51
|
Lin Y, Yao Y, Zhang W, Fang Q, Zhang L, Zhang Y, Xu Y. Applications of upconversion nanoparticles in cellular optogenetics. Acta Biomater 2021; 135:1-12. [PMID: 34461347 DOI: 10.1016/j.actbio.2021.08.035] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 08/07/2021] [Accepted: 08/23/2021] [Indexed: 11/16/2022]
Abstract
Upconversion-mediated optogenetics is an emerging powerful technique to remotely control and manipulate the deep-tissue protein functions and signaling pathway activation. This technique uses lanthanide upconversion nanoparticles (UCNPs) as light transducers and through near-infrared light to indirectly activate the traditional optogenetic proteins. With the merits of high spatiotemporal resolution and minimal invasiveness, this technique enables cell-type specific manipulation of cellular activities in deep tissues as well as in living animals. In this review, we introduce the latest development of optogenetic modules and UCNPs, with emphasis on the integration of UCNPs with cellular optogenetics and their biomedical applications on the control of neural/brain activity, cancer therapy and cardiac optogenetics in vivo. Furthermore, we analyze the current developed strategies to optimize and advance the upconversion-mediated optogenetics and discuss the remaining challenges of its further applications in biomedical study and clinical translational research. STATEMENT OF SIGNIFICANCE: Optogenetics harnesses photoactivatable proteins to optically stimulate and control intracellular activities. UCNPs-mediated NIR-activatable optogenetics uses lanthanide upconversion nanoparticles (UCNPs) as light transducers and utilizes near-infrared (NIR) light to indirectly activate the traditional optogenetic proteins. The integration of UCNPs with cellular optogenetics has showed great promise in biomedical applications in regulating neural/brain activity, cancer therapy and cardiac optogenetics in vivo. The evolution and optimization of functional UCNPs and the discovery and engineering of novel optogenetic modules would both contribute to the advance of such unique hybrid technology, which may lead to discoveries in biomedical research and provide new treatments for human diseases.
Collapse
Affiliation(s)
- Yinyan Lin
- Department of Biomedical Engineering, Key Laboratory of Biomedical Engineering of Ministry of Education, State Key Laboratory of Modern Optical Instrumentation, Zhejiang Provincial Key Laboratory of Cardio-Cerebral Vascular Detection Technology and Medicinal Effectiveness Appraisal, Zhejiang University, Hangzhou 310027, China
| | - Yuanfa Yao
- Department of Biomedical Engineering, Key Laboratory of Biomedical Engineering of Ministry of Education, State Key Laboratory of Modern Optical Instrumentation, Zhejiang Provincial Key Laboratory of Cardio-Cerebral Vascular Detection Technology and Medicinal Effectiveness Appraisal, Zhejiang University, Hangzhou 310027, China
| | - Wanmei Zhang
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Qiuyu Fang
- Department of Biomedical Engineering, Key Laboratory of Biomedical Engineering of Ministry of Education, State Key Laboratory of Modern Optical Instrumentation, Zhejiang Provincial Key Laboratory of Cardio-Cerebral Vascular Detection Technology and Medicinal Effectiveness Appraisal, Zhejiang University, Hangzhou 310027, China
| | - Luhao Zhang
- Department of Biomedical Engineering, Key Laboratory of Biomedical Engineering of Ministry of Education, State Key Laboratory of Modern Optical Instrumentation, Zhejiang Provincial Key Laboratory of Cardio-Cerebral Vascular Detection Technology and Medicinal Effectiveness Appraisal, Zhejiang University, Hangzhou 310027, China
| | - Yan Zhang
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China.
| | - Yingke Xu
- Department of Biomedical Engineering, Key Laboratory of Biomedical Engineering of Ministry of Education, State Key Laboratory of Modern Optical Instrumentation, Zhejiang Provincial Key Laboratory of Cardio-Cerebral Vascular Detection Technology and Medicinal Effectiveness Appraisal, Zhejiang University, Hangzhou 310027, China; Department of Endocrinology, The Affiliated Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China.
| |
Collapse
|
52
|
Identification of pimavanserin tartrate as a potent Ca 2+-calcineurin-NFAT pathway inhibitor for glioblastoma therapy. Acta Pharmacol Sin 2021; 42:1860-1874. [PMID: 34363007 PMCID: PMC8563877 DOI: 10.1038/s41401-021-00724-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Accepted: 06/21/2021] [Indexed: 02/07/2023] Open
Abstract
Glioblastoma multiforme (GBM) is the most common and malignant type of primary brain tumor, and 95% of patients die within 2 years after diagnosis. In this study, aiming to overcome chemoresistance to the first-line drug temozolomide (TMZ), we carried out research to discover a novel alternative drug targeting the oncogenic NFAT signaling pathway for GBM therapy. To accelerate the drug's clinical application, we took advantage of a drug repurposing strategy to identify novel NFAT signaling pathway inhibitors. After screening a set of 93 FDA-approved drugs with simple structures, we identified pimavanserin tartrate (PIM), an effective 5-HT2A receptor inverse agonist used for the treatment of Parkinson's disease-associated psychiatric symptoms, as having the most potent inhibitory activity against the NFAT signaling pathway. Further study revealed that PIM suppressed STIM1 puncta formation to inhibit store-operated calcium entry (SOCE) and subsequent NFAT activity. In cellula, PIM significantly suppressed the proliferation, migration, division, and motility of U87 glioblastoma cells, induced G1/S phase arrest and promoted apoptosis. In vivo, the growth of subcutaneous and orthotopic glioblastoma xenografts was markedly suppressed by PIM. Unbiased omics studies revealed the novel molecular mechanism of PIM's antitumor activity, which included suppression of the ATR/CDK2/E2F axis, MYC, and AuroraA/B signaling. Interestingly, the genes upregulated by PIM were largely associated with cholesterol homeostasis, which may contribute to PIM's side effects and should be given more attention. Our study identified store-operated calcium channels as novel targets of PIM and was the first to systematically highlight the therapeutic potential of pimavanserin tartrate for glioblastoma.
Collapse
|
53
|
Drees C, Rühl P, Czerny J, Chandra G, Bajorath J, Haase M, Heinemann SH, Piehler J. Diffraction-Unlimited Photomanipulation at the Plasma Membrane via Specifically Targeted Upconversion Nanoparticles. NANO LETTERS 2021; 21:8025-8034. [PMID: 34519216 DOI: 10.1021/acs.nanolett.1c02267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Engineered UCNP are used to trigger rapid photoconversion of the fluorescent protein Dendra2 with nanoscopic precision and over longer distances in mammalian cells. By exploiting the synergy of high-level thulium doping with core-shell design and elevated excitation intensities, intense UCNP emission is achieved, allowing fast photoconversion of Dendra2 with <10 nm resolution. A tailored biocompatible surface coating and functionalization with a derivate of green fluorescent protein (GFP) for recognition of antiGFP nanobodies are developed. Highly specific targeting of UCNP to fusion proteins of antiGFP on the surface of mammalian cells is demonstrated. UCNP bound to extracellular Dendra2 enable rapid photoconversion selectively in molecular proximity and thus unambiguous detection of cytokine receptor dimerization in the plasma membrane and in endosomes. Remarkably, UCNPs are also suited for manipulating intracellular Dendra2 across the plasma membrane. This study thus establishes UCNP-controlled photomanipulation with nanoscale precision, opening exciting opportunities for bioanalytical applications in cell biology.
Collapse
Affiliation(s)
- Christoph Drees
- Department of Biology/Chemistry and Center of Cellular Nanoanalytics (CellNanOs), Osnabrück University, 49076 Osnabrück, Germany
| | - Philipp Rühl
- Center for Molecular Biomedicine, Department of Biophysics, Friedrich Schiller University Jena and Jena University Hospital, 07745 Jena, Germany
| | - Jacqueline Czerny
- Department of Biology/Chemistry and Center of Cellular Nanoanalytics (CellNanOs), Osnabrück University, 49076 Osnabrück, Germany
| | - Gemini Chandra
- Center for Molecular Biomedicine, Department of Biophysics, Friedrich Schiller University Jena and Jena University Hospital, 07745 Jena, Germany
| | - Janosch Bajorath
- Department of Biology/Chemistry and Center of Cellular Nanoanalytics (CellNanOs), Osnabrück University, 49076 Osnabrück, Germany
| | - Markus Haase
- Department of Biology/Chemistry and Center of Cellular Nanoanalytics (CellNanOs), Osnabrück University, 49076 Osnabrück, Germany
| | - Stefan H Heinemann
- Center for Molecular Biomedicine, Department of Biophysics, Friedrich Schiller University Jena and Jena University Hospital, 07745 Jena, Germany
| | - Jacob Piehler
- Department of Biology/Chemistry and Center of Cellular Nanoanalytics (CellNanOs), Osnabrück University, 49076 Osnabrück, Germany
| |
Collapse
|
54
|
Patel M, Meenu M, Pandey JK, Kumar P, Patel R. Recent development in upconversion nanoparticles and their application in optogenetics: A review. J RARE EARTH 2021. [DOI: 10.1016/j.jre.2021.10.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
55
|
Liu X, Chen H, Wang Y, Si Y, Zhang H, Li X, Zhang Z, Yan B, Jiang S, Wang F, Weng S, Xu W, Zhao D, Zhang J, Zhang F. Near-infrared manipulation of multiple neuronal populations via trichromatic upconversion. Nat Commun 2021; 12:5662. [PMID: 34580314 PMCID: PMC8476604 DOI: 10.1038/s41467-021-25993-7] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 09/08/2021] [Indexed: 11/23/2022] Open
Abstract
Using multi-color visible lights for independent optogenetic manipulation of multiple neuronal populations offers the ability for sophisticated brain functions and behavior dissection. To mitigate invasive fiber insertion, infrared light excitable upconversion nanoparticles (UCNPs) with deep tissue penetration have been implemented in optogenetics. However, due to the chromatic crosstalk induced by the multiple emission peaks, conventional UCNPs or their mixture cannot independently activate multiple targeted neuronal populations. Here, we report NIR multi-color optogenetics by the well-designed trichromatic UCNPs with excitation-specific luminescence. The blue, green and red color emissions can be separately tuned by switching excitation wavelength to match respective spectral profiles of optogenetic proteins ChR2, C1V1 and ChrimsonR, which enables selective activation of three distinct neuronal populations. Such stimulation with tunable intensity can not only activate distinct neuronal populations selectively, but also achieve transcranial selective modulation of the motion behavior of awake-mice, which opens up a possibility of multi-color upconversion optogenetics. Conventional upconversion nanoparticles (UCNPs) cannot activate multiple neuron populations independently using optogenetics. Here the authors report trichromatic UCNPs with excitation-specific luminescence to allow activation of three distinct neuronal populations in the brain of awake mice.
Collapse
Affiliation(s)
- Xuan Liu
- Department of Chemistry, Institutes of Brain Science, State Key Laboratory of Molecular Engineering of Polymers, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Fudan University, Shanghai, 200433, P. R. China
| | - Heming Chen
- Department of Chemistry, Institutes of Brain Science, State Key Laboratory of Molecular Engineering of Polymers, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Fudan University, Shanghai, 200433, P. R. China
| | - Yiting Wang
- Department of Chemistry, Institutes of Brain Science, State Key Laboratory of Molecular Engineering of Polymers, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Fudan University, Shanghai, 200433, P. R. China
| | - Yueguang Si
- Department of Chemistry, Institutes of Brain Science, State Key Laboratory of Molecular Engineering of Polymers, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Fudan University, Shanghai, 200433, P. R. China
| | - Hongxin Zhang
- Department of Chemistry, Institutes of Brain Science, State Key Laboratory of Molecular Engineering of Polymers, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Fudan University, Shanghai, 200433, P. R. China
| | - Xiaomin Li
- Department of Chemistry, Institutes of Brain Science, State Key Laboratory of Molecular Engineering of Polymers, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Fudan University, Shanghai, 200433, P. R. China.
| | - Zhengcheng Zhang
- Department of Chemistry, Institutes of Brain Science, State Key Laboratory of Molecular Engineering of Polymers, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Fudan University, Shanghai, 200433, P. R. China
| | - Biao Yan
- Department of Chemistry, Institutes of Brain Science, State Key Laboratory of Molecular Engineering of Polymers, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Fudan University, Shanghai, 200433, P. R. China
| | - Su Jiang
- Department of Hand Surgery, Huashan Hospital, Priority Among Priorities of Shanghai Municipal Clinical Medicine Center, National Clinical Research Center for Aging and Medicine, Department of Hand and Upper Extremity Surgery, Jing'an District Central Hospital of Shanghai, Shanghai, 200040, China
| | - Fei Wang
- Department of Hand Surgery, Huashan Hospital, Priority Among Priorities of Shanghai Municipal Clinical Medicine Center, National Clinical Research Center for Aging and Medicine, Department of Hand and Upper Extremity Surgery, Jing'an District Central Hospital of Shanghai, Shanghai, 200040, China
| | - Shijun Weng
- Department of Chemistry, Institutes of Brain Science, State Key Laboratory of Molecular Engineering of Polymers, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Fudan University, Shanghai, 200433, P. R. China
| | - Wendong Xu
- Department of Chemistry, Institutes of Brain Science, State Key Laboratory of Molecular Engineering of Polymers, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Fudan University, Shanghai, 200433, P. R. China.,Department of Hand Surgery, Huashan Hospital, Priority Among Priorities of Shanghai Municipal Clinical Medicine Center, National Clinical Research Center for Aging and Medicine, Department of Hand and Upper Extremity Surgery, Jing'an District Central Hospital of Shanghai, Shanghai, 200040, China
| | - Dongyuan Zhao
- Department of Chemistry, Institutes of Brain Science, State Key Laboratory of Molecular Engineering of Polymers, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Fudan University, Shanghai, 200433, P. R. China
| | - Jiayi Zhang
- Department of Chemistry, Institutes of Brain Science, State Key Laboratory of Molecular Engineering of Polymers, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Fudan University, Shanghai, 200433, P. R. China.
| | - Fan Zhang
- Department of Chemistry, Institutes of Brain Science, State Key Laboratory of Molecular Engineering of Polymers, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Fudan University, Shanghai, 200433, P. R. China.
| |
Collapse
|
56
|
Manoilov KY, Verkhusha VV, Shcherbakova DM. A guide to the optogenetic regulation of endogenous molecules. Nat Methods 2021; 18:1027-1037. [PMID: 34446923 DOI: 10.1038/s41592-021-01240-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Accepted: 07/09/2021] [Indexed: 12/26/2022]
Abstract
Genetically encoded tools for the regulation of endogenous molecules (RNA, DNA elements and protein) are needed to study and control biological processes with minimal interference caused by protein overexpression and overactivation of signaling pathways. Here we focus on light-controlled optogenetic tools (OTs) that allow spatiotemporally precise regulation of gene expression and protein function. To control endogenous molecules, OTs combine light-sensing modules from natural photoreceptors with specific protein or nucleic acid binders. We discuss OT designs and group OTs according to the principles of their regulation. We outline characteristics of OT performance, discuss considerations for their use in vivo and review available OTs and their applications in cells and in vivo. Finally, we provide a brief outlook on the development of OTs.
Collapse
Affiliation(s)
- Kyrylo Yu Manoilov
- Department of Anatomy and Structural Biology and Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Vladislav V Verkhusha
- Department of Anatomy and Structural Biology and Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY, USA. .,Medicum, Faculty of Medicine, University of Helsinki, Helsinki, Finland. .,Science Center for Genetics and Life Sciences, Sirius University of Science and Technology, Sochi, Russia.
| | - Daria M Shcherbakova
- Department of Anatomy and Structural Biology and Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY, USA.
| |
Collapse
|
57
|
Miller IC, Zamat A, Sun LK, Phuengkham H, Harris AM, Gamboa L, Yang J, Murad JP, Priceman SJ, Kwong GA. Enhanced intratumoural activity of CAR T cells engineered to produce immunomodulators under photothermal control. Nat Biomed Eng 2021; 5:1348-1359. [PMID: 34385695 DOI: 10.1038/s41551-021-00781-2] [Citation(s) in RCA: 77] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Accepted: 07/11/2021] [Indexed: 12/17/2022]
Abstract
Treating solid malignancies with chimeric antigen receptor (CAR) T cells typically results in poor responses. Immunomodulatory biologics delivered systemically can augment the cells' activity, but off-target toxicity narrows the therapeutic window. Here we show that the activity of intratumoural CAR T cells can be controlled photothermally via synthetic gene switches that trigger the expression of transgenes in response to mild temperature elevations (to 40-42 °C). In vitro, heating engineered primary human T cells for 15-30 min led to over 60-fold-higher expression of a reporter transgene without affecting the cells' proliferation, migration and cytotoxicity. In mice, CAR T cells photothermally heated via gold nanorods produced a transgene only within the tumours. In mouse models of adoptive transfer, the systemic delivery of CAR T cells followed by intratumoural production, under photothermal control, of an interleukin-15 superagonist or a bispecific T cell engager bearing an NKG2D receptor redirecting T cells against NKG2D ligands enhanced antitumour activity and mitigated antigen escape. Localized photothermal control of the activity of engineered T cells may enhance their safety and efficacy.
Collapse
Affiliation(s)
- Ian C Miller
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, Atlanta, GA, USA
| | - Ali Zamat
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, Atlanta, GA, USA
| | - Lee-Kai Sun
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, Atlanta, GA, USA
| | - Hathaichanok Phuengkham
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, Atlanta, GA, USA
| | - Adrian M Harris
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, Atlanta, GA, USA
| | - Lena Gamboa
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, Atlanta, GA, USA
| | - Jason Yang
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope, Duarte, CA, USA
| | - John P Murad
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope, Duarte, CA, USA
| | - Saul J Priceman
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope, Duarte, CA, USA.,Department of Immuno-Oncology, Beckman Research Institute of City of Hope, Duarte, CA, USA
| | - Gabriel A Kwong
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, Atlanta, GA, USA. .,Institute for Electronics and Nanotechnology, Georgia Institute of Technology, Atlanta, GA, USA. .,Parker H. Petit Institute of Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA. .,Integrated Cancer Research Center, Georgia Institute of Technology, Atlanta, GA, USA. .,Georgia Immunoengineering Consortium, Emory University and Georgia Institute of Technology, Atlanta, GA, USA. .,Winship Cancer Institute, Emory University, Atlanta, GA, USA.
| |
Collapse
|
58
|
Circularly permuted LOV2 as a modular photoswitch for optogenetic engineering. Nat Chem Biol 2021; 17:915-923. [PMID: 33958793 DOI: 10.1038/s41589-021-00792-9] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Accepted: 04/02/2021] [Indexed: 02/02/2023]
Abstract
Plant-based photosensors, such as the light-oxygen-voltage sensing domain 2 (LOV2) from oat phototropin 1, can be modularly wired into cell signaling networks to remotely control protein activity and physiological processes. However, the applicability of LOV2 is hampered by the limited choice of available caging surfaces and its preference to accommodate the effector domains downstream of the C-terminal Jα helix. Here, we engineered a set of LOV2 circular permutants (cpLOV2) with additional caging capabilities, thereby expanding the repertoire of genetically encoded photoswitches to accelerate the design of optogenetic devices. We demonstrate the use of cpLOV2-based optogenetic tools to reversibly gate ion channels, antagonize CRISPR-Cas9-mediated genome engineering, control protein subcellular localization, reprogram transcriptional outputs, elicit cell suicide and generate photoactivatable chimeric antigen receptor T cells for inducible tumor cell killing. Our approach is widely applicable for engineering other photoreceptors to meet the growing need of optogenetic tools tailored for biomedical and biotechnological applications.
Collapse
|
59
|
Tiffner A, Derler I. Isoform-Specific Properties of Orai Homologues in Activation, Downstream Signaling, Physiology and Pathophysiology. Int J Mol Sci 2021; 22:8020. [PMID: 34360783 PMCID: PMC8347056 DOI: 10.3390/ijms22158020] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 07/21/2021] [Accepted: 07/23/2021] [Indexed: 11/21/2022] Open
Abstract
Ca2+ ion channels are critical in a variety of physiological events, including cell growth, differentiation, gene transcription and apoptosis. One such essential entry pathway for calcium into the cell is the Ca2+ release-activated Ca2+ (CRAC) channel. It consists of the Ca2+ sensing protein, stromal interaction molecule 1 (STIM1) located in the endoplasmic reticulum (ER) and a Ca2+ ion channel Orai in the plasma membrane. The Orai channel family includes three homologues Orai1, Orai2 and Orai3. While Orai1 is the "classical" Ca2+ ion channel within the CRAC channel complex and plays a universal role in the human body, there is increasing evidence that Orai2 and Orai3 are important in specific physiological and pathophysiological processes. This makes them an attractive target in drug discovery, but requires a detailed understanding of the three Orai channels and, in particular, their differences. Orai channel activation is initiated via Ca2+ store depletion, which is sensed by STIM1 proteins, and induces their conformational change and oligomerization. Upon STIM1 coupling, Orai channels activate to allow Ca2+ permeation into the cell. While this activation mechanism is comparable among the isoforms, they differ by a number of functional and structural properties due to non-conserved regions in their sequences. In this review, we summarize the knowledge as well as open questions in our current understanding of the three isoforms in terms of their structure/function relationship, downstream signaling and physiology as well as pathophysiology.
Collapse
Affiliation(s)
| | - Isabella Derler
- Institute of Biophysics, JKU Life Science Center, Johannes Kepler University Linz, A-4020 Linz, Austria;
| |
Collapse
|
60
|
Kazzaz SA, Baraniak JH, Zhou Y, Gill DL. Remote light-activation of native Orai channels. Cell Res 2021; 31:727-729. [PMID: 33654210 PMCID: PMC8249468 DOI: 10.1038/s41422-021-00484-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Affiliation(s)
- Sarah A Kazzaz
- Department of Cellular and Molecular Physiology, The Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA
| | - James H Baraniak
- Department of Cellular and Molecular Physiology, The Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA
| | - Yandong Zhou
- Department of Cellular and Molecular Physiology, The Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA
| | - Donald L Gill
- Department of Cellular and Molecular Physiology, The Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA.
| |
Collapse
|
61
|
Cheng P, Tian X, Tang W, Cheng J, Bao J, Wang H, Zheng S, Wang Y, Wei X, Chen T, Feng H, Xue T, Goda K, He H. Direct control of store-operated calcium channels by ultrafast laser. Cell Res 2021; 31:758-772. [PMID: 33469157 PMCID: PMC8249419 DOI: 10.1038/s41422-020-00463-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2020] [Accepted: 12/21/2020] [Indexed: 01/30/2023] Open
Abstract
Ca2+ channels are essential to cell birth, life, and death. They can be externally activated by optogenetic tools, but this requires robust introduction of exogenous optogenetic genes for expression of photosensitive proteins in biological systems. Here we present femtoSOC, a method for direct control of Ca2+ channels solely by ultrafast laser without the need for optogenetic tools or any other exogenous reagents. Specifically, by focusing and scanning wavelength-tuned low-power femtosecond laser pulses on the plasma membrane for multiphoton excitation, we directly induced Ca2+ influx in cultured cells. Mechanistic study reveals that photoexcited flavins covalently bind cysteine residues in Orai1 via thioether bonds, which facilitates Orai1 polymerization to form store-operated calcium channels (SOCs) independently of STIM1, a protein generally participating in SOC formation, enabling all-optical activation of Ca2+ influx and downstream signaling pathways. Moreover, we used femtoSOC to demonstrate direct neural activation both in brain slices in vitro and in intact brains of living mice in vivo in a spatiotemporal-specific manner, indicating potential utility of femtoSOC.
Collapse
Affiliation(s)
- Pan Cheng
- grid.16821.3c0000 0004 0368 8293School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200030 China
| | - Xiaoying Tian
- grid.16821.3c0000 0004 0368 8293School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200030 China
| | - Wanyi Tang
- grid.16821.3c0000 0004 0368 8293School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200030 China
| | - Juan Cheng
- grid.59053.3a0000000121679639School of life science, the University of Science and Technology of China, Hefei, Anhui 230026 China ,grid.186775.a0000 0000 9490 772XDepartment of Physiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui 230032 China
| | - Jin Bao
- grid.59053.3a0000000121679639School of life science, the University of Science and Technology of China, Hefei, Anhui 230026 China
| | - Haipeng Wang
- grid.16821.3c0000 0004 0368 8293School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200030 China
| | - Sisi Zheng
- grid.20513.350000 0004 1789 9964Beijing Key Laboratory of Gene Resource and Molecular Development, College of Life Sciences, Beijing Normal University, Beijing, 100875 China
| | - Youjun Wang
- grid.20513.350000 0004 1789 9964Beijing Key Laboratory of Gene Resource and Molecular Development, College of Life Sciences, Beijing Normal University, Beijing, 100875 China
| | - Xunbin Wei
- grid.16821.3c0000 0004 0368 8293School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200030 China
| | - Tunan Chen
- grid.410570.70000 0004 1760 6682Institute of Neurosurgery, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038 China
| | - Hua Feng
- grid.410570.70000 0004 1760 6682Institute of Neurosurgery, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038 China
| | - Tian Xue
- grid.59053.3a0000000121679639School of life science, the University of Science and Technology of China, Hefei, Anhui 230026 China
| | - Keisuke Goda
- grid.26999.3d0000 0001 2151 536XDepartment of Chemistry, University of Tokyo, Tokyo, 113-0033 Japan ,grid.49470.3e0000 0001 2331 6153Institute of Technological Sciences, Wuhan University, Wuhan, Hubei 430072 China ,grid.19006.3e0000 0000 9632 6718Department of Bioengineering, University of California, Los Angeles, CA 90095 USA
| | - Hao He
- grid.16821.3c0000 0004 0368 8293School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200030 China
| |
Collapse
|
62
|
He L, Huang Z, Huang K, Chen R, Nguyen NT, Wang R, Cai X, Huang Z, Siwko S, Walker JR, Han G, Zhou Y, Jing J. Optogenetic Control of Non-Apoptotic Cell Death. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:2100424. [PMID: 34540558 PMCID: PMC8438606 DOI: 10.1002/advs.202100424] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 03/08/2021] [Indexed: 05/20/2023]
Abstract
Herein, a set of optogenetic tools (designated LiPOP) that enable photoswitchable necroptosis and pyroptosis in live cells with varying kinetics, is introduced. The LiPOP tools allow reconstruction of the key molecular steps involved in these two non-apoptotic cell death pathways by harnessing the power of light. Further, the use of LiPOPs coupled with upconversion nanoparticles or bioluminescence is demonstrated to achieve wireless optogenetic or chemo-optogenetic killing of cancer cells in multiple mouse tumor models. LiPOPs can trigger necroptotic and pyroptotic cell death in cultured prokaryotic or eukaryotic cells and in living animals, and set the stage for studying the role of non-apoptotic cell death pathways during microbial infection and anti-tumor immunity.
Collapse
Affiliation(s)
- Lian He
- Center for Translational Cancer ResearchInstitute of Biosciences and TechnologyTexas A&M UniversityHoustonTX77030USA
| | - Zixian Huang
- Department of Oral and Maxillofacial SurgerySun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouGuangdong510120China
| | - Kai Huang
- Department of Biochemistry and Molecular PharmacologyUniversity of Massachusetts Medical SchoolWorcesterMA01605USA
| | - Rui Chen
- Department of Oral and Maxillofacial SurgerySun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouGuangdong510120China
| | - Nhung T. Nguyen
- Center for Translational Cancer ResearchInstitute of Biosciences and TechnologyTexas A&M UniversityHoustonTX77030USA
| | - Rui Wang
- Center for Translational Cancer ResearchInstitute of Biosciences and TechnologyTexas A&M UniversityHoustonTX77030USA
| | - Xiaoli Cai
- Center for Epigenetics and Disease PreventionInstitute of Biosciences and TechnologyTexas A&M UniversityHoustonTX77030USA
| | - Zhiquan Huang
- Department of Oral and Maxillofacial SurgerySun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouGuangdong510120China
| | - Stefan Siwko
- Center for Translational Cancer ResearchInstitute of Biosciences and TechnologyTexas A&M UniversityHoustonTX77030USA
| | | | - Gang Han
- Department of Biochemistry and Molecular PharmacologyUniversity of Massachusetts Medical SchoolWorcesterMA01605USA
| | - Yubin Zhou
- Center for Translational Cancer ResearchInstitute of Biosciences and TechnologyDepartment of Translational Medical SciencesCollege of MedicineTexas A&M UniversityHoustonTX77030USA
| | - Ji Jing
- The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital)Institute of Basic Medicine and Cancer (IBMC)Chinese Academy of SciencesHangzhouZhejiang310022China
| |
Collapse
|
63
|
Tan P, He L, Zhou Y. Engineering Supramolecular Organizing Centers for Optogenetic Control of Innate Immune Responses. Adv Biol (Weinh) 2021; 5:e2000147. [PMID: 34028210 PMCID: PMC8144545 DOI: 10.1002/adbi.202000147] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 09/18/2020] [Indexed: 12/20/2022]
Abstract
The spatiotemporal organization of oligomeric protein complexes, such as the supramolecular organizing centers (SMOCs) made of MyDDosome and MAVSome, is essential for transcriptional activation of host inflammatory responses and immunometabolism. Light-inducible assembly of MyDDosome and MAVSome is presented herein to induce activation of nuclear factor-kB and type-I interferons. Engineering of SMOCs and the downstream transcription factor permits programmable and customized innate immune operations in a light-dependent manner. These synthetic molecular tools will likely enable optical and user-defined modulation of innate immunity at a high spatiotemporal resolution to facilitate mechanistic studies of distinct modes of innate immune activations and potential intervention of immune disorders and cancer.
Collapse
Affiliation(s)
- Peng Tan
- Institute of Biosciences and Technology, College of Medicine, Texas A&M University, Houston, TX, 77030, USA
| | - Lian He
- Institute of Biosciences and Technology, College of Medicine, Texas A&M University, Houston, TX, 77030, USA
| | - Yubin Zhou
- Institute of Biosciences and Technology, College of Medicine, Texas A&M University, Houston, TX, 77030, USA
- Department of Translational Medical Sciences, College of Medicine, Texas A&M University, Houston, TX, 77030, USA
| |
Collapse
|
64
|
Li L, He L, Wu B, Yu C, Zhao H, Zhou Y, Wang J, Zhu L. Structural Determinants for Light-Dependent Membrane Binding of a Photoswitchable Polybasic Domain. ACS Synth Biol 2021; 10:542-551. [PMID: 33689308 DOI: 10.1021/acssynbio.0c00571] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
OptoPB is an optogenetic tool engineered by fusion of the phosphoinositide (PI)-binding polybasic domain of Rit1 (Rit-PB) to a photoreactive light-oxygen-voltage (LOV) domain. OptoPB selectively and reversibly binds the plasma membrane (PM) under blue light excitation, and in the dark, it releases back to the cytoplasm. However, the molecular mechanism of optical regulation and lipid recognition is still unclear. Here using nuclear magnetic resonance (NMR) spectroscopy, liposome pulldown assay, and surface plasmon resonance (SPR), we find that OptoPB binds to membrane mimetics containing di- or triphosphorylated phosphatidylinositols, particularly phosphatidylinositol 4,5-bisphosphate (PI(4,5)P2), an acidic phospholipid predominantly located in the eukaryotic PM. In the dark, steric hindrance prevented this protein-membrane interaction, while 470 nm blue light illumination activated it. NMR titration and site-directed mutagenesis revealed that both cationic and hydrophobic Rit-PB residues are essential to the membrane interaction, indicating that OptoPB binds the membrane via a specific PI(4,5)P2-dependent mechanism.
Collapse
Affiliation(s)
- Ling Li
- High Magnetic Field Laboratory, CAS Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, China
- University of Science and Technology of China, Hefei 230026, China
| | - Lian He
- Center for Translational Cancer Research, Institute of Biosciences and Technology, Texas A&M University, Houston, Texas 77030, United States
| | - Bo Wu
- High Magnetic Field Laboratory, CAS Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, China
| | - Chuandi Yu
- High Magnetic Field Laboratory, CAS Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, China
- University of Science and Technology of China, Hefei 230026, China
| | - Hongxin Zhao
- High Magnetic Field Laboratory, CAS Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, China
| | - Yubin Zhou
- Center for Translational Cancer Research, Institute of Biosciences and Technology, Texas A&M University, Houston, Texas 77030, United States
| | - Junfeng Wang
- High Magnetic Field Laboratory, CAS Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, China
- University of Science and Technology of China, Hefei 230026, China
- Institute of Physical Science and Information Technology, Anhui University, Hefei 230031, China
| | - Lei Zhu
- High Magnetic Field Laboratory, CAS Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, China
| |
Collapse
|
65
|
Lee YT, Chen R, Zhou Y, He L. Optogenetic control of calcium influx in mammalian cells. Methods Enzymol 2021; 654:255-270. [PMID: 34120716 DOI: 10.1016/bs.mie.2021.02.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Optogenetics combines optics and genetics to enable non-invasive interrogation of cell physiology at an unprecedented high spatiotemporal resolution. Here, we introduce Opto-CRAC as a set of genetically-encoded calcium actuators (GECAs) engineered from the calcium release-activated calcium (CRAC) channel, which has been tailored for optical control of calcium entry and calcium-dependent physiological responses in non-excitable cells and tissues. We describe a detailed protocol for applying Opto-CRAC as an optogenetic tool to achieve photo-tunable control over intracellular calcium signals and calcium-dependent gene expression in mammalian cells.
Collapse
Affiliation(s)
- Yi-Tsang Lee
- Center for Translational Cancer Research, Institute of Biosciences and Technology, Texas A&M University, Houston, TX, United States
| | - Rui Chen
- Center for Translational Cancer Research, Institute of Biosciences and Technology, Texas A&M University, Houston, TX, United States
| | - Yubin Zhou
- Center for Translational Cancer Research, Institute of Biosciences and Technology, Texas A&M University, Houston, TX, United States.
| | - Lian He
- Center for Translational Cancer Research, Institute of Biosciences and Technology, Texas A&M University, Houston, TX, United States.
| |
Collapse
|
66
|
Li H, Wang X, Ohulchanskyy TY, Chen G. Lanthanide-Doped Near-Infrared Nanoparticles for Biophotonics. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2021; 33:e2000678. [PMID: 32638426 DOI: 10.1002/adma.202000678] [Citation(s) in RCA: 69] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 03/20/2020] [Accepted: 04/10/2020] [Indexed: 05/27/2023]
Abstract
Light in the near-infrared (NIR) spectral region is increasingly utilized in bioapplications, providing deeper penetration in biological tissues owing to the lower absorption and scattering in comparison with light in the visible range. Lanthanide-doped luminescent nanoparticles with excitation and/or emission in the NIR range have recently attracted tremendous attention as one of the prime candidates for noninvasive biological applications due to their unique optical properties, such as large Stokes shift, spectrally sharp luminescence emissions, long luminescence lifetimes, and excellent photostability. Herein, recent advances of lanthanide-doped nanoparticles with NIR upconversion or downshifting luminescence and their uses in cutting-edge biophotonic applications are presented. A set of efficient strategies for overcoming the fundamental limit of low luminescence brightness of lanthanide-doped nanoparticles is introduced. An in-depth literature review of their state-of-art biophotonics applications is also included, showing their superiority for high-resolution imaging, single-nanoparticle-level detection, and efficacy for tissue-penetrating diagnostics and therapeutics.
Collapse
Affiliation(s)
- Hui Li
- MIIT Key Laboratory of Critical Materials Technology for New Energy Conversion and Storage, School of Chemistry and Chemical Engineering and Key Laboratory of Micro-Systems and Micro-Structures, Ministry of Education and State Key Laboratory of Urban Water, Resource and Environment, Harbin Institute of Technology, Harbin, 150001, P. R. China
| | - Xin Wang
- MIIT Key Laboratory of Critical Materials Technology for New Energy Conversion and Storage, School of Chemistry and Chemical Engineering and Key Laboratory of Micro-Systems and Micro-Structures, Ministry of Education and State Key Laboratory of Urban Water, Resource and Environment, Harbin Institute of Technology, Harbin, 150001, P. R. China
| | - Tymish Y Ohulchanskyy
- College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen, Guangdong Province, 518060, P. R. China
| | - Guanying Chen
- MIIT Key Laboratory of Critical Materials Technology for New Energy Conversion and Storage, School of Chemistry and Chemical Engineering and Key Laboratory of Micro-Systems and Micro-Structures, Ministry of Education and State Key Laboratory of Urban Water, Resource and Environment, Harbin Institute of Technology, Harbin, 150001, P. R. China
| |
Collapse
|
67
|
Wang T, He L, Jing J, Lan T, Hong T, Wang F, Huang Y, Ma G, Zhou Y. Caffeine-Operated Synthetic Modules for Chemogenetic Control of Protein Activities by Life Style. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:2002148. [PMID: 33552855 PMCID: PMC7856909 DOI: 10.1002/advs.202002148] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 10/05/2020] [Indexed: 06/12/2023]
Abstract
A genetically encoded caffeine-operated synthetic module (COSMO) is introduced herein as a robust chemically induced dimerization (CID) system. COSMO enables chemogenetic manipulation of biological processes by caffeine and its metabolites, as well as caffeinated beverages, including coffee, tea, soda, and energy drinks. This CID tool, evolved from an anti-caffeine nanobody via cell-based high-throughput screening, permits caffeine-inducible gating of calcium channels, tumor killing via necroptosis, growth factors-independent activation of tyrosine receptor kinase signaling, and enhancement of nanobody-mediated antigen recognition for the severe acute respiratory distress coronavirus 2 (SARS-CoV-2) spike protein. Further rationalized engineering of COSMO leads to 34-217-fold enhancement in caffeine sensitivity (EC50 = 16.9 nanomolar), which makes it among the most potent CID systems like the FK506 binding protein (FKBP)-FKBP rapamycin binding domain (FRB)-rapamycin complex. Furthermore, bivalent COSMO (biCOMSO) connected with a long linker favors intramolecular dimerization and acts as a versatile precision switch when inserted in host proteins to achieve tailored function. Given the modularity and high transferability of COMSO and biCOSMO, these chemical biology tools are anticipated to greatly accelerate the development of therapeutic cells and biologics that can be switched on and off by caffeinated beverages commonly consumed in the daily life.
Collapse
Affiliation(s)
- Tianlu Wang
- Center for Translational Cancer ResearchInstitute of Biosciences and TechnologyTexas A&M UniversityHoustonTX77030USA
| | - Lian He
- Center for Translational Cancer ResearchInstitute of Biosciences and TechnologyTexas A&M UniversityHoustonTX77030USA
| | - Ji Jing
- Center for Translational Cancer ResearchInstitute of Biosciences and TechnologyTexas A&M UniversityHoustonTX77030USA
| | - Tien‐Hung Lan
- Center for Translational Cancer ResearchInstitute of Biosciences and TechnologyTexas A&M UniversityHoustonTX77030USA
| | - Tingting Hong
- Center for Epigenetics and Disease PreventionInstitute of Biosciences and TechnologyTexas A&M UniversityHoustonTX77030USA
| | - Fen Wang
- Center for Translational Cancer ResearchInstitute of Biosciences and TechnologyTexas A&M UniversityHoustonTX77030USA
| | - Yun Huang
- Center for Epigenetics and Disease PreventionInstitute of Biosciences and TechnologyTexas A&M UniversityHoustonTX77030USA
| | - Guolin Ma
- Center for Translational Cancer ResearchInstitute of Biosciences and TechnologyTexas A&M UniversityHoustonTX77030USA
| | - Yubin Zhou
- Center for Translational Cancer ResearchInstitute of Biosciences and TechnologyTexas A&M UniversityHoustonTX77030USA
- Department of Translational Medical SciencesCollege of MedicineTexas A&M UniversityHoustonTX77030USA
| |
Collapse
|
68
|
Walker JS, Hing ZA, Harrington B, Baumhardt J, Ozer HG, Lehman A, Giacopelli B, Beaver L, Williams K, Skinner JN, Cempre CB, Sun Q, Shacham S, Stromberg BR, Summers MK, Abruzzo LV, Rassenti L, Kipps TJ, Parikh S, Kay NE, Rogers KA, Woyach JA, Coppola V, Chook YM, Oakes C, Byrd JC, Lapalombella R. Recurrent XPO1 mutations alter pathogenesis of chronic lymphocytic leukemia. J Hematol Oncol 2021; 14:17. [PMID: 33451349 PMCID: PMC7809770 DOI: 10.1186/s13045-021-01032-2] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2020] [Accepted: 01/01/2021] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Exportin 1 (XPO1/CRM1) is a key mediator of nuclear export with relevance to multiple cancers, including chronic lymphocytic leukemia (CLL). Whole exome sequencing has identified hot-spot somatic XPO1 point mutations which we found to disrupt highly conserved biophysical interactions in the NES-binding groove, conferring novel cargo-binding abilities and forcing cellular mis-localization of critical regulators. However, the pathogenic role played by change-in-function XPO1 mutations in CLL is not fully understood. METHODS We performed a large, multi-center retrospective analysis of CLL cases (N = 1286) to correlate nonsynonymous mutations in XPO1 (predominantly E571K or E571G; n = 72) with genetic and epigenetic features contributing to the overall outcomes in these patients. We then established a mouse model with over-expression of wildtype (wt) or mutant (E571K or E571G) XPO1 restricted to the B cell compartment (Eµ-XPO1). Eµ-XPO1 mice were then crossed with the Eµ-TCL1 CLL mouse model. Lastly, we determined crystal structures of XPO1 (wt or E571K) bound to several selective inhibitors of nuclear export (SINE) molecules (KPT-185, KPT-330/Selinexor, and KPT-8602/Eltanexor). RESULTS We report that nonsynonymous mutations in XPO1 associate with high risk genetic and epigenetic features and accelerated CLL progression. Using the newly-generated Eµ-XPO1 mouse model, we found that constitutive B-cell over-expression of wt or mutant XPO1 could affect development of a CLL-like disease in aged mice. Furthermore, concurrent B-cell expression of XPO1 with E571K or E571G mutations and TCL1 accelerated the rate of leukemogenesis relative to that of Eµ-TCL1 mice. Lastly, crystal structures of E571 or E571K-XPO1 bound to SINEs, including Selinexor, are highly similar, suggesting that the activity of this class of compounds will not be affected by XPO1 mutations at E571 in patients with CLL. CONCLUSIONS These findings indicate that mutations in XPO1 at E571 can drive leukemogenesis by priming the pre-neoplastic lymphocytes for acquisition of additional genetic and epigenetic abnormalities that collectively result in neoplastic transformation.
Collapse
Affiliation(s)
- Janek S Walker
- Division of Hematology, Department of Internal Medicine, The Ohio State University, 460 OSUCCC, 410 West 12th Avenue, Columbus, OH, 43210, USA
| | - Zachary A Hing
- Division of Hematology, Department of Internal Medicine, The Ohio State University, 460 OSUCCC, 410 West 12th Avenue, Columbus, OH, 43210, USA
| | - Bonnie Harrington
- Division of Hematology, Department of Internal Medicine, The Ohio State University, 460 OSUCCC, 410 West 12th Avenue, Columbus, OH, 43210, USA
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH, USA
| | - Jordan Baumhardt
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Hatice Gulcin Ozer
- Department of Biomedical Informatics, The Ohio State University, Columbus, OH, USA
| | - Amy Lehman
- Center for Biostatistics, Department of Biomedical Informatics, The Ohio State University, Columbus, OH, USA
| | - Brian Giacopelli
- Division of Hematology, Department of Internal Medicine, The Ohio State University, 460 OSUCCC, 410 West 12th Avenue, Columbus, OH, 43210, USA
| | - Larry Beaver
- Division of Hematology, Department of Internal Medicine, The Ohio State University, 460 OSUCCC, 410 West 12th Avenue, Columbus, OH, 43210, USA
| | - Katie Williams
- Division of Hematology, Department of Internal Medicine, The Ohio State University, 460 OSUCCC, 410 West 12th Avenue, Columbus, OH, 43210, USA
| | - Jordan N Skinner
- Division of Hematology, Department of Internal Medicine, The Ohio State University, 460 OSUCCC, 410 West 12th Avenue, Columbus, OH, 43210, USA
| | - Casey B Cempre
- Division of Hematology, Department of Internal Medicine, The Ohio State University, 460 OSUCCC, 410 West 12th Avenue, Columbus, OH, 43210, USA
| | - Qingxiang Sun
- Department of Pathology and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | | | - Benjamin R Stromberg
- Department of Radiation Oncology, Arthur G James Comprehensive Cancer Center and Richard L. Solove Research Institute, The Ohio State University, Columbus, OH, USA
| | - Matthew K Summers
- Department of Radiation Oncology, Arthur G James Comprehensive Cancer Center and Richard L. Solove Research Institute, The Ohio State University, Columbus, OH, USA
| | - Lynne V Abruzzo
- Department of Pathology, The Ohio State University, Columbus, OH, USA
| | - Laura Rassenti
- Department of Medicine, Division of Hematology, University of California-San Diego School of Medicine, San Diego, CA, USA
| | - Thomas J Kipps
- Department of Medicine, Division of Hematology, University of California-San Diego School of Medicine, San Diego, CA, USA
| | - Sameer Parikh
- Division of Hematology, Department of Medicine, Mayo Clinic, Rochester, MN, USA
| | - Neil E Kay
- Division of Hematology, Department of Medicine, Mayo Clinic, Rochester, MN, USA
| | - Kerry A Rogers
- Division of Hematology, Department of Internal Medicine, The Ohio State University, 460 OSUCCC, 410 West 12th Avenue, Columbus, OH, 43210, USA
| | - Jennifer A Woyach
- Division of Hematology, Department of Internal Medicine, The Ohio State University, 460 OSUCCC, 410 West 12th Avenue, Columbus, OH, 43210, USA
| | - Vincenzo Coppola
- Department of Cancer Biology and Genetics, The Ohio State University College of Medicine, Columbus, OH, USA
- Genetically Engineered Mouse Modeling Core, The Ohio State University and Arthur G. James Comprehensive Cancer Center, Columbus, OH, USA
| | - Yuh Min Chook
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Christopher Oakes
- Division of Hematology, Department of Internal Medicine, The Ohio State University, 460 OSUCCC, 410 West 12th Avenue, Columbus, OH, 43210, USA
| | - John C Byrd
- Division of Hematology, Department of Internal Medicine, The Ohio State University, 460 OSUCCC, 410 West 12th Avenue, Columbus, OH, 43210, USA
- Division of Medicinal Chemistry, College of Pharmacy, The Ohio State University, Columbus, OH, USA
| | - Rosa Lapalombella
- Division of Hematology, Department of Internal Medicine, The Ohio State University, 460 OSUCCC, 410 West 12th Avenue, Columbus, OH, 43210, USA.
| |
Collapse
|
69
|
He L, Wang L, Zeng H, Tan P, Ma G, Zheng S, Li Y, Sun L, Dou F, Siwko S, Huang Y, Wang Y, Zhou Y. Engineering of a bona fide light-operated calcium channel. Nat Commun 2021; 12:164. [PMID: 33431868 PMCID: PMC7801460 DOI: 10.1038/s41467-020-20425-4] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 11/25/2020] [Indexed: 12/17/2022] Open
Abstract
The current optogenetic toolkit lacks a robust single-component Ca2+-selective ion channel tailored for remote control of Ca2+ signaling in mammals. Existing tools are either derived from engineered channelrhodopsin variants without strict Ca2+ selectivity or based on the stromal interaction molecule 1 (STIM1) that might crosstalk with other targets. Here, we describe the design of a light-operated Ca2+ channel (designated LOCa) by inserting a plant-derived photosensory module into the intracellular loop of an engineered ORAI1 channel. LOCa displays biophysical features reminiscent of the ORAI1 channel, which enables precise optical control over Ca2+ signals and hallmark Ca2+-dependent physiological responses. Furthermore, we demonstrate the use of LOCa to modulate aberrant hematopoietic stem cell self-renewal, transcriptional programming, cell suicide, as well as neurodegeneration in a Drosophila model of amyloidosis. Existing optogenetic methods to induce calcium mobilisation lack selectivity and specificity. Here, the authors design and engineer a single-component light-operated calcium channel to provide optical control over calcium signals and calcium-dependent physiological responses: LOCa.
Collapse
Affiliation(s)
- Lian He
- Center for Translational Cancer Research, Institute of Biosciences and Technology, Texas A&M University, Houston, TX, USA
| | - Liuqing Wang
- Beijing Key Laboratory of Gene Resource and Molecular Development, College of Life Sciences, Beijing Normal University, Beijing, China
| | - Hongxiang Zeng
- Center for Epigenetics and Disease Prevention, Institute of Biosciences and Technology, Texas A&M University, Houston, TX, USA
| | - Peng Tan
- Center for Translational Cancer Research, Institute of Biosciences and Technology, Texas A&M University, Houston, TX, USA
| | - Guolin Ma
- Center for Translational Cancer Research, Institute of Biosciences and Technology, Texas A&M University, Houston, TX, USA
| | - Sisi Zheng
- Beijing Key Laboratory of Gene Resource and Molecular Development, College of Life Sciences, Beijing Normal University, Beijing, China
| | - Yaxin Li
- Beijing Key Laboratory of Gene Resource and Molecular Development, College of Life Sciences, Beijing Normal University, Beijing, China
| | - Lin Sun
- Beijing Key Laboratory of Gene Resource and Molecular Development, College of Life Sciences, Beijing Normal University, Beijing, China
| | - Fei Dou
- Beijing Key Laboratory of Genetic Engineering Drugs and Biotechnology, College of Life Sciences, Beijing Normal University, Beijing, China
| | - Stefan Siwko
- Center for Translational Cancer Research, Institute of Biosciences and Technology, Texas A&M University, Houston, TX, USA
| | - Yun Huang
- Center for Epigenetics and Disease Prevention, Institute of Biosciences and Technology, Texas A&M University, Houston, TX, USA. .,Department of Translational Medical Sciences, College of Medicine, Texas A&M University, Houston, TX, USA.
| | - Youjun Wang
- Beijing Key Laboratory of Gene Resource and Molecular Development, College of Life Sciences, Beijing Normal University, Beijing, China.
| | - Yubin Zhou
- Center for Translational Cancer Research, Institute of Biosciences and Technology, Texas A&M University, Houston, TX, USA. .,Department of Translational Medical Sciences, College of Medicine, Texas A&M University, Houston, TX, USA.
| |
Collapse
|
70
|
The Orai Pore Opening Mechanism. Int J Mol Sci 2021; 22:ijms22020533. [PMID: 33430308 PMCID: PMC7825772 DOI: 10.3390/ijms22020533] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Revised: 12/30/2020] [Accepted: 01/02/2021] [Indexed: 02/07/2023] Open
Abstract
Cell survival and normal cell function require a highly coordinated and precise regulation of basal cytosolic Ca2+ concentrations. The primary source of Ca2+ entry into the cell is mediated by the Ca2+ release-activated Ca2+ (CRAC) channel. Its action is stimulated in response to internal Ca2+ store depletion. The fundamental constituents of CRAC channels are the Ca2+ sensor, stromal interaction molecule 1 (STIM1) anchored in the endoplasmic reticulum, and a highly Ca2+-selective pore-forming subunit Orai1 in the plasma membrane. The precise nature of the Orai1 pore opening is currently a topic of intensive research. This review describes how Orai1 gating checkpoints in the middle and cytosolic extended transmembrane regions act together in a concerted manner to ensure an opening-permissive Orai1 channel conformation. In this context, we highlight the effects of the currently known multitude of Orai1 mutations, which led to the identification of a series of gating checkpoints and the determination of their role in diverse steps of the Orai1 activation cascade. The synergistic action of these gating checkpoints maintains an intact pore geometry, settles STIM1 coupling, and governs pore opening. We describe the current knowledge on Orai1 channel gating mechanisms and summarize still open questions of the STIM1-Orai1 machinery.
Collapse
|
71
|
Bacsa B, Graziani A, Krivic D, Wiedner P, Malli R, Rauter T, Tiapko O, Groschner K. Pharmaco-Optogenetic Targeting of TRPC Activity Allows for Precise Control Over Mast Cell NFAT Signaling. Front Immunol 2021; 11:613194. [PMID: 33391284 PMCID: PMC7775509 DOI: 10.3389/fimmu.2020.613194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Accepted: 11/17/2020] [Indexed: 11/13/2022] Open
Abstract
Canonical transient receptor potential (TRPC) channels are considered as elements of the immune cell Ca2+ handling machinery. We therefore hypothesized that TRPC photopharmacology may enable uniquely specific modulation of immune responses. Utilizing a recently established TRPC3/6/7 selective, photochromic benzimidazole agonist OptoBI-1, we set out to test this concept for mast cell NFAT signaling. RBL-2H3 mast cells were found to express TRPC3 and TRPC7 mRNA but lacked appreciable Ca2+/NFAT signaling in response to OptoBI-1 photocycling. Genetic modification of the cells by introduction of single recombinant TRPC isoforms revealed that exclusively TRPC6 expression generated OptoBI-1 sensitivity suitable for opto-chemical control of NFAT1 activity. Expression of any of three benzimidazole-sensitive TRPC isoforms (TRPC3/6/7) reconstituted plasma membrane TRPC conductances in RBL cells, and expression of TRPC6 or TRPC7 enabled light-mediated generation of temporally defined Ca2+ signaling patterns. Nonetheless, only cells overexpressing TRPC6 retained essentially low basal levels of NFAT activity and displayed rapid and efficient NFAT nuclear translocation upon OptoBI-1 photocycling. Hence, genetic modification of the mast cells' TRPC expression pattern by the introduction of TRPC6 enables highly specific opto-chemical control over Ca2+ transcription coupling in these immune cells.
Collapse
Affiliation(s)
- Bernadett Bacsa
- Gottfried-Schatz-Research-Center-Biophysics, Medical University of Graz, Graz, Austria
| | - Annarita Graziani
- Gottfried-Schatz-Research-Center-Biophysics, Medical University of Graz, Graz, Austria
| | - Denis Krivic
- Gottfried-Schatz-Research-Center-Biophysics, Medical University of Graz, Graz, Austria
| | - Patrick Wiedner
- Gottfried-Schatz-Research-Center-Biophysics, Medical University of Graz, Graz, Austria
| | - Roland Malli
- Gottfried-Schatz-Research-Center-Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria
| | - Thomas Rauter
- Gottfried-Schatz-Research-Center-Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria
| | - Oleksandra Tiapko
- Gottfried-Schatz-Research-Center-Biophysics, Medical University of Graz, Graz, Austria
| | - Klaus Groschner
- Gottfried-Schatz-Research-Center-Biophysics, Medical University of Graz, Graz, Austria
| |
Collapse
|
72
|
Abstract
Phototherapy, including photodynamic therapy and photothermal therapy, exploits light to activate photo-reactions that kill cancer cells. Recent studies show that phototherapy can not only kill irradiated tumor cells, but also elicit a tumor specific immune response. This phenomenon breaks the limitations of conventional phototherapy, and has reinvigorated phototherapy-related research in the era of cancer immunotherapy. Nanoparticles play essential roles in this new campaign for allowing simultaneous delivery of photo-reactive agents and immune modulators. Some nanoparticles are potent adjuvants on their own and can augment anticancer immunity to fight off tumor relapse and metastasis. In this review, we summarize recent advances on exploiting nanoparticle-based photodynamic therapy and photothermal therapy for cancer immunotherapy, with an emphasis on nanoplatform design and functions.
Collapse
|
73
|
Qian Y, Cosio DMO, Piatkevich KD, Aufmkolk S, Su WC, Celiker OT, Schohl A, Murdock MH, Aggarwal A, Chang YF, Wiseman PW, Ruthazer ES, Boyden ES, Campbell RE. Improved genetically encoded near-infrared fluorescent calcium ion indicators for in vivo imaging. PLoS Biol 2020; 18:e3000965. [PMID: 33232322 PMCID: PMC7723245 DOI: 10.1371/journal.pbio.3000965] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 12/08/2020] [Accepted: 10/29/2020] [Indexed: 12/15/2022] Open
Abstract
Near-infrared (NIR) genetically encoded calcium ion (Ca2+) indicators (GECIs) can provide advantages over visible wavelength fluorescent GECIs in terms of reduced phototoxicity, minimal spectral cross talk with visible light excitable optogenetic tools and fluorescent probes, and decreased scattering and absorption in mammalian tissues. Our previously reported NIR GECI, NIR-GECO1, has these advantages but also has several disadvantages including lower brightness and limited fluorescence response compared to state-of-the-art visible wavelength GECIs, when used for imaging of neuronal activity. Here, we report 2 improved NIR GECI variants, designated NIR-GECO2 and NIR-GECO2G, derived from NIR-GECO1. We characterized the performance of the new NIR GECIs in cultured cells, acute mouse brain slices, and Caenorhabditis elegans and Xenopus laevis in vivo. Our results demonstrate that NIR-GECO2 and NIR-GECO2G provide substantial improvements over NIR-GECO1 for imaging of neuronal Ca2+ dynamics. This study describes improved genetically encoded near-infrared fluorescent calcium ion indicators, demonstrating that they enable robust detection of neuronal activity in cultured cells, rodent brain slices, Caenorhabditis elegans, and Xenopus laevis.
Collapse
Affiliation(s)
- Yong Qian
- Department of Chemistry, University of Alberta, Edmonton, Alberta, Canada
- Departments of Biological Engineering, Media Arts and Sciences, Brain and Cognitive Sciences, McGovern Institute, Koch Institute, Center for Neurobiological Engineering, MIT, and Howard Hughes Medical Institute, Cambridge, Massachusetts, United States of America
| | - Danielle M. Orozco Cosio
- Departments of Biological Engineering, Media Arts and Sciences, Brain and Cognitive Sciences, McGovern Institute, Koch Institute, Center for Neurobiological Engineering, MIT, and Howard Hughes Medical Institute, Cambridge, Massachusetts, United States of America
| | - Kiryl D. Piatkevich
- Departments of Biological Engineering, Media Arts and Sciences, Brain and Cognitive Sciences, McGovern Institute, Koch Institute, Center for Neurobiological Engineering, MIT, and Howard Hughes Medical Institute, Cambridge, Massachusetts, United States of America
- School of Life Sciences, Westlake University, Hangzhou, China
| | - Sarah Aufmkolk
- Montreal Neurological Institute-Hospital, Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada
- Department of Chemistry, McGill University, Montreal, Quebec, Canada
- Department of Genetics, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Wan-Chi Su
- LumiSTAR Biotechnology, Nangang District, Taipei City, Taiwan
| | - Orhan T. Celiker
- Departments of Biological Engineering, Media Arts and Sciences, Brain and Cognitive Sciences, McGovern Institute, Koch Institute, Center for Neurobiological Engineering, MIT, and Howard Hughes Medical Institute, Cambridge, Massachusetts, United States of America
| | - Anne Schohl
- Montreal Neurological Institute-Hospital, Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada
| | - Mitchell H. Murdock
- Departments of Biological Engineering, Media Arts and Sciences, Brain and Cognitive Sciences, McGovern Institute, Koch Institute, Center for Neurobiological Engineering, MIT, and Howard Hughes Medical Institute, Cambridge, Massachusetts, United States of America
| | - Abhi Aggarwal
- Howard Hughes Medical Institute, Janelia Research Campus, Ashburn, Virginia, United States of America
| | - Yu-Fen Chang
- LumiSTAR Biotechnology, Nangang District, Taipei City, Taiwan
| | - Paul W. Wiseman
- Department of Chemistry, McGill University, Montreal, Quebec, Canada
- Department of Physics, McGill University, Montreal, Quebec, Canada
| | - Edward S. Ruthazer
- Montreal Neurological Institute-Hospital, Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada
| | - Edward S. Boyden
- Departments of Biological Engineering, Media Arts and Sciences, Brain and Cognitive Sciences, McGovern Institute, Koch Institute, Center for Neurobiological Engineering, MIT, and Howard Hughes Medical Institute, Cambridge, Massachusetts, United States of America
| | - Robert E. Campbell
- Department of Chemistry, University of Alberta, Edmonton, Alberta, Canada
- Department of Chemistry, The University of Tokyo, Tokyo, Japan
- * E-mail:
| |
Collapse
|
74
|
Mathony J, Niopek D. Enlightening Allostery: Designing Switchable Proteins by Photoreceptor Fusion. Adv Biol (Weinh) 2020; 5:e2000181. [PMID: 33107225 DOI: 10.1002/adbi.202000181] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 10/01/2020] [Indexed: 11/05/2022]
Abstract
Optogenetics harnesses natural photoreceptors to non-invasively control selected processes in cells with previously unmet spatiotemporal precision. Linking the activity of a protein of choice to the conformational state of a photosensor domain through allosteric coupling represents a powerful method for engineering light-responsive proteins. It enables the design of compact and highly potent single-component optogenetic systems with fast on- and off-switching kinetics. However, designing protein-photoreceptor chimeras, in which structural changes of the photoreceptor are effectively propagated to the fused effector protein, is a challenging engineering problem and often relies on trial and error. Here, recent advances in the design and application of optogenetic allosteric switches are reviewed. First, an overview of existing optogenetic tools based on inducible allostery is provided and their utility for cell biology applications is highlighted. Focusing on light-oxygen-voltage domains, a widely applied class of small blue light sensors, the available strategies for engineering light-dependent allostery are presented and their individual advantages and limitations are highlighted. Finally, high-throughput screening technologies based on comprehensive insertion libraries, which could accelerate the creation of stimulus-responsive receptor-protein chimeras for use in optogenetics and beyond, are discussed.
Collapse
Affiliation(s)
- Jan Mathony
- Department of Biology and Centre for Synthetic Biology, Technische Universität Darmstadt, Schnittspahnstrasse 12, Darmstadt, 64287, Germany.,BZH graduate school, Heidelberg University, Im Neuheimer Feld 328, Heidelberg, 69120, Germany
| | - Dominik Niopek
- Department of Biology and Centre for Synthetic Biology, Technische Universität Darmstadt, Schnittspahnstrasse 12, Darmstadt, 64287, Germany
| |
Collapse
|
75
|
Hongdusit A, Liechty ET, Fox JM. Optogenetic interrogation and control of cell signaling. Curr Opin Biotechnol 2020; 66:195-206. [PMID: 33053496 DOI: 10.1016/j.copbio.2020.07.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 07/11/2020] [Accepted: 07/13/2020] [Indexed: 02/05/2023]
Abstract
Signaling networks control the flow of information through biological systems and coordinate the chemical processes that constitute cellular life. Optogenetic actuators - genetically encoded proteins that undergo light-induced changes in activity or conformation - are useful tools for probing signaling networks over time and space. They have permitted detailed dissections of cellular proliferation, differentiation, motility, and death, and enabled the assembly of synthetic systems with applications in areas as diverse as photography, chemical synthesis, and medicine. In this review, we provide a brief introduction to optogenetic systems and describe their application to molecular-level analyses of cell signaling. Our discussion highlights important research achievements and speculates on future opportunities to exploit optogenetic systems in the study and assembly of complex biochemical networks.
Collapse
Affiliation(s)
- Akarawin Hongdusit
- Department of Chemical and Biological Engineering, University of Colorado, Boulder, CO, 80303, USA
| | - Evan T Liechty
- Department of Chemical and Biological Engineering, University of Colorado, Boulder, CO, 80303, USA
| | - Jerome M Fox
- Department of Chemical and Biological Engineering, University of Colorado, Boulder, CO, 80303, USA.
| |
Collapse
|
76
|
Wang Q, Fan H, Li F, Skeeters SS, Krishnamurthy VV, Song Y, Zhang K. Optical control of ERK and AKT signaling promotes axon regeneration and functional recovery of PNS and CNS in Drosophila. eLife 2020; 9:57395. [PMID: 33021199 PMCID: PMC7567606 DOI: 10.7554/elife.57395] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 09/15/2020] [Indexed: 12/17/2022] Open
Abstract
Neuroregeneration is a dynamic process synergizing the functional outcomes of multiple signaling circuits. Channelrhodopsin-based optogenetics shows the feasibility of stimulating neural repair but does not pin down specific signaling cascades. Here, we utilized optogenetic systems, optoRaf and optoAKT, to delineate the contribution of the ERK and AKT signaling pathways to neuroregeneration in live Drosophila larvae. We showed that optoRaf or optoAKT activation not only enhanced axon regeneration in both regeneration-competent and -incompetent sensory neurons in the peripheral nervous system but also allowed temporal tuning and proper guidance of axon regrowth. Furthermore, optoRaf and optoAKT differ in their signaling kinetics during regeneration, showing a gated versus graded response, respectively. Importantly in the central nervous system, their activation promotes axon regrowth and functional recovery of the thermonociceptive behavior. We conclude that non-neuronal optogenetics targets damaged neurons and signaling subcircuits, providing a novel strategy in the intervention of neural damage with improved precision. Most cells have a built-in regeneration signaling program that allows them to divide and repair. But, in the cells of the central nervous system, which are called neurons, this program is ineffective. This is why accidents and illnesses affecting the brain and spinal cord can cause permanent damage. Reactivating regeneration in neurons could help them repair, but it is not easy. Certain small molecules can switch repair signaling programs back on. Unfortunately, these molecules diffuse easily through tissues, spreading around the body and making it hard to target individual damaged cells. This both hampers research into neuronal repair and makes treatments directed at healing damage to the nervous system more likely to have side-effects. It is unclear whether reactivating regeneration signaling in individual neurons is possible. One way to address this question is to use optogenetics. This technique uses genetic engineering to fuse proteins that are light-sensitive to proteins responsible for relaying signals in the cell. When specific wavelengths of light hit the light-sensitive proteins, the fused signaling proteins switch on, leading to the activation of any proteins they control, for example, those involved in regeneration. Wang et al. used optogenetic tools to determine if light can help repair neurons in fruit fly larvae. First, a strong laser light was used to damage an individual neuron in a fruit fly larva that had been genetically modified so that blue light would activate the regeneration program in its neurons. Then, Wang et al. illuminated the cell with dim blue light, switching on the regeneration program. Not only did this allow the neuron to repair itself, it also allowed the light to guide its regeneration. By focusing the blue light on the damaged end of the neuron, it was possible to guide the direction of the cell's growth as it regenerated. Regeneration programs in flies and mammals involve similar signaling proteins, but blue light does not penetrate well into mammalian tissues. This means that further research into LEDs that can be implanted may be necessary before neuronal repair experiments can be performed in mammals. In any case, the ability to focus treatment on individual neurons paves the way for future work into the regeneration of the nervous system, and the combination of light and genetics could reveal more about how repair signals work.
Collapse
Affiliation(s)
- Qin Wang
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, The Children's Hospital of Philadelphia, Philadelphia, United States.,Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, United States
| | - Huaxun Fan
- Department of Biochemistry, Urbana, United States
| | - Feng Li
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, The Children's Hospital of Philadelphia, Philadelphia, United States.,Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, United States
| | | | | | - Yuanquan Song
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, The Children's Hospital of Philadelphia, Philadelphia, United States.,Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, United States
| | - Kai Zhang
- Department of Biochemistry, Urbana, United States.,Neuroscience Program, Urbana, United States.,Center for Biophysics and Quantitative Biology, Urbana, United States.,Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, United States
| |
Collapse
|
77
|
Abstract
Store-operated calcium entry (SOCE) through Orai ion channels is an intracellular signaling pathway that is initiated by ligand-induced depletion of calcium from the endoplasmic reticulum (ER) store. The molecular link between SOCE and ER store depletion is thereby provided by a distinct class of single pass ER transmembrane proteins known as stromal interaction molecules (STIM). STIM proteins are equipped with a precise N-terminal calcium sensing domain that enables them to react to changes of the ER luminal calcium concentration. Additionally, a C-terminal coiled-coil domain permits relaying of signals to Orai ion channels via direct physical interaction. In this review, we provide a brief introduction to STIM proteins with a focus on structure and function and give an overview of recent developments in the field of STIM research.
Collapse
Affiliation(s)
- Marc Fahrner
- Institute of Biophysics, Johannes Kepler University Linz, Gruberstrasse 40, 4020 Linz, Austria
| | - Herwig Grabmayr
- Institute of Biophysics, Johannes Kepler University Linz, Gruberstrasse 40, 4020 Linz, Austria
| | - Christoph Romanin
- Institute of Biophysics, Johannes Kepler University Linz, Gruberstrasse 40, 4020 Linz, Austria
| |
Collapse
|
78
|
Hu M, Han Q, Lyu L, Tong Y, Dong S, Loh ZH, Xing B. Luminescent molecules towards precise cellular event regulation. Chem Commun (Camb) 2020; 56:10231-10234. [PMID: 32749396 DOI: 10.1039/d0cc01923b] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
A unique lanthanide complex which responds to near-infrared (NIR) stimulation was developed for remote regulation of cellular events. This molecule can be localized specifically on the cell surface. Upon NIR stimulation, strong emission of the complex can successfully modulate the activities of light-gated membrane channels and regulate the ion flux in vivo.
Collapse
Affiliation(s)
- Ming Hu
- Division of Chemistry and Biological Chemistry, School of Physical & Mathematical Sciences, Nanyang Technological University, 21 Nanyang link, 637371, Singapore.
| | | | | | | | | | | | | |
Collapse
|
79
|
Zhang Y, Wiesholler LM, Rabie H, Jiang P, Lai J, Hirsch T, Lee KB. Remote Control of Neural Stem Cell Fate Using NIR-Responsive Photoswitching Upconversion Nanoparticle Constructs. ACS APPLIED MATERIALS & INTERFACES 2020; 12:40031-40041. [PMID: 32805826 DOI: 10.1021/acsami.0c10145] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Light-mediated remote control of stem cell fate, such as proliferation, differentiation, and migration, can bring a significant impact on stem cell biology and regenerative medicine. Current UV/vis-mediated control approaches are limited in terms of nonspecific absorption, poor tissue penetration, and phototoxicity. Upconversion nanoparticle (UCNP)-based near-infrared (NIR)-mediated control systems have gained increasing attention for vast applications with minimal nonspecific absorption, good penetration depth, and minimal phototoxicity from NIR excitations. Specifically, 808 nm NIR-responsive upconversion nanomaterials have shown clear advantages for biomedical applications owing to diminished heating effects and better tissue penetration. Herein, a novel 808 nm NIR-mediated control method for stem cell differentiation has been developed using multishell UCNPs, which are optimized for upconverting 808 nm NIR light to UV emission. The locally generated UV emissions further toggle photoswitching polymer capping ligands to achieve spatiotemporally controlled small-molecule release. More specifically, with 808 nm NIR excitation, stem cell differentiation factors can be released to guide neural stem cell (NSC) differentiation in a highly controlled manner. Given the challenges in stem cell behavior control, the developed 808 nm NIR-responsive UCNP-based approach to control stem cell differentiation can represent a new tool for studying single-molecule roles in stem cell and developmental biology.
Collapse
Affiliation(s)
- Yixiao Zhang
- Department of Chemistry and Chemical Biology, Rutgers, The State University of New Jersey, Piscataway, New Jersey 08854, United States
| | - Lisa M Wiesholler
- Institute of Analytical Chemistry, Chemo- and Biosensors, University of Regensburg, 93040 Regensburg, Germany
| | - Hudifah Rabie
- Department of Chemistry and Chemical Biology, Rutgers, The State University of New Jersey, Piscataway, New Jersey 08854, United States
| | - Pengfei Jiang
- Department of Chemistry and Chemical Biology, Rutgers, The State University of New Jersey, Piscataway, New Jersey 08854, United States
| | - Jinping Lai
- Department of Chemistry and Chemical Biology, Rutgers, The State University of New Jersey, Piscataway, New Jersey 08854, United States
| | - Thomas Hirsch
- Institute of Analytical Chemistry, Chemo- and Biosensors, University of Regensburg, 93040 Regensburg, Germany
| | - Ki-Bum Lee
- Department of Chemistry and Chemical Biology, Rutgers, The State University of New Jersey, Piscataway, New Jersey 08854, United States
| |
Collapse
|
80
|
Tristán-Manzano M, Justicia-Lirio P, Maldonado-Pérez N, Cortijo-Gutiérrez M, Benabdellah K, Martin F. Externally-Controlled Systems for Immunotherapy: From Bench to Bedside. Front Immunol 2020; 11:2044. [PMID: 33013864 PMCID: PMC7498544 DOI: 10.3389/fimmu.2020.02044] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Accepted: 07/28/2020] [Indexed: 12/27/2022] Open
Abstract
Immunotherapy is a very promising therapeutic approach against cancer that is particularly effective when combined with gene therapy. Immuno-gene therapy approaches have led to the approval of four advanced therapy medicinal products (ATMPs) for the treatment of p53-deficient tumors (Gendicine and Imlygic), refractory acute lymphoblastic leukemia (Kymriah) and large B-cell lymphomas (Yescarta). In spite of these remarkable successes, immunotherapy is still associated with severe side effects for CD19+ malignancies and is inefficient for solid tumors. Controlling transgene expression through an externally administered inductor is envisioned as a potent strategy to improve safety and efficacy of immunotherapy. The aim is to develop smart immunogene therapy-based-ATMPs, which can be controlled by the addition of innocuous drugs or agents, allowing the clinicians to manage the intensity and durability of the therapy. In the present manuscript, we will review the different inducible, versatile and externally controlled gene delivery systems that have been developed and their applications to the field of immunotherapy. We will highlight the advantages and disadvantages of each system and their potential applications in clinics.
Collapse
Affiliation(s)
- María Tristán-Manzano
- Gene and Cell Therapy Unit, Genomic Medicine Department, Pfizer-University of Granada-Junta de Andalucía Centre for Genomics and Oncological Research (GENYO), Granada, Spain
| | - Pedro Justicia-Lirio
- Gene and Cell Therapy Unit, Genomic Medicine Department, Pfizer-University of Granada-Junta de Andalucía Centre for Genomics and Oncological Research (GENYO), Granada, Spain.,LentiStem Biotech, Pfizer-University of Granada-Junta de Andalucía Centre for Genomics and Oncological Research (GENYO), Granada, Spain
| | - Noelia Maldonado-Pérez
- Gene and Cell Therapy Unit, Genomic Medicine Department, Pfizer-University of Granada-Junta de Andalucía Centre for Genomics and Oncological Research (GENYO), Granada, Spain
| | - Marina Cortijo-Gutiérrez
- Gene and Cell Therapy Unit, Genomic Medicine Department, Pfizer-University of Granada-Junta de Andalucía Centre for Genomics and Oncological Research (GENYO), Granada, Spain
| | - Karim Benabdellah
- Gene and Cell Therapy Unit, Genomic Medicine Department, Pfizer-University of Granada-Junta de Andalucía Centre for Genomics and Oncological Research (GENYO), Granada, Spain
| | - Francisco Martin
- Gene and Cell Therapy Unit, Genomic Medicine Department, Pfizer-University of Granada-Junta de Andalucía Centre for Genomics and Oncological Research (GENYO), Granada, Spain
| |
Collapse
|
81
|
Meško M, Lebar T, Dekleva P, Jerala R, Benčina M. Engineering and Rewiring of a Calcium-Dependent Signaling Pathway. ACS Synth Biol 2020; 9:2055-2065. [PMID: 32643923 PMCID: PMC7467823 DOI: 10.1021/acssynbio.0c00133] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
![]()
An important feature of synthetic
biological circuits is their
response to physicochemical signals, which enables the external control
of cellular processes. Calcium-dependent regulation is an attractive
approach for achieving such control, as diverse stimuli induce calcium
influx by activating membrane channel receptors. Most calcium-dependent
gene circuits use the endogenous nuclear factor of activated T-cells
(NFAT) signaling pathway. Here, we employed engineered NFAT transcription
factors to induce the potent and robust activation of exogenous gene
expression in HEK293T cells. Furthermore, we designed a calcium-dependent
transcription factor that does not interfere with NFAT-regulated promoters
and potently activates transcription in several mammalian cell types.
Additionally, we demonstrate that coupling the circuit to a calcium-selective
ion channel resulted in capsaicin- and temperature-controlled gene
expression. This engineered calcium-dependent signaling pathway enables
tightly controlled regulation of gene expression through different
stimuli in mammalian cells and is versatile, adaptable, and useful
for a wide range of therapeutic and diagnostic applications.
Collapse
Affiliation(s)
- Maja Meško
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, Hajdrihova 19, SI-1001 Ljubljana, Slovenia
- Interfaculty Doctoral Study of Biomedicine, University of Ljubljana, SI-1000 Ljubljana, Slovenia
| | - Tina Lebar
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, Hajdrihova 19, SI-1001 Ljubljana, Slovenia
| | - Petra Dekleva
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, Hajdrihova 19, SI-1001 Ljubljana, Slovenia
| | - Roman Jerala
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, Hajdrihova 19, SI-1001 Ljubljana, Slovenia
- EN-FIST Centre of Excellence, Trg Osvobodilne fronte 13, SI-1000 Ljubljana, Slovenia
| | - Mojca Benčina
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, Hajdrihova 19, SI-1001 Ljubljana, Slovenia
- EN-FIST Centre of Excellence, Trg Osvobodilne fronte 13, SI-1000 Ljubljana, Slovenia
| |
Collapse
|
82
|
Zhao J, Ellis-Davies GCR. Intracellular photoswitchable neuropharmacology driven by luminescence from upconverting nanoparticles. Chem Commun (Camb) 2020; 56:9445-9448. [PMID: 32761019 PMCID: PMC7812838 DOI: 10.1039/d0cc03956j] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Photoswitchable drugs are small-molecule optical probes that undergo chromatically selective control of drug efficacy using, most often, UV-visible light. Here we report that luminescence produced by near-infrared stimulation of NaYF4:TmYb nanoparticles can be used for "remote control" of an azobenzene-based photochromic ion channel blocker of neurons in living brain slices.
Collapse
Affiliation(s)
- Jun Zhao
- Department of Neuroscience, Mount Sinai School of Medicine, New York, NY 10029, USA.
| | | |
Collapse
|
83
|
Yu Y, Yang X, Reghu S, Kaul SC, Wadhwa R, Miyako E. Photothermogenetic inhibition of cancer stemness by near-infrared-light-activatable nanocomplexes. Nat Commun 2020; 11:4117. [PMID: 32807785 PMCID: PMC7431860 DOI: 10.1038/s41467-020-17768-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Accepted: 07/17/2020] [Indexed: 02/06/2023] Open
Abstract
Strategies for eradicating cancer stem cells (CSCs) are urgently required because CSCs are resistant to anticancer drugs and cause treatment failure, relapse and metastasis. Here, we show that photoactive functional nanocarbon complexes exhibit unique characteristics, such as homogeneous particle morphology, high water dispersibility, powerful photothermal conversion, rapid photoresponsivity and excellent photothermal stability. In addition, the present biologically permeable second near-infrared (NIR-II) light-induced nanocomplexes photo-thermally trigger calcium influx into target cells overexpressing the transient receptor potential vanilloid family type 2 (TRPV2). This combination of nanomaterial design and genetic engineering effectively eliminates cancer cells and suppresses stemness of cancer cells in vitro and in vivo. Finally, in molecular analyses of mechanisms, we show that inhibition of cancer stemness involves calcium-mediated dysregulation of the Wnt/β-catenin signalling pathway. The present technological concept may lead to innovative therapies to address the global issue of refractory cancers. Cancer stem cells (CSCs) are known to induce chemotherapy resistance, and cause tumour relapse and metastasis. Here, the authors develop photoactive nanocarbon complexes with second near-infrared photothermal ability to target cancer cells overexpressing the receptor TRPV2 and show it to suppress CSCs through dysregulation of the Wnt/β-catenin signalling pathway.
Collapse
Affiliation(s)
- Yue Yu
- Graduate School of Advanced Science and Technology, Japan Advanced Institute of Science and Technology, 1-1 Asahidai, Nomi, Ishikawa, 923-1292, Japan.,Biomedical Research Institute, National Institute of Advanced Industrial Science & Technology (AIST), Ikeda, 563-8577, Japan
| | - Xi Yang
- Graduate School of Advanced Science and Technology, Japan Advanced Institute of Science and Technology, 1-1 Asahidai, Nomi, Ishikawa, 923-1292, Japan
| | - Sheethal Reghu
- Graduate School of Advanced Science and Technology, Japan Advanced Institute of Science and Technology, 1-1 Asahidai, Nomi, Ishikawa, 923-1292, Japan
| | - Sunil C Kaul
- AIST-INDIA DAILAB, DBT-AIST International Center for Translational & Environmental Research (DAICENTER), Cellular and Molecular Biotechnology Research Institute, AIST, Tsukuba, 305-8565, Japan
| | - Renu Wadhwa
- AIST-INDIA DAILAB, DBT-AIST International Center for Translational & Environmental Research (DAICENTER), Cellular and Molecular Biotechnology Research Institute, AIST, Tsukuba, 305-8565, Japan
| | - Eijiro Miyako
- Graduate School of Advanced Science and Technology, Japan Advanced Institute of Science and Technology, 1-1 Asahidai, Nomi, Ishikawa, 923-1292, Japan.
| |
Collapse
|
84
|
Nguyen NT, Ma G, Zhou Y, Jing J. Optogenetic approaches to control Ca 2+-modulated physiological processes. CURRENT OPINION IN PHYSIOLOGY 2020; 17:187-196. [PMID: 33184610 DOI: 10.1016/j.cophys.2020.08.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
As a versatile intracellular second messenger, calcium ion (Ca2+) regulates a plethora of physiological processes. To achieve precise control over Ca2+ signals in living cells and organisms, a set of optogenetic tools have recently been crafted by engineering photosensitive domains into intracellular signaling proteins, G-protein coupled receptors (GPCRs), receptor tyrosine kinases (RTKs), and Ca2+ channels. We highlight herein the optogenetic engineering strategies, kinetic properties, advantages and limitations of these genetically-encoded Ca2+ channel actuators (GECAs) and modulators. In parallel, we present exemplary applications in both excitable and non-excitable cells and tissues. Furthermore, we briefly discuss potential solutions for wireless optogenetics to accelerate the in vivo applications of GECAs under physiological conditions, with an emphasis on integrating near-infrared (NIR) light-excitable upconversion nanoparticles (UCNPs) and bioluminescence with optogenetics.
Collapse
Affiliation(s)
- Nhung T Nguyen
- Center for Translational Cancer Research, Institute of Biosciences and Technology, Texas A&M University, Houston, TX 77030, USA
| | - Guolin Ma
- Center for Translational Cancer Research, Institute of Biosciences and Technology, Texas A&M University, Houston, TX 77030, USA
| | - Yubin Zhou
- Center for Translational Cancer Research, Institute of Biosciences and Technology, Texas A&M University, Houston, TX 77030, USA.,Department of Translational Medical Sciences, College of Medicine, Texas A&M University, Houston, TX 77030, USA
| | - Ji Jing
- Center for Translational Cancer Research, Institute of Biosciences and Technology, Texas A&M University, Houston, TX 77030, USA
| |
Collapse
|
85
|
Near-infrared photocontrolled therapeutic release via upconversion nanocomposites. J Control Release 2020; 324:104-123. [DOI: 10.1016/j.jconrel.2020.05.011] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 04/30/2020] [Accepted: 05/05/2020] [Indexed: 12/12/2022]
|
86
|
A non-invasive far-red light-induced split-Cre recombinase system for controllable genome engineering in mice. Nat Commun 2020; 11:3708. [PMID: 32709899 PMCID: PMC7381682 DOI: 10.1038/s41467-020-17530-9] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Accepted: 07/03/2020] [Indexed: 12/19/2022] Open
Abstract
The Cre-loxP recombination system is a powerful tool for genetic manipulation. However, there are widely recognized limitations with chemically inducible Cre-loxP systems, and the UV and blue-light induced systems have phototoxicity and minimal capacity for deep tissue penetration. Here, we develop a far-red light-induced split Cre-loxP system (FISC system) based on a bacteriophytochrome optogenetic system and split-Cre recombinase, enabling optogenetical regulation of genome engineering in vivo solely by utilizing a far-red light (FRL). The FISC system exhibits low background and no detectable photocytotoxicity, while offering efficient FRL-induced DNA recombination. Our in vivo studies showcase the strong organ-penetration capacity of FISC system, markedly outperforming two blue-light-based Cre systems for recombination induction in the liver. Demonstrating its strong clinical relevance, we successfully deploy a FISC system using adeno-associated virus (AAV) delivery. Thus, the FISC system expands the optogenetic toolbox for DNA recombination to achieve spatiotemporally controlled, non-invasive genome engineering in living systems. Current light-inducible Cre-loxP systems have minimal capacity for deep tissue penetration. Here, the authors present a far-red light-induced split Cre-loxP system for in vivo genome engineering.
Collapse
|
87
|
Zhao J, Li B, Ma J, Jin W, Ma X. Photoactivatable RNA N 6 -Methyladenosine Editing with CRISPR-Cas13. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2020; 16:e1907301. [PMID: 32583968 DOI: 10.1002/smll.201907301] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/14/2019] [Revised: 04/27/2020] [Indexed: 06/11/2023]
Abstract
RNA has important and diverse biological roles, but the molecular methods to manipulate it spatiotemporally are limited. Here, an engineered photoactivatable RNA N6 -methyladenosine (m6 A) editing system with CRISPR-Cas13 is designed to direct specific m6 A editing. Light-inducible heterodimerizing proteins CIBN and CRY2PHR are fused to catalytically inactive PguCas13 (dCas13) and m6 A effectors, respectively. This system, referred to as PAMEC, enables the spatiotemporal control of m6 A editing in response to blue light. Further optimization of this system to create a highly efficient version, known as PAMECR , allows the manipulation of multiple genes robustly and simultaneously. When coupled with an upconversion nanoparticle film, the optogenetic operation window is extended from the visible range to tissue-penetrable near-infrared wavelengths, which offers an appealing avenue to remotely control RNA editing. These results show that PAMEC is a promising optogenetic platform for flexible and efficient targeting of RNA, with broad applicability for epitranscriptome engineering, imaging, and future therapeutic development.
Collapse
Affiliation(s)
- Jie Zhao
- Department of Orthopedics, Tianjin University Tianjin Hospital, Tianjin, 300211, China
| | - Bing Li
- Department of Orthopedics, Tianjin University Tianjin Hospital, Tianjin, 300211, China
| | - Jianxiong Ma
- Orthopedic Research Institute, Tianjin University Tianjin Hospital, Tianjin, 300050, China
| | - Weilin Jin
- Institute of Nano Biomedicine and Engineering, Shanghai Engineering Center for Intelligent Diagnosis and Treatment Instrument, Department of Instrument Science and Engineering, Key Laboratory for Thin Film and Microfabrication Technology of Ministry of Education, School of Electronic Information and Electronic Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
- National Center for Translational Medicine, Collaborative Innovational Center for System Biology, Shanghai Jiao Tong University, Shanghai, 200240, China
- Shaanxi Key Laboratory of Brain Disorders & Institute of Basic and Translational Medicine, Xi'an Medical University, Xi'an, 710021, China
| | - Xinlong Ma
- Department of Orthopedics, Tianjin University Tianjin Hospital, Tianjin, 300211, China
| |
Collapse
|
88
|
Ma G, Zhou Y. A STIMulating journey into optogenetic engineering. Cell Calcium 2020; 88:102197. [PMID: 32402855 PMCID: PMC7609480 DOI: 10.1016/j.ceca.2020.102197] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2020] [Accepted: 03/23/2020] [Indexed: 01/27/2023]
Abstract
Genetically-encoded calcium actuators (GECAs) stemmed from STIM1 have enabled optical activation of endogenous ORAI1 channels in both excitable and non-excitable tissues. These GECAs offer new non-invasive means to probe the structure-function relations of calcium channels and wirelessly control the behavior of awake mice.
Collapse
Affiliation(s)
- Guolin Ma
- Center for Translational Cancer Research, Institute of Biosciences and Technology, Texas A&M University, Houston, TX 77030, USA.
| | - Yubin Zhou
- Center for Translational Cancer Research, Institute of Biosciences and Technology, Texas A&M University, Houston, TX 77030, USA.
| |
Collapse
|
89
|
Yang X, Ma G, Zheng S, Qin X, Li X, Du L, Wang Y, Zhou Y, Li M. Optical Control of CRAC Channels Using Photoswitchable Azopyrazoles. J Am Chem Soc 2020; 142:9460-9470. [DOI: 10.1021/jacs.0c02949] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Affiliation(s)
- Xingye Yang
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology, School of Pharmacy, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Guolin Ma
- Center for Translational Cancer Research, Institute of Biosciences and Technology, Texas A&M University, Houston, Texas 77030, United States
| | - Sisi Zheng
- Beijing Key Laboratory of Gene Resource and Molecular Development, College of Life Sciences, Beijing Normal University, Beijing 100875, China
| | - Xiaojun Qin
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology, School of Pharmacy, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Xiang Li
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology, School of Pharmacy, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Lupei Du
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology, School of Pharmacy, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Youjun Wang
- Beijing Key Laboratory of Gene Resource and Molecular Development, College of Life Sciences, Beijing Normal University, Beijing 100875, China
| | - Yubin Zhou
- Center for Translational Cancer Research, Institute of Biosciences and Technology, Texas A&M University, Houston, Texas 77030, United States
| | - Minyong Li
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology, School of Pharmacy, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
- Helmholtz International Lab, State Key Laboratory of Microbial Technology, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250100, China
| |
Collapse
|
90
|
Leippe P, Broichhagen J, Cailliau K, Mougel A, Morel M, Dissous C, Trauner D, Vicogne J. Transformation of Receptor Tyrosine Kinases into Glutamate Receptors and Photoreceptors. Angew Chem Int Ed Engl 2020. [DOI: 10.1002/ange.201915352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Affiliation(s)
- Philipp Leippe
- Department of ChemistryLudwig-Maximilians-Universität München and Munich Center for Integrated Protein Science Butenandtstrasse 5–13 81377 München Germany
- Present address: Department of Chemical BiologyMax Planck Institute for Medical Research Jahnstr. 29 69120 Heidelberg Germany
| | - Johannes Broichhagen
- Department of ChemistryLudwig-Maximilians-Universität München and Munich Center for Integrated Protein Science Butenandtstrasse 5–13 81377 München Germany
- Present address: Department of Chemical BiologyForschungsinstitut für Molekulare Pharmakologie Robert-Rössle Str. 10 13125 Berlin Germany
| | - Katia Cailliau
- CNRS UMR 8576University of Lille Villeneuve d'Asq France
| | - Alexandra Mougel
- Univ. LilleCNRSInserm, CHU LilleInstitut Pasteur de LilleU1019—UMR 8204, Center for Infection and Immunity of Lille (CIIL) 59000 Lille France
| | - Marion Morel
- Department of Biochemistry and Molecular BiologyBoonshoft School of MedicineWright State University Dayton OH 45435 USA
| | - Colette Dissous
- Univ. LilleCNRSInserm, CHU LilleInstitut Pasteur de LilleU1019—UMR 8204, Center for Infection and Immunity of Lille (CIIL) 59000 Lille France
| | - Dirk Trauner
- Department of ChemistryLudwig-Maximilians-Universität München and Munich Center for Integrated Protein Science Butenandtstrasse 5–13 81377 München Germany
- Department of ChemistrySilver Center for Arts and ScienceNew York University 100 Washington Square East New York NY 10003 USA
| | - Jérôme Vicogne
- Univ. LilleCNRSInserm, CHU LilleInstitut Pasteur de LilleU1019—UMR 8204, Center for Infection and Immunity of Lille (CIIL) 59000 Lille France
| |
Collapse
|
91
|
Chen G, Cao Y, Tang Y, Yang X, Liu Y, Huang D, Zhang Y, Li C, Wang Q. Advanced Near-Infrared Light for Monitoring and Modulating the Spatiotemporal Dynamics of Cell Functions in Living Systems. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2020; 7:1903783. [PMID: 32328436 PMCID: PMC7175256 DOI: 10.1002/advs.201903783] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Revised: 02/06/2020] [Indexed: 05/07/2023]
Abstract
Light-based technique, including optical imaging and photoregulation, has become one of the most important tools for both fundamental research and clinical practice, such as cell signal sensing, cancer diagnosis, tissue engineering, drug delivery, visual regulation, neuromodulation, and disease treatment. In particular, low energy near-infrared (NIR, 700-1700 nm) light possesses lower phototoxicity and higher tissue penetration depth in living systems as compared with ultraviolet/visible light, making it a promising tool for in vivo applications. Currently, the NIR light-based imaging and photoregulation strategies have offered a possibility to real-time sense and/or modulate specific cellular events in deep tissues with subcellular accuracy. Herein, the recent progress with respect to NIR light for monitoring and modulating the spatiotemporal dynamics of cell functions in living systems are summarized. In particular, the applications of NIR light-based techniques in cancer theranostics, regenerative medicine, and neuroscience research are systematically introduced and discussed. In addition, the challenges and prospects for NIR light-based cell sensing and regulating techniques are comprehensively discussed.
Collapse
Affiliation(s)
- Guangcun Chen
- CAS Key Laboratory of Nano‐Bio InterfaceDivision of Nanobiomedicine and i‐LabCAS Center for Excellence in Brain ScienceSuzhou Institute of Nano‐Tech and Nano‐BionicsChinese Academy of SciencesSuzhou215123China
- Suzhou Key Laboratory of Functional Molecular Imaging TechnologySuzhou Institute of Nano‐Tech and Nano‐BionicsChinese Academy of SciencesSuzhou215123China
- School of Nano‐Tech and Nano‐BionicsUniversity of Science and Technology of ChinaHefei230026China
| | - Yuheng Cao
- CAS Key Laboratory of Nano‐Bio InterfaceDivision of Nanobiomedicine and i‐LabCAS Center for Excellence in Brain ScienceSuzhou Institute of Nano‐Tech and Nano‐BionicsChinese Academy of SciencesSuzhou215123China
- Suzhou Key Laboratory of Functional Molecular Imaging TechnologySuzhou Institute of Nano‐Tech and Nano‐BionicsChinese Academy of SciencesSuzhou215123China
| | - Yanxing Tang
- CAS Key Laboratory of Nano‐Bio InterfaceDivision of Nanobiomedicine and i‐LabCAS Center for Excellence in Brain ScienceSuzhou Institute of Nano‐Tech and Nano‐BionicsChinese Academy of SciencesSuzhou215123China
- Suzhou Key Laboratory of Functional Molecular Imaging TechnologySuzhou Institute of Nano‐Tech and Nano‐BionicsChinese Academy of SciencesSuzhou215123China
| | - Xue Yang
- CAS Key Laboratory of Nano‐Bio InterfaceDivision of Nanobiomedicine and i‐LabCAS Center for Excellence in Brain ScienceSuzhou Institute of Nano‐Tech and Nano‐BionicsChinese Academy of SciencesSuzhou215123China
- Suzhou Key Laboratory of Functional Molecular Imaging TechnologySuzhou Institute of Nano‐Tech and Nano‐BionicsChinese Academy of SciencesSuzhou215123China
- School of Nano‐Tech and Nano‐BionicsUniversity of Science and Technology of ChinaHefei230026China
| | - Yongyang Liu
- CAS Key Laboratory of Nano‐Bio InterfaceDivision of Nanobiomedicine and i‐LabCAS Center for Excellence in Brain ScienceSuzhou Institute of Nano‐Tech and Nano‐BionicsChinese Academy of SciencesSuzhou215123China
- Suzhou Key Laboratory of Functional Molecular Imaging TechnologySuzhou Institute of Nano‐Tech and Nano‐BionicsChinese Academy of SciencesSuzhou215123China
- School of Nano‐Tech and Nano‐BionicsUniversity of Science and Technology of ChinaHefei230026China
| | - Dehua Huang
- CAS Key Laboratory of Nano‐Bio InterfaceDivision of Nanobiomedicine and i‐LabCAS Center for Excellence in Brain ScienceSuzhou Institute of Nano‐Tech and Nano‐BionicsChinese Academy of SciencesSuzhou215123China
- Suzhou Key Laboratory of Functional Molecular Imaging TechnologySuzhou Institute of Nano‐Tech and Nano‐BionicsChinese Academy of SciencesSuzhou215123China
- School of Nano‐Tech and Nano‐BionicsUniversity of Science and Technology of ChinaHefei230026China
| | - Yejun Zhang
- CAS Key Laboratory of Nano‐Bio InterfaceDivision of Nanobiomedicine and i‐LabCAS Center for Excellence in Brain ScienceSuzhou Institute of Nano‐Tech and Nano‐BionicsChinese Academy of SciencesSuzhou215123China
- Suzhou Key Laboratory of Functional Molecular Imaging TechnologySuzhou Institute of Nano‐Tech and Nano‐BionicsChinese Academy of SciencesSuzhou215123China
- School of Nano‐Tech and Nano‐BionicsUniversity of Science and Technology of ChinaHefei230026China
| | - Chunyan Li
- CAS Key Laboratory of Nano‐Bio InterfaceDivision of Nanobiomedicine and i‐LabCAS Center for Excellence in Brain ScienceSuzhou Institute of Nano‐Tech and Nano‐BionicsChinese Academy of SciencesSuzhou215123China
- Suzhou Key Laboratory of Functional Molecular Imaging TechnologySuzhou Institute of Nano‐Tech and Nano‐BionicsChinese Academy of SciencesSuzhou215123China
- School of Nano‐Tech and Nano‐BionicsUniversity of Science and Technology of ChinaHefei230026China
| | - Qiangbin Wang
- CAS Key Laboratory of Nano‐Bio InterfaceDivision of Nanobiomedicine and i‐LabCAS Center for Excellence in Brain ScienceSuzhou Institute of Nano‐Tech and Nano‐BionicsChinese Academy of SciencesSuzhou215123China
- Suzhou Key Laboratory of Functional Molecular Imaging TechnologySuzhou Institute of Nano‐Tech and Nano‐BionicsChinese Academy of SciencesSuzhou215123China
- School of Nano‐Tech and Nano‐BionicsUniversity of Science and Technology of ChinaHefei230026China
- College of Materials Sciences and Opto‐Electronic TechnologyUniversity of Chinese Academy of SciencesBeijing100049China
| |
Collapse
|
92
|
Leippe P, Broichhagen J, Cailliau K, Mougel A, Morel M, Dissous C, Trauner D, Vicogne J. Transformation of Receptor Tyrosine Kinases into Glutamate Receptors and Photoreceptors. Angew Chem Int Ed Engl 2020; 59:6720-6723. [DOI: 10.1002/anie.201915352] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Indexed: 11/07/2022]
Affiliation(s)
- Philipp Leippe
- Department of ChemistryLudwig-Maximilians-Universität München and Munich Center for Integrated Protein Science Butenandtstrasse 5–13 81377 München Germany
- Present address: Department of Chemical BiologyMax Planck Institute for Medical Research Jahnstr. 29 69120 Heidelberg Germany
| | - Johannes Broichhagen
- Department of ChemistryLudwig-Maximilians-Universität München and Munich Center for Integrated Protein Science Butenandtstrasse 5–13 81377 München Germany
- Present address: Department of Chemical BiologyForschungsinstitut für Molekulare Pharmakologie Robert-Rössle Str. 10 13125 Berlin Germany
| | - Katia Cailliau
- CNRS UMR 8576University of Lille Villeneuve d'Asq France
| | - Alexandra Mougel
- Univ. LilleCNRSInserm, CHU LilleInstitut Pasteur de LilleU1019—UMR 8204, Center for Infection and Immunity of Lille (CIIL) 59000 Lille France
| | - Marion Morel
- Department of Biochemistry and Molecular BiologyBoonshoft School of MedicineWright State University Dayton OH 45435 USA
| | - Colette Dissous
- Univ. LilleCNRSInserm, CHU LilleInstitut Pasteur de LilleU1019—UMR 8204, Center for Infection and Immunity of Lille (CIIL) 59000 Lille France
| | - Dirk Trauner
- Department of ChemistryLudwig-Maximilians-Universität München and Munich Center for Integrated Protein Science Butenandtstrasse 5–13 81377 München Germany
- Department of ChemistrySilver Center for Arts and ScienceNew York University 100 Washington Square East New York NY 10003 USA
| | - Jérôme Vicogne
- Univ. LilleCNRSInserm, CHU LilleInstitut Pasteur de LilleU1019—UMR 8204, Center for Infection and Immunity of Lille (CIIL) 59000 Lille France
| |
Collapse
|
93
|
Paoletti P, Ellis-Davies GCR, Mourot A. Optical control of neuronal ion channels and receptors. Nat Rev Neurosci 2020; 20:514-532. [PMID: 31289380 DOI: 10.1038/s41583-019-0197-2] [Citation(s) in RCA: 118] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Light-controllable tools provide powerful means to manipulate and interrogate brain function with relatively low invasiveness and high spatiotemporal precision. Although optogenetic approaches permit neuronal excitation or inhibition at the network level, other technologies, such as optopharmacology (also known as photopharmacology) have emerged that provide molecular-level control by endowing light sensitivity to endogenous biomolecules. In this Review, we discuss the challenges and opportunities of photocontrolling native neuronal signalling pathways, focusing on ion channels and neurotransmitter receptors. We describe existing strategies for rendering receptors and channels light sensitive and provide an overview of the neuroscientific insights gained from such approaches. At the crossroads of chemistry, protein engineering and neuroscience, optopharmacology offers great potential for understanding the molecular basis of brain function and behaviour, with promises for future therapeutics.
Collapse
Affiliation(s)
- Pierre Paoletti
- Institut de Biologie de l'Ecole Normale Supérieure (IBENS), Ecole Normale Supérieure, CNRS, INSERM, Université PSL, Paris, France.
| | | | - Alexandre Mourot
- Neuroscience Paris Seine-Institut de Biologie Paris Seine (NPS-IBPS), CNRS, INSERM, Sorbonne Université, Paris, France.
| |
Collapse
|
94
|
Lei Z, Ling X, Mei Q, Fu S, Zhang J, Zhang Y. An Excitation Navigating Energy Migration of Lanthanide Ions in Upconversion Nanoparticles. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2020; 32:e1906225. [PMID: 31961986 DOI: 10.1002/adma.201906225] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/22/2019] [Revised: 11/25/2019] [Indexed: 06/10/2023]
Abstract
Upconversion nanoparticles (UCNPs) doped with lanthanide ions that possess ladder-like energy levels can give out multiple emissions at specific ultra-violet or visible wavelengths irrespective of excitation light. However, precisely controlling energy migration processes between different energy levels of the same lanthanide ion to generate switchable emissions remains elusive. Herein, a novel dumbbell-shaped UCNP is reported with upconverted red emission switched to green emission when excitation wavelength changed from 980 to 808 nm. The sensitizer Yb ions are doped with activator Er ions and energy modulator Mn ions in NaYF4 core nanocrystal coated with an inner NaYF4 :Yb shell to generate red emission after harvesting 980 nm excitation light, while an outer NaNdF4 :Yb shell is coated to form a dumbbell shape to generate green emission upon 808 nm excitation. Such specially designed UCNPs with switchable green and red emissions are further explored for imaging of latent fingerprint and detection of explosive residues in the fingerprint simultaneously. This work suggests a novel research interest in fine-tuning of upconversion emissions through precisely controlling energy migration processes of the same lanthanide activator ion. Furthermore, use of these nanoparticles in other applications such as simultaneous dual-color imaging or orthogonal bidirectional photoactivation can be explored.
Collapse
Affiliation(s)
- Zhendong Lei
- NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore, Singapore, 117583, Singapore
- Department of Biomedical Engineering, Faculty of Engineering, National University of Singapore, Singapore, 117583, Singapore
| | - Xiao Ling
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, Anhui, 230009, China
| | - Qingsong Mei
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, Anhui, 230009, China
| | - Shuai Fu
- School of Environmental and Chemical Engineering, Shanghai University, Shanghai, 200444, China
| | - Jing Zhang
- School of Environmental and Chemical Engineering, Shanghai University, Shanghai, 200444, China
| | - Yong Zhang
- NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore, Singapore, 117583, Singapore
- Department of Biomedical Engineering, Faculty of Engineering, National University of Singapore, Singapore, 117583, Singapore
- School of Environmental and Chemical Engineering, Shanghai University, Shanghai, 200444, China
| |
Collapse
|
95
|
Ma G, He L, Liu S, Xie J, Huang Z, Jing J, Lee YT, Wang R, Luo H, Han W, Huang Y, Zhou Y. Optogenetic engineering to probe the molecular choreography of STIM1-mediated cell signaling. Nat Commun 2020; 11:1039. [PMID: 32098964 PMCID: PMC7042325 DOI: 10.1038/s41467-020-14841-9] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Accepted: 02/06/2020] [Indexed: 02/07/2023] Open
Abstract
Genetically encoded photoswitches have enabled spatial and temporal control of cellular events to achieve tailored functions in living cells, but their applications to probe the structure-function relations of signaling proteins are still underexplored. We illustrate herein the incorporation of various blue light-responsive photoreceptors into modular domains of the stromal interaction molecule 1 (STIM1) to manipulate protein activity and faithfully recapitulate STIM1-mediated signaling events. Capitalizing on these optogenetic tools, we identify the molecular determinants required to mediate protein oligomerization, intramolecular conformational switch, and protein-target interactions. In parallel, we have applied these synthetic devices to enable light-inducible gating of calcium channels, conformational switch, dynamic protein-microtubule interactions and assembly of membrane contact sites in a reversible manner. Our optogenetic engineering approach can be broadly applied to aid the mechanistic dissection of cell signaling, as well as non-invasive interrogation of physiological processes with high precision. Optogenetic tools have been used to control cellular behaviours but their use to probe structure-function relations of signalling proteins are underexplored. Here the authors engineer optogenetic modules into STIM1 to dissect molecular details of STIM1-mediated signalling and control various cellular events.
Collapse
Affiliation(s)
- Guolin Ma
- Center for Translational Cancer Research, Institute of Biosciences and Technology, Texas A&M University, Houston, TX, 77030, USA
| | - Lian He
- Center for Translational Cancer Research, Institute of Biosciences and Technology, Texas A&M University, Houston, TX, 77030, USA
| | - Shuzhong Liu
- Center for Translational Cancer Research, Institute of Biosciences and Technology, Texas A&M University, Houston, TX, 77030, USA.,Department of Gastroenterology, Key Laboratory of Hubei Province for Digestive System Disease, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Jiansheng Xie
- Center for Translational Cancer Research, Institute of Biosciences and Technology, Texas A&M University, Houston, TX, 77030, USA.,Department of Medical Oncology, Laboratory of Cancer Biology, Institute of Clinical Science, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Zixian Huang
- Center for Translational Cancer Research, Institute of Biosciences and Technology, Texas A&M University, Houston, TX, 77030, USA.,Department of Oral and Maxillofacial Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510120, China
| | - Ji Jing
- Center for Translational Cancer Research, Institute of Biosciences and Technology, Texas A&M University, Houston, TX, 77030, USA
| | - Yi-Tsang Lee
- Center for Translational Cancer Research, Institute of Biosciences and Technology, Texas A&M University, Houston, TX, 77030, USA
| | - Rui Wang
- Center for Translational Cancer Research, Institute of Biosciences and Technology, Texas A&M University, Houston, TX, 77030, USA
| | - Hesheng Luo
- Department of Gastroenterology, Key Laboratory of Hubei Province for Digestive System Disease, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Weidong Han
- Department of Medical Oncology, Laboratory of Cancer Biology, Institute of Clinical Science, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China.
| | - Yun Huang
- Center for Epigenetics and Disease Prevention, Institute of Biosciences and Technology, Texas A&M University, Houston, TX, 77030, USA.
| | - Yubin Zhou
- Center for Translational Cancer Research, Institute of Biosciences and Technology, Texas A&M University, Houston, TX, 77030, USA. .,Department of Translational Medical Sciences, College of Medicine, Texas A&M University, Houston, TX, 77030, USA.
| |
Collapse
|
96
|
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
|
97
|
Kim S, Kyung T, Chung JH, Kim N, Keum S, Lee J, Park H, Kim HM, Lee S, Shin HS, Do Heo W. Non-invasive optical control of endogenous Ca 2+ channels in awake mice. Nat Commun 2020; 11:210. [PMID: 31924789 PMCID: PMC6954201 DOI: 10.1038/s41467-019-14005-4] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2019] [Accepted: 12/05/2019] [Indexed: 02/08/2023] Open
Abstract
Optogenetic approaches for controlling Ca2+ channels provide powerful means for modulating diverse Ca2+-specific biological events in space and time. However, blue light-responsive photoreceptors are, in principle, considered inadequate for deep tissue stimulation unless accompanied by optic fiber insertion. Here, we present an ultra-light-sensitive optogenetic Ca2+ modulator, named monSTIM1 encompassing engineered cryptochrome2 for manipulating Ca2+ signaling in the brain of awake mice through non-invasive light delivery. Activation of monSTIM1 in either excitatory neurons or astrocytes of mice brain is able to induce Ca2+-dependent gene expression without any mechanical damage in the brain. Furthermore, we demonstrate that non-invasive Ca2+ modulation in neurons can be sufficiently and effectively translated into changes in behavioral phenotypes of awake mice. Optogenetic applications in the brain of live animals often require the use of optic fibers due to poor tissue-penetration of blue light. Here the authors present monSTIM1, an improved high sensitivity optogenetic tool able to modulate Ca2+ signaling in the brain of awake mice using non-invasive light stimulation.
Collapse
Affiliation(s)
- Sungsoo Kim
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Taeyoon Kyung
- Center for Cognition and Sociality, Institute for Basic Science (IBS), Daejeon, Republic of Korea.,David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Jae-Hee Chung
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Nury Kim
- Center for Cognition and Sociality, Institute for Basic Science (IBS), Daejeon, Republic of Korea
| | - Sehoon Keum
- Center for Cognition and Sociality, Institute for Basic Science (IBS), Daejeon, Republic of Korea
| | - Jinsu Lee
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Hyerim Park
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Ho Min Kim
- Center for Biomolecular and Cellular Structure, Institute for Basic Science (IBS), Daejeon, Republic of Korea.,Graduate School of Medical Science & Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Sangkyu Lee
- Center for Cognition and Sociality, Institute for Basic Science (IBS), Daejeon, Republic of Korea.
| | - Hee-Sup Shin
- Center for Cognition and Sociality, Institute for Basic Science (IBS), Daejeon, Republic of Korea.
| | - Won Do Heo
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea. .,Center for Cognition and Sociality, Institute for Basic Science (IBS), Daejeon, Republic of Korea. .,KAIST Institute for the BioCentury, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea.
| |
Collapse
|
98
|
Butorac C, Krizova A, Derler I. Review: Structure and Activation Mechanisms of CRAC Channels. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1131:547-604. [PMID: 31646526 DOI: 10.1007/978-3-030-12457-1_23] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Ca2+ release activated Ca2+ (CRAC) channels represent a primary pathway for Ca2+ to enter non-excitable cells. The two key players in this process are the stromal interaction molecule (STIM), a Ca2+ sensor embedded in the membrane of the endoplasmic reticulum, and Orai, a highly Ca2+ selective ion channel located in the plasma membrane. Upon depletion of the internal Ca2+ stores, STIM is activated, oligomerizes, couples to and activates Orai. This review provides an overview of novel findings about the CRAC channel activation mechanisms, structure and gating. In addition, it highlights, among diverse STIM and Orai mutants, also the disease-related mutants and their implications.
Collapse
Affiliation(s)
- Carmen Butorac
- Institute of Biophysics, Johannes Kepler University of Linz, Linz, Austria
| | - Adéla Krizova
- Institute of Biophysics, Johannes Kepler University of Linz, Linz, Austria
| | - Isabella Derler
- Institute of Biophysics, Johannes Kepler University of Linz, Linz, Austria.
| |
Collapse
|
99
|
Blue light-triggered optogenetic system for treating uveal melanoma. Oncogene 2019; 39:2118-2124. [PMID: 31811271 DOI: 10.1038/s41388-019-1119-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 11/07/2019] [Accepted: 11/11/2019] [Indexed: 11/09/2022]
Abstract
Uveal melanoma is the most common intraocular primary malignancy in adults and has been considered a fatal disease for decades. Optogenetics is an emerging technique that can control the activation of signaling components via irradiation with visible light. The clinical translation of optogenetics has been limited because of the need for surgical implantation of electrodes and relatively shallow tissue penetration. As visible light easily penetrates the eyes, we hypothesized that an optogenetics approach can be an effective treatment of uveal melanoma without surgery. In this study, we evaluated the feasibility of this strategy by using a genetically encoded optogenetic system based on reversible blue light-induced binding pairs between Fas-CIB1-EGFP and CRY2-mCherry-FADD. Subretinal injection of B16 cells was performed to create a uveal melanoma model. Plasmids pairs were co-transfected into B16 cells. We found that blue light irradiation dynamically controlled the translocation of FADD to Fas on the plasma membrane and induced the apoptosis of B16 cells transfected with the optogenetic nanosystem in vitro. Moreover, the blue light-controlled optogenetic nanosystem suppressed the growth of uveal melanoma in vivo by inducing apoptosis. These results suggest that light-controlled optogenetic therapy can be used as a potential novel therapeutic strategy for uveal melanoma.
Collapse
|
100
|
Mondal P, Krishnamurthy VV, Sharum SR, Haack N, Zhou H, Cheng J, Yang J, Zhang K. Repurposing Protein Degradation for Optogenetic Modulation of Protein Activities. ACS Synth Biol 2019; 8:2585-2592. [PMID: 31600062 DOI: 10.1021/acssynbio.9b00285] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Non-neuronal optogenetic approaches empower precise regulation of protein dynamics in live cells but often require target-specific protein engineering. To address this challenge, we developed a generalizable light-modulated protein stabilization system (GLIMPSe) to control the intracellular protein level independent of its functionality. We applied GLIMPSe to control two distinct classes of proteins: mitogen-activated protein kinase phosphatase 3 (MKP3), a negative regulator of the extracellular signal-regulated kinase (ERK) pathway, and a constitutively active form of MEK (CA MEK), a positive regulator of the same pathway. Kinetics study showed that light-induced protein stabilization could be achieved within 30 min of blue light stimulation. GLIMPSe enables target-independent optogenetic control of protein activities and therefore minimizes the systematic variation embedded within different photoactivatable proteins. Overall, GLIMPSe promises to achieve light-mediated post-translational stabilization of a wide array of target proteins in live cells.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Jing Yang
- Department of Comparative Biosciences, University of Illinois at Urbana−Champaign, 2001 S Lincoln Avenue, Urbana, Illinois 61802, United States
| | | |
Collapse
|