101
|
Davis HC, Kang S, Lee JH, Shin TH, Putterman H, Cheon J, Shapiro MG. Nanoscale Heat Transfer from Magnetic Nanoparticles and Ferritin in an Alternating Magnetic Field. Biophys J 2020; 118:1502-1510. [PMID: 32061270 PMCID: PMC7091488 DOI: 10.1016/j.bpj.2020.01.028] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Revised: 01/16/2020] [Accepted: 01/21/2020] [Indexed: 11/28/2022] Open
Abstract
Recent suggestions of nanoscale heat confinement on the surface of synthetic and biogenic magnetic nanoparticles during heating by radio frequency-alternating magnetic fields have generated intense interest because of the potential utility of this phenomenon for noninvasive control of biomolecular and cellular function. However, such confinement would represent a significant departure from the classical heat transfer theory. Here, we report an experimental investigation of nanoscale heat confinement on the surface of several types of iron oxide nanoparticles commonly used in biological research, using an all-optical method devoid of the potential artifacts present in previous studies. By simultaneously measuring the fluorescence of distinct thermochromic dyes attached to the particle surface or dissolved in the surrounding fluid during radio frequency magnetic stimulation, we found no measurable difference between the nanoparticle surface temperature and that of the surrounding fluid for three distinct nanoparticle types. Furthermore, the metalloprotein ferritin produced no temperature increase on the protein surface nor in the surrounding fluid. Experiments mimicking the designs of previous studies revealed potential sources of the artifacts. These findings inform the use of magnetic nanoparticle hyperthermia in engineered cellular and molecular systems.
Collapse
Affiliation(s)
- Hunter C Davis
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, California
| | - Sunghwi Kang
- Center for NanoMedicine,Yonsei-Institute for Basic Science, Seoul, Republic of Korea
| | - Jae-Hyun Lee
- Center for NanoMedicine,Yonsei-Institute for Basic Science, Seoul, Republic of Korea
| | - Tae-Hyun Shin
- Center for NanoMedicine,Yonsei-Institute for Basic Science, Seoul, Republic of Korea
| | - Harry Putterman
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, California
| | - Jinwoo Cheon
- Center for NanoMedicine,Yonsei-Institute for Basic Science, Seoul, Republic of Korea
| | - Mikhail G Shapiro
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, California; Center for NanoMedicine,Yonsei-Institute for Basic Science, Seoul, Republic of Korea.
| |
Collapse
|
102
|
Lin FC, Zink JI. Probing the Local Nanoscale Heating Mechanism of a Magnetic Core in Mesoporous Silica Drug-Delivery Nanoparticles Using Fluorescence Depolarization. J Am Chem Soc 2020; 142:5212-5220. [PMID: 32091888 DOI: 10.1021/jacs.9b13082] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
In the presence of an alternating magnetic field (AMF), a superparamagnetic iron oxide nanoparticle (SPION) generates heat. Understanding the local heating mechanism of a SPION in suspension and in a mesoporous silica nanoparticle (MSN) will advance the design of hyperthermia-based nanotheranostics and AMF-stimulated drug delivery in biomedical applications. The AMF-induced heating of single-domain SPION can be explained by the Néel relaxation (reorientation of the magnetization) or the Brownian relaxation (motion of the particle). The latter is investigated using fluorescence depolarization based on detecting the mobility-dependent polarization anisotropy (r) of two luminescence emission bands at different wavelengths corresponded to the europium-doped luminescent SPION (EuSPION) core and the silica-based intrinsically emitting shell of the core-shell MSN. The fluorescence depolarization experiments are carried out with both the free and the silica-encapsulated SPION nanoparticles with and without application of the AMF. The r value of a EuSPION core-mesoporous silica shell in the presence of the AMF does not change, indicating that no additional rotational motion of the core-shell nanoparticles is induced by the AMF, disproving the contribution of Brownian heating and thus supporting Néel relaxation as the dominant heating mechanism.
Collapse
Affiliation(s)
- Fang-Chu Lin
- Department of Chemistry and Biochemistry, University of California-Los Angeles, Los Angeles, California 90095, United States.,California NanoSystems Institute, University of California-Los Angeles, Los Angeles, California 90095, United States
| | - Jeffrey I Zink
- Department of Chemistry and Biochemistry, University of California-Los Angeles, Los Angeles, California 90095, United States.,California NanoSystems Institute, University of California-Los Angeles, Los Angeles, California 90095, United States
| |
Collapse
|
103
|
Gamboa L, Zamat AH, Kwong GA. Synthetic immunity by remote control. Theranostics 2020; 10:3652-3667. [PMID: 32206114 PMCID: PMC7069089 DOI: 10.7150/thno.41305] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Accepted: 02/03/2020] [Indexed: 12/11/2022] Open
Abstract
Cell-based immunotherapies, such as T cells engineered with chimeric antigen receptors (CARs), have the potential to cure patients of disease otherwise refractory to conventional treatments. Early-on-treatment and long-term durability of patient responses depend critically on the ability to control the potency of adoptively transferred T cells, as overactivation can lead to complications like cytokine release syndrome, and immunosuppression can result in ineffective responses to therapy. Drugs or biologics (e.g., cytokines) that modulate immune activity are limited by mass transport barriers that reduce the local effective drug concentration, and lack site or target cell specificity that results in toxicity. Emerging technologies that enable site-targeted, remote control of key T cell functions - including proliferation, antigen-sensing, and target-cell killing - have the potential to increase treatment precision and safety profile. These technologies are broadly applicable to other immune cells to expand immune cell therapies across many cancers and diseases. In this review, we highlight the opportunities, challenges and the current state-of-the-art for remote control of synthetic immunity.
Collapse
Affiliation(s)
- Lena Gamboa
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, Atlanta, GA 30332, USA
| | - Ali H. Zamat
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, Atlanta, GA 30332, USA
| | - Gabriel A. Kwong
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, Atlanta, GA 30332, USA
- Institute for Electronics and Nanotechnology, Georgia Institute of Technology, Atlanta, GA 30332, USA
- Parker H. Petit Institute of Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332, USA
- Integrated Cancer Research Center, Georgia Institute of Technology, Atlanta, GA 30332, USA
- Georgia Immunoengineering Consortium, Emory University and Georgia Institute of Technology, Atlanta, GA 30332, USA
| |
Collapse
|
104
|
Liu X, Zhang Y, Wang Y, Zhu W, Li G, Ma X, Zhang Y, Chen S, Tiwari S, Shi K, Zhang S, Fan HM, Zhao YX, Liang XJ. Comprehensive understanding of magnetic hyperthermia for improving antitumor therapeutic efficacy. Theranostics 2020; 10:3793-3815. [PMID: 32206123 PMCID: PMC7069093 DOI: 10.7150/thno.40805] [Citation(s) in RCA: 268] [Impact Index Per Article: 67.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Accepted: 01/13/2020] [Indexed: 12/20/2022] Open
Abstract
Magnetic hyperthermia (MH) has been introduced clinically as an alternative approach for the focal treatment of tumors. MH utilizes the heat generated by the magnetic nanoparticles (MNPs) when subjected to an alternating magnetic field (AMF). It has become an important topic in the nanomedical field due to their multitudes of advantages towards effective antitumor therapy such as high biosafety, deep tissue penetration, and targeted selective tumor killing. However, in order for MH to progress and to realize its paramount potential as an alternative choice for cancer treatment, tremendous challenges have to be overcome. Thus, the efficiency of MH therapy needs enhancement. In its recent 60-year of history, the field of MH has focused primarily on heating using MNPs for therapeutic applications. Increasing the thermal conversion efficiency of MNPs is the fundamental strategy for improving therapeutic efficacy. Recently, emerging experimental evidence indicates that MNPs-MH produces nano-scale heat effects without macroscopic temperature rise. A deep understanding of the effect of this localized induction heat for the destruction of subcellular/cellular structures further supports the efficacy of MH in improving therapeutic therapy. In this review, the currently available strategies for improving the antitumor therapeutic efficacy of MNPs-MH will be discussed. Firstly, the recent advancements in engineering MNP size, composition, shape, and surface to significantly improve their energy dissipation rates will be explored. Secondly, the latest studies depicting the effect of local induction heat for selectively disrupting cells/intracellular structures will be examined. Thirdly, strategies to enhance the therapeutics by combining MH therapy with chemotherapy, radiotherapy, immunotherapy, photothermal/photodynamic therapy (PDT), and gene therapy will be reviewed. Lastly, the prospect and significant challenges in MH-based antitumor therapy will be discussed. This review is to provide a comprehensive understanding of MH for improving antitumor therapeutic efficacy, which would be of utmost benefit towards guiding the users and for the future development of MNPs-MH towards successful application in medicine.
Collapse
Affiliation(s)
- Xiaoli Liu
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education; School of Medicine, Northwest University, Xi'an 710069, China
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, No. 11, First North Road, Zhongguancun, Beijing 100190, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yifan Zhang
- Key Laboratory of Synthetic and Natural Functional Molecule Chemistry of the Ministry of Education, College of Chemistry and Materials Science, Northwest University, Xi'an 710127, China
| | - Yanyun Wang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education; School of Medicine, Northwest University, Xi'an 710069, China
| | - Wenjing Zhu
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education; School of Medicine, Northwest University, Xi'an 710069, China
| | - Galong Li
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education; School of Medicine, Northwest University, Xi'an 710069, China
| | - Xiaowei Ma
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, No. 11, First North Road, Zhongguancun, Beijing 100190, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yihan Zhang
- Key Laboratory of Synthetic and Natural Functional Molecule Chemistry of the Ministry of Education, College of Chemistry and Materials Science, Northwest University, Xi'an 710127, China
| | - Shizhu Chen
- Beijing General Pharmaceutical Corporation, Beijing 100101, China
- The National Institutes of Pharmaceutical R&D Co., Ltd., China Resources Pharmaceutical Group Limited, Beijing 102206, China
| | - Shivani Tiwari
- Key Laboratory of Synthetic and Natural Functional Molecule Chemistry of the Ministry of Education, College of Chemistry and Materials Science, Northwest University, Xi'an 710127, China
| | - Kejian Shi
- Beijing Institute of Traumatology and Orthopaedics, Beijing 100035, China
| | - Shouwen Zhang
- Neurophysiology Department, Beijing ChaoYang Emergency Medical Center, Beijing 100122, China
| | - Hai Ming Fan
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education; School of Medicine, Northwest University, Xi'an 710069, China
- Key Laboratory of Synthetic and Natural Functional Molecule Chemistry of the Ministry of Education, College of Chemistry and Materials Science, Northwest University, Xi'an 710127, China
| | - Yong Xiang Zhao
- National Center for International Research of Biotargeting Theranostics, Guangxi Key Laboratory of Biotargeting Theranostics, Collaborative Innovation Center for Targeting Tumour Theranostics and Therapy, Guangxi Medical University, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| | - Xing-Jie Liang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, No. 11, First North Road, Zhongguancun, Beijing 100190, China; University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
105
|
Abstract
Developments of new strategies to restore vision and improving on current strategies by harnessing new advancements in material and electrical sciences, and biological and genetic-based technologies are of upmost health priorities around the world. Federal and private entities are spending billions of dollars on visual prosthetics technologies. This review describes the most current and state-of-the-art bioengineering technologies to restore vision. This includes a thorough description of traditional electrode-based visual prosthetics that have improved substantially since early prototypes. Recent advances in molecular and synthetic biology have transformed vision-assisted technologies; For example, optogenetic technologies that introduce light-responsive proteins offer excellent resolution but cortical applications are restricted by fiber implantation and tissue damage. Other stimulation modalities, such as magnetic fields, have been explored to achieve non-invasive neuromodulation. Miniature magnetic coils are currently being developed to activate select groups of neurons. Magnetically-responsive nanoparticles or exogenous proteins can significantly enhance the coupling between external electromagnetic devices and any neurons affiliated with these modifications. The need to minimize cytotoxic effects for nanoparticle-based therapies will likely restrict the number of usable materials. Nevertheless, advances in identifying and utilizing proteins that respond to magnetic fields may lead to non-invasive, cell-specific stimulation and may overcome many of the limitations that currently exist with other methods. Finally, sensory substitution systems also serve as viable visual prostheses by converting visual input to auditory and somatosensory stimuli. This review also discusses major challenges in the field and offers bioengineering strategies to overcome those.
Collapse
Affiliation(s)
- Alexander Farnum
- Department of Biomedical Engineering, College of Engineering, Michigan State University, East Lansing, MI, United States
- Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, United States
| | - Galit Pelled
- Department of Biomedical Engineering, College of Engineering, Michigan State University, East Lansing, MI, United States
- Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, United States
- Department of Radiology, College of Human Medicine, Michigan State University, East Lansing, MI, United States
| |
Collapse
|
106
|
Chandrasekharan P, Tay ZW, Hensley D, Zhou XY, Fung BKL, Colson C, Lu Y, Fellows BD, Huynh Q, Saayujya C, Yu E, Orendorff R, Zheng B, Goodwill P, Rinaldi C, Conolly S. Using magnetic particle imaging systems to localize and guide magnetic hyperthermia treatment: tracers, hardware, and future medical applications. Am J Cancer Res 2020; 10:2965-2981. [PMID: 32194849 PMCID: PMC7053197 DOI: 10.7150/thno.40858] [Citation(s) in RCA: 80] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Accepted: 01/27/2020] [Indexed: 01/07/2023] Open
Abstract
Magnetic fluid hyperthermia (MFH) treatment makes use of a suspension of superparamagnetic iron oxide nanoparticles, administered systemically or locally, in combination with an externally applied alternating magnetic field, to ablate target tissue by generating heat through a process called induction. The heat generated above the mammalian euthermic temperature of 37°C induces apoptotic cell death and/or enhances the susceptibility of the target tissue to other therapies such as radiation and chemotherapy. While most hyperthermia techniques currently in development are targeted towards cancer treatment, hyperthermia is also used to treat restenosis, to remove plaques, to ablate nerves and to alleviate pain by increasing regional blood flow. While RF hyperthermia can be directed invasively towards the site of treatment, non-invasive localization of heat through induction is challenging. In this review, we discuss recent progress in the field of RF magnetic fluid hyperthermia and introduce a new diagnostic imaging modality called magnetic particle imaging that allows for a focused theranostic approach encompassing treatment planning, treatment monitoring and spatially localized inductive heating.
Collapse
Affiliation(s)
- Prashant Chandrasekharan
- University of California Berkeley, Department of Bioengineering, Berkeley, CA 94720, United States,✉ Corresponding author: E-mail: ; Phone: +1 (510) 642 3420
| | - Zhi Wei Tay
- University of California Berkeley, Department of Bioengineering, Berkeley, CA 94720, United States
| | - Daniel Hensley
- Magnetic Insight, Inc., Alameda, CA 94501, United States
| | - Xinyi Y Zhou
- University of California Berkeley, Department of Bioengineering, Berkeley, CA 94720, United States
| | - Barry KL Fung
- University of California Berkeley, Department of Bioengineering, Berkeley, CA 94720, United States
| | - Caylin Colson
- University of California Berkeley, Department of Bioengineering, Berkeley, CA 94720, United States
| | - Yao Lu
- University of California Berkeley, Department of Bioengineering, Berkeley, CA 94720, United States
| | - Benjamin D Fellows
- University of California Berkeley, Department of Bioengineering, Berkeley, CA 94720, United States
| | - Quincy Huynh
- Department of Electrical Engineering and Computer Sciences, University of California, Berkeley, CA 94720, United States
| | - Chinmoy Saayujya
- Department of Electrical Engineering and Computer Sciences, University of California, Berkeley, CA 94720, United States
| | - Elaine Yu
- Magnetic Insight, Inc., Alameda, CA 94501, United States
| | - Ryan Orendorff
- Magnetic Insight, Inc., Alameda, CA 94501, United States
| | - Bo Zheng
- University of California Berkeley, Department of Bioengineering, Berkeley, CA 94720, United States
| | | | - Carlos Rinaldi
- University of Florida, J. Crayton Pruitt Family Department of Biomedical Engineering and Department of Chemical Engineering, FL, 32611 United States
| | - Steven Conolly
- University of California Berkeley, Department of Bioengineering, Berkeley, CA 94720, United States,Department of Electrical Engineering and Computer Sciences, University of California, Berkeley, CA 94720, United States
| |
Collapse
|
107
|
Song Y, Li D, Lu Y, Jiang K, Yang Y, Xu Y, Dong L, Yan X, Ling D, Yang X, Yu SH. Ferrimagnetic mPEG-b-PHEP copolymer micelles loaded with iron oxide nanocubes and emodin for enhanced magnetic hyperthermia–chemotherapy. Natl Sci Rev 2020; 7:723-736. [PMID: 34692091 PMCID: PMC8289054 DOI: 10.1093/nsr/nwz201] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 11/18/2019] [Accepted: 12/02/2019] [Indexed: 02/01/2023] Open
Abstract
As a non-invasive therapeutic method without penetration-depth limitation, magnetic hyperthermia therapy (MHT) under alternating magnetic field (AMF) is a clinically promising thermal therapy. However, the poor heating conversion efficiency and lack of stimulus–response obstruct the clinical application of magnetofluid-mediated MHT. Here, we develop a ferrimagnetic polyethylene glycol-poly(2-hexoxy-2-oxo-1,3,2-dioxaphospholane) (mPEG-b-PHEP) copolymer micelle loaded with hydrophobic iron oxide nanocubes and emodin (denoted as EMM). Besides an enhanced magnetic resonance (MR) contrast ability (r2 = 271 mM−1 s−1) due to the high magnetization, the specific absorption rate (2518 W/g at 35 kA/m) and intrinsic loss power (6.5 nHm2/kg) of EMM are dozens of times higher than the clinically available iron oxide nanoagents (Feridex and Resovist), indicating the high heating conversion efficiency. Furthermore, this composite micelle with a flowable core exhibits a rapid response to magnetic hyperthermia, leading to an AMF-activated supersensitive drug release. With the high magnetic response, thermal sensitivity and magnetic targeting, this supersensitive ferrimagnetic nanocomposite realizes an above 70% tumor cell killing effect at an extremely low dosage (10 μg Fe/mL), and the tumors on mice are completely eliminated after the combined MHT–chemotherapy.
Collapse
Affiliation(s)
- Yonghong Song
- Key Laboratory of Advanced Catalytic Materials and Reaction Engineering, School of Chemistry and Chemical Engineering, Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, Hefei University of Technology, Hefei 230009, China
| | - Dongdong Li
- Institutes for Life Sciences, School of Medicine, South China University of Technology, Guangzhou 510006, China
| | - Yang Lu
- Key Laboratory of Advanced Catalytic Materials and Reaction Engineering, School of Chemistry and Chemical Engineering, Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, Hefei University of Technology, Hefei 230009, China
| | - Kun Jiang
- Key Laboratory of Advanced Catalytic Materials and Reaction Engineering, School of Chemistry and Chemical Engineering, Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, Hefei University of Technology, Hefei 230009, China
| | - Yi Yang
- Key Laboratory of Advanced Catalytic Materials and Reaction Engineering, School of Chemistry and Chemical Engineering, Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, Hefei University of Technology, Hefei 230009, China
| | - Yunjun Xu
- Division of Nanomaterials & Chemistry, Hefei National Laboratory for Physical Sciences at the Microscale, CAS Center for Excellence in Nanoscience, Hefei Science Center of CAS, Department of Chemistry, Institute of Biomimetic Materials & Chemistry, University of Science and Technology of China, Hefei 230026, China
| | - Liang Dong
- Division of Nanomaterials & Chemistry, Hefei National Laboratory for Physical Sciences at the Microscale, CAS Center for Excellence in Nanoscience, Hefei Science Center of CAS, Department of Chemistry, Institute of Biomimetic Materials & Chemistry, University of Science and Technology of China, Hefei 230026, China
| | - Xu Yan
- Key Laboratory of Advanced Catalytic Materials and Reaction Engineering, School of Chemistry and Chemical Engineering, Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, Hefei University of Technology, Hefei 230009, China
| | - Daishun Ling
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Key Laboratory of Biomedical Engineering of the Ministry of Education, Zhejiang University, Hangzhou 310058, China
| | - Xianzhu Yang
- Institutes for Life Sciences, School of Medicine, South China University of Technology, Guangzhou 510006, China
| | - Shu-Hong Yu
- Division of Nanomaterials & Chemistry, Hefei National Laboratory for Physical Sciences at the Microscale, CAS Center for Excellence in Nanoscience, Hefei Science Center of CAS, Department of Chemistry, Institute of Biomimetic Materials & Chemistry, University of Science and Technology of China, Hefei 230026, China
| |
Collapse
|
108
|
Mannaris C, Yang C, Carugo D, Owen J, Lee JY, Nwokeoha S, Seth A, Teo BM. Acoustically responsive polydopamine nanodroplets: A novel theranostic agent. ULTRASONICS SONOCHEMISTRY 2020; 60:104782. [PMID: 31539725 DOI: 10.1016/j.ultsonch.2019.104782] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Revised: 08/21/2019] [Accepted: 09/06/2019] [Indexed: 05/06/2023]
Abstract
Ultrasound-induced cavitation has been used as a tool of enhancing extravasation and tissue penetration of anticancer agents in tumours. Initiating cavitation in tissue however, requires high acoustic intensities that are neither safe nor easy to achieve with current clinical systems. The use of cavitation nuclei can however lower the acoustic intensities required to initiate cavitation and the resulting bio-effects in situ. Microbubbles, solid gas-trapping nanoparticles, and phase shift nanodroplets are some examples in a growing list of proposed cavitation nuclei. Besides the ability to lower the cavitation threshold, stability, long circulation times, biocompatibility and biodegradability, are some of the desirable characteristics that a clinically applicable cavitation agent should possess. In this study, we present a novel formulation of ultrasound-triggered phase transition sub-micrometer sized nanodroplets (~400 nm) stabilised with a biocompatible polymer, polydopamine (PDA). PDA offers some important benefits: (1) facile fabrication, as dopamine monomers are directly polymerised on the nanodroplets, (2) high polymer biocompatibility, and (3) ease of functionalisation with other molecules such as drugs or targeting species. We demonstrate that the acoustic intensities required to initiate inertial cavitation can all be achieved with existing clinical ultrasound systems. Cell viability and haemolysis studies show that nanodroplets are biocompatible. Our results demonstrate the great potential of PDA nanodroplets as an acoustically active nanodevice, which is highly valuable for biomedical applications including drug delivery and treatment monitoring.
Collapse
Affiliation(s)
- Christophoros Mannaris
- Institute of Biomedical Engineering, Old Road Campus Research Building, University of Oxford, Oxford OX3 7DQ, UK.
| | - Chuanxu Yang
- Key Laboratory of Colloid and Interface Chemistry, Ministry of Education, Shandong University, Jinan 250100, China
| | - Dario Carugo
- Institute of Biomedical Engineering, Old Road Campus Research Building, University of Oxford, Oxford OX3 7DQ, UK; Mechatronics and Bioengineering Science Research Groups, Faculty of Engineering and the Environment, University of Southampton, Southampton, UK
| | - Joshua Owen
- Institute of Biomedical Engineering, Old Road Campus Research Building, University of Oxford, Oxford OX3 7DQ, UK
| | - Jeong Yu Lee
- Institute of Biomedical Engineering, Old Road Campus Research Building, University of Oxford, Oxford OX3 7DQ, UK
| | - Sandra Nwokeoha
- Institute of Biomedical Engineering, Old Road Campus Research Building, University of Oxford, Oxford OX3 7DQ, UK
| | - Anjali Seth
- Institute of Biomedical Engineering, Old Road Campus Research Building, University of Oxford, Oxford OX3 7DQ, UK
| | - Boon Mian Teo
- Institute of Biomedical Engineering, Old Road Campus Research Building, University of Oxford, Oxford OX3 7DQ, UK; Key Laboratory of Colloid and Interface Chemistry, Ministry of Education, Shandong University, Jinan 250100, China; School of Chemistry, Clayton Campus, Monash University Victoria, 3800, Australia.
| |
Collapse
|
109
|
Tong S, Zhu H, Bao G. Magnetic Iron Oxide Nanoparticles for Disease Detection and Therapy. MATERIALS TODAY (KIDLINGTON, ENGLAND) 2019; 31:86-99. [PMID: 32831620 PMCID: PMC7441585 DOI: 10.1016/j.mattod.2019.06.003] [Citation(s) in RCA: 88] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
Magnetic iron oxide nanoparticles (MIONs) are among the first generation of nanomaterials that have advanced to clinic use. A broad range of biomedical techniques has been developed by combining the versatile nanomagnetism of MIONs with various forms of applied magnetic fields. MIONs can generate imaging contrast and provide mechanical/thermal energy in vivo in response to an external magnetic field, a special feature that distinguishes MIONs from other nanomaterials. These properties offer unique opportunities for nanomaterials engineering in biomedical research and clinical interventions. The past few decades have witnessed the evolution of the applications of MIONs from conventional drug delivery and hyperthermia to the regulation of molecular and cellular processes in the body. Here we review the most recent development in this field, including clinical studies of MIONs and the emerging techniques that may contribute to future innovation in medicine.
Collapse
Affiliation(s)
- Sheng Tong
- Department of Bioengineering, Rice University, Houston, Texas 77030, USA
| | - Haibao Zhu
- Department of Bioengineering, Rice University, Houston, Texas 77030, USA
| | - Gang Bao
- Department of Bioengineering, Rice University, Houston, Texas 77030, USA
| |
Collapse
|
110
|
Chen Y, Liang Y, Jiang P, Li F, Yu B, Yan F. Lipid/PLGA Hybrid Microbubbles as a Versatile Platform for Noninvasive Image-Guided Targeted Drug Delivery. ACS APPLIED MATERIALS & INTERFACES 2019; 11:41842-41852. [PMID: 31633326 DOI: 10.1021/acsami.9b10188] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
Microbubbles (MBs) have recently emerged as promising theranostic carriers for ultrasound contrast imaging and drug delivery. However, conventional lipid-based MBs have a poor drug encapsulation efficiency, and polymer-based MBs show a weak capability in contrast imaging and ultrasound-triggered drug release. Here, we developed a novel type of multiporous lipid/PLGA hybrid MBs (lipid/PLGA MBs) that solved the dilemma of MBs as imaging agents and drug carriers. The lipid/PLGA MBs were designed through regulating the elasticity of the bubble shells using lipids to incorporate into the PLGA shells and ammonium bicarbonate as a gas-generating agent. The softened shells and the porous bubble structure make them be able to generate stronger harmonic signals and be more vulnerable to ultrasound irradiation, leading to their excellent performance in ultrasound contrast imaging and ultrasound-triggered MB destruction in vitro and in vivo. By using doxorubicin (Dox) as a model drug, the Dox-loaded lipid/PLGA MBs (Dox-lipid/PLGA MBs) were prepared and achieved a high drug encapsulation efficiency. The real-time tracking of drug delivery and on-command controlled drug release by ultrasound were successfully realized in the tumor-bearing mice. A significantly enhanced tumor growth inhibition effect could be observed when using Dox-lipid/PLGA MBs combined with ultrasound irradiation, compared with free Dox and Dox-lipid/PLGA MBs without ultrasound. Our study provides an innovative multifunctional platform of MBs for ultrasound contrast imaging and drug delivery applications.
Collapse
Affiliation(s)
- Yan Chen
- Ultrasound Medical Center , Zhujiang Hospital of Southern Medical University , Guangzhou 510282 , China
| | - Yangbiao Liang
- Ultrasound Medical Center , Zhujiang Hospital of Southern Medical University , Guangzhou 510282 , China
| | - Peng Jiang
- Paul C. Lauterbur Research Center for Biomedical Imaging, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology , Chinese Academy of Sciences , Shenzhen 518055 , China
| | - Fei Li
- Paul C. Lauterbur Research Center for Biomedical Imaging, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology , Chinese Academy of Sciences , Shenzhen 518055 , China
| | - Bo Yu
- Ultrasound Medical Center , Zhujiang Hospital of Southern Medical University , Guangzhou 510282 , China
| | - Fei Yan
- Paul C. Lauterbur Research Center for Biomedical Imaging, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology , Chinese Academy of Sciences , Shenzhen 518055 , China
| |
Collapse
|
111
|
Xiong R, Zhang W, Zhang Y, Zhang Y, Chen Y, He Y, Fan H. Remote and real time control of an FVIO-enzyme hybrid nanocatalyst using magnetic stimulation. NANOSCALE 2019; 11:18081-18089. [PMID: 31343649 DOI: 10.1039/c9nr04289j] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Remote modulation of nanoscale biochemical processes in a living system using magnetic stimulation is appealing but is restricted by the lack of a highly efficient nanomediator which can deliver timely and effective response to biological molecules under an external magnetic field. Herein, we report the development of a novel nanocatalyst based on a ferrimagnetic vortex-domain nanoring (FVIO)-enzyme hybrid that enables real-time modulation of enzymatic catalysis under an alternating magnetic field (AMF). The role of the FVIO is to provide localized heating immediately upon exposure to an AMF, which efficiently and selectively promotes the activity of conjugated enzymes on the surface. The reaction rate of the as-fabricated FVIO-β-Gal hybrid was shown to be boosted up to 180% of its initial value by localized heat generated under an AMF of 550 Oe in less than 2 s and without heating up the bulk solution. Moreover, the degree of activity acceleration was shown to be tunable by increasing the strength of the AMF. The concept of remote magnetic stimulation of enzymatic reactions has been further applied to other enzymes (e.g. FVIO-KPC and FVIO-GOx), demonstrating the general applicability of this strategy. Since almost all metabolic processes in cells rely on enzymatic catalysis to sustain life, the FVIO-enzyme system developed in this work provides a valuable nanoplatform for spatiotemporally manipulating biochemical reactions, which might pave the way for future remote manipulation of living organisms.
Collapse
Affiliation(s)
- Ran Xiong
- College of Chemistry and Materials Science, Key Laboratory of Synthetic and Natural Functional Molecule Chemistry of Ministry of Education, Northwest University, 1 Xue Fu Avenue, Xi'an, 710127, Shaanxi, China.
| | | | | | | | | | | | | |
Collapse
|
112
|
Rao S, Chen R, LaRocca AA, Christiansen MG, Senko AW, Shi CH, Chiang PH, Varnavides G, Xue J, Zhou Y, Park S, Ding R, Moon J, Feng G, Anikeeva P. Remotely controlled chemomagnetic modulation of targeted neural circuits. NATURE NANOTECHNOLOGY 2019; 14:967-973. [PMID: 31427746 PMCID: PMC6778020 DOI: 10.1038/s41565-019-0521-z] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 07/03/2019] [Indexed: 05/20/2023]
Abstract
Connecting neural circuit output to behaviour can be facilitated by the precise chemical manipulation of specific cell populations1,2. Engineered receptors exclusively activated by designer small molecules enable manipulation of specific neural pathways3,4. However, their application to studies of behaviour has thus far been hampered by a trade-off between the low temporal resolution of systemic injection versus the invasiveness of implanted cannulae or infusion pumps2. Here, we developed a remotely controlled chemomagnetic modulation-a nanomaterials-based technique that permits the pharmacological interrogation of targeted neural populations in freely moving subjects. The heat dissipated by magnetic nanoparticles (MNPs) in the presence of alternating magnetic fields (AMFs) triggers small-molecule release from thermally sensitive lipid vesicles with a 20 s latency. Coupled with the chemogenetic activation of engineered receptors, this technique permits the control of specific neurons with temporal and spatial precision. The delivery of chemomagnetic particles to the ventral tegmental area (VTA) allows the remote modulation of motivated behaviour in mice. Furthermore, this chemomagnetic approach activates endogenous circuits by enabling the regulated release of receptor ligands. Applied to an endogenous dopamine receptor D1 (DRD1) agonist in the nucleus accumbens (NAc), a brain area involved in mediating social interactions, chemomagnetic modulation increases sociability in mice. By offering a temporally precise control of specified ligand-receptor interactions in neurons, this approach may facilitate molecular neuroscience studies in behaving organisms.
Collapse
Affiliation(s)
- Siyuan Rao
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA
- Simons Center for Social Brain, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Ritchie Chen
- Simons Center for Social Brain, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Ava A LaRocca
- Department of Materials Science and Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Michael G Christiansen
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Materials Science and Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Health Sciences and Technology at the Swiss Federal Institute of Technology in Zürich (ETHZ), Zürich, Switzerland
| | - Alexander W Senko
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Materials Science and Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Cindy H Shi
- Department of Materials Science and Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Po-Han Chiang
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Georgios Varnavides
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Materials Science and Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
| | - Jian Xue
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Yang Zhou
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Seongjun Park
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Ruihua Ding
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
| | - Junsang Moon
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Materials Science and Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Guoping Feng
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Polina Anikeeva
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Department of Materials Science and Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA.
| |
Collapse
|
113
|
Reply to: Magneto is ineffective in controlling electrical properties of cerebellar Purkinje cells, Assessing the utility of Magneto to control neuronal excitability in the somatosensory cortex and Revaluation of magnetic properties of Magneto. Nat Neurosci 2019; 23:1051-1054. [PMID: 31570860 DOI: 10.1038/s41593-019-0472-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Accepted: 07/23/2019] [Indexed: 01/16/2023]
|
114
|
Tastanova A, Folcher M, Müller M, Camenisch G, Ponti A, Horn T, Tikhomirova MS, Fussenegger M. Synthetic biology-based cellular biomedical tattoo for detection of hypercalcemia associated with cancer. Sci Transl Med 2019; 10:10/437/eaap8562. [PMID: 29669854 DOI: 10.1126/scitranslmed.aap8562] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2017] [Accepted: 03/29/2018] [Indexed: 12/12/2022]
Abstract
Diagnosis marks the beginning of any successful therapy. Because many medical conditions progress asymptomatically over extended periods of time, their timely diagnosis remains difficult, and this adversely affects patient prognosis. Focusing on hypercalcemia associated with cancer, we aimed to develop a synthetic biology-inspired biomedical tattoo using engineered cells that would (i) monitor long-term blood calcium concentration, (ii) detect onset of mild hypercalcemia, and (iii) respond via subcutaneous accumulation of the black pigment melanin to form a visible tattoo. For this purpose, we designed cells containing an ectopically expressed calcium-sensing receptor rewired to a synthetic signaling cascade that activates expression of transgenic tyrosinase, which produces melanin in response to persistently increased blood Ca2+ We confirmed that the melanin-generated color change produced by this biomedical tattoo could be detected with the naked eye and optically quantified. The system was validated in wild-type mice bearing subcutaneously implanted encapsulated engineered cells. All animals inoculated with hypercalcemic breast and colon adenocarcinoma cells developed tattoos, whereas no tattoos were seen in animals inoculated with normocalcemic tumor cells. All tumor-bearing animals remained asymptomatic throughout the 38-day experimental period. Although hypercalcemia is also associated with other pathologies, our findings demonstrate that it is possible to detect hypercalcemia associated with cancer in murine models using this cell-based diagnostic strategy.
Collapse
Affiliation(s)
- Aizhan Tastanova
- Department of Biosystems Science and Engineering, ETH Zurich, Mattenstrasse 26, CH-4058 Basel, Switzerland
| | - Marc Folcher
- Department of Biosystems Science and Engineering, ETH Zurich, Mattenstrasse 26, CH-4058 Basel, Switzerland
| | - Marius Müller
- Department of Biosystems Science and Engineering, ETH Zurich, Mattenstrasse 26, CH-4058 Basel, Switzerland
| | - Gieri Camenisch
- Department of Biosystems Science and Engineering, ETH Zurich, Mattenstrasse 26, CH-4058 Basel, Switzerland
| | - Aaron Ponti
- Department of Biosystems Science and Engineering, ETH Zurich, Mattenstrasse 26, CH-4058 Basel, Switzerland
| | - Thomas Horn
- Department of Biosystems Science and Engineering, ETH Zurich, Mattenstrasse 26, CH-4058 Basel, Switzerland
| | - Maria S Tikhomirova
- 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
|
115
|
Sardoiwala MN, Srivastava AK, Karmakar S, Roy Choudhury S. Nanostructure Endows Neurotherapeutic Potential in Optogenetics: Current Development and Future Prospects. ACS Chem Neurosci 2019; 10:3375-3385. [PMID: 31244053 DOI: 10.1021/acschemneuro.9b00246] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Optogenetics have evolved as a promising tool to control the processes at a cellular level via photons. Specially, it confers a specific control over cellular function through real-time cytomodulation even in freely moving animals. Neuronal stimulation is prerequisite for deep tissue light penetration or insertion of optrode for light illumination to the neurons that have been proven to be compromised due to poor light penetration and invasiveness of the procedure, respectively. In this review, the application of nanotechnology is being elaborated by the use of metal nanoparticles (AuNPs), upconversion nanocrystals (UCNPs), and quantum dots (CdSe) for targeting particular organs or tissues, and their potential to emit a specific light on excitation to overcome the limitations associated with earlier methods has been elucidated. The optothermal and magnetothermal properties, photoluminescence, and higher photostability of nanomaterials are explored in context of therapeutic applicability of optogenetics. The nanostructure characteristics and specific ion channel targeting have shown promising therapeutic potential against neurodegenerative disorders (Alzheimer's, Parkinson's, Huntington's), epilepsy, and blindness. This review compiles mechanical and optical characteristics of nanomaterials that endow superior optogenetic therapeutic potentials to cure immedicable infirmities.
Collapse
Affiliation(s)
| | - Anup K. Srivastava
- Institute of Nano Science and Technology, Habitat Centre, Phase-10, Mohali, Punjab 160062, India
| | - Surajit Karmakar
- Institute of Nano Science and Technology, Habitat Centre, Phase-10, Mohali, Punjab 160062, India
| | - Subhasree Roy Choudhury
- Institute of Nano Science and Technology, Habitat Centre, Phase-10, Mohali, Punjab 160062, India
| |
Collapse
|
116
|
Barbic M. Possible magneto-mechanical and magneto-thermal mechanisms of ion channel activation in magnetogenetics. eLife 2019; 8:45807. [PMID: 31373554 PMCID: PMC6693891 DOI: 10.7554/elife.45807] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Accepted: 07/28/2019] [Indexed: 01/11/2023] Open
Abstract
The palette of tools for perturbation of neural activity is continually expanding. On the forefront of this expansion is magnetogenetics, where ion channels are genetically engineered to be closely coupled to the iron-storage protein ferritin. Initial reports on magnetogenetics have sparked a vigorous debate on the plausibility of physical mechanisms of ion channel activation by means of external magnetic fields. The criticism leveled against magnetogenetics as being physically implausible is based on the specific assumptions about the magnetic spin configurations of iron in ferritin. I consider here a wider range of possible spin configurations of iron in ferritin and the consequences these might have in magnetogenetics. I propose several new magneto-mechanical and magneto-thermal mechanisms of ion channel activation that may clarify some of the mysteries that presently challenge our understanding of the reported biological experiments. Finally, I present some additional puzzles that will require further theoretical and experimental investigation.
Collapse
Affiliation(s)
- Mladen Barbic
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, United States
| |
Collapse
|
117
|
Li J, Duan H, Pu K. Nanotransducers for Near-Infrared Photoregulation in Biomedicine. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2019; 31:e1901607. [PMID: 31199021 DOI: 10.1002/adma.201901607] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Revised: 04/05/2019] [Indexed: 06/09/2023]
Abstract
Photoregulation, which utilizes light to remotely control biological events, provides a precise way to decipher biology and innovate in medicine; however, its potential is limited by the shallow tissue penetration and/or phototoxicity of ultraviolet (UV)/visible light that are required to match the optical responses of endogenous photosensitive substances. Thereby, biologically friendly near-infrared (NIR) light with improved tissue penetration is desired for photoregulation. Since there are a few endogenous biomolecules absorbing or emitting light in the NIR region, the development of molecular transducers is essential to convert NIR light into the cues for regulation of biological events. In this regard, optical nanomaterials able to convert NIR light into UV/visible light, heat, or free radicals are suitable for this task. Here, the recent developments of optical nanotransducers for NIR-light-mediated photoregulation in medicine are summarized. The emerging applications, including photoregulation of neural activity, gene expression, and visual systems, as well as photochemical tissue bonding, are highlighted, along with the design principles of nanotransducers. Moreover, the current challenges and perspectives in this field are discussed.
Collapse
Affiliation(s)
- Jingchao Li
- School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore, 637457, Singapore
| | - Hongwei Duan
- School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore, 637457, Singapore
| | - Kanyi Pu
- School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore, 637457, Singapore
| |
Collapse
|
118
|
Liu Y, Charpin-El Hamri G, Ye H, Fussenegger M. A synthetic free fatty acid-regulated transgene switch in mammalian cells and mice. Nucleic Acids Res 2019; 46:9864-9874. [PMID: 30219861 PMCID: PMC6182168 DOI: 10.1093/nar/gky805] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2018] [Accepted: 08/28/2018] [Indexed: 02/06/2023] Open
Abstract
Trigger-inducible transgene expression systems are utilized in biopharmaceutical manufacturing and also to enable controlled release of therapeutic agents in vivo. We considered that free fatty acids (FFAs), which are dietary components, signaling molecules and important biomarkers, would be attractive candidates as triggers for novel transgene switches with many potential applications, e.g. in future gene- and cell-based therapies. To develop such a switch, we rewired the signal pathway of human G-protein coupled receptor 40 to a chimeric promoter triggering gene expression through an increase of intracellular calcium concentration. This synthetic gene switch is responsive to physiologically relevant FFA concentrations in different mammalian cell types grown in culture or in a bioreactor, or implanted into mice. Animal recipients of microencapsulated sensor cells containing this switch exhibited significant transgene induction following consumption of dietary fat (such as Swiss cheese) or under hyperlipidaemic conditions, including obesity, diabetes and lipodystrophy.
Collapse
Affiliation(s)
- Ying Liu
- Department of Biosystems Science and Engineering, ETH Zurich, Mattenstrasse 26, CH-4058 Basel, Switzerland
| | - Ghislaine Charpin-El Hamri
- Département Génie Biologique, Université Claude Bernard 1, 43 Boulevard du 11 Novembre 1918, F-69100 Villeurbanne, France
| | - Haifeng Ye
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Dongchuan Road 500, Shanghai 200241, China
| | - 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
|
119
|
Berthoud HR, Neuhuber WL. Vagal mechanisms as neuromodulatory targets for the treatment of metabolic disease. Ann N Y Acad Sci 2019; 1454:42-55. [PMID: 31268181 PMCID: PMC6810744 DOI: 10.1111/nyas.14182] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 05/23/2019] [Accepted: 06/05/2019] [Indexed: 12/30/2022]
Abstract
With few effective treatments available, the global rise of metabolic diseases, including obesity, type 2 diabetes mellitus, and cardiovascular disease, seems unstoppable. Likely caused by an obesogenic environment interacting with genetic susceptibility, the pathophysiology of obesity and metabolic diseases is highly complex and involves crosstalk between many organs and systems, including the brain. The vagus nerve is in a key position to bidirectionally link several peripheral metabolic organs with the brain and is increasingly targeted for neuromodulation therapy to treat metabolic disease. Here, we review the basics of vagal functional anatomy and its implications for vagal neuromodulation therapies. We find that most existing vagal neuromodulation techniques either ignore or misinterpret the rich functional specificity of both vagal efferents and afferents as demonstrated by a large body of literature. This lack of specificity of manipulating vagal fibers is likely the reason for the relatively poor beneficial long‐term effects of such therapies. For these therapies to become more effective, rigorous validation of all physiological endpoints and optimization of stimulation parameters as well as electrode placements will be necessary. However, given the large number of function‐specific fibers in any vagal branch, genetically guided neuromodulation techniques are more likely to succeed.
Collapse
Affiliation(s)
- Hans-Rudolf Berthoud
- Neurobiology of Nutrition and Metabolism Department, Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, Louisiana
| | - Winfried L Neuhuber
- Institut fur Anatomie und Zellbiologie, Friedrich-Alexander Universität Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
120
|
Kashida S, Wang DO, Saito H, Gueroui Z. Nanoparticle-based local translation reveals mRNA as a translation-coupled scaffold with anchoring function. Proc Natl Acad Sci U S A 2019; 116:13346-13351. [PMID: 31217293 PMCID: PMC6613171 DOI: 10.1073/pnas.1900310116] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The spatial regulation of messenger RNA (mRNA) translation is central to cellular functions and relies on numerous complex processes. Biomimetic approaches could bypass these endogenous complex processes, improve our comprehension of the regulation, and allow for controlling local translation regulations and functions. However, the causality between local translation and nascent protein function remains elusive. Here, we developed a nanoparticle (NP)-based strategy to magnetically control mRNA spatial patterns in mammalian cell extracts and investigate how local translation impacts nascent protein localization and function. By monitoring the translation of the magnetically localized mRNAs, we show that mRNA-NP complexes operate as a source for the continuous production of proteins from defined positions. By applying this approach to actin-binding proteins, we triggered the local formation of actin cytoskeletons and identified the minimal requirements for spatial control of the actin filament network. In addition, our bottom-up approach identified a role for mRNA as a translation-coupled scaffold for the function of nascent N-terminal protein domains. Our approach will serve as a platform for regulating mRNA localization and investigating the function of nascent protein domains during translation.
Collapse
Affiliation(s)
- Shunnichi Kashida
- PASTEUR, Département de chimie, École normale supérieure, Paris Sciences et Lettres (PSL) University, Sorbonne Université, CNRS, 75005 Paris, France
| | - Dan Ohtan Wang
- Institute for Integrated Cell-Material Sciences, Kyoto University, 606-8501 Kyoto, Japan
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning, 110016, People's Republic of China
| | - Hirohide Saito
- Department of Life Science Frontiers, Center for iPS Cell Research and Application, Kyoto University, 606-8507 Kyoto, Japan
| | - Zoher Gueroui
- PASTEUR, Département de chimie, École normale supérieure, Paris Sciences et Lettres (PSL) University, Sorbonne Université, CNRS, 75005 Paris, France;
| |
Collapse
|
121
|
Zhang D, Zhang C, Lan S, Huang Y, Liu J, Li J, Liu X, Yang H. Near-Infrared Light Activated Thermosensitive Ion Channel to Remotely Control Transgene System for Thrombolysis Therapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2019; 15:e1901176. [PMID: 31094078 DOI: 10.1002/smll.201901176] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 04/25/2019] [Indexed: 06/09/2023]
Abstract
Current antithrombotic therapeutic strategies often suffer from severe post-thrombotic syndromes (PTS), inconvenient daily subcutaneous injections for a long time and short circulation times accompanied by a dose-dependent risk of intracranial hemorrhage. Aiming at noninvasive, on-demand, and sustained antithrombotic therapy, a new thrombolysis approach based on the transgene system has been developed to remotely and precisely control the expression of urokinase plasminogen activator (uPA) by bioengineered cells for antithrombotic therapy both in vitro and in vivo. In this design, the near-infrared (NIR) light could activate the expression of the thermosensitive TRPV1 channel in response to photothermal responsive nanotransducers to trigger the synthetic signaling pathway to secret uPA. By encapsulating bioengineered cells in injectable hydrogel to ensure long-term survival and convenience for injection, the engineered cells could noninvasively and precisely control the production of uPA protein in situ via an NIR laser to significantly enhance the thrombolysis therapeutic effects by spatiotemporally controlling the local temperature, in both the microfluidic blood circulation mimic and the murine tail thrombus model. This novel thrombolysis approach could overcome some key limitations that are associated with conventional antithrombotic therapy, thus opening a new direction for developing remotely and precisely controllable continuous thrombolysis through artificially designed signaling.
Collapse
Affiliation(s)
- Da Zhang
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, Fujian Provincial Key Laboratory of Analysis and Detection Technology for Food Safety, State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou, 350116, P. R. China
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, 350025, P. R. China
| | - Cuilin Zhang
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, 350025, P. R. China
| | - Shanyou Lan
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, 350025, P. R. China
| | - Yanbing Huang
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, 350025, P. R. China
| | - Jingfeng Liu
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, 350025, P. R. China
| | - Juan Li
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, Fujian Provincial Key Laboratory of Analysis and Detection Technology for Food Safety, State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou, 350116, P. R. China
- Institute of Molecular Medicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200240, P. R. China
| | - Xiaolong Liu
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, 350025, P. R. China
| | - Huanghao Yang
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, Fujian Provincial Key Laboratory of Analysis and Detection Technology for Food Safety, State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou, 350116, P. R. China
| |
Collapse
|
122
|
Theoretical Analysis for Wireless Magnetothermal Deep Brain Stimulation Using Commercial Nanoparticles. Int J Mol Sci 2019; 20:ijms20122873. [PMID: 31212841 PMCID: PMC6627245 DOI: 10.3390/ijms20122873] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Revised: 06/09/2019] [Accepted: 06/10/2019] [Indexed: 12/22/2022] Open
Abstract
A wireless magnetothermal stimulation (WMS) is suggested as a fast, tetherless, and implanted device-free stimulation method using low-radio frequency (100 kHz to 1 MHz) alternating magnetic fields (AMF). As magnetic nanoparticles (MNPs) can transduce alternating magnetic fields into heat, they are targeted to a region of the brain expressing the temperature-sensitive ion channel (TRPV1). The local temperature of the targeted area is increased up to 44 °C to open the TRPV1 channels and cause an influx of Ca2+ sensitive promoter, which can activate individual neurons inside the brain. The WMS has initially succeeded in showing the potential of thermomagnetics for the remote control of neural cell activity with MNPs that are internally targeted to the brain. In this paper, by using the steady-state temperature rise defined by Fourier’s law, the bio-heat equation, and COMSOL Multiphysics software, we investigate most of the basic parameters such as the specific loss power (SLP) of MNPs, the injection volume of magnetic fluid, stimulation and cooling times, and cytotoxic effects at high temperatures (43–44 °C) to provide a realizable design guideline for WMS.
Collapse
|
123
|
White BD, Duan C, Townley HE. Nanoparticle Activation Methods in Cancer Treatment. Biomolecules 2019; 9:E202. [PMID: 31137744 PMCID: PMC6572460 DOI: 10.3390/biom9050202] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 05/17/2019] [Accepted: 05/22/2019] [Indexed: 12/25/2022] Open
Abstract
In this review, we intend to highlight the progress which has been made in recent years around different types of smart activation nanosystems for cancer treatment. Conventional treatment methods, such as chemotherapy or radiotherapy, suffer from a lack of specific targeting and consequent off-target effects. This has led to the development of smart nanosystems which can effect specific regional and temporal activation. In this review, we will discuss the different methodologies which have been designed to permit activation at the tumour site. These can be divided into mechanisms which take advantage of the differences between healthy cells and cancer cells to trigger activation, and those which activate by a mechanism extrinsic to the cell or tumour environment.
Collapse
Affiliation(s)
- Benjamin D White
- Department of Engineering Science, Oxford University, Parks Road, OX1 3PJ, Oxford, UK.
| | - Chengchen Duan
- Nuffield department of Women's and Reproductive Health, Oxford University John Radcliffe Hospital, Headington, Oxford, OX3 9DU, UK.
| | - Helen E Townley
- Department of Engineering Science, Oxford University, Parks Road, OX1 3PJ, Oxford, UK.
- Nuffield department of Women's and Reproductive Health, Oxford University John Radcliffe Hospital, Headington, Oxford, OX3 9DU, UK.
| |
Collapse
|
124
|
Wang S, Zhang B, Su L, Nie W, Han D, Han G, Zhang H, Chong C, Tan J. Subcellular distributions of iron oxide nanoparticles in rat brains affected by different surface modifications. J Biomed Mater Res A 2019; 107:1988-1998. [PMID: 31067350 DOI: 10.1002/jbm.a.36711] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2018] [Revised: 03/07/2019] [Accepted: 05/02/2019] [Indexed: 12/31/2022]
Abstract
The impact of the surface modification on the subcellular distribution of nanoparticles in the brain remains elusive. The nanoparticles prepared by conjugating polyethylene glycol and maleic anhydride-coated superparamagnetic iron oxide nanoparticles (Mal-SPIONs) with bovine serum albumin (BSA/Mal-SPIONs) and with Arg-Gly-Asp peptide (RGD/Mal-SPIONs) were injected into the rat substantia nigra. Observation of transmission electron microscopy (TEM) samples obtained 24 h after perfusion showed that abundant RGD/Mal-SPIONs accumulated in the myelin sheath, dendrites, axon terminals and mitochondria, and on cell membranes in the brain tissue near the injection site. For rats injected with BSA/Mal-SPIONs, a few nanoparticles accumulated in the myelin sheath, axon terminals, endoplasmic reticulum, mitochondria, Golgi, and lysosomes of neurons and glial cells while least SPIONs in rats injected with Mal-SPIONs were found. TEM pictures showed some Mal-SPIONs were expelled out of the brain. RGD/Mal-SPIONs diffused extensively to the thalamus, frontal cortex, temporal lobe, olfactory bulb, and brain stem after injection. Only a few BSA/Mal-SPIONs diffused to the afore-mentioned brain areas. This work reveals different surface modifications on the iron oxide nanoparticles play crucial roles in their distribution and diffusion in the rat brains.
Collapse
Affiliation(s)
- Sheng Wang
- Key Laboratory of New Processing Technology for Nonferrous Metal and Materials, College of Materials Science and Engineering, Guilin University of Technology, Guilin, China.,Guangxi Key Laboratory of Optical and Electronic Materials and Devices, College of Materials Science and Engineering, Guilin University of Technology, Guilin, China
| | - Baolin Zhang
- Key Laboratory of New Processing Technology for Nonferrous Metal and Materials, College of Materials Science and Engineering, Guilin University of Technology, Guilin, China.,Guangxi Key Laboratory of Optical and Electronic Materials and Devices, College of Materials Science and Engineering, Guilin University of Technology, Guilin, China
| | - Lichao Su
- Key Laboratory of New Processing Technology for Nonferrous Metal and Materials, College of Materials Science and Engineering, Guilin University of Technology, Guilin, China.,Guangxi Key Laboratory of Optical and Electronic Materials and Devices, College of Materials Science and Engineering, Guilin University of Technology, Guilin, China
| | - Wan Nie
- Key Laboratory of New Processing Technology for Nonferrous Metal and Materials, College of Materials Science and Engineering, Guilin University of Technology, Guilin, China.,Guangxi Key Laboratory of Optical and Electronic Materials and Devices, College of Materials Science and Engineering, Guilin University of Technology, Guilin, China
| | - Dong Han
- Key Laboratory of New Processing Technology for Nonferrous Metal and Materials, College of Materials Science and Engineering, Guilin University of Technology, Guilin, China.,Guangxi Key Laboratory of Optical and Electronic Materials and Devices, College of Materials Science and Engineering, Guilin University of Technology, Guilin, China
| | - Guihua Han
- Key Laboratory of New Processing Technology for Nonferrous Metal and Materials, College of Materials Science and Engineering, Guilin University of Technology, Guilin, China.,Guangxi Key Laboratory of Optical and Electronic Materials and Devices, College of Materials Science and Engineering, Guilin University of Technology, Guilin, China
| | - Hao Zhang
- Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin Medical University, Guilin, China
| | - Chuangang Chong
- Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin Medical University, Guilin, China
| | - Jie Tan
- Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin Medical University, Guilin, China
| |
Collapse
|
125
|
Thang DC, Wang Z, Lu X, Xing B. Precise cell behaviors manipulation through light-responsive nano-regulators: recent advance and perspective. Theranostics 2019; 9:3308-3340. [PMID: 31244956 PMCID: PMC6567964 DOI: 10.7150/thno.33888] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Accepted: 04/08/2019] [Indexed: 02/07/2023] Open
Abstract
Nanotechnology-assisted spatiotemporal manipulation of biological events holds great promise in advancing the practice of precision medicine in healthcare systems. The progress in internal and/or external stimuli-responsive nanoplatforms for highly specific cellular regulations and theranostic controls offer potential clinical translations of the revolutionized nanomedicine. To successfully implement this new paradigm, the emerging light-responsive nanoregulators with unparalleled precise cell functions manipulation have gained intensive attention, providing UV-Vis light-triggered photocleavage or photoisomerization studies, as well as near-infrared (NIR) light-mediated deep-tissue applications for stimulating cellular signal cascades and treatment of mortal diseases. This review discusses current developments of light-activatable nanoplatforms for modulations of various cellular events including neuromodulations, stem cell monitoring, immunomanipulation, cancer therapy, and other biological target intervention. In summary, the propagation of light-controlled nanomedicine would place a bright prospect for future medicine.
Collapse
Affiliation(s)
- Do Cong Thang
- Division of Chemistry and Biological Chemistry, School of Physical & Mathematical Sciences, Nanyang Technological University, Singapore 637371, Singapore
| | - Zhimin Wang
- Division of Chemistry and Biological Chemistry, School of Physical & Mathematical Sciences, Nanyang Technological University, Singapore 637371, Singapore
| | - Xiaoling Lu
- International Nanobody Research Center of Guangxi, Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Bengang Xing
- Sino-Singapore International Joint Research Institute (SSIJRI), Guangzhou 510000, China
- Division of Chemistry and Biological Chemistry, School of Physical & Mathematical Sciences, Nanyang Technological University, Singapore 637371, Singapore
| |
Collapse
|
126
|
Stanley SA, Friedman JM. Electromagnetic Regulation of Cell Activity. Cold Spring Harb Perspect Med 2019; 9:cshperspect.a034322. [PMID: 30249601 DOI: 10.1101/cshperspect.a034322] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The ability to observe the effects of rapidly and reversibly regulating cell activity in targeted cell populations has provided numerous physiologic insights. Over the last decade, a wide range of technologies have emerged for regulating cellular activity using optical, chemical, and, more recently, electromagnetic modalities. Electromagnetic fields can freely penetrate cells and tissue and their energy can be absorbed by metal particles. When released, the absorbed energy can in turn gate endogenous or engineered receptors and ion channels to regulate cell activity. In this manner, electromagnetic fields acting on external nanoparticles have been used to exert mechanical forces on cell membranes and organelles to generate heat and interact with thermally activated proteins or to induce receptor aggregation and intracellular signaling. More recently, technologies using genetically encoded nanoparticles composed of the iron storage protein, ferritin, have been used for targeted, temporal control of cell activity in vitro and in vivo. These tools provide a means for noninvasively modulating gene expression, intracellular organelles, such as endosomes, and whole-cell activity both in vitro and in freely moving animals. The use of magnetic fields interacting with external or genetically encoded nanoparticles thus provides a rapid noninvasive means for regulating cell activity.
Collapse
Affiliation(s)
- Sarah A Stanley
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029.,Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York 10029
| | - Jeffrey M Friedman
- Laboratory of Molecular Genetics, Rockefeller University, New York, New York 10065.,Howard Hughes Medical Institute, New York, New York 10065
| |
Collapse
|
127
|
Ito A, Teranishi R, Kamei K, Yamaguchi M, Ono A, Masumoto S, Sonoda Y, Horie M, Kawabe Y, Kamihira M. Magnetically triggered transgene expression in mammalian cells by localized cellular heating of magnetic nanoparticles. J Biosci Bioeng 2019; 128:355-364. [PMID: 30962099 DOI: 10.1016/j.jbiosc.2019.03.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 03/07/2019] [Accepted: 03/11/2019] [Indexed: 10/27/2022]
Abstract
To develop a remote control system of transgene expression through localized cellular heating of magnetic nanoparticles, a heat-inducible transgene expression system was introduced into mammalian cells. Cells were labeled with magnetic nanoparticles and exposed to an alternating magnetic field. The magnetically labeled cells expressed the transgene in a monolayer and multilayered cell sheets in which cells were heated around the magnetic nanoparticles without an apparent temperature increase in the culture medium. Magnetic cells were also generated by genetically engineering with a ferritin gene, and transgene expression could be induced by exposure to an alternating magnetic field. This approach may be applicable to the development of novel gene therapies in cell-based medicine.
Collapse
Affiliation(s)
- Akira Ito
- Department of Chemical Engineering, Faculty of Engineering, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan
| | - Ryoji Teranishi
- Department of Chemical Engineering, Faculty of Engineering, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan
| | - Kazuki Kamei
- Department of Chemical Engineering, Faculty of Engineering, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan
| | - Masaki Yamaguchi
- Department of Chemical Engineering, Faculty of Engineering, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan
| | - Akihiko Ono
- Graduate School of Systems Life Sciences, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan
| | - Shinya Masumoto
- Department of Chemical Engineering, Faculty of Engineering, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan
| | - Yuto Sonoda
- Department of Chemical Engineering, Faculty of Engineering, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan
| | - Masanobu Horie
- Division of Biochemical Engineering, Radioisotope Research Center, Kyoto University, Yoshida Konoe-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Yoshinori Kawabe
- Department of Chemical Engineering, Faculty of Engineering, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan
| | - Masamichi Kamihira
- Department of Chemical Engineering, Faculty of Engineering, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan; Graduate School of Systems Life Sciences, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan.
| |
Collapse
|
128
|
Abstract
Magnetic fields pass through tissue undiminished and without producing harmful effects, motivating their use as a wireless, minimally invasive means to control neural activity. Here, we review mechanisms and techniques coupling magnetic fields to changes in electrochemical potentials across neuronal membranes. Biological magnetoreception, although incompletely understood, is discussed as a potential source of inspiration. The emergence of magnetic properties in materials is reviewed to clarify the distinction between biomolecules containing transition metals and ferrite nanoparticles that exhibit significant net moments. We describe recent developments in the use of magnetic nanomaterials as transducers converting magnetic stimuli to forms readily perceived by neurons and discuss opportunities for multiplexed and bidirectional control as well as the challenges posed by delivery to the brain. The variety of magnetic field conditions and mechanisms by which they can be coupled to neuronal signaling cascades highlights the desirability of continued interchange between magnetism physics and neurobiology.
Collapse
Affiliation(s)
- Michael G Christiansen
- Department of Health Sciences and Technology, Swiss Federal Institute of Technology, 8093 Zürich, Switzerland
| | - Alexander W Senko
- Department of Materials Science and Engineering, Research Laboratory of Electronics, and McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA;
| | - Polina Anikeeva
- Department of Materials Science and Engineering, Research Laboratory of Electronics, and McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA;
| |
Collapse
|
129
|
Roet M, Hescham SA, Jahanshahi A, Rutten BPF, Anikeeva PO, Temel Y. Progress in neuromodulation of the brain: A role for magnetic nanoparticles? Prog Neurobiol 2019; 177:1-14. [PMID: 30878723 DOI: 10.1016/j.pneurobio.2019.03.002] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Revised: 03/05/2019] [Accepted: 03/07/2019] [Indexed: 12/19/2022]
Abstract
The field of neuromodulation is developing rapidly. Current techniques, however, are still limited as they i) either depend on permanent implants, ii) require invasive procedures, iii) are not cell-type specific, iv) involve slow pharmacokinetics or v) have a restricted penetration depth making it difficult to stimulate regions deep within the brain. Refinements into the different fields of neuromodulation are thus needed. In this review, we will provide background information on the different techniques of neuromodulation discussing their latest refinements and future potentials including the implementation of nanoparticles (NPs). In particular we will highlight the usage of magnetic nanoparticles (MNPs) as transducers in advanced neuromodulation. When exposed to an alternating magnetic field (AMF), certain MNPs can generate heat through hysteresis. This MNP heating has been promising in the field of cancer therapy and has recently been introduced as a method for remote and wireless neuromodulation. This indicates that MNPs may aid in the exploration of brain functions via neuromodulation and may eventually be applied for treatment of neuropsychiatric disorders. We will address the materials chemistry of MNPs, their biomedical applications, their delivery into the brain, their mechanisms of stimulation with emphasis on MNP heating and their remote control in living tissue. The final section compares and discusses the parameters used for MNP heating in brain cancer treatment and neuromodulation. Concluding, using MNPs for nanomaterial-mediated neuromodulation seem promising in a variety of techniques and could be applied for different neuropsychiatric disorders when more extensively investigated.
Collapse
Affiliation(s)
- Milaine Roet
- School for Mental Health and Neuroscience, Department of Neurosurgery, Maastricht University, Maastricht, 6200, MD, The Netherlands; European Graduate School of Neuroscience (EURON), The Netherlands
| | - Sarah-Anna Hescham
- School for Mental Health and Neuroscience, Department of Neurosurgery, Maastricht University, Maastricht, 6200, MD, The Netherlands; European Graduate School of Neuroscience (EURON), The Netherlands
| | - Ali Jahanshahi
- School for Mental Health and Neuroscience, Department of Neurosurgery, Maastricht University, Maastricht, 6200, MD, The Netherlands; European Graduate School of Neuroscience (EURON), The Netherlands
| | - Bart P F Rutten
- School for Mental Health and Neuroscience, Department of Psychiatry and Neuropsychology, Maastricht University, Maastricht, 6200, MD, The Netherlands; European Graduate School of Neuroscience (EURON), The Netherlands
| | - Polina O Anikeeva
- Department of Materials Science and Engineering, Department of Brain and Cognitive Sciences, Research Laboratory of Electronics, McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, 02139, MA, United States of America
| | - Yasin Temel
- School for Mental Health and Neuroscience, Department of Neurosurgery, Maastricht University, Maastricht, 6200, MD, The Netherlands; European Graduate School of Neuroscience (EURON), The Netherlands; Department of Neurosurgery, Maastricht University Medical Center, Maastricht, 6202, AZ, The Netherlands.
| |
Collapse
|
130
|
Andres J, Blomeier T, Zurbriggen MD. Synthetic Switches and Regulatory Circuits in Plants. PLANT PHYSIOLOGY 2019; 179:862-884. [PMID: 30692218 PMCID: PMC6393786 DOI: 10.1104/pp.18.01362] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Accepted: 01/18/2019] [Indexed: 05/20/2023]
Abstract
Synthetic biology is an established but ever-growing interdisciplinary field of research currently revolutionizing biomedicine studies and the biotech industry. The engineering of synthetic circuitry in bacterial, yeast, and animal systems prompted considerable advances for the understanding and manipulation of genetic and metabolic networks; however, their implementation in the plant field lags behind. Here, we review theoretical-experimental approaches to the engineering of synthetic chemical- and light-regulated (optogenetic) switches for the targeted interrogation and control of cellular processes, including existing applications in the plant field. We highlight the strategies for the modular assembly of genetic parts into synthetic circuits of different complexity, ranging from Boolean logic gates and oscillatory devices up to semi- and fully synthetic open- and closed-loop molecular and cellular circuits. Finally, we explore potential applications of these approaches for the engineering of novel functionalities in plants, including understanding complex signaling networks, improving crop productivity, and the production of biopharmaceuticals.
Collapse
Affiliation(s)
- Jennifer Andres
- Institute of Synthetic Biology and CEPLAS, University of Düsseldorf, 40225 Duesseldorf, Germany
| | - Tim Blomeier
- Institute of Synthetic Biology and CEPLAS, University of Düsseldorf, 40225 Duesseldorf, Germany
| | - Matias D Zurbriggen
- Institute of Synthetic Biology and CEPLAS, University of Düsseldorf, 40225 Duesseldorf, Germany
| |
Collapse
|
131
|
Shahrajabian M, Hormozi-Nezhad MR. Chemiluminometric fingerprints for identification of plasmonic nanoparticles. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2019; 209:85-94. [PMID: 30359853 DOI: 10.1016/j.saa.2018.10.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Revised: 09/06/2018] [Accepted: 10/14/2018] [Indexed: 06/08/2023]
Abstract
Development of a convenient and inexpensive method for identification and detection of nanoparticles (NPs) is of great interest. In this work, we have developed a novel and simple chemiluminescence based sensor array, with its sensing mechanism mimicking that of olfactory and gustatory systems for discriminating a set of NPs. The proposed method is based on the enhancement effect of NPs on luminol-oxidant CL intensity by their catalytic effect. Three kinds of oxidant including H2O2, AgNO3, and K3Fe(CN)6 were used as sensor elements and NPs exhibited diverse enhancing responses to different oxidant-luminol CL systems producing unique response patterns that were identified through heat map and chemometric methods, including linear discriminant analysis (LDA) and hierarchical cluster analysis (HCA). Five NPs have been well distinguished at various concentrations. In addition, this method clearly revealed a linear relationship between CL signal values and the concentrations of NPs for the quantitative detection of NPs. We believe that this type of CL sensor array can open a new way for facile discrimination and detection of different kinds of NPs.
Collapse
Affiliation(s)
- Maryam Shahrajabian
- Department of Chemistry, Sharif University of Technology, Tehran 11155-9516, Iran
| | - Mohammad Reza Hormozi-Nezhad
- Department of Chemistry, Sharif University of Technology, Tehran 11155-9516, Iran; Institute for Nanoscience and Nanotechnology, Sharif University of Technology, Tehran, Iran.
| |
Collapse
|
132
|
Ferdinandus, Arai S. The ABC Guide to Fluorescent Toolsets for the Development of Future Biomaterials. Front Bioeng Biotechnol 2019; 7:5. [PMID: 30729108 PMCID: PMC6351439 DOI: 10.3389/fbioe.2019.00005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Accepted: 01/07/2019] [Indexed: 01/06/2023] Open
Abstract
In recent decades, diversified approaches using nanoparticles or nano-structured scaffolds have been applied to drug delivery and tissue engineering. Thanks to recent interdisciplinary studies, the materials developed have been intensively evaluated at animal level. Despite these efforts, less attention has been paid to what is really going on at the subcellular level during the interaction between a nanomaterial and a cell. As the proposed concept becomes more complex, the need for investigation of the dynamics of these materials at the cellular level becomes more prominent. For a deeper understanding of cellular events, fluorescent imaging techniques have been a powerful means whereby spatiotemporal information related to cellular events can be visualized as detectable fluorescent signals. To date, several excellent review papers have summarized the use of fluorescent imaging toolsets in cellular biology. However, applying these toolsets becomes a laborious process for those who are not familiar with imaging studies to engage with owing to the skills gap between them and cell biologists. This review aims to highlight the valuable essentials of fluorescent imaging as a tool for the development of effective biomaterials by introducing some cases including photothermal and photodynamic therapies. This distilled information will be a convenient short-cut for those who are keen to fabricate next generation biomaterials.
Collapse
Affiliation(s)
- Ferdinandus
- Waseda Bioscience Research Institute in Singapore, Singapore, Singapore
| | - Satoshi Arai
- Research Institute for Science and Engineering, Waseda University, Tokyo, Japan.,PRIME-AMED, Tokyo, Japan
| |
Collapse
|
133
|
Duret G, Polali S, Anderson ED, Bell AM, Tzouanas CN, Avants BW, Robinson JT. Magnetic Entropy as a Proposed Gating Mechanism for Magnetogenetic Ion Channels. Biophys J 2019; 116:454-468. [PMID: 30665695 PMCID: PMC6369444 DOI: 10.1016/j.bpj.2019.01.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Revised: 12/22/2018] [Accepted: 01/02/2019] [Indexed: 12/25/2022] Open
Abstract
Magnetically sensitive ion channels would allow researchers to better study how specific brain cells affect behavior in freely moving animals; however, recent reports of "magnetogenetic" ion channels based on biogenic ferritin nanoparticles have been questioned because known biophysical mechanisms cannot explain experimental observations. Here, we reproduce a weak magnetically mediated calcium response in HEK cells expressing a previously published TRPV4-ferritin fusion protein. We find that this magnetic sensitivity is attenuated when we reduce the temperature sensitivity of the channel but not when we reduce the mechanical sensitivity of the channel, suggesting that the magnetic sensitivity of this channel is thermally mediated. As a potential mechanism for this thermally mediated magnetic response, we propose that changes in the magnetic entropy of the ferritin particle can generate heat via the magnetocaloric effect and consequently gate the associated temperature-sensitive ion channel. Unlike other forms of magnetic heating, the magnetocaloric mechanism can cool magnetic particles during demagnetization. To test this prediction, we constructed a magnetogenetic channel based on the cold-sensitive TRPM8 channel. Our observation of a magnetic response in cold-gated channels is consistent with the magnetocaloric hypothesis. Together, these new data and our proposed mechanism of action provide additional resources for understanding how ion channels could be activated by low-frequency magnetic fields.
Collapse
Affiliation(s)
- Guillaume Duret
- Department of Electrical and Computer Engineering, Rice University, Houston, Texas
| | - Sruthi Polali
- Department of Electrical and Computer Engineering, Rice University, Houston, Texas; Applied Physics Program, Rice University, Houston, Texas
| | - Erin D Anderson
- Department of Bioengineering, Rice University, Houston, Texas
| | - A Martin Bell
- Department of Electrical and Computer Engineering, Rice University, Houston, Texas; Applied Physics Program, Rice University, Houston, Texas
| | | | - Benjamin W Avants
- Department of Electrical and Computer Engineering, Rice University, Houston, Texas
| | - Jacob T Robinson
- Department of Electrical and Computer Engineering, Rice University, Houston, Texas; Department of Bioengineering, Rice University, Houston, Texas; Applied Physics Program, Rice University, Houston, Texas; Department of Neuroscience, Baylor College of Medicine, Houston, Texas.
| |
Collapse
|
134
|
Mansouri M, Strittmatter T, Fussenegger M. Light-Controlled Mammalian Cells and Their Therapeutic Applications in Synthetic Biology. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2019; 6:1800952. [PMID: 30643713 PMCID: PMC6325585 DOI: 10.1002/advs.201800952] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Revised: 08/21/2018] [Indexed: 05/12/2023]
Abstract
The ability to remote control the expression of therapeutic genes in mammalian cells in order to treat disease is a central goal of synthetic biology-inspired therapeutic strategies. Furthermore, optogenetics, a combination of light and genetic sciences, provides an unprecedented ability to use light for precise control of various cellular activities with high spatiotemporal resolution. Recent work to combine optogenetics and therapeutic synthetic biology has led to the engineering of light-controllable designer cells, whose behavior can be regulated precisely and noninvasively. This Review focuses mainly on non-neural optogenetic systems, which are often used in synthetic biology, and their applications in genetic programing of mammalian cells. Here, a brief overview of the optogenetic tool kit that is available to build light-sensitive mammalian cells is provided. Then, recently developed strategies for the control of designer cells with specific biological functions are summarized. Recent translational applications of optogenetically engineered cells are also highlighted, ranging from in vitro basic research to in vivo light-controlled gene therapy. Finally, current bottlenecks, possible solutions, and future prospects for optogenetics in synthetic biology are discussed.
Collapse
Affiliation(s)
- Maysam Mansouri
- Department of Biosystems Science and EngineeringETH ZurichMattenstrasse 26CH‐4058BaselSwitzerland
| | - Tobias Strittmatter
- Department of Biosystems Science and EngineeringETH ZurichMattenstrasse 26CH‐4058BaselSwitzerland
| | - Martin Fussenegger
- Department of Biosystems Science and EngineeringETH ZurichMattenstrasse 26CH‐4058BaselSwitzerland
- Faculty of ScienceUniversity of BaselMattenstrasse 26CH‐4058BaselSwitzerland
| |
Collapse
|
135
|
Miyazaki T, Chowdhury S, Yamashita T, Matsubara T, Yawo H, Yuasa H, Yamanaka A. Large Timescale Interrogation of Neuronal Function by Fiberless Optogenetics Using Lanthanide Micro-particles. Cell Rep 2019; 26:1033-1043.e5. [DOI: 10.1016/j.celrep.2019.01.001] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Revised: 10/08/2018] [Accepted: 12/27/2018] [Indexed: 12/20/2022] Open
|
136
|
Han G, Zhang B, Zhang H, Han D, Tan J, Yang B. The synthesis and characterization of glutathione-modified superparamagnetic iron oxide nanoparticles and their distribution in rat brains after injection in substantia nigra. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2018; 30:5. [PMID: 30569308 DOI: 10.1007/s10856-018-6209-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Accepted: 12/11/2018] [Indexed: 06/09/2023]
Abstract
Glutathione-modified superparamagnetic iron oxide nanoparticles (GSH-SPIONs) were prepared by conjugating glutathione (GSH) on the surface of the PEG (Polyethylene glycol)/PEI (polyethyleneimine)-SPIONs which were synthesized by thermal decomposition method. Thermogravimetric analysis showed that the mass fraction of GSH on the surface of SPIONs was 30.64 wt%. GSH-SPIONs in PBS were injected into the substantia nigra of rat brains. The subcellular distributions of the nanoparticles in the brains was examined by the transmission electron microscope (TEM). A remarkable amount of GSH-SPIONs were found in vesicles inside cell bodies and axons, and in mitochondria. TEM pictures show that GSH-SPIONs enter the neuronal cells by endocytosis and travel through axoplasmic transport. GSH-SPIONs have great potential as drug delivery agents in the brain to treat diseases or study brain function via mitochondria-targeting way or axoplasmic transport way.
Collapse
Affiliation(s)
- Guihua Han
- Key Laboratory of Nonferrous and Materials Processing Technology, Ministry of Education; Guangxi Key Laboratory of Optical and Electronic Materials and Devices; College of Materials Science and Engineering, Guilin University of Technology, Jian Gan Road 12, 541004, Guilin, China
| | - Baolin Zhang
- Key Laboratory of Nonferrous and Materials Processing Technology, Ministry of Education; Guangxi Key Laboratory of Optical and Electronic Materials and Devices; College of Materials Science and Engineering, Guilin University of Technology, Jian Gan Road 12, 541004, Guilin, China.
| | - Hao Zhang
- Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin Medical University, 109 North 2nd Huan Cheng Road, 541004, Guilin, China
| | - Dong Han
- Key Laboratory of Nonferrous and Materials Processing Technology, Ministry of Education; Guangxi Key Laboratory of Optical and Electronic Materials and Devices; College of Materials Science and Engineering, Guilin University of Technology, Jian Gan Road 12, 541004, Guilin, China
| | - Jie Tan
- Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin Medical University, 109 North 2nd Huan Cheng Road, 541004, Guilin, China.
| | - Boning Yang
- Guangxi Collaborative Innovation Center for Biomedicine and Department of Human Anatomy, Guangxi Medical University, Shuang Yong Road 22, 530000, Nanning, China.
| |
Collapse
|
137
|
Perspectives of RAS and RHEB GTPase Signaling Pathways in Regenerating Brain Neurons. Int J Mol Sci 2018; 19:ijms19124052. [PMID: 30558189 PMCID: PMC6321366 DOI: 10.3390/ijms19124052] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 12/05/2018] [Accepted: 12/13/2018] [Indexed: 12/29/2022] Open
Abstract
Cellular activation of RAS GTPases into the GTP-binding “ON” state is a key switch for regulating brain functions. Molecular protein structural elements of rat sarcoma (RAS) and RAS homolog protein enriched in brain (RHEB) GTPases involved in this switch are discussed including their subcellular membrane localization for triggering specific signaling pathways resulting in regulation of synaptic connectivity, axonal growth, differentiation, migration, cytoskeletal dynamics, neural protection, and apoptosis. A beneficial role of neuronal H-RAS activity is suggested from cellular and animal models of neurodegenerative diseases. Recent experiments on optogenetic regulation offer insights into the spatiotemporal aspects controlling RAS/mitogen activated protein kinase (MAPK) or phosphoinositide-3 kinase (PI3K) pathways. As optogenetic manipulation of cellular signaling in deep brain regions critically requires penetration of light through large distances of absorbing tissue, we discuss magnetic guidance of re-growing axons as a complementary approach. In Parkinson’s disease, dopaminergic neuronal cell bodies degenerate in the substantia nigra. Current human trials of stem cell-derived dopaminergic neurons must take into account the inability of neuronal axons navigating over a large distance from the grafted site into striatal target regions. Grafting dopaminergic precursor neurons directly into the degenerating substantia nigra is discussed as a novel concept aiming to guide axonal growth by activating GTPase signaling through protein-functionalized intracellular magnetic nanoparticles responding to external magnets.
Collapse
|
138
|
Gold nanoparticles-enhanced ion-transmission mass spectrometry for highly sensitive detection of chemical warfare agent simulants. Talanta 2018; 190:403-409. [PMID: 30172526 DOI: 10.1016/j.talanta.2018.08.021] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Revised: 07/31/2018] [Accepted: 08/05/2018] [Indexed: 01/01/2023]
Abstract
Gold nanoparticles (AuNPs)-embedded paper was coupled with ion-transmission mass spectrometry (MS) to enable the highly sensitive detection of chemical warfare agent (CWA) simulants in solutions. With the assistance of a low-temperature plasma (LTP) probe, we found that AuNPs were capable to enhance the ionization efficiencies of target analytes, with MS signal intensities surprisingly undergone an 800-fold increase under optimized conditions. The interaction between AuNPs and the radiofrequency electromagnetic field was believed to promote the desorption/ionization process, resulting in the unusual signal enhancement phenomenon. Based on this finding, we established a method for the rapid analysis of two simulants of nerve agents, dimethyl methylphosphonate (DMMP) and diisopropyl methylphosphonate (DIMP), with a dynamic range from 0.5 ng/mL to 100 ng/mL and detection limits of 0.1 ng/mL and 0.3 ng/mL, respectively. As sample pretreatments have been eliminated, the developed strategy is particularly promising for the on-site detection of CWAs considering its simple and rapid analytical workflow.
Collapse
|
139
|
Long X, Zhang SJ. Commentary: MagR Alone Is Insufficient to Confer Cellular Calcium Responses to Magnetic Stimulation. Front Neural Circuits 2018; 12:97. [PMID: 30483065 PMCID: PMC6240619 DOI: 10.3389/fncir.2018.00097] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Accepted: 10/11/2018] [Indexed: 01/02/2023] Open
Affiliation(s)
- Xiaoyang Long
- Department of Neurosurgery, Xinqiao Hospital, Army Medical University, Chongqing, China.,Department of Biomedical Engineering, School of Medicine, Tsinghua University, Beijing, China.,College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, China
| | - Sheng-Jia Zhang
- Department of Neurosurgery, Xinqiao Hospital, Army Medical University, Chongqing, China
| |
Collapse
|
140
|
Alvarsson A, Stanley SA. Remote control of glucose-sensing neurons to analyze glucose metabolism. Am J Physiol Endocrinol Metab 2018; 315:E327-E339. [PMID: 29812985 PMCID: PMC6171010 DOI: 10.1152/ajpendo.00469.2017] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Revised: 04/25/2018] [Accepted: 05/23/2018] [Indexed: 12/13/2022]
Abstract
The central nervous system relies on a continual supply of glucose, and must be able to detect glucose levels and regulate peripheral organ functions to ensure that its energy requirements are met. Specialized glucose-sensing neurons, first described half a century ago, use glucose as a signal and modulate their firing rates as glucose levels change. Glucose-excited neurons are activated by increasing glucose concentrations, while glucose-inhibited neurons increase their firing rate as glucose concentrations fall and decrease their firing rate as glucose concentrations rise. Glucose-sensing neurons are present in multiple brain regions and are highly expressed in hypothalamic regions, where they are involved in functions related to glucose homeostasis. However, the roles of glucose-sensing neurons in healthy and disease states remain poorly understood. Technologies that can rapidly and reversibly activate or inhibit defined neural populations provide invaluable tools to investigate how specific neural populations regulate metabolism and other physiological roles. Optogenetics has high temporal and spatial resolutions, requires implants for neural stimulation, and is suitable for modulating local neural populations. Chemogenetics, which requires injection of a synthetic ligand, can target both local and widespread populations. Radio- and magnetogenetics offer rapid neural activation in localized or widespread neural populations without the need for implants or injections. These tools will allow us to better understand glucose-sensing neurons and their metabolism-regulating circuits.
Collapse
Affiliation(s)
- Alexandra Alvarsson
- Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai , New York, New York
| | - Sarah A Stanley
- Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai , New York, New York
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai , New York, New York
| |
Collapse
|
141
|
Bossio C, Abdel Aziz I, Tullii G, Zucchetti E, Debellis D, Zangoli M, Di Maria F, Lanzani G, Antognazza MR. Photocatalytic Activity of Polymer Nanoparticles Modulates Intracellular Calcium Dynamics and Reactive Oxygen Species in HEK-293 Cells. Front Bioeng Biotechnol 2018; 6:114. [PMID: 30211158 PMCID: PMC6119808 DOI: 10.3389/fbioe.2018.00114] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Accepted: 07/20/2018] [Indexed: 11/13/2022] Open
Abstract
Optical modulation of living cells activity by light-absorbing exogenous materials is gaining increasing interest, due to the possibility both to achieve high spatial and temporal resolution with a minimally invasive and reversible technique and to avoid the need of viral transfection with light-sensitive proteins. In this context, conjugated polymers represent ideal candidates for photo-transduction, due to their excellent optoelectronic and biocompatibility properties. In this work, we demonstrate that organic polymer nanoparticles, based on poly(3-hexylthiophene) conjugated polymer, establish a functional interaction with an in vitro cell model (Human Embryonic Kidney cells, HEK-293). They display photocatalytic activity in aqueous environment and, once internalized within the cell cytosol, efficiently generate reactive oxygen species (ROS) upon visible light excitation, without affecting cell viability. Interestingly, light-activated ROS generation deterministically triggers modulation of intracellular calcium ion flux, successfully controlled at the single cell level. In perspective, the capability of polymer NPs to produce ROS and to modulate Ca2+ dynamics by illumination on-demand, at non-toxic levels, may open the path to the study of biological processes with a gene-less approach and unprecedented spatio-temporal resolution, as well as to the development of new biotechnology tools for cell optical modulation.
Collapse
Affiliation(s)
- Caterina Bossio
- Center for Nano Science and Technology@PoliMi, Istituto Italiano di Tecnologia, Milan, Italy
| | - Ilaria Abdel Aziz
- Center for Nano Science and Technology@PoliMi, Istituto Italiano di Tecnologia, Milan, Italy
- Dipartimento di Fisica, Politecnico di Milano, Milan, Italy
| | - Gabriele Tullii
- Center for Nano Science and Technology@PoliMi, Istituto Italiano di Tecnologia, Milan, Italy
- Dipartimento di Fisica, Politecnico di Milano, Milan, Italy
| | - Elena Zucchetti
- Center for Nano Science and Technology@PoliMi, Istituto Italiano di Tecnologia, Milan, Italy
- Dipartimento di Fisica, Politecnico di Milano, Milan, Italy
| | - Doriana Debellis
- Electron Microscopy Facility, Istituto Italiano di Tecnologia, Genova, Italy
| | - Mattia Zangoli
- Institute for Organic Synthesis and Photoreactivity, CNR-ISOF, Bologna, Italy
| | - Francesca Di Maria
- Institute for Organic Synthesis and Photoreactivity, CNR-ISOF, Bologna, Italy
| | - Guglielmo Lanzani
- Center for Nano Science and Technology@PoliMi, Istituto Italiano di Tecnologia, Milan, Italy
- Dipartimento di Fisica, Politecnico di Milano, Milan, Italy
| | - Maria Rosa Antognazza
- Center for Nano Science and Technology@PoliMi, Istituto Italiano di Tecnologia, Milan, Italy
| |
Collapse
|
142
|
He S, Zhang H, Liu Y, Sun F, Yu X, Li X, Zhang L, Wang L, Mao K, Wang G, Lin Y, Han Z, Sabirianov R, Zeng H. Maximizing Specific Loss Power for Magnetic Hyperthermia by Hard-Soft Mixed Ferrites. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2018; 14:e1800135. [PMID: 29931802 DOI: 10.1002/smll.201800135] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Revised: 04/03/2018] [Indexed: 05/22/2023]
Abstract
Maximized specific loss power and intrinsic loss power approaching theoretical limits for alternating-current (AC) magnetic-field heating of nanoparticles are reported. This is achieved by engineering the effective magnetic anisotropy barrier of nanoparticles via alloying of hard and soft ferrites. 22 nm Co0.03 Mn0.28 Fe2.7 O4 /SiO2 nanoparticles reach a specific loss power value of 3417 W g-1metal at a field of 33 kA m-1 and 380 kHz. Biocompatible Zn0.3 Fe2.7 O4 /SiO2 nanoparticles achieve specific loss power of 500 W g-1metal and intrinsic loss power of 26.8 nHm2 kg-1 at field parameters of 7 kA m-1 and 380 kHz, below the clinical safety limit. Magnetic bone cement achieves heating adequate for bone tumor hyperthermia, incorporating an ultralow dosage of just 1 wt% of nanoparticles. In cellular hyperthermia experiments, these nanoparticles demonstrate high cell death rate at low field parameters. Zn0.3 Fe2.7 O4 /SiO2 nanoparticles show cell viabilities above 97% at concentrations up to 500 µg mL-1 within 48 h, suggesting toxicity lower than that of magnetite.
Collapse
Affiliation(s)
- Shuli He
- Department of Physics, Capital Normal University, Beijing, 10048, China
- Department of Physics, University at Buffalo, SUNY, Buffalo, NY, 14260, USA
- Beijing Advanced Innovation Center for Imaging Technology, Beijing, 10048, China
| | - Hongwang Zhang
- Department of Physics, University at Buffalo, SUNY, Buffalo, NY, 14260, USA
| | - Yihao Liu
- Department of Physics, Capital Normal University, Beijing, 10048, China
- Chinese PLA General Hospital, Beijing, 10048, China
| | - Fan Sun
- Department of Physics, University at Buffalo, SUNY, Buffalo, NY, 14260, USA
| | - Xiang Yu
- Department of Physics, Capital Normal University, Beijing, 10048, China
| | - Xueyan Li
- Department of Physics, Capital Normal University, Beijing, 10048, China
| | - Li Zhang
- Department of Physics, Capital Normal University, Beijing, 10048, China
| | - Lichen Wang
- Department of Physics, Capital Normal University, Beijing, 10048, China
| | - Keya Mao
- Chinese PLA General Hospital, Beijing, 10048, China
| | - Gangshi Wang
- Chinese PLA General Hospital, Beijing, 10048, China
| | - Yunjuan Lin
- Chinese PLA General Hospital, Beijing, 10048, China
| | | | - Renat Sabirianov
- Department of Physics, University of Nebraska-Omaha, Omaha, NE, 68182, USA
| | - Hao Zeng
- Department of Physics, University at Buffalo, SUNY, Buffalo, NY, 14260, USA
| |
Collapse
|
143
|
Krishnan V, Park SA, Shin SS, Alon L, Tressler CM, Stokes W, Banerjee J, Sorrell ME, Tian Y, Fridman GY, Celnik P, Pevsner J, Guggino WB, Gilad AA, Pelled G. Wireless control of cellular function by activation of a novel protein responsive to electromagnetic fields. Sci Rep 2018; 8:8764. [PMID: 29884813 PMCID: PMC5993716 DOI: 10.1038/s41598-018-27087-9] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Accepted: 05/24/2018] [Indexed: 11/26/2022] Open
Abstract
The Kryptopterus bicirrhis (glass catfish) is known to respond to electromagnetic fields (EMF). Here we tested its avoidance behavior in response to static and alternating magnetic fields stimulation. Using expression cloning we identified an electromagnetic perceptive gene (EPG) from the K. bicirrhis encoding a protein that responds to EMF. This EPG gene was cloned and expressed in mammalian cells, neuronal cultures and in rat’s brain. Immunohistochemistry showed that the expression of EPG is confined to the mammalian cell membrane. Calcium imaging in mammalian cells and cultured neurons expressing EPG demonstrated that remote activation by EMF significantly increases intracellular calcium concentrations, indicative of cellular excitability. Moreover, wireless magnetic activation of EPG in rat motor cortex induced motor evoked responses of the contralateral forelimb in vivo. Here we report on the development of a new technology for remote, non-invasive modulation of cell function.
Collapse
Affiliation(s)
- Vijai Krishnan
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, Maryland, 21205, USA.,Russell H. Morgan Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21205, USA.,Department of Biomedical Engineering, Michigan State University, East Lansing, Michigan, 48823, USA.,The Institute of Quantitative Health Science and Engineering, Michigan State University, East Lansing, Michigan, 48823, USA
| | - Sarah A Park
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, Maryland, 21205, USA.,Russell H. Morgan Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21205, USA
| | - Samuel S Shin
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, Maryland, 21205, USA.,Russell H. Morgan Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21205, USA
| | - Lina Alon
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, Maryland, 21205, USA.,Russell H. Morgan Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21205, USA
| | - Caitlin M Tressler
- Russell H. Morgan Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21205, USA.,Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21205, USA
| | - William Stokes
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, Maryland, 21205, USA
| | - Jineta Banerjee
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, Maryland, 21205, USA.,Russell H. Morgan Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21205, USA
| | - Mary E Sorrell
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, Maryland, 21205, USA.,Russell H. Morgan Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21205, USA
| | - Yuemin Tian
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21205, USA
| | - Gene Y Fridman
- Department of Otolaryngology, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21205, USA
| | - Pablo Celnik
- Department of Physical Medicine and Rehabilitation, The Johns Hopkins University School of Medicine, Baltimore, Maryland, 21287, USA
| | - Jonathan Pevsner
- Department of Neurology, Kennedy Krieger Institute, Baltimore, Maryland, 21205, USA
| | - William B Guggino
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21205, USA
| | - Assaf A Gilad
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, Maryland, 21205, USA. .,Russell H. Morgan Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21205, USA. .,Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21205, USA. .,Department of Biomedical Engineering, Michigan State University, East Lansing, Michigan, 48823, USA. .,The Institute of Quantitative Health Science and Engineering, Michigan State University, East Lansing, Michigan, 48823, USA. .,Department of Radiology, Michigan State University, East Lansing, Michigan, 48823, USA.
| | - Galit Pelled
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, Maryland, 21205, USA. .,Russell H. Morgan Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21205, USA. .,Department of Biomedical Engineering, Michigan State University, East Lansing, Michigan, 48823, USA. .,The Institute of Quantitative Health Science and Engineering, Michigan State University, East Lansing, Michigan, 48823, USA. .,Department of Radiology, Michigan State University, East Lansing, Michigan, 48823, USA.
| |
Collapse
|
144
|
Luo Z, Weiss DE, Liu Q, Tian B. Biomimetic Approaches Toward Smart Bio-hybrid Systems. NANO RESEARCH 2018; 11:3009-3030. [PMID: 30906509 PMCID: PMC6430233 DOI: 10.1007/s12274-018-2004-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Revised: 01/21/2018] [Accepted: 01/23/2018] [Indexed: 05/30/2023]
Abstract
Bio-integrated materials and devices can blur the interfaces between living and artificial systems. Microfluidics, bioelectronics and engineered nanostructures, with close interactions with biology at the cellular or tissue levels, have already yielded a spectrum of new applications. Many new designs emerge, including those of organ-on-a-chip systems, biodegradable implants, electroceutical devices, minimally invasive neuro-prosthetic tools, and soft robotics. In this review, we highlight a few recent advances on the fabrication and application of the smart bio-hybrid systems, with a particular emphasis on the three-dimensional (3D) bio-integrated devices that mimick the 3D feature of tissue scaffolds. Moreover, neurons integrated with engineered nanostructures for wireless neuromodulation and dynamic neural output will be briefly discussed. We will also go over the progress in the construction of cell-enabled soft robotics, where a tight coupling of the synthetic and biological parts is crucial for efficient functions. Finally, we summarize the approaches for enhancing bio-integration with biomimetic micro- and nanostructures.
Collapse
Affiliation(s)
- Zhiqiang Luo
- College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei 430074, PR China
| | - Dara E. Weiss
- Department of Chemistry, The University of Chicago, Chicago, IL 60637, USA
| | - Qingyun Liu
- College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei 430074, PR China
| | - Bozhi Tian
- Department of Chemistry, The University of Chicago, Chicago, IL 60637, USA
- The James Franck Institute, The University of Chicago, Chicago, IL 60637, USA
- The Institute for Biophysical Dynamics, The University of Chicago, Chicago, IL 60637, USA
| |
Collapse
|
145
|
Jackson R, Patrick PS, Page K, Powell MJ, Lythgoe MF, Miodownik MA, Parkin IP, Carmalt CJ, Kalber TL, Bear JC. Chemically Treated 3D Printed Polymer Scaffolds for Biomineral Formation. ACS OMEGA 2018; 3:4342-4351. [PMID: 29732454 PMCID: PMC5928486 DOI: 10.1021/acsomega.8b00219] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Accepted: 03/26/2018] [Indexed: 06/08/2023]
Abstract
We present the synthesis of nylon-12 scaffolds by 3D printing and demonstrate their versatility as matrices for cell growth, differentiation, and biomineral formation. We demonstrate that the porous nature of the printed parts makes them ideal for the direct incorporation of preformed nanomaterials or material precursors, leading to nanocomposites with very different properties and environments for cell growth. Additives such as those derived from sources such as tetraethyl orthosilicate applied at a low temperature promote successful cell growth, due partly to the high surface area of the porous matrix. The incorporation of presynthesized iron oxide nanoparticles led to a material that showed rapid heating in response to an applied ac magnetic field, an excellent property for use in gene expression and, with further improvement, chemical-free sterilization. These methods also avoid changing polymer feedstocks and contaminating or even damaging commonly used selective laser sintering printers. The chemically treated 3D printed matrices presented herein have great potential for use in addressing current issues surrounding bone grafting, implants, and skeletal repair, and a wide variety of possible incorporated material combinations could impact many other areas.
Collapse
Affiliation(s)
- Richard
J. Jackson
- UCL
Healthcare Biomagnetics Laboratory, The
Royal Institution of Great Britain, 21 Albemarle Street, London W1S 4BS, U.K.
| | - P. Stephen Patrick
- Centre
for Advanced Biomedical Imaging (CABI), Department of Medicine and
Institute of Child Health, University College
London, London WC1E 6DD, U.K.
| | - Kristopher Page
- Materials
Chemistry Centre, Department of Chemistry, University College London, 20 Gordon Street, London WC1H 0AJ, U.K.
| | - Michael J. Powell
- Materials
Chemistry Centre, Department of Chemistry, University College London, 20 Gordon Street, London WC1H 0AJ, U.K.
| | - Mark F. Lythgoe
- Centre
for Advanced Biomedical Imaging (CABI), Department of Medicine and
Institute of Child Health, University College
London, London WC1E 6DD, U.K.
| | - Mark A. Miodownik
- Department
of Mechanical Engineering, University College
London, London WC1E 7JE, U.K.
| | - Ivan P. Parkin
- Materials
Chemistry Centre, Department of Chemistry, University College London, 20 Gordon Street, London WC1H 0AJ, U.K.
| | - Claire J. Carmalt
- Materials
Chemistry Centre, Department of Chemistry, University College London, 20 Gordon Street, London WC1H 0AJ, U.K.
| | - Tammy L. Kalber
- Centre
for Advanced Biomedical Imaging (CABI), Department of Medicine and
Institute of Child Health, University College
London, London WC1E 6DD, U.K.
| | - Joseph C. Bear
- School
of Life Science, Pharmacy & Chemistry, Kingston University London, Penrhyn Road, Kingston upon Thames, Surrey KT1 2EE, U.K.
| |
Collapse
|
146
|
Magnetic nanoparticles based cancer therapy: current status and applications. SCIENCE CHINA-LIFE SCIENCES 2018; 61:400-414. [DOI: 10.1007/s11427-017-9271-1] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/23/2017] [Accepted: 01/15/2018] [Indexed: 12/11/2022]
|
147
|
Soni B, Nimsarkar P, Mol M, Saha B, Singh S. Systems-synthetic biology in understanding the complexities and simple devices in immunology. Cytokine 2018; 108:60-66. [PMID: 29579544 DOI: 10.1016/j.cyto.2018.03.029] [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: 11/20/2017] [Revised: 03/16/2018] [Accepted: 03/19/2018] [Indexed: 10/17/2022]
Abstract
Systems and synthetic biology in the coming era has the ability to manipulate, stimulate and engineer cells to counteract the pathogenic immune response. The inherent biological complexities associated with the creation of a device allow capitalizing the biotechnological resources either by simply administering a recombinant cytokine or just reprogramming the immune cells. The strategy outlined, adopted and discussed may mark the beginning with promising therapeutics based on the principles of synthetic immunology.
Collapse
Affiliation(s)
- Bhavnita Soni
- National Centre for Cell Science, NCCS Complex, Ganeshkhind, SP Pune University Campus, Pune 411007, India
| | - Prajakta Nimsarkar
- National Centre for Cell Science, NCCS Complex, Ganeshkhind, SP Pune University Campus, Pune 411007, India
| | - Milsee Mol
- National Centre for Cell Science, NCCS Complex, Ganeshkhind, SP Pune University Campus, Pune 411007, India
| | - Bhaskar Saha
- National Centre for Cell Science, NCCS Complex, Ganeshkhind, SP Pune University Campus, Pune 411007, India
| | - Shailza Singh
- National Centre for Cell Science, NCCS Complex, Ganeshkhind, SP Pune University Campus, Pune 411007, India.
| |
Collapse
|
148
|
Lim S, Choi B, Lee SY, Lee S, Nahm HH, Kim YH, Kim T, Park JG, Lee J, Hong J, Kwon SG, Hyeon T. Microscopic States and the Verwey Transition of Magnetite Nanocrystals Investigated by Nuclear Magnetic Resonance. NANO LETTERS 2018; 18:1745-1750. [PMID: 29461844 DOI: 10.1021/acs.nanolett.7b04866] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
57Fe nuclear magnetic resonance (NMR) of magnetite nanocrystals ranging in size from 7 nm to 7 μm is measured. The line width of the NMR spectra changes drastically around 120 K, showing microscopic evidence of the Verwey transition. In the region above the transition temperature, the line width of the spectrum increases and the spin-spin relaxation time decreases as the nanocrystal size decreases. The line-width broadening indicates the significant deformation of magnetic structure and reduction of charge order compared to bulk crystals, even when the structural distortion is unobservable. The reduction of the spin-spin relaxation time is attributed to the suppressed polaron hopping conductivity in ferromagnetic metals, which is a consequence of the enhanced electron-phonon coupling in the quantum-confinement regime. Our results show that the magnetic distortion occurs in the entire nanocrystal and does not comply with the simple model of the core-shell binary structure with a sharp boundary.
Collapse
|
149
|
Mosabbir AA, Truong K. Genetically Encoded Circuit for Remote Regulation of Cell Migration by Magnetic Fields. ACS Synth Biol 2018; 7:718-726. [PMID: 29343055 DOI: 10.1021/acssynbio.7b00415] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Magnetoreception can be generally defined as the ability to transduce the effects of a magnetic field into a cellular response. Magnetic stimulation at the cellular level is particularly attractive due to its ability for deep penetration and minimal invasiveness, allowing remote regulation of engineered biological processes. Previously, a magnetic-responsive genetic circuit was engineered using the transient receptor potential vanilloid 1 (TRPV1) and the iron containing ferritin protein (i.e., the TF circuit). In this study, we combined the TF circuit with a Ca2+ activated RhoA protein (CaRQ) to allow a magnetic field to remotely regulate cell migration. Cells expressing the TF circuit and CaRQ exhibited consistent dynamic protrusions, leading to migration along a porous membrane, directed spreading in response to a magnetic field gradient, as well as wound healing. This work offers a compelling interface for programmable electrical devices to control the migration of living systems for potential applications in cell-based therapy.
Collapse
Affiliation(s)
- Abdullah A. Mosabbir
- Institute
of Biomaterials and Biomedical Engineering, University of Toronto, 164 College Street, Toronto, Ontario M5S 3G9, Canada
| | - Kevin Truong
- Institute
of Biomaterials and Biomedical Engineering, University of Toronto, 164 College Street, Toronto, Ontario M5S 3G9, Canada
- Edward
S. Rogers Sr. Department of Electrical and Computer Engineering, University of Toronto, 10 King’s College Circle, Toronto, Ontario M5S 3G4, Canada
| |
Collapse
|
150
|
Mechanogenetics for the remote and noninvasive control of cancer immunotherapy. Proc Natl Acad Sci U S A 2018; 115:992-997. [PMID: 29343642 DOI: 10.1073/pnas.1714900115] [Citation(s) in RCA: 162] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
While cell-based immunotherapy, especially chimeric antigen receptor (CAR)-expressing T cells, is becoming a paradigm-shifting therapeutic approach for cancer treatment, there is a lack of general methods to remotely and noninvasively regulate genetics in live mammalian cells and animals for cancer immunotherapy within confined local tissue space. To address this limitation, we have identified a mechanically sensitive Piezo1 ion channel (mechanosensor) that is activatable by ultrasound stimulation and integrated it with engineered genetic circuits (genetic transducer) in live HEK293T cells to convert the ultrasound-activated Piezo1 into transcriptional activities. We have further engineered the Jurkat T-cell line and primary T cells (peripheral blood mononuclear cells) to remotely sense the ultrasound wave and transduce it into transcriptional activation for the CAR expression to recognize and eradicate target tumor cells. This approach is modular and can be extended for remote-controlled activation of different cell types with high spatiotemporal precision for therapeutic applications.
Collapse
|