect are unclear and controversial. Theoretical calculations suggest that the heat produced by RF fields is likely orders of magnitude weaker than needed for channel activation. Using the FeRIC (Ferritin iron Redistribution to Ion Channels) system, we have uncovered a mechanism of activation of ferritin-tagged channels via a biochemical pathway initiated by RF disturbance of ferritin and mediated by ferritin-associated iron. We show that, in cells expressing TRPVFeRIC channels, RF increases the levels of the labile iron pool in a ferritin-dependent manner. Free iron participates in chemical reactions, producing reactive oxygen species and oxidized lipids that ultimately activate the TRPVFeRIC channels. This biochemical pathway predicts a similar RF-induced activation of other lipid-sensitive TRP channels and may guide future magnetogenetic designs.
Collapse
|
49
|
Functional Expression of TRPV1 Ion Channel in the Canine Peripheral Blood Mononuclear Cells. Int J Mol Sci 2021; 22:ijms22063177. [PMID: 33804707 PMCID: PMC8003907 DOI: 10.3390/ijms22063177] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 03/12/2021] [Accepted: 03/17/2021] [Indexed: 12/25/2022] Open
Abstract
TRPV1, known as a capsaicin receptor, is the best-described transient receptor potential (TRP) ion channel. Recently, it was shown to be expressed by non-excitable cells such as lymphocytes. However, the data regarding the functional expression of the TRPV1 channel in the immune cells are often contradictory. In the present study, we performed a phylogenetical analysis of the canine TRP ion channels, we assessed the expression of TRPV1 in the canine peripheral blood mononuclear cells (PBMC) by qPCR and Western blot, and we determined the functionality of TRPV1 by whole-cell patch-clamp recordings and calcium assay. We found high expression of TRPV2, -M2, and -M7 in the canine PBMCs, while expression of TRPV1, -V4 and, -M5 was relatively low. We confirmed that TRPV1 is expressed on the protein level in the PBMC and it localizes in the plasma membrane. The whole-cell patch-clamp recording revealed that capsaicin application caused a significant increase in the current density. Similarly, the results from the calcium assay show a dose-dependent increase in intracellular calcium level in the presence of capsaicin that was partially abolished by capsazepine. Our study confirms the expression of TRPV1 ion channel on both mRNA and protein levels in the canine PBMC and indicates that the ion channel is functional.
Collapse
|
50
|
Kim S, Uroz M, Bays JL, Chen CS. Harnessing Mechanobiology for Tissue Engineering. Dev Cell 2021; 56:180-191. [PMID: 33453155 PMCID: PMC7855912 DOI: 10.1016/j.devcel.2020.12.017] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 11/10/2020] [Accepted: 12/22/2020] [Indexed: 12/13/2022]
Abstract
A primary challenge in tissue engineering is to recapitulate both the structural and functional features of whole tissues and organs. In vivo, patterning of the body plan and constituent tissues emerges from the carefully orchestrated interactions between the transcriptional programs that give rise to cell types and the mechanical forces that drive the bending, twisting, and extensions critical to morphogenesis. Substantial recent progress in mechanobiology-understanding how mechanics regulate cell behaviors and what cellular machineries are responsible-raises the possibility that one can begin to use these insights to help guide the strategy and design of functional engineered tissues. In this perspective, we review and propose the development of different approaches, from providing appropriate extracellular mechanical cues to interfering with cellular mechanosensing machinery, to aid in controlling cell and tissue structure and function.
Collapse
Affiliation(s)
- Sudong Kim
- Department of Biomedical Engineering and the Biological Design Center, Boston University, Boston, MA 02215, USA; The Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA 02115, USA
| | - Marina Uroz
- Department of Biomedical Engineering and the Biological Design Center, Boston University, Boston, MA 02215, USA; The Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA 02115, USA
| | - Jennifer L Bays
- Department of Biomedical Engineering and the Biological Design Center, Boston University, Boston, MA 02215, USA; The Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA 02115, USA
| | - Christopher S Chen
- Department of Biomedical Engineering and the Biological Design Center, Boston University, Boston, MA 02215, USA; The Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA 02115, USA.
| |
Collapse
|