1
|
Liu W, Lu D, Jia S, Yang Y, Meng F, Du Y, Yang Y, Yuan L, Nan Y. Molecular mechanism of Gancao Xiexin Decoction regulating EMT and suppressing hepatic metastasis of gastric cancer via the TGF-β1/SMAD pathway. JOURNAL OF ETHNOPHARMACOLOGY 2025; 342:119430. [PMID: 39900270 DOI: 10.1016/j.jep.2025.119430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 01/16/2025] [Accepted: 01/28/2025] [Indexed: 02/05/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Gastric cancer (GC) is a highly malignant tumor of the digestive tract, posing a significant menace to human health. Gancao Xiexin Decoction (GCXXD), being a traditional Chinese medicine (TCM), has a good effect on inhibiting the proliferation and metastasis of GC. However, its mechanisms still need further investigation. AIM OF STUDY To investigate the mechanism by which GCXXD inhibits GC metastasis through network pharmacology, and to verify through in vivo and in vitro experiments. MATERIALS AND METHODS The TCMSP and GEO databases, in combination with UPLC-MS/MS techniques, were employed to identify the hub genes, active ingredients, and critical pathways of GCXXD in the treatment of GC. Subsequently, molecular docking was conducted on both the hub genes and the core components. Finally, based on the results of the bioinformatics analysis, the role of GCXXD in inhibiting liver metastasis of GC was elucidated through in vivo and in vitro experiments, including scratch assays, Transwell assays, HE staining, immunohistochemistry, in vivo live imaging, qRT-PCR, and Western blotting. RESULTS Utilizing UPLC-MS/MS and network pharmacology, we identified 20 active ingredients and 5 hub targets in the treatment of GC by GCXXD. Through KEGG analyses, GCXXD treatment of GC could through the TGF-beta pathway. In vivo and in vitro experiments, GCXXD downregulated the mRNA and protein expression level of hub genes involved in the TGF-β1/SMAD pathway and the EMT process. Additionally, GCXXD significantly reduced the incidence of liver metastases in GC. CONCLUSION GCXXD inhibited EMT via blocking the TGF-β1/SMAD pathway, which suppressed GC cell growth and liver metastasis. This study provides data to support the treatment of liver metastasis in GC with TCM and holds significant importance for the research and development of new anticancer drugs.
Collapse
Affiliation(s)
- Wenjing Liu
- Key Laboratory of Ningxia Ethnomedicine Modernization, Ministry of Education, Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - Doudou Lu
- School of Clinical Medicine, Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - Shumin Jia
- Key Laboratory of Ningxia Ethnomedicine Modernization, Ministry of Education, Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - Yating Yang
- The Second Hospital of Chinese Medicine of BAO JI City, Baoji, 721300, Xian, China
| | - Fandi Meng
- Traditional Chinese Medicine College, Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - Yuhua Du
- College of Pharmacy, Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - Yi Yang
- College of Pharmacy, Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - Ling Yuan
- College of Pharmacy, Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - Yi Nan
- Key Laboratory of Ningxia Ethnomedicine Modernization, Ministry of Education, Ningxia Medical University, Yinchuan, 750004, Ningxia, China; Traditional Chinese Medicine College, Ningxia Medical University, Yinchuan, 750004, Ningxia, China.
| |
Collapse
|
2
|
Gangadaran P, Onkar A, Rajendran RL, Goenka A, Oh JM, Khan F, Nagarajan AK, Muthu S, Krishnan A, Hong CM, Ahn BC. Noninvasive in vivo imaging of macrophages: understanding tumor microenvironments and delivery of therapeutics. Biomark Res 2025; 13:20. [PMID: 39865337 PMCID: PMC11770947 DOI: 10.1186/s40364-025-00735-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Accepted: 01/21/2025] [Indexed: 01/28/2025] Open
Abstract
Macrophages are pivotal in the body's defense and response to inflammation. They are present in significant numbers and are widely implicated in various diseases, including cancer. While molecular and histological techniques have advanced our understanding of macrophage biology, their precise function within the cancerous microenvironments remains underexplored. Enhancing our knowledge of macrophages and the dynamics of their extracellular vesicles (EVs) in cancer development can potentially improve therapeutic management. Notably, macrophages have also been harnessed to deliver drugs. Noninvasive in vivo molecular imaging of macrophages is crucial for investigating intricate cellular processes, comprehending the underlying mechanisms of diseases, tracking cells and EVs' migration, and devising macrophage-dependent drug-delivery systems in living organisms. Thus, in vivo imaging of macrophages has become an indispensable tool in biomedical research. The integration of multimodal imaging approaches and the continued development of novel contrast agents hold promise for overcoming current limitations and expanding the applications of macrophage imaging. This study comprehensively reviews several methods for labeling macrophages and various imaging modalities, assessing the merits and drawbacks of each approach. The review concludes by offering insights into the applicability of molecular imaging techniques for real time monitoring of macrophages in preclinical and clinical scenarios.
Collapse
Affiliation(s)
- Prakash Gangadaran
- BK21 FOUR KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, Department of Biomedical Sciences, School of Medicine, Kyungpook National University, Daegu, 41944, Korea
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu, 41944, Korea
- Cardiovascular Research Institute, Kyungpook National University, Daegu, 41944, Republic of Korea
| | - Akanksha Onkar
- Department of Laboratory Medicine, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Ramya Lakshmi Rajendran
- BK21 FOUR KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, Department of Biomedical Sciences, School of Medicine, Kyungpook National University, Daegu, 41944, Korea
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu, 41944, Korea
- Cardiovascular Research Institute, Kyungpook National University, Daegu, 41944, Republic of Korea
| | - Anshika Goenka
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA, 30322, USA
| | - Ji Min Oh
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu, 41944, Korea
- Cardiovascular Research Institute, Kyungpook National University, Daegu, 41944, Republic of Korea
| | - Fatima Khan
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44195, USA
| | - ArulJothi Kandasamy Nagarajan
- Department of Genetic Engineering, SRM Institute of Science and Technology, Kattankulathur, Chengalpattu, 603203, Tamilnadu, India
| | - Sathish Muthu
- Department of Orthopaedics, Government Medical College, Tamil Nadu, 639004, Karur, India
- Department of Biotechnology, Faculty of Engineering, Karpagam Academy of Higher Education, Tamil Nadu, 641021, Coimbatore, India
| | - Anand Krishnan
- Precision Medicine and Integrated Nano-Diagnostics (P-MIND) Research Group, Office of the Dean, Faculty of Health Sciences, University of the Free State, Bloemfontein, 9300, South Africa
| | - Chae Moon Hong
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu, 41944, Korea.
- Cardiovascular Research Institute, Kyungpook National University, Daegu, 41944, Republic of Korea.
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, 41944, Korea.
| | - Byeong-Cheol Ahn
- BK21 FOUR KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, Department of Biomedical Sciences, School of Medicine, Kyungpook National University, Daegu, 41944, Korea.
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu, 41944, Korea.
- Cardiovascular Research Institute, Kyungpook National University, Daegu, 41944, Republic of Korea.
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, 41944, Korea.
| |
Collapse
|
3
|
Tian X, Zhang Y, Ai HW. PEGylated ATP-Independent Luciferins for Noninvasive High-Sensitivity High-Speed Bioluminescence Imaging. ACS Chem Biol 2025; 20:128-136. [PMID: 39714242 PMCID: PMC11744661 DOI: 10.1021/acschembio.4c00601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 11/17/2024] [Accepted: 12/12/2024] [Indexed: 12/24/2024]
Abstract
Bioluminescence imaging (BLI) is a powerful, noninvasive imaging method for animal studies. NanoLuc luciferase and its derivatives are attractive bioluminescent reporters recognized for their efficient photon production and ATP independence. However, utilizing them for animal imaging poses notable challenges. Low substrate solubility has been a prominent problem, limiting in vivo brightness, while the susceptibility of luciferins to auto-oxidation by molecular oxygen in air increases handling complexity and poses an obstacle to obtaining consistent results. To address these issues, we developed a range of caged PEGylated luciferins with increased auto-oxidation resistance and water solubility of up to 25 mM, resulting in substantial in vivo bioluminescence increases in mouse models. This advancement has created the brightest and most sensitive luciferase-luciferin combination, enabling high-speed video-rate imaging of freely moving mice with brain-expressed luciferase. These innovative substrates offer new possibilities for investigating a wide range of biological processes and are poised to become invaluable resources for chemical, biological, and biomedical fields.
Collapse
Affiliation(s)
- Xiaodong Tian
- Department
of Molecular Physiology and Biological Physics, University of Virginia School of Medicine, Charlottesville, Virginia 22908, United States
- Center
for Membrane and Cell Physiology, University
of Virginia School of Medicine, Charlottesville, Virginia 22908, United States
| | - Yiyu Zhang
- Department
of Molecular Physiology and Biological Physics, University of Virginia School of Medicine, Charlottesville, Virginia 22908, United States
- Center
for Membrane and Cell Physiology, University
of Virginia School of Medicine, Charlottesville, Virginia 22908, United States
| | - Hui-Wang Ai
- Department
of Molecular Physiology and Biological Physics, University of Virginia School of Medicine, Charlottesville, Virginia 22908, United States
- Center
for Membrane and Cell Physiology, University
of Virginia School of Medicine, Charlottesville, Virginia 22908, United States
- The
UVA Comprehensive Cancer Center, University
of Virginia, Charlottesville, Virginia 22908, United States
| |
Collapse
|
4
|
Boitet M, Achek A, Bouchenaki K, Grailhe R. BrightMice: a low-cost do-it-yourself instrument, designed for in vivo fluorescence mouse imaging. Sci Rep 2024; 14:22685. [PMID: 39349676 PMCID: PMC11442974 DOI: 10.1038/s41598-024-73130-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 09/13/2024] [Indexed: 10/04/2024] Open
Abstract
In vivo fluorescent imaging represents a potent means for real-time probe quantification, facilitating insights into disease pathophysiology and therapeutic responses. Nonetheless, accurate signal quantification remains challenging due to inherent factors like light scattering and tissue absorption. Existing imaging systems, though sophisticated, often entail high costs and are typically restricted to well-funded laboratory settings. This study introduces BrightMice, an innovative in vivo fluorescent imaging system that harnesses 3D printing and consumer-grade digital cameras. Tailored for various fluorophores such as EYFP and E2-crimson, the system showcases both adaptability and effectiveness in detecting in vivo fluorescent signals in several reporter mouse strains. Comparative analyses against commercial instruments confirm BrightMice's sensitivity and underscore its potential to democratize in vivo fluorescence imaging. By providing a cost-effective and accessible solution, BrightMice stands to benefit diverse research environments.
Collapse
Affiliation(s)
- Maylis Boitet
- Technology Development Platform, Institut Pasteur Korea, 16, Daewangpangyo-ro 712beon-gil, Bundang-gu, Seongnam-si, 13488, Republic of Korea
- Division of Bio-Medical Science & Technology, Korea University of Science and Technology, 217 Gajeong-ro Yuseong-gu, Daejeon, Republic of Korea
- Core Technology Platforms, New York University Abu Dhabi, P.O. Box 129188, Abu Dhabi, United Arab Emirates
| | - Asma Achek
- Technology Development Platform, Institut Pasteur Korea, 16, Daewangpangyo-ro 712beon-gil, Bundang-gu, Seongnam-si, 13488, Republic of Korea
| | | | - Regis Grailhe
- Technology Development Platform, Institut Pasteur Korea, 16, Daewangpangyo-ro 712beon-gil, Bundang-gu, Seongnam-si, 13488, Republic of Korea.
- Division of Bio-Medical Science & Technology, Korea University of Science and Technology, 217 Gajeong-ro Yuseong-gu, Daejeon, Republic of Korea.
- Smart-MD, Institut Pasteur Korea, Seongnam, Republic of Korea.
| |
Collapse
|
5
|
Ramos-Gonzalez MR, Sirpu Natesh N, Rachagani S, Amos-Landgraf J, Shirwan H, Yolcu ES, Gomez-Gutierrez JG. Establishment of Translational Luciferase-Based Cancer Models to Evaluate Antitumoral Therapies. Int J Mol Sci 2024; 25:10418. [PMID: 39408747 PMCID: PMC11476533 DOI: 10.3390/ijms251910418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 09/24/2024] [Accepted: 09/25/2024] [Indexed: 10/19/2024] Open
Abstract
Luciferase (luc) bioluminescence (BL) is the most used light-emitting protein that has been engineered to be expressed in multiple cancer cell lines, allowing for the detection of tumor nodules in vivo as it can penetrate most tissues. The goal of this study was to develop an oncolytic adenovirus (OAd)-resistant human triple-negative breast cancer (TNBC) that could express luciferase. Thus, when combining an OAd with chemotherapies or targeted therapies, we would be able to monitor the ability of these compounds to enhance OAd antitumor efficacy using BL in real time. The TNBC cell line HCC1937 was stably transfected with the plasmid pGL4.50[luc2/CMV/Hygro] (HCC1937/luc2). Once established, HCC1937/luc2 was orthotopically implanted in the 4th mammary gland fat pad of NSG (non-obese diabetic severe combined immunodeficiency disease gamma) female mice. Bioluminescence imaging (BLI) revealed that the HCC1937/luc2 cell line developed orthotopic breast tumor and lung metastasis over time. However, the integration of luc plasmid modified the HCC1937 phenotype, making HCC1937/luc2 more sensitive to OAdmCherry compared to the parental cell line and blunting the interferon (IFN) antiviral response. Testing two additional luc cell lines revealed that this was not a universal response; however, proper controls would need to be evaluated, as the integration of luciferase could affect the cells' response to different treatments.
Collapse
Affiliation(s)
- Martin R. Ramos-Gonzalez
- Roy Blunt NextGen Precision Health Institute, University of Missouri, Columbia, MO 65211, USA; (M.R.R.-G.); (N.S.N.); (S.R.); (H.S.); (E.S.Y.)
- Ellis Fischel Cancer Center, School of Medicine, University of Missouri, Columbia, MO 65212, USA;
| | - Nagabhishek Sirpu Natesh
- Roy Blunt NextGen Precision Health Institute, University of Missouri, Columbia, MO 65211, USA; (M.R.R.-G.); (N.S.N.); (S.R.); (H.S.); (E.S.Y.)
- Department of Veterinary Medicine and Surgery, University of Missouri, Columbia, MO 65211, USA
| | - Satyanarayana Rachagani
- Roy Blunt NextGen Precision Health Institute, University of Missouri, Columbia, MO 65211, USA; (M.R.R.-G.); (N.S.N.); (S.R.); (H.S.); (E.S.Y.)
- Ellis Fischel Cancer Center, School of Medicine, University of Missouri, Columbia, MO 65212, USA;
- Department of Veterinary Medicine and Surgery, University of Missouri, Columbia, MO 65211, USA
| | - James Amos-Landgraf
- Ellis Fischel Cancer Center, School of Medicine, University of Missouri, Columbia, MO 65212, USA;
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO 65211, USA
| | - Haval Shirwan
- Roy Blunt NextGen Precision Health Institute, University of Missouri, Columbia, MO 65211, USA; (M.R.R.-G.); (N.S.N.); (S.R.); (H.S.); (E.S.Y.)
- Ellis Fischel Cancer Center, School of Medicine, University of Missouri, Columbia, MO 65212, USA;
- Department of Pediatrics, School of Medicine, University of Missouri, Columbia, MO 65211, USA
| | - Esma S. Yolcu
- Roy Blunt NextGen Precision Health Institute, University of Missouri, Columbia, MO 65211, USA; (M.R.R.-G.); (N.S.N.); (S.R.); (H.S.); (E.S.Y.)
- Ellis Fischel Cancer Center, School of Medicine, University of Missouri, Columbia, MO 65212, USA;
- Department of Pediatrics, School of Medicine, University of Missouri, Columbia, MO 65211, USA
| | - Jorge G. Gomez-Gutierrez
- Roy Blunt NextGen Precision Health Institute, University of Missouri, Columbia, MO 65211, USA; (M.R.R.-G.); (N.S.N.); (S.R.); (H.S.); (E.S.Y.)
- Ellis Fischel Cancer Center, School of Medicine, University of Missouri, Columbia, MO 65212, USA;
- Department of Pediatrics, School of Medicine, University of Missouri, Columbia, MO 65211, USA
| |
Collapse
|
6
|
Alahmari S, Schultz A, Albrecht J, Tagal V, Siddiqui Z, Prabhakaran S, El Naqa I, Anderson A, Heiser L, Andor N. Cell identity revealed by precise cell cycle state mapping links data modalities. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.04.610488. [PMID: 39282313 PMCID: PMC11398313 DOI: 10.1101/2024.09.04.610488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 09/21/2024]
Abstract
Several methods for cell cycle inference from sequencing data exist and are widely adopted. In contrast, methods for classification of cell cycle state from imaging data are scarce. We have for the first time integrated sequencing and imaging derived cell cycle pseudo-times for assigning 449 imaged cells to 693 sequenced cells at an average resolution of 3.4 and 2.4 cells for sequencing and imaging data respectively. Data integration revealed thousands of pathways and organelle features that are correlated with each other, including several previously known interactions and novel associations. The ability to assign the transcriptome state of a profiled cell to its closest living relative, which is still actively growing and expanding opens the door for genotype-phenotype mapping at single cell resolution forward in time.
Collapse
Affiliation(s)
- Saeed Alahmari
- Department of Computer Science, Najran University, Najran 66462, Saudi Arabia
| | - Andrew Schultz
- Department of Integrated Mathematical Oncology, H Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Jordan Albrecht
- Department of Integrated Mathematical Oncology, H Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Vural Tagal
- Department of Integrated Mathematical Oncology, H Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Zaid Siddiqui
- Department of Radiation Oncology, Baylor College of Medicine, Houston, TX, USA
| | - Sandhya Prabhakaran
- Department of Integrated Mathematical Oncology, H Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Issam El Naqa
- Department of Machine Learning, H Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Alexander Anderson
- Department of Integrated Mathematical Oncology, H Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Laura Heiser
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, OR, USA
| | - Noemi Andor
- Department of Integrated Mathematical Oncology, H Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| |
Collapse
|
7
|
Foulke JG, Chen L, Chang H, McManus CE, Tian F, Gu Z. Optimizing Ex Vivo CAR-T Cell-Mediated Cytotoxicity Assay through Multimodality Imaging. Cancers (Basel) 2024; 16:2497. [PMID: 39061136 PMCID: PMC11274748 DOI: 10.3390/cancers16142497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 07/02/2024] [Accepted: 07/06/2024] [Indexed: 07/28/2024] Open
Abstract
CAR-T cell-based therapies have demonstrated remarkable efficacy in treating malignant cancers, especially liquid tumors, and are increasingly being evaluated in clinical trials for solid tumors. With the FDA's initiative to advance alternative methods for drug discovery and development, full human ex vivo assays are increasingly essential for precision CAR-T development. However, prevailing ex vivo CAR-T cell-mediated cytotoxicity assays are limited by their use of radioactive materials, lack of real-time measurement, low throughput, and inability to automate, among others. To address these limitations, we optimized the assay using multimodality imaging methods, including bioluminescence, impedance tracking, phase contrast, and fluorescence, to track CAR-T cells co-cultured with CD19, CD20, and HER2 luciferase reporter cancer cells in real-time. Additionally, we varied the ratio of CAR-T cells to cancer cells to determine optimal cytotoxicity readouts. Our findings demonstrated that the CAR-T cell group effectively attacked cancer cells, and the optimized assay provided superior temporal and spatial precision measurements of ex vivo CAR-T killing of cancer cells, confirming the reliability, consistency, and high throughput of the optimized assay.
Collapse
Affiliation(s)
| | | | | | | | - Fang Tian
- American Type Culture Collection (ATCC), Manassas, VA 20110, USA
| | - Zhizhan Gu
- American Type Culture Collection (ATCC), Manassas, VA 20110, USA
| |
Collapse
|
8
|
Merolla A, Michetti C, Moschetta M, Vacca F, Ciano L, Emionite L, Astigiano S, Romei A, Horenkamp S, Berglund K, Gross RE, Cesca F, Colombo E, Benfenati F. A pH-sensitive closed-loop nanomachine to control hyperexcitability at the single neuron level. Nat Commun 2024; 15:5609. [PMID: 38965228 PMCID: PMC11224301 DOI: 10.1038/s41467-024-49941-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Accepted: 06/20/2024] [Indexed: 07/06/2024] Open
Abstract
Epilepsy affects 1% of the general population and 30% of patients are resistant to antiepileptic drugs. Although optogenetics is an efficient antiepileptic strategy, the difficulty of illuminating deep brain areas poses translational challenges. Thus, the search of alternative light sources is strongly needed. Here, we develop pH-sensitive inhibitory luminopsin (pHIL), a closed-loop chemo-optogenetic nanomachine composed of a luciferase-based light generator, a fluorescent sensor of intracellular pH (E2GFP), and an optogenetic actuator (halorhodopsin) for silencing neuronal activity. Stimulated by coelenterazine, pHIL experiences bioluminescence resonance energy transfer between luciferase and E2GFP which, under conditions of acidic pH, activates halorhodopsin. In primary neurons, pHIL senses the intracellular pH drop associated with hyperactivity and optogenetically aborts paroxysmal activity elicited by the administration of convulsants. The expression of pHIL in hippocampal pyramidal neurons is effective in decreasing duration and increasing latency of pilocarpine-induced tonic-clonic seizures upon in vivo coelenterazine administration, without affecting higher brain functions. The same treatment is effective in markedly decreasing seizure manifestations in a murine model of genetic epilepsy. The results indicate that pHIL represents a potentially promising closed-loop chemo-optogenetic strategy to treat drug-refractory epilepsy.
Collapse
Affiliation(s)
- Assunta Merolla
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Genova, Italy
- IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Caterina Michetti
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Genova, Italy
- Department of Experimental Medicine, University of Genova, Genova, Italy
| | - Matteo Moschetta
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Genova, Italy
- IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Francesca Vacca
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Genova, Italy
| | - Lorenzo Ciano
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Genova, Italy
- Department of Experimental Medicine, University of Genova, Genova, Italy
| | | | | | - Alessandra Romei
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Genova, Italy
| | - Simone Horenkamp
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Genova, Italy
| | - Ken Berglund
- Department of Neurosurgery, Emory University School of Medicine, Atlanta, GA, USA
| | - Robert E Gross
- Department of Neurosurgery, Emory University School of Medicine, Atlanta, GA, USA
| | - Fabrizia Cesca
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Genova, Italy.
- Department of Life Sciences, University of Trieste, Trieste, Italy.
| | - Elisabetta Colombo
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Genova, Italy.
- IRCCS Ospedale Policlinico San Martino, Genova, Italy.
| | - Fabio Benfenati
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Genova, Italy
- IRCCS Ospedale Policlinico San Martino, Genova, Italy
| |
Collapse
|
9
|
Ohlendorf R, Li N, Phi Van VD, Schwalm M, Ke Y, Dawson M, Jiang Y, Das S, Stallings B, Zheng WT, Jasanoff A. Imaging bioluminescence by detecting localized haemodynamic contrast from photosensitized vasculature. Nat Biomed Eng 2024; 8:775-786. [PMID: 38730257 PMCID: PMC12121613 DOI: 10.1038/s41551-024-01210-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 03/30/2024] [Indexed: 05/12/2024]
Abstract
Bioluminescent probes are widely used to monitor biomedically relevant processes and cellular targets in living animals. However, the absorption and scattering of visible light by tissue drastically limit the depth and resolution of the detection of luminescence. Here we show that bioluminescent sources can be detected with magnetic resonance imaging by leveraging the light-mediated activation of vascular cells expressing a photosensitive bacterial enzyme that causes the conversion of bioluminescent emission into local changes in haemodynamic contrast. In the brains of rats with photosensitized vasculature, we used magnetic resonance imaging to volumetrically map bioluminescent xenografts and cell populations virally transduced to express luciferase. Detecting bioluminescence-induced haemodynamic signals from photosensitized vasculature will extend the applications of bioluminescent probes.
Collapse
Affiliation(s)
- Robert Ohlendorf
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Max Planck Institute for Biological Cybernetics, Tubingen, Germany
| | - Nan Li
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Advanced Imaging Research Center and Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Valerie Doan Phi Van
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Miriam Schwalm
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Yuting Ke
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Miranda Dawson
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Ying Jiang
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Sayani Das
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Brenna Stallings
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Wen Ting Zheng
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Alan Jasanoff
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Department of Nuclear Science and Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
| |
Collapse
|
10
|
Islam MM, Rahman MF, Islam A, Afroz MS, Mamun MA, Rahman MM, Maniruzzaman M, Xu L, Sakamoto T, Takahashi Y, Sato T, Kahyo T, Setou M. Elucidating Gender-Specific Distribution of Imipramine, Chloroquine, and Their Metabolites in Mice Kidney Tissues through AP-MALDI-MSI. Int J Mol Sci 2024; 25:4840. [PMID: 38732055 PMCID: PMC11084644 DOI: 10.3390/ijms25094840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 04/24/2024] [Accepted: 04/26/2024] [Indexed: 05/13/2024] Open
Abstract
Knowledge of gender-specific drug distributions in different organs are of great importance for personalized medicine and reducing toxicity. However, such drug distributions have not been well studied. In this study, we investigated potential differences in the distribution of imipramine and chloroquine, as well as their metabolites, between male and female kidneys. Kidneys were collected from mice treated with imipramine or chloroquine and then subjected to atmospheric pressure matrix-assisted laser desorption ionization-mass spectrometry imaging (AP-MALDI-MSI). We observed differential distributions of the drugs and their metabolites between male and female kidneys. Imipramine showed prominent distributions in the cortex and medulla in male and female kidneys, respectively. Desipramine, one of the metabolites of imipramine, showed significantly higher (*** p < 0.001) distributions in the medulla of the male kidney compared to that of the female kidney. Chloroquine and its metabolites were accumulated in the pelvis of both male and female kidneys. Interestingly, they showed a characteristic distribution in the medulla of the female kidney, while almost no distributions were observed in the same areas of the male kidney. For the first time, our study revealed that the distributions of imipramine, chloroquine, and their metabolites were different in male and female kidneys.
Collapse
Affiliation(s)
- Md. Monirul Islam
- Department of Cellular and Molecular Anatomy, Hamamatsu University School of Medicine, 1-20-1 Handayama, Chuo-Ku, Hamamatsu City 431-3192, Shizuoka, Japan; (M.M.I.); (M.F.R.); (A.I.); (M.S.A.); (M.A.M.); (T.S.); (Y.T.); (T.S.); (T.K.)
- Institute of Food and Radiation Biology, Atomic Energy Research Establishment, Bangladesh Atomic Energy Commission, Dhaka 1349, Bangladesh
| | - Md Foyzur Rahman
- Department of Cellular and Molecular Anatomy, Hamamatsu University School of Medicine, 1-20-1 Handayama, Chuo-Ku, Hamamatsu City 431-3192, Shizuoka, Japan; (M.M.I.); (M.F.R.); (A.I.); (M.S.A.); (M.A.M.); (T.S.); (Y.T.); (T.S.); (T.K.)
| | - Ariful Islam
- Department of Cellular and Molecular Anatomy, Hamamatsu University School of Medicine, 1-20-1 Handayama, Chuo-Ku, Hamamatsu City 431-3192, Shizuoka, Japan; (M.M.I.); (M.F.R.); (A.I.); (M.S.A.); (M.A.M.); (T.S.); (Y.T.); (T.S.); (T.K.)
- Department of Biochemistry and Microbiology, School of Health and Life Sciences, North South University, Bashundhara, Dhaka 1229, Bangladesh
- Preppers Co., Ltd., Hamamatsu City 431-3192, Shizuoka, Japan
| | - Mst. Sayela Afroz
- Department of Cellular and Molecular Anatomy, Hamamatsu University School of Medicine, 1-20-1 Handayama, Chuo-Ku, Hamamatsu City 431-3192, Shizuoka, Japan; (M.M.I.); (M.F.R.); (A.I.); (M.S.A.); (M.A.M.); (T.S.); (Y.T.); (T.S.); (T.K.)
| | - Md. Al Mamun
- Department of Cellular and Molecular Anatomy, Hamamatsu University School of Medicine, 1-20-1 Handayama, Chuo-Ku, Hamamatsu City 431-3192, Shizuoka, Japan; (M.M.I.); (M.F.R.); (A.I.); (M.S.A.); (M.A.M.); (T.S.); (Y.T.); (T.S.); (T.K.)
- Preppers Co., Ltd., Hamamatsu City 431-3192, Shizuoka, Japan
| | - Md. Muedur Rahman
- Department of Cellular and Molecular Anatomy, Hamamatsu University School of Medicine, 1-20-1 Handayama, Chuo-Ku, Hamamatsu City 431-3192, Shizuoka, Japan; (M.M.I.); (M.F.R.); (A.I.); (M.S.A.); (M.A.M.); (T.S.); (Y.T.); (T.S.); (T.K.)
- Preppers Co., Ltd., Hamamatsu City 431-3192, Shizuoka, Japan
| | - Md Maniruzzaman
- Department of Cellular and Molecular Anatomy, Hamamatsu University School of Medicine, 1-20-1 Handayama, Chuo-Ku, Hamamatsu City 431-3192, Shizuoka, Japan; (M.M.I.); (M.F.R.); (A.I.); (M.S.A.); (M.A.M.); (T.S.); (Y.T.); (T.S.); (T.K.)
| | - Lili Xu
- Department of Cellular and Molecular Anatomy, Hamamatsu University School of Medicine, 1-20-1 Handayama, Chuo-Ku, Hamamatsu City 431-3192, Shizuoka, Japan; (M.M.I.); (M.F.R.); (A.I.); (M.S.A.); (M.A.M.); (T.S.); (Y.T.); (T.S.); (T.K.)
| | - Takumi Sakamoto
- Department of Cellular and Molecular Anatomy, Hamamatsu University School of Medicine, 1-20-1 Handayama, Chuo-Ku, Hamamatsu City 431-3192, Shizuoka, Japan; (M.M.I.); (M.F.R.); (A.I.); (M.S.A.); (M.A.M.); (T.S.); (Y.T.); (T.S.); (T.K.)
- Preppers Co., Ltd., Hamamatsu City 431-3192, Shizuoka, Japan
| | - Yutaka Takahashi
- Department of Cellular and Molecular Anatomy, Hamamatsu University School of Medicine, 1-20-1 Handayama, Chuo-Ku, Hamamatsu City 431-3192, Shizuoka, Japan; (M.M.I.); (M.F.R.); (A.I.); (M.S.A.); (M.A.M.); (T.S.); (Y.T.); (T.S.); (T.K.)
- Preppers Co., Ltd., Hamamatsu City 431-3192, Shizuoka, Japan
- International Mass Imaging Center, Hamamatsu University School of Medicine, 1-20-1 Handayama, Chuo-Ku, Hamamatsu City 431-3192, Shizuoka, Japan
| | - Tomohito Sato
- Department of Cellular and Molecular Anatomy, Hamamatsu University School of Medicine, 1-20-1 Handayama, Chuo-Ku, Hamamatsu City 431-3192, Shizuoka, Japan; (M.M.I.); (M.F.R.); (A.I.); (M.S.A.); (M.A.M.); (T.S.); (Y.T.); (T.S.); (T.K.)
- International Mass Imaging Center, Hamamatsu University School of Medicine, 1-20-1 Handayama, Chuo-Ku, Hamamatsu City 431-3192, Shizuoka, Japan
| | - Tomoaki Kahyo
- Department of Cellular and Molecular Anatomy, Hamamatsu University School of Medicine, 1-20-1 Handayama, Chuo-Ku, Hamamatsu City 431-3192, Shizuoka, Japan; (M.M.I.); (M.F.R.); (A.I.); (M.S.A.); (M.A.M.); (T.S.); (Y.T.); (T.S.); (T.K.)
- International Mass Imaging Center, Hamamatsu University School of Medicine, 1-20-1 Handayama, Chuo-Ku, Hamamatsu City 431-3192, Shizuoka, Japan
| | - Mitsutoshi Setou
- Department of Cellular and Molecular Anatomy, Hamamatsu University School of Medicine, 1-20-1 Handayama, Chuo-Ku, Hamamatsu City 431-3192, Shizuoka, Japan; (M.M.I.); (M.F.R.); (A.I.); (M.S.A.); (M.A.M.); (T.S.); (Y.T.); (T.S.); (T.K.)
- Preppers Co., Ltd., Hamamatsu City 431-3192, Shizuoka, Japan
- International Mass Imaging Center, Hamamatsu University School of Medicine, 1-20-1 Handayama, Chuo-Ku, Hamamatsu City 431-3192, Shizuoka, Japan
- Department of Systems Molecular Anatomy, Institute for Medical Photonics Research, Preeminent Medical Photonics Education and Research Center, 1-20-1 Handayama, Chuo-Ku, Hamamatsu City 431-3192, Shizuoka, Japan
| |
Collapse
|
11
|
Haider MT, Freytag V, Krause L, Spethmann T, Gosau T, Beine MC, Knies C, Schröder-Schwarz J, Horn M, Riecken K, Lange T. Comparison of ex vivo bioluminescence imaging, Alu-qPCR and histology for the quantification of spontaneous lung and bone metastases in subcutaneous xenograft mouse models. Clin Exp Metastasis 2024; 41:103-115. [PMID: 38353934 PMCID: PMC10972982 DOI: 10.1007/s10585-024-10268-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 01/16/2024] [Indexed: 03/28/2024]
Abstract
Bioluminescence imaging (BLI) is a non-invasive state-of-the-art-method for longitudinal tracking of tumor cells in mice. The technique is commonly used to determine bone metastatic burden in vivo and also suitable ex vivo to detect even smallest bone micro-metastases in spontaneous metastasis xenograft models. However, it is unclear to which extent ex vivo BLI correlates with alternative methods for metastasis quantification. Here, we compared ex vivo BLI, human DNA-based Alu-qPCR, and histology for the quantification of bone vs. lung metastases, which are amongst the most common sites of metastasis in prostate cancer (PCa) patients and spontaneous PCa xenograft models. Data from 93 immunodeficient mice were considered, each of which were subcutaneously injected with luciferase/RGB-labeled human PCa PC-3 cells. The primary tumors were resected at ~ 0.75 cm³ and mice were sacrificed ~ 3 weeks after surgery and immediately examined by ex vivo BLI. Afterwards, the right lungs and hind limbs with the higher BLI signal (BLIHi bone) were processed for histology, whereas the left lung lobes and hind limbs with the lower BLI signal (BLILo bone) were prepared for Alu-qPCR. Our data demonstrate remarkable differences in the correlation coefficients of the different methods for lung metastasis detection (r ~ 0.8) vs. bone metastasis detection (r ~ 0.4). However, the BLI values of the BLIHi and BLILo bones correlated very strongly (r ~ 0.9), indicating that the method per se was reliable under identical limitations; the overall level of metastasis to contralateral bones was astonishingly similar. Instead, the level of lung metastasis only weakly to moderately correlated with the level of bone metastasis formation. Summarized, we observed a considerable discrepancy between ex vivo BLI and histology/Alu-qPCR in the quantification of bone metastases, which was not observed in the case of lung metastases. Future studies using ex vivo BLI for bone metastasis quantification should combine multiple methods to accurately determine metastatic load in bone samples.
Collapse
Affiliation(s)
- Marie-Therese Haider
- Institute of Anatomy and Experimental Morphology, University Medical Center Hamburg- Eppendorf, Martinistrasse 52, Hamburg, 20246, Germany
| | - Vera Freytag
- Institute of Anatomy and Experimental Morphology, University Medical Center Hamburg- Eppendorf, Martinistrasse 52, Hamburg, 20246, Germany
| | - Linda Krause
- Institute of Medical Biometry and Epidemiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Tanja Spethmann
- Institute of Anatomy and Experimental Morphology, University Medical Center Hamburg- Eppendorf, Martinistrasse 52, Hamburg, 20246, Germany
| | - Tobias Gosau
- Institute of Anatomy and Experimental Morphology, University Medical Center Hamburg- Eppendorf, Martinistrasse 52, Hamburg, 20246, Germany
| | - Mia C Beine
- Institute of Anatomy and Experimental Morphology, University Medical Center Hamburg- Eppendorf, Martinistrasse 52, Hamburg, 20246, Germany
| | - Christine Knies
- Institute of Anatomy and Experimental Morphology, University Medical Center Hamburg- Eppendorf, Martinistrasse 52, Hamburg, 20246, Germany
| | - Jennifer Schröder-Schwarz
- Institute of Anatomy and Experimental Morphology, University Medical Center Hamburg- Eppendorf, Martinistrasse 52, Hamburg, 20246, Germany
| | - Michael Horn
- Core Facility In Vivo Optical Imaging, University Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Mildred Scheel Cancer Career Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Kristoffer Riecken
- Research Department Cell and Gene Therapy, Department of Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Tobias Lange
- Institute of Anatomy and Experimental Morphology, University Medical Center Hamburg- Eppendorf, Martinistrasse 52, Hamburg, 20246, Germany.
- Institute of Anatomy I, University Hospital Jena, Teichgraben 7, Jena, 07743, Germany.
- Comprehensive Cancer Center Central Germany (CCCG), Ulm, Germany.
| |
Collapse
|
12
|
Porta-de-la-Riva M, Morales-Curiel LF, Carolina Gonzalez A, Krieg M. Bioluminescence as a functional tool for visualizing and controlling neuronal activity in vivo. NEUROPHOTONICS 2024; 11:024203. [PMID: 38348359 PMCID: PMC10861157 DOI: 10.1117/1.nph.11.2.024203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 01/18/2024] [Accepted: 01/19/2024] [Indexed: 02/15/2024]
Abstract
The use of bioluminescence as a reporter for physiology in neuroscience is as old as the discovery of the calcium-dependent photon emission of aequorin. Over the years, luciferases have been largely replaced by fluorescent reporters, but recently, the field has seen a renaissance of bioluminescent probes, catalyzed by unique developments in imaging technology, bioengineering, and biochemistry to produce luciferases with previously unseen colors and intensity. This is not surprising as the advantages of bioluminescence make luciferases very attractive for noninvasive, longitudinal in vivo observations without the need of an excitation light source. Here, we review how the development of dedicated and specific sensor-luciferases afforded, among others, transcranial imaging of calcium and neurotransmitters, or cellular metabolites and physical quantities such as forces and membrane voltage. Further, the increased versatility and light output of luciferases have paved the way for a new field of functional bioluminescence optogenetics, in which the photon emission of the luciferase is coupled to the gating of a photosensor, e.g., a channelrhodopsin and we review how they have been successfully used to engineer synthetic neuronal connections. Finally, we provide a primer to consider important factors in setting up functional bioluminescence experiments, with a particular focus on the genetic model Caenorhabditis elegans, and discuss the leading challenges that the field needs to overcome to regain a competitive advantage over fluorescence modalities. Together, our paper caters to experienced users of bioluminescence as well as novices who would like to experience the advantages of luciferases in their own hand.
Collapse
Affiliation(s)
- Montserrat Porta-de-la-Riva
- ICFO—Institut de Ciències Fotòniques, The Barcelona Institute of Science and Technology, Castelldefels, Barcelona, Spain
| | - Luis-Felipe Morales-Curiel
- ICFO—Institut de Ciències Fotòniques, The Barcelona Institute of Science and Technology, Castelldefels, Barcelona, Spain
| | - Adriana Carolina Gonzalez
- ICFO—Institut de Ciències Fotòniques, The Barcelona Institute of Science and Technology, Castelldefels, Barcelona, Spain
| | - Michael Krieg
- ICFO—Institut de Ciències Fotòniques, The Barcelona Institute of Science and Technology, Castelldefels, Barcelona, Spain
| |
Collapse
|
13
|
Klein E, Marsh S, Becker J, Andermann M, Lehtinen M, Moore CI. BioLuminescent OptoGenetics in the choroid plexus: integrated opto- and chemogenetic control in vivo. NEUROPHOTONICS 2024; 11:024210. [PMID: 38948888 PMCID: PMC11213259 DOI: 10.1117/1.nph.11.2.024210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 05/30/2024] [Accepted: 05/30/2024] [Indexed: 07/02/2024]
Abstract
Significance The choroid plexus (ChP) epithelial network displays diverse dynamics, including propagating calcium waves and individuated fluctuations in single cells. These rapid events underscore the possibility that ChP dynamics may reflect behaviorally relevant and clinically important changes in information processing and signaling. Optogenetic and chemogenetic tools provide spatiotemporally precise and sustained approaches for testing such dynamics in vivo. Here, we describe the feasibility of a novel combined opto- and chemogenetic tool, BioLuminescent-OptoGenetics (BL-OG), for the ChP in vivo. In the "LuMinOpsin" (LMO) BL-OG strategy, a luciferase is tethered to an adjacent optogenetic element. This molecule allows chemogenetic activation when the opsin is driven by light produced through luciferase binding a small molecule (luciferin) or by conventional optogenetic light sources and BL-OG report of activation through light production. Aim To test the viability of BL-OG/LMO for ChP control. Approach Using transgenic and Cre-directed targeting to the ChP, we expressed LMO3 (a Gaussia luciferase-VChR1 fusion), a highly effective construct in neural systems. In mice expressing LMO3 in ChP, we directly imaged BL light production following multiple routes of coelenterazine (CTZ: luciferin) administration using an implanted cannula system. We also used home-cage videography with Deep LabCut analysis to test for any impact of repeated CTZ administration on basic health and behavioral indices. Results Multiple routes of CTZ administration drove BL photon production, including intracerebroventricular, intravenous, and intraperitoneal injection. Intravenous administration resulted in fast "flash" kinetics that diminished in seconds to minutes, and intraperitoneal administration resulted in slow rising activity that sustained hours. Mice showed no consistent impact of 1 week of intraperitoneal CTZ administration on weight, drinking, motor behavior, or sleep/wake cycles. Conclusions BL-OG/LMO provides unique advantages for testing the role of ChP dynamics in biological processes.
Collapse
Affiliation(s)
- Eric Klein
- Brown University, Providence, Rhode Island, United States
| | - Sophie Marsh
- Brown University, Providence, Rhode Island, United States
| | - Jordan Becker
- Brown University, Providence, Rhode Island, United States
| | - Mark Andermann
- Beth Israel Deaconess Medical Center Harvard, Boston, Massachusetts, United States
| | - Maria Lehtinen
- Brown University, Providence, Rhode Island, United States
- Boston Children’s Hospital, Boston, Massachusetts, United States
| | | |
Collapse
|
14
|
Torres Chavez AG, McKenna MK, Balasubramanian K, Riffle L, Patel NL, Kalen JD, St. Croix B, Leen AM, Bajgain P. A dual-luciferase bioluminescence system for the assessment of cellular therapies. MOLECULAR THERAPY. ONCOLOGY 2024; 32:200763. [PMID: 38596291 PMCID: PMC10869576 DOI: 10.1016/j.omton.2024.200763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 12/17/2023] [Accepted: 01/05/2024] [Indexed: 04/11/2024]
Abstract
Bioluminescence imaging is a well-established platform for evaluating engineered cell therapies in preclinical studies. However, despite the discovery of new luciferases and substrates, optimal combinations to simultaneously monitor two cell populations remain limited. This makes the functional assessment of cellular therapies cumbersome and expensive, especially in preclinical in vivo models. In this study, we explored the potential of using a green bioluminescence-emitting click beetle luciferase, CBG99, and a red bioluminescence-emitting firefly luciferase mutant, Akaluc, together to simultaneously monitor two cell populations. Using various chimeric antigen receptor T cells and tumor pairings, we demonstrate that these luciferases are suitable for real-time tracking of two cell types using 2D and 3D cultures in vitro and experimental models in vivo. Our data show the broad compatibility of this dual-luciferase (duo-luc) system with multiple bioluminescence detection equipment ranging from benchtop spectrophotometers to live animal imaging systems. Although this study focused on investigating complex CAR T cells and tumor cell interactions, this duo-luc system has potential utility for the simultaneous monitoring of any two cellular components-for example, to unravel the impact of a specific genetic variant on clonal dominance in a mixed population of tumor cells.
Collapse
Affiliation(s)
| | - Mary K. McKenna
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX 77030, USA
| | | | - Lisa Riffle
- Small Animal Imaging Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Frederick, MD 21702, USA
| | - Nimit L. Patel
- Small Animal Imaging Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Frederick, MD 21702, USA
| | - Joseph D. Kalen
- Small Animal Imaging Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Frederick, MD 21702, USA
| | - Brad St. Croix
- Tumor Angiogenesis Unit, Mouse Cancer Genetics Program, Center for Cancer Research, National Cancer Institute, Frederick, MD 21702, USA
| | - Ann M. Leen
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX 77030, USA
| | - Pradip Bajgain
- Tumor Angiogenesis Unit, Mouse Cancer Genetics Program, Center for Cancer Research, National Cancer Institute, Frederick, MD 21702, USA
| |
Collapse
|
15
|
Goker S, Lee S, Hong CI. Monitoring Circadian Oscillations with a Bioluminescence Reporter in Organoids. J Vis Exp 2024:10.3791/66381. [PMID: 38436407 PMCID: PMC11551969 DOI: 10.3791/66381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/05/2024] Open
Abstract
Most living organisms possess circadian rhythms, which are biological processes that occur within a period of approximately 24 h and regulate a diverse repertoire of cellular and physiological processes ranging from sleep-wake cycles to metabolism. This clock mechanism entrains the organism based on environmental changes and coordinates the temporal regulation of molecular and physiological events. Previously, it was demonstrated that autonomous circadian rhythms are maintained even at the single-cell level using cell lines such as NIH3T3 fibroblasts, which were instrumental in uncovering the mechanisms of circadian rhythms. However, these cell lines are homogeneous cultures lacking multicellularity and robust intercellular communications. In the past decade, extensive work has been performed on the development, characterization, and application of 3D organoids, which are in vitro multicellular systems that resemble in vivo morphological structures and functions. This paper describes a protocol for detecting circadian rhythms using a bioluminescent reporter in human intestinal enteroids, which enables the investigation of circadian rhythms in multicellular systems in vitro.
Collapse
Affiliation(s)
- Sevde Goker
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine
| | - Suengwon Lee
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine
| | - Christian I Hong
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine;
| |
Collapse
|
16
|
Fu Q, Yang X, Wang M, Zhu K, Wang Y, Song J. Activatable Probes for Ratiometric Imaging of Endogenous Biomarkers In Vivo. ACS NANO 2024; 18:3916-3968. [PMID: 38258800 DOI: 10.1021/acsnano.3c10659] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
Dynamic variations in the concentration and abnormal distribution of endogenous biomarkers are strongly associated with multiple physiological and pathological states. Therefore, it is crucial to design imaging systems capable of real-time detection of dynamic changes in biomarkers for the accurate diagnosis and effective treatment of diseases. Recently, ratiometric imaging has emerged as a widely used technique for sensing and imaging of biomarkers due to its advantage of circumventing the limitations inherent to conventional intensity-dependent signal readout methods while also providing built-in self-calibration for signal correction. Here, the recent progress of ratiometric probes and their applications in sensing and imaging of biomarkers are outlined. Ratiometric probes are classified according to their imaging mechanisms, and ratiometric photoacoustic imaging, ratiometric optical imaging including photoluminescence imaging and self-luminescence imaging, ratiometric magnetic resonance imaging, and dual-modal ratiometric imaging are discussed. The applications of ratiometric probes in the sensing and imaging of biomarkers such as pH, reactive oxygen species (ROS), reactive nitrogen species (RNS), glutathione (GSH), gas molecules, enzymes, metal ions, and hypoxia are discussed in detail. Additionally, this Review presents an overview of challenges faced in this field along with future research directions.
Collapse
Affiliation(s)
- Qinrui Fu
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, Shandong 266021, China
| | - Xiao Yang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, Shandong 266021, China
| | - Mengzhen Wang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, Shandong 266021, China
| | - Kang Zhu
- State Key Laboratory of Chemical Resource Engineering, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Yin Wang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, Shandong 266021, China
| | - Jibin Song
- State Key Laboratory of Chemical Resource Engineering, Beijing University of Chemical Technology, Beijing, 100029, China
| |
Collapse
|
17
|
Heath NG, Segal DJ. CRISPR-Based Split Luciferase as a Biosensor for Unique DNA Sequences In Situ. Methods Mol Biol 2024; 2784:285-299. [PMID: 38502493 DOI: 10.1007/978-1-0716-3766-1_19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/21/2024]
Abstract
To date, CRISPR-based DNA targeting approaches have typically used fusion proteins between full fluorescent reporters and catalytically inactive Cas9 (dCas9) for imaging rather than detection of endogenous genomic DNA sequences. A promising alternative strategy for DNA targeting is the direct biosensing of user-defined sequences at single copy with single-cell resolution. Our recently described DNA biosensing approach using a dual fusion protein biosensor comprised of two independently optimized fragments of NanoLuc luciferase (NLuc) directionally fused to dCas9 paired with user-defined single-guide RNAs (sgRNAs) could allow users to sensitively detect unique copies of a target sequence in individual living cells using common laboratory equipment such as a microscope or a luminescence-equipped microplate reader. Here we describe a protocol for using such a DNA biosensor noninvasively in situ.
Collapse
Affiliation(s)
- Nicholas G Heath
- Genome Center and Department of Biochemistry and Molecular Medicine, University of California, Davis, Davis, CA, USA
- Integrative Genetics and Genomics, University of California, Davis, Davis, CA, USA
- Innovative Genomics Institute, University of California, Berkeley, CA, USA
| | - David J Segal
- Genome Center and Department of Biochemistry and Molecular Medicine, University of California, Davis, Davis, CA, USA.
- Integrative Genetics and Genomics, University of California, Davis, Davis, CA, USA.
- Innovative Genomics Institute, University of California, Berkeley, CA, USA.
| |
Collapse
|
18
|
Araújo-Gomes N, Zambito G, Johnbosco C, Calejo I, Leijten J, Löwik C, Karperien M, Mezzanotte L, Teixeira LM. Bioluminescence imaging on-chip platforms for non-invasive high-content bioimaging. Biosens Bioelectron 2023; 237:115510. [PMID: 37442028 DOI: 10.1016/j.bios.2023.115510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 06/09/2023] [Accepted: 07/01/2023] [Indexed: 07/15/2023]
Abstract
Incorporating non-invasive biosensing features in organ-on-chip models is of paramount importance for a wider implementation of these advanced in vitro microfluidic platforms. Optical biosensors, based on Bioluminescence Imaging (BLI), enable continuous, non-invasive, and in-situ imaging of cells, tissues or miniaturized organs without the drawbacks of conventional fluorescence imaging. Here, we report the first-of-its-kind integration and optimization of BLI in microfluidic chips, for non-invasive imaging of multiple biological readouts. The cell line HEK293T-GFP was engineered to express NanoLuc® luciferase under the control of a constitutive promoter and were cultured on-chip in 3D, in standard ECM-like hydrogels, to assess optimal cell detection conditions. Using real-time in-vitro dual-color microscopy, Bioluminescence (BL) and fluorescence (FL) were detectable using distinct imaging setups. Detection of the bioluminescent signals were observed at single cell resolution on-chip 20 min post-addition of Furimazine substrate and under perfusion. All hydrogels enabled BLI with higher signal-to-noise ratios as compared to fluorescence. For instance, agarose gels showed a ∼5-fold greater BL signal over background after injection of the substrate as compared to the FL signal. The use of BLI with microfluidic chip technologies opens up the potential for simultaneous in situ detection with continuous monitoring of multicolor cell reporters. Moreover, this can be achieved in a non-invasive manner. BL has great promise as a highly desirable biosensor for studying organ-on-chip platforms.
Collapse
Affiliation(s)
- Nuno Araújo-Gomes
- Department of Developmental Bioengineering, Technical Medical Centre, University of Twente, Enschede, the Netherlands
| | - Giorgia Zambito
- Department of Radiology and Nuclear Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands; Department of Molecular Genetics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Castro Johnbosco
- Department of Developmental Bioengineering, Technical Medical Centre, University of Twente, Enschede, the Netherlands
| | - Isabel Calejo
- Department of Developmental Bioengineering, Technical Medical Centre, University of Twente, Enschede, the Netherlands
| | - Jeroen Leijten
- Department of Developmental Bioengineering, Technical Medical Centre, University of Twente, Enschede, the Netherlands
| | - Clemens Löwik
- Department of Radiology and Nuclear Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands; Department of Molecular Genetics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Marcel Karperien
- Department of Developmental Bioengineering, Technical Medical Centre, University of Twente, Enschede, the Netherlands
| | - Laura Mezzanotte
- Department of Radiology and Nuclear Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands; Department of Molecular Genetics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands.
| | - Liliana Moreira Teixeira
- Department of Advanced Organ Bioengineering and Therapeutics, University of Twente, Enschede, the Netherlands.
| |
Collapse
|
19
|
Hersh J, Yang YP, Roberts E, Bilbao D, Tao W, Pollack A, Daunert S, Deo SK. Targeted Bioluminescent Imaging of Pancreatic Ductal Adenocarcinoma Using Nanocarrier-Complexed EGFR-Binding Affibody-Gaussia Luciferase Fusion Protein. Pharmaceutics 2023; 15:1976. [PMID: 37514162 PMCID: PMC10384630 DOI: 10.3390/pharmaceutics15071976] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 07/14/2023] [Accepted: 07/17/2023] [Indexed: 07/30/2023] Open
Abstract
In vivo imaging has enabled impressive advances in biological research, both preclinical and clinical, and researchers have an arsenal of imaging methods available. Bioluminescence imaging is an advantageous method for in vivo studies that allows for the simple acquisition of images with low background signals. Researchers have increasingly been looking for ways to improve bioluminescent imaging for in vivo applications, which we sought to achieve by developing a bioluminescent probe that could specifically target cells of interest. We chose pancreatic ductal adenocarcinoma (PDAC) as the disease model because it is the most common type of pancreatic cancer and has an extremely low survival rate. We targeted the epidermal growth factor receptor (EGFR), which is frequently overexpressed in pancreatic cancer cells, using an EGFR-specific affibody to selectively identify PDAC cells and delivered a Gaussia luciferase (GLuc) bioluminescent protein for imaging by engineering a fusion protein with both the affibody and the bioluminescent protein. This fusion protein was then complexed with a G5-PAMAM dendrimer nanocarrier. The dendrimer was used to improve the protein stability in vivo and increase signal strength. Our targeted bioluminescent complex had an enhanced uptake into PDAC cells in vitro and localized to PDAC tumors in vivo in pancreatic cancer xenograft mice. The bioluminescent complexes could delineate the tumor shape, identify multiple masses, and locate metastases. Through this work, an EGFR-targeted bioluminescent-dendrimer complex enabled the straightforward identification and imaging of pancreatic cancer cells in vivo in preclinical models. This argues for the targeted nanocarrier-mediated delivery of bioluminescent proteins as a way to improve in vivo bioluminescent imaging.
Collapse
Affiliation(s)
- Jessica Hersh
- Department of Biochemistry & Molecular Biology, Leonard M. Miller School of Medicine, University of Miami, Miami, FL 33136, USA; (J.H.); (Y.-P.Y.); (S.D.)
- The Dr. John T. McDonald Foundation Bionanotechnology Institute, University of Miami, Miami, FL 33136, USA
- Sylvester Comprehensive Cancer Center, Leonard M. Miller School of Medicine, University of Miami, Miami, FL 33136, USA; (E.R.); (D.B.); (W.T.); (A.P.)
| | - Yu-Ping Yang
- Department of Biochemistry & Molecular Biology, Leonard M. Miller School of Medicine, University of Miami, Miami, FL 33136, USA; (J.H.); (Y.-P.Y.); (S.D.)
- The Dr. John T. McDonald Foundation Bionanotechnology Institute, University of Miami, Miami, FL 33136, USA
- Sylvester Comprehensive Cancer Center, Leonard M. Miller School of Medicine, University of Miami, Miami, FL 33136, USA; (E.R.); (D.B.); (W.T.); (A.P.)
| | - Evan Roberts
- Sylvester Comprehensive Cancer Center, Leonard M. Miller School of Medicine, University of Miami, Miami, FL 33136, USA; (E.R.); (D.B.); (W.T.); (A.P.)
| | - Daniel Bilbao
- Sylvester Comprehensive Cancer Center, Leonard M. Miller School of Medicine, University of Miami, Miami, FL 33136, USA; (E.R.); (D.B.); (W.T.); (A.P.)
- Department of Pathology and Laboratory Medicine, Leonard M. Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Wensi Tao
- Sylvester Comprehensive Cancer Center, Leonard M. Miller School of Medicine, University of Miami, Miami, FL 33136, USA; (E.R.); (D.B.); (W.T.); (A.P.)
- Department of Radiation Oncology, Leonard M. Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Alan Pollack
- Sylvester Comprehensive Cancer Center, Leonard M. Miller School of Medicine, University of Miami, Miami, FL 33136, USA; (E.R.); (D.B.); (W.T.); (A.P.)
- Department of Radiation Oncology, Leonard M. Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Sylvia Daunert
- Department of Biochemistry & Molecular Biology, Leonard M. Miller School of Medicine, University of Miami, Miami, FL 33136, USA; (J.H.); (Y.-P.Y.); (S.D.)
- The Dr. John T. McDonald Foundation Bionanotechnology Institute, University of Miami, Miami, FL 33136, USA
- Sylvester Comprehensive Cancer Center, Leonard M. Miller School of Medicine, University of Miami, Miami, FL 33136, USA; (E.R.); (D.B.); (W.T.); (A.P.)
| | - Sapna K. Deo
- Department of Biochemistry & Molecular Biology, Leonard M. Miller School of Medicine, University of Miami, Miami, FL 33136, USA; (J.H.); (Y.-P.Y.); (S.D.)
- The Dr. John T. McDonald Foundation Bionanotechnology Institute, University of Miami, Miami, FL 33136, USA
- Sylvester Comprehensive Cancer Center, Leonard M. Miller School of Medicine, University of Miami, Miami, FL 33136, USA; (E.R.); (D.B.); (W.T.); (A.P.)
| |
Collapse
|
20
|
Costa AC, Alves PM, Monteiro FJ, Salgado C. Interactions between Dental MSCs and Biomimetic Composite Scaffold during Bone Remodeling Followed by In Vivo Real-Time Bioimaging. Int J Mol Sci 2023; 24:ijms24031827. [PMID: 36768151 PMCID: PMC9915259 DOI: 10.3390/ijms24031827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 01/06/2023] [Accepted: 01/14/2023] [Indexed: 01/18/2023] Open
Abstract
Oral-maxillofacial tumor removal can generate critical bone defects and major problems for patients, causing dysfunctionalities and affecting oral competencies such as mastication, swallowing, and breathing. The association of novel biomaterials and cell therapies in tissue engineering strategies could offer new strategies to promote osteomucosa healing. This study focused on the development of a bioengineered construct loaded with human dental follicle cells (MSCs). To increase the bioconstruct integration to the surrounding tissue, a novel and comprehensive approach was designed combining an injectable biomimetic hydrogel and dental stem cells (hDFMSCs) expressing luminescence/fluorescence for semi-quantitative tissue imaging in live animals. This in vivo model with human MSCs was based on an intramembranous bone regeneration process (IMO). Biologically, the biocomposite based on collagen/nanohydroxyapatite filled with cell-loaded osteopontin-fibrin hydrogel (Coll/nanoHA OPN-Fb) exhibited a high cellular proliferation rate, increased bone extracellular matrix deposition (osteopontin) and high ALP activity, indicating an early osteogenic differentiation. Thus, the presence of human OPN enhanced hDFMSC adhesion, migration, and spatial distribution within the 3D matrix. The developed 3D bioconstruct provided the necessary pro-regenerative effect to modulate the biological response, precisely fitting the bone defect with fine-tuned adjustment to the surrounding original structure and promoting oral osteomucosa tissue regeneration. We were also able to track the cells in vivo and evaluate their behavior (migration, proliferation, and differentiation), providing a glimpse into bone regeneration and helping in the optimization of patient-specific therapies.
Collapse
Affiliation(s)
- Ana Catarina Costa
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal
- Instituto Nacional de Engenharia Biomédica (INEB), Rua Alfredo Allen, 208, 4200-135 Porto, Portugal
- Faculdade de Engenharia, Universidade do Porto, Rua Dr. Roberto Frias, s/n, 4200-465 Porto, Portugal
| | - Patrícia Mafalda Alves
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal
- Instituto Nacional de Engenharia Biomédica (INEB), Rua Alfredo Allen, 208, 4200-135 Porto, Portugal
- Faculdade de Medicina Dentária, Universidade do Porto, Rua Dr. Manuel Pereira da Silva, 4200-393 Porto, Portugal
| | - Fernando Jorge Monteiro
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal
- Instituto Nacional de Engenharia Biomédica (INEB), Rua Alfredo Allen, 208, 4200-135 Porto, Portugal
- Faculdade de Engenharia, Universidade do Porto, Rua Dr. Roberto Frias, s/n, 4200-465 Porto, Portugal
- Porto Comprehensive Cancer Center (P.CCC), R. Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal
| | - Christiane Salgado
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal
- Instituto Nacional de Engenharia Biomédica (INEB), Rua Alfredo Allen, 208, 4200-135 Porto, Portugal
- Correspondence:
| |
Collapse
|
21
|
Morales-Curiel LF, Gonzalez AC, Castro-Olvera G, Lin LCL, El-Quessny M, Porta-de-la-Riva M, Severino J, Morera LB, Venturini V, Ruprecht V, Ramallo D, Loza-Alvarez P, Krieg M. Volumetric imaging of fast cellular dynamics with deep learning enhanced bioluminescence microscopy. Commun Biol 2022; 5:1330. [PMID: 36463346 PMCID: PMC9719505 DOI: 10.1038/s42003-022-04292-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 11/23/2022] [Indexed: 12/05/2022] Open
Abstract
Bioluminescence microscopy is an appealing alternative to fluorescence microscopy, because it does not depend on external illumination, and consequently does neither produce spurious background autofluorescence, nor perturb intrinsically photosensitive processes in living cells and animals. The low photon emission of known luciferases, however, demands long exposure times that are prohibitive for imaging fast biological dynamics. To increase the versatility of bioluminescence microscopy, we present an improved low-light microscope in combination with deep learning methods to image extremely photon-starved samples enabling subsecond exposures for timelapse and volumetric imaging. We apply our method to image subcellular dynamics in mouse embryonic stem cells, epithelial morphology during zebrafish development, and DAF-16 FoxO transcription factor shuttling from the cytoplasm to the nucleus under external stress. Finally, we concatenate neural networks for denoising and light-field deconvolution to resolve intracellular calcium dynamics in three dimensions of freely moving Caenorhabditis elegans.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Jacqueline Severino
- Center for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Laura Battle Morera
- Center for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Valeria Venturini
- Center for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Verena Ruprecht
- Center for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
- ICREA, Pg. Lluis Companys 23, 08010, Barcelona, Spain
| | - Diego Ramallo
- ICFO, Institut de Ciencies Fotòniques, Castelldefels, Spain
| | | | - Michael Krieg
- ICFO, Institut de Ciencies Fotòniques, Castelldefels, Spain.
| |
Collapse
|
22
|
Gregor C. Generation of bright autobioluminescent bacteria by chromosomal integration of the improved lux operon ilux2. Sci Rep 2022; 12:19039. [PMID: 36351939 PMCID: PMC9646698 DOI: 10.1038/s41598-022-22068-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 10/10/2022] [Indexed: 11/10/2022] Open
Abstract
The bacterial bioluminescence system enables the generation of light by living cells without the requirement of an external luciferin. Due to the relatively low light emission, many applications of bioluminescence imaging would benefit from an increase in brightness of this system. In this report, a new approach of mutagenesis and screening of the involved proteins is described that is based on the identification of mutants with improved properties under rate-limiting reaction conditions. Multiple rounds of screening in Escherichia coli resulted in the operon ilux2 that contains 26 new mutations in the fatty acid reductase complex which provides the aldehyde substrate for the bioluminescence reaction. Chromosomal integration of ilux2 yielded an autonomously bioluminescent E. coli strain with sixfold increased brightness compared to the previously described ilux operon. The ilux2 strain produces sufficient signal for the robust detection of individual cells and enables highly sensitive long-term imaging of bacterial propagation without a selection marker.
Collapse
Affiliation(s)
- Carola Gregor
- Department of NanoBiophotonics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany.
- Department of Optical Nanoscopy, Institut für Nanophotonik Göttingen e.V., Göttingen, Germany.
- Cluster of Excellence "Multiscale Bioimaging: From Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Göttingen, Germany.
| |
Collapse
|
23
|
Improved Locomotor Recovery in a Rat Model of Spinal Cord Injury by BioLuminescent-OptoGenetic (BL-OG) Stimulation with an Enhanced Luminopsin. Int J Mol Sci 2022; 23:ijms232112994. [PMID: 36361784 PMCID: PMC9656028 DOI: 10.3390/ijms232112994] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 10/09/2022] [Accepted: 10/19/2022] [Indexed: 11/27/2022] Open
Abstract
Irrespective of the many strategies focused on dealing with spinal cord injury (SCI), there is still no way to restore motor function efficiently or an adequate regenerative therapy. One promising method that could potentially prove highly beneficial for rehabilitation in patients is to re-engage specific neuronal populations of the spinal cord following SCI. Targeted activation may maintain and strengthen existing neuronal connections and/or facilitate the reorganization and development of new connections. BioLuminescent-OptoGenetics (BL-OG) presents an avenue to non-invasively and specifically stimulate neurons; genetically targeted neurons express luminopsins (LMOs), light-emitting luciferases tethered to light-sensitive channelrhodopsins that are activated by adding the luciferase substrate coelenterazine (CTZ). This approach employs ion channels for current conduction while activating the channels through treatment with the small molecule CTZ, thus allowing non-invasive stimulation of all targeted neurons. We previously showed the efficacy of this approach for improving locomotor recovery following severe spinal cord contusion injury in rats expressing the excitatory luminopsin 3 (LMO3) under control of a pan-neuronal and motor-neuron-specific promoter with CTZ applied through a lateral ventricle cannula. The goal of the present study was to test a new generation of LMOs based on opsins with higher light sensitivity which will allow for peripheral delivery of the CTZ. In this construct, the slow-burn Gaussia luciferase variant (sbGLuc) is fused to the opsin CheRiff, creating LMO3.2. Taking advantage of the high light sensitivity of this opsin, we stimulated transduced lumbar neurons after thoracic SCI by intraperitoneal application of CTZ, allowing for a less invasive treatment. The efficacy of this non-invasive BioLuminescent-OptoGenetic approach was confirmed by improved locomotor function. This study demonstrates that peripheral delivery of the luciferin CTZ can be used to activate LMOs expressed in spinal cord neurons that employ an opsin with increased light sensitivity.
Collapse
|
24
|
Torio EA, Ressler VT, Kincaid VA, Hurst R, Hall MP, Encell LP, Zimmerman K, Forsyth SK, Rehrauer WM, Accola MA, Hsu CC, Machleidt T, Dart ML. Development of a rapid, simple, and sensitive point-of-care technology platform utilizing ternary NanoLuc. Front Microbiol 2022; 13:970233. [PMID: 36386626 PMCID: PMC9643700 DOI: 10.3389/fmicb.2022.970233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 10/06/2022] [Indexed: 12/02/2022] Open
Abstract
Point-of-care tests are highly valuable in providing fast results for medical decisions for greater flexibility in patient care. Many diagnostic tests, such as ELISAs, that are commonly used within clinical laboratory settings require trained technicians, laborious workflows, and complex instrumentation hindering their translation into point-of-care applications. Herein, we demonstrate the use of a homogeneous, bioluminescent-based, split reporter platform that enables a simple, sensitive, and rapid method for analyte detection in clinical samples. We developed this point-of-care application using an optimized ternary, split-NanoLuc luciferase reporter system that consists of two small reporter peptides added as appendages to analyte-specific affinity reagents. A bright, stable bioluminescent signal is generated as the affinity reagents bind to the analyte, allowing for proximity-induced complementation between the two reporter peptides and the polypeptide protein, in addition to the furimazine substrate. Through lyophilization of the stabilized reporter system with the formulated substrate, we demonstrate a shelf-stable, all-in-one, add-and-read analyte-detection system for use in complex sample matrices at the point-of-care. We highlight the modularity of this platform using two distinct SARS-CoV-2 model systems: SARS-CoV-2 N-antigen detection for active infections and anti-SARS-CoV-2 antibodies for immunity status detection using chemically conjugated or genetically fused affinity reagents, respectively. This technology provides a simple and standardized method to develop rapid, robust, and sensitive analyte-detection assays with flexible assay formatting making this an ideal platform for research, clinical laboratory, as well as point-of-care applications utilizing a simple handheld luminometer.
Collapse
Affiliation(s)
| | | | | | - Robin Hurst
- Promega Corporation, Madison, WI, United States
| | - Mary P Hall
- Promega Corporation, Madison, WI, United States
| | | | | | | | - William M Rehrauer
- Department of Pathology and Laboratory Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, United States
- University of Wisconsin Hospital and Clinics, Clinical Laboratories, Madison, WI, United States
| | - Molly A Accola
- University of Wisconsin Hospital and Clinics, Clinical Laboratories, Madison, WI, United States
| | | | | | | |
Collapse
|
25
|
Xiong Y, Zhang Y, Li Z, Reza MS, Li X, Tian X, Ai HW. Engineered Amber-Emitting Nano Luciferase and Its Use for Immunobioluminescence Imaging In Vivo. J Am Chem Soc 2022; 144:14101-14111. [PMID: 35913786 PMCID: PMC9425369 DOI: 10.1021/jacs.2c02320] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The NanoLuc luciferase (NLuc) and its furimazine (FRZ) substrate have revolutionized bioluminescence (BL) assays and imaging. However, the use of the NLuc-FRZ luciferase-luciferin pair for mammalian tissue imaging is hindered by the low tissue penetration of the emitting blue photons. Here, we present the development of an NLuc mutant, QLuc, which catalyzes the oxidation of a synthetic QTZ luciferin for bright and red-shifted emission peaking at ∼585 nm. Compared to other small single-domain NLuc mutants, this amber-light-emitting luciferase exhibited improved performance for imaging deep-tissue targets in live mice. Leveraging this novel bioluminescent reporter, we further pursued in vivo immunobioluminescence imaging (immunoBLI), which used a fusion protein of a single-chain variable antibody fragment (scFv) and QLuc for molecular imaging of tumor-associated antigens in a xenograft mouse model. As one of the most red-shifted NLuc variants, we expect QLuc to find broad applications in noninvasive mammalian imaging. Moreover, the immunoBLI method complements immunofluorescence imaging and immuno-positron emission tomography (immunoPET), serving as a convenient and nonradioactive molecular imaging tool for animal models in basic and preclinical research.
Collapse
Affiliation(s)
- Ying Xiong
- Department of Molecular Physiology and Biological Physics, School of Medicine, University of Virginia, Charlottesville, VA, 22908, USA
- Center for Membrane and Cell Physiology, School of Medicine, University of Virginia, Charlottesville, VA, 22908, USA
| | - Yiyu Zhang
- Department of Molecular Physiology and Biological Physics, School of Medicine, University of Virginia, Charlottesville, VA, 22908, USA
- Center for Membrane and Cell Physiology, School of Medicine, University of Virginia, Charlottesville, VA, 22908, USA
| | - Zefan Li
- Department of Molecular Physiology and Biological Physics, School of Medicine, University of Virginia, Charlottesville, VA, 22908, USA
- Center for Membrane and Cell Physiology, School of Medicine, University of Virginia, Charlottesville, VA, 22908, USA
| | - Md Shamim Reza
- Department of Molecular Physiology and Biological Physics, School of Medicine, University of Virginia, Charlottesville, VA, 22908, USA
- Center for Membrane and Cell Physiology, School of Medicine, University of Virginia, Charlottesville, VA, 22908, USA
| | - Xinyu Li
- Department of Molecular Physiology and Biological Physics, School of Medicine, University of Virginia, Charlottesville, VA, 22908, USA
- Center for Membrane and Cell Physiology, School of Medicine, University of Virginia, Charlottesville, VA, 22908, USA
| | - Xiaodong Tian
- Department of Molecular Physiology and Biological Physics, School of Medicine, University of Virginia, Charlottesville, VA, 22908, USA
- Center for Membrane and Cell Physiology, School of Medicine, University of Virginia, Charlottesville, VA, 22908, USA
| | - Hui-wang Ai
- Department of Molecular Physiology and Biological Physics, School of Medicine, University of Virginia, Charlottesville, VA, 22908, USA
- Center for Membrane and Cell Physiology, School of Medicine, University of Virginia, Charlottesville, VA, 22908, USA
- The UVA Comprehensive Cancer Center, University of Virginia, Charlottesville, VA, 22908, USA
| |
Collapse
|
26
|
Zhao F, Li X, Liu J, Zhang D, Diao H, Lin D. Establishment of stable expression of firefly luciferase and EGFP in a canine inflammatory mammary carcinoma cell line and tumor-bearing model in nude mice. Front Vet Sci 2022; 9:935005. [PMID: 35982924 PMCID: PMC9378969 DOI: 10.3389/fvets.2022.935005] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Accepted: 07/04/2022] [Indexed: 11/17/2022] Open
Abstract
Canine inflammatory mammary carcinoma (CIMC) is a type of canine malignant mammary tumor with a poor prognosis and high mortality. We transduced firefly luciferase and enhanced green fluorescent protein (EGFP) into CHMp, a CIMC cell line, and established CHMp-Luc-EGFP cells. We investigated the characteristics of this cell line in vitro and in vivo. CHMp-Luc-EGFP was passaged continuously 75 times, with stable expression of luciferase and EGFP. Compared with the wild-type, CHMp-Luc-EGFP had similar proliferation, metastasis, histopathology characteristics, and expression of E-cadherin, N-cadherin, and Ki-67. A tumor-bearing model was established by implantation of CHMp-Luc-EGFP cells, and the dynamic changes of tumors were visualized and quantified using the IVIS imaging system. In summary, the cell line we established could reflect the biological characteristics of CHMp cells, visualize the tumor progression in vivo, and provide a powerful tool for the study of CIMC.
Collapse
Affiliation(s)
- Fangying Zhao
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Xinqiu Li
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Jiayue Liu
- Key Laboratory of Animal Pathogen Infection and Immunology of Fujian Province, College of Animal Science (College of Bee Science), Fujian Agriculture and Forestry University, Fuzhou, China
| | - Di Zhang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, China Agricultural University, Beijing, China
- *Correspondence: Di Zhang
| | - Hongxiu Diao
- Key Laboratory of Animal Pathogen Infection and Immunology of Fujian Province, College of Animal Science (College of Bee Science), Fujian Agriculture and Forestry University, Fuzhou, China
- Hongxiu Diao
| | - Degui Lin
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, China Agricultural University, Beijing, China
- Degui Lin
| |
Collapse
|
27
|
Tian X, Zhang Y, Li X, Xiong Y, Wu T, Ai HW. A luciferase prosubstrate and a red bioluminescent calcium indicator for imaging neuronal activity in mice. Nat Commun 2022; 13:3967. [PMID: 35803917 PMCID: PMC9270435 DOI: 10.1038/s41467-022-31673-x] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 06/28/2022] [Indexed: 01/25/2023] Open
Abstract
Although fluorescent indicators have been broadly utilized for monitoring bioactivities, fluorescence imaging, when applied to mammals, is limited to superficial targets or requires invasive surgical procedures. Thus, there is emerging interest in developing bioluminescent indicators for noninvasive mammalian imaging. Bioluminescence imaging (BLI) of neuronal activity is highly desired but hindered by insufficient photons needed to digitalize fast brain activities. In this work, we develop a luciferase prosubstrate deliverable at an increased dose and activated in vivo by nonspecific esterase. We further engineer a bright, bioluminescent indicator with robust responsiveness to calcium ions (Ca2+) and appreciable emission above 600 nm. Integration of these advantageous components enables the imaging of the activity of neuronal ensembles in awake mice minimally invasively with excellent signal-to-background and subsecond temporal resolution. This study thus establishes a paradigm for studying brain function in health and disease.
Collapse
Affiliation(s)
- Xiaodong Tian
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, VA, 22908, USA
- Center for Membrane and Cell Physiology, University of Virginia, Charlottesville, VA, 22908, USA
| | - Yiyu Zhang
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, VA, 22908, USA
- Center for Membrane and Cell Physiology, University of Virginia, Charlottesville, VA, 22908, USA
| | - Xinyu Li
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, VA, 22908, USA
- Center for Membrane and Cell Physiology, University of Virginia, Charlottesville, VA, 22908, USA
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, China
| | - Ying Xiong
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, VA, 22908, USA
- Center for Membrane and Cell Physiology, University of Virginia, Charlottesville, VA, 22908, USA
| | - Tianchen Wu
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, VA, 22908, USA
- Center for Membrane and Cell Physiology, University of Virginia, Charlottesville, VA, 22908, USA
| | - Hui-Wang Ai
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, VA, 22908, USA.
- Center for Membrane and Cell Physiology, University of Virginia, Charlottesville, VA, 22908, USA.
- The UVA Comprehensive Cancer Center, University of Virginia, Charlottesville, VA, 22908, USA.
| |
Collapse
|
28
|
Vervaeke P, Borgos SE, Sanders NN, Combes F. Regulatory guidelines and preclinical tools to study the biodistribution of RNA therapeutics. Adv Drug Deliv Rev 2022; 184:114236. [PMID: 35351470 PMCID: PMC8957368 DOI: 10.1016/j.addr.2022.114236] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 02/09/2022] [Accepted: 03/23/2022] [Indexed: 12/27/2022]
Abstract
The success of the messenger RNA-based COVID-19 vaccines of Moderna and Pfizer/BioNTech marks the beginning of a new chapter in modern medicine. However, the rapid rise of mRNA therapeutics has resulted in a regulatory framework that is somewhat lagging. The current guidelines either do not apply, do not mention RNA therapeutics, or do not have widely accepted definitions. This review describes the guidelines for preclinical biodistribution studies of mRNA/siRNA therapeutics and highlights the relevant differences for mRNA vaccines. We also discuss the role of in vivo RNA imaging techniques and other assays to fulfill and/or complement the regulatory requirements. Specifically, quantitative whole-body autoradiography, microautoradiography, mass spectrometry-based assays, hybridization techniques (FISH, bDNA), PCR-based methods, in vivo fluorescence imaging, and in vivo bioluminescence imaging, are discussed. We conclude that this new and rapidly evolving class of medicines demands a multi-layered approach to fully understand its biodistribution and in vivo characteristics.
Collapse
Affiliation(s)
- P Vervaeke
- Laboratory of Gene Therapy, Department of Veterinary and Biosciences, Faculty of Veterinary Medicine, Ghent University, Heidestraat 19, B-9820 Merelbeke, Belgium
| | - S E Borgos
- SINTEF Industry, Dept. of Biotechnology and Nanomedicine, Research Group Mass Spectrometry, Sem Sælands v. 2A, N-7034 Trondheim, Norway
| | - N N Sanders
- Laboratory of Gene Therapy, Department of Veterinary and Biosciences, Faculty of Veterinary Medicine, Ghent University, Heidestraat 19, B-9820 Merelbeke, Belgium.
| | - F Combes
- SINTEF Industry, Dept. of Biotechnology and Nanomedicine, Research Group Mass Spectrometry, Sem Sælands v. 2A, N-7034 Trondheim, Norway.
| |
Collapse
|
29
|
Challa S, Ryu KW, Whitaker AL, Abshier JC, Camacho CV, Kraus WL. Development and characterization of new tools for detecting poly(ADP-ribose) in vitro and in vivo. eLife 2022; 11:e72464. [PMID: 35476036 PMCID: PMC9045816 DOI: 10.7554/elife.72464] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Accepted: 03/23/2022] [Indexed: 12/13/2022] Open
Abstract
ADP-ribosylation (ADPRylation) is a reversible post-translation modification resulting in the covalent attachment of ADP-ribose (ADPR) moieties on substrate proteins. Naturally occurring protein motifs and domains, including WWEs, PBZs, and macrodomains, act as 'readers' for protein-linked ADPR. Although recombinant, antibody-like ADPR detection reagents containing these readers have facilitated the detection of ADPR, they are limited in their ability to capture the dynamic nature of ADPRylation. Herein, we describe and characterize a set of poly(ADP-ribose) (PAR) Trackers (PAR-Ts)-optimized dimerization-dependent or split-protein reassembly PAR sensors in which a naturally occurring PAR binding domain, WWE, was fused to both halves of dimerization-dependent GFP (ddGFP) or split Nano Luciferase (NanoLuc), respectively. We demonstrate that these new tools allow the detection and quantification of PAR levels in extracts, living cells, and living tissues with greater sensitivity, as well as temporal and spatial precision. Importantly, these sensors detect changes in cellular ADPR levels in response to physiological cues (e.g., hormone-dependent induction of adipogenesis without DNA damage), as well as xenograft tumor tissues in living mice. Our results indicate that PAR Trackers have broad utility for detecting ADPR in many different experimental and biological systems.
Collapse
Affiliation(s)
- Sridevi Challa
- Laboratory of Signaling and Gene Regulation, Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical CenterDallasUnited States
- Division of Basic Research, Department of Obstetrics and Gynecology, University of Texas Southwestern Medical CenterDallasUnited States
| | - Keun W Ryu
- Laboratory of Signaling and Gene Regulation, Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical CenterDallasUnited States
- Division of Basic Research, Department of Obstetrics and Gynecology, University of Texas Southwestern Medical CenterDallasUnited States
| | - Amy L Whitaker
- Laboratory of Signaling and Gene Regulation, Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical CenterDallasUnited States
- Division of Basic Research, Department of Obstetrics and Gynecology, University of Texas Southwestern Medical CenterDallasUnited States
- Program in Genetics, Development, and Disease, Graduate School of Biomedical Sciences, University of Texas Southwestern Medical CenterDallasUnited States
| | - Jonathan C Abshier
- Laboratory of Signaling and Gene Regulation, Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical CenterDallasUnited States
- Division of Basic Research, Department of Obstetrics and Gynecology, University of Texas Southwestern Medical CenterDallasUnited States
| | - Cristel V Camacho
- Laboratory of Signaling and Gene Regulation, Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical CenterDallasUnited States
- Division of Basic Research, Department of Obstetrics and Gynecology, University of Texas Southwestern Medical CenterDallasUnited States
| | - W Lee Kraus
- Laboratory of Signaling and Gene Regulation, Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical CenterDallasUnited States
- Division of Basic Research, Department of Obstetrics and Gynecology, University of Texas Southwestern Medical CenterDallasUnited States
| |
Collapse
|
30
|
Heath NG, O’Geen H, Halmai NB, Corn JE, Segal DJ. Imaging Unique DNA Sequences in Individual Cells Using a CRISPR-Cas9-Based, Split Luciferase Biosensor. Front Genome Ed 2022; 4:867390. [PMID: 35403097 PMCID: PMC8990833 DOI: 10.3389/fgeed.2022.867390] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Accepted: 03/09/2022] [Indexed: 11/13/2022] Open
Abstract
An extensive arsenal of biosensing tools has been developed based on the clustered regularly interspaced short palindromic repeat (CRISPR) platform, including those that detect specific DNA sequences both in vitro and in live cells. To date, DNA imaging approaches have traditionally used full fluorescent reporter-based fusion probes. Such “always-on” probes differentiate poorly between bound and unbound probe and are unable to sensitively detect unique copies of a target sequence in individual cells. Herein we describe a DNA biosensor that provides a sensitive readout for such low-copy DNA sequences through proximity-mediated reassembly of two independently optimized fragments of NanoLuc luciferase (NLuc), a small, bright luminescent reporter. Applying this “turn-on” probe in live cells, we demonstrate an application not easily achieved by fluorescent reporter-based probes, detection of individual endogenous genomic loci using standard epifluorescence microscopy. This approach could enable detection of gene edits during ex vivo editing procedures and should be a useful platform for many other live cell DNA biosensing applications.
Collapse
Affiliation(s)
- Nicholas G. Heath
- Genome Center and Department of Biochemistry and Molecular Medicine, University of California, Davis, Davis, CA, United States
- Integrative Genetics and Genomics, University of California, Davis, Davis, CA, United States
- Innovative Genomics Institute, University of California, Berkeley, Berkeley, CA, United States
| | - Henriette O’Geen
- Genome Center and Department of Biochemistry and Molecular Medicine, University of California, Davis, Davis, CA, United States
- Innovative Genomics Institute, University of California, Berkeley, Berkeley, CA, United States
| | - Nicole B. Halmai
- Genome Center and Department of Biochemistry and Molecular Medicine, University of California, Davis, Davis, CA, United States
- Innovative Genomics Institute, University of California, Berkeley, Berkeley, CA, United States
| | - Jacob E. Corn
- Innovative Genomics Institute, University of California, Berkeley, Berkeley, CA, United States
- Department of Biology, ETH, Zürich, Switzerland
| | - David J. Segal
- Genome Center and Department of Biochemistry and Molecular Medicine, University of California, Davis, Davis, CA, United States
- Integrative Genetics and Genomics, University of California, Davis, Davis, CA, United States
- Innovative Genomics Institute, University of California, Berkeley, Berkeley, CA, United States
- *Correspondence: David J. Segal,
| |
Collapse
|
31
|
Abdelfattah AS, Ahuja S, Akkin T, Allu SR, Brake J, Boas DA, Buckley EM, Campbell RE, Chen AI, Cheng X, Čižmár T, Costantini I, De Vittorio M, Devor A, Doran PR, El Khatib M, Emiliani V, Fomin-Thunemann N, Fainman Y, Fernandez-Alfonso T, Ferri CGL, Gilad A, Han X, Harris A, Hillman EMC, Hochgeschwender U, Holt MG, Ji N, Kılıç K, Lake EMR, Li L, Li T, Mächler P, Miller EW, Mesquita RC, Nadella KMNS, Nägerl UV, Nasu Y, Nimmerjahn A, Ondráčková P, Pavone FS, Perez Campos C, Peterka DS, Pisano F, Pisanello F, Puppo F, Sabatini BL, Sadegh S, Sakadzic S, Shoham S, Shroff SN, Silver RA, Sims RR, Smith SL, Srinivasan VJ, Thunemann M, Tian L, Tian L, Troxler T, Valera A, Vaziri A, Vinogradov SA, Vitale F, Wang LV, Uhlířová H, Xu C, Yang C, Yang MH, Yellen G, Yizhar O, Zhao Y. Neurophotonic tools for microscopic measurements and manipulation: status report. NEUROPHOTONICS 2022; 9:013001. [PMID: 35493335 PMCID: PMC9047450 DOI: 10.1117/1.nph.9.s1.013001] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/30/2023]
Abstract
Neurophotonics was launched in 2014 coinciding with the launch of the BRAIN Initiative focused on development of technologies for advancement of neuroscience. For the last seven years, Neurophotonics' agenda has been well aligned with this focus on neurotechnologies featuring new optical methods and tools applicable to brain studies. While the BRAIN Initiative 2.0 is pivoting towards applications of these novel tools in the quest to understand the brain, this status report reviews an extensive and diverse toolkit of novel methods to explore brain function that have emerged from the BRAIN Initiative and related large-scale efforts for measurement and manipulation of brain structure and function. Here, we focus on neurophotonic tools mostly applicable to animal studies. A companion report, scheduled to appear later this year, will cover diffuse optical imaging methods applicable to noninvasive human studies. For each domain, we outline the current state-of-the-art of the respective technologies, identify the areas where innovation is needed, and provide an outlook for the future directions.
Collapse
Affiliation(s)
- Ahmed S. Abdelfattah
- Brown University, Department of Neuroscience, Providence, Rhode Island, United States
| | - Sapna Ahuja
- University of Pennsylvania, Perelman School of Medicine, Department of Biochemistry and Biophysics, Philadelphia, Pennsylvania, United States
- University of Pennsylvania, School of Arts and Sciences, Department of Chemistry, Philadelphia, Pennsylvania, United States
| | - Taner Akkin
- University of Minnesota, Department of Biomedical Engineering, Minneapolis, Minnesota, United States
| | - Srinivasa Rao Allu
- University of Pennsylvania, Perelman School of Medicine, Department of Biochemistry and Biophysics, Philadelphia, Pennsylvania, United States
- University of Pennsylvania, School of Arts and Sciences, Department of Chemistry, Philadelphia, Pennsylvania, United States
| | - Joshua Brake
- Harvey Mudd College, Department of Engineering, Claremont, California, United States
| | - David A. Boas
- Boston University, Department of Biomedical Engineering, Boston, Massachusetts, United States
| | - Erin M. Buckley
- Georgia Institute of Technology and Emory University, Wallace H. Coulter Department of Biomedical Engineering, Atlanta, Georgia, United States
- Emory University, Department of Pediatrics, Atlanta, Georgia, United States
| | - Robert E. Campbell
- University of Tokyo, Department of Chemistry, Tokyo, Japan
- University of Alberta, Department of Chemistry, Edmonton, Alberta, Canada
| | - Anderson I. Chen
- Boston University, Department of Biomedical Engineering, Boston, Massachusetts, United States
| | - Xiaojun Cheng
- Boston University, Department of Biomedical Engineering, Boston, Massachusetts, United States
| | - Tomáš Čižmár
- Institute of Scientific Instruments of the Czech Academy of Sciences, Brno, Czech Republic
| | - Irene Costantini
- University of Florence, European Laboratory for Non-Linear Spectroscopy, Department of Biology, Florence, Italy
- National Institute of Optics, National Research Council, Rome, Italy
| | - Massimo De Vittorio
- Istituto Italiano di Tecnologia, Center for Biomolecular Nanotechnologies, Arnesano, Italy
| | - Anna Devor
- Boston University, Department of Biomedical Engineering, Boston, Massachusetts, United States
- Massachusetts General Hospital, Harvard Medical School, Athinoula A. Martinos Center for Biomedical Imaging, Charlestown, Massachusetts, United States
| | - Patrick R. Doran
- Boston University, Department of Biomedical Engineering, Boston, Massachusetts, United States
| | - Mirna El Khatib
- University of Pennsylvania, Perelman School of Medicine, Department of Biochemistry and Biophysics, Philadelphia, Pennsylvania, United States
- University of Pennsylvania, School of Arts and Sciences, Department of Chemistry, Philadelphia, Pennsylvania, United States
| | | | - Natalie Fomin-Thunemann
- Boston University, Department of Biomedical Engineering, Boston, Massachusetts, United States
| | - Yeshaiahu Fainman
- University of California San Diego, Department of Electrical and Computer Engineering, La Jolla, California, United States
| | - Tomas Fernandez-Alfonso
- University College London, Department of Neuroscience, Physiology and Pharmacology, London, United Kingdom
| | - Christopher G. L. Ferri
- University of California San Diego, Departments of Neurosciences, La Jolla, California, United States
| | - Ariel Gilad
- The Hebrew University of Jerusalem, Institute for Medical Research Israel–Canada, Department of Medical Neurobiology, Faculty of Medicine, Jerusalem, Israel
| | - Xue Han
- Boston University, Department of Biomedical Engineering, Boston, Massachusetts, United States
| | - Andrew Harris
- Weizmann Institute of Science, Department of Brain Sciences, Rehovot, Israel
| | | | - Ute Hochgeschwender
- Central Michigan University, Department of Neuroscience, Mount Pleasant, Michigan, United States
| | - Matthew G. Holt
- University of Porto, Instituto de Investigação e Inovação em Saúde (i3S), Porto, Portugal
| | - Na Ji
- University of California Berkeley, Department of Physics, Berkeley, California, United States
| | - Kıvılcım Kılıç
- Boston University, Department of Biomedical Engineering, Boston, Massachusetts, United States
| | - Evelyn M. R. Lake
- Yale School of Medicine, Department of Radiology and Biomedical Imaging, New Haven, Connecticut, United States
| | - Lei Li
- California Institute of Technology, Andrew and Peggy Cherng Department of Medical Engineering, Department of Electrical Engineering, Pasadena, California, United States
| | - Tianqi Li
- University of Minnesota, Department of Biomedical Engineering, Minneapolis, Minnesota, United States
| | - Philipp Mächler
- Boston University, Department of Biomedical Engineering, Boston, Massachusetts, United States
| | - Evan W. Miller
- University of California Berkeley, Departments of Chemistry and Molecular & Cell Biology and Helen Wills Neuroscience Institute, Berkeley, California, United States
| | | | | | - U. Valentin Nägerl
- Interdisciplinary Institute for Neuroscience University of Bordeaux & CNRS, Bordeaux, France
| | - Yusuke Nasu
- University of Tokyo, Department of Chemistry, Tokyo, Japan
| | - Axel Nimmerjahn
- Salk Institute for Biological Studies, Waitt Advanced Biophotonics Center, La Jolla, California, United States
| | - Petra Ondráčková
- Institute of Scientific Instruments of the Czech Academy of Sciences, Brno, Czech Republic
| | - Francesco S. Pavone
- National Institute of Optics, National Research Council, Rome, Italy
- University of Florence, European Laboratory for Non-Linear Spectroscopy, Department of Physics, Florence, Italy
| | - Citlali Perez Campos
- Columbia University, Zuckerman Mind Brain Behavior Institute, New York, United States
| | - Darcy S. Peterka
- Columbia University, Zuckerman Mind Brain Behavior Institute, New York, United States
| | - Filippo Pisano
- Istituto Italiano di Tecnologia, Center for Biomolecular Nanotechnologies, Arnesano, Italy
| | - Ferruccio Pisanello
- Istituto Italiano di Tecnologia, Center for Biomolecular Nanotechnologies, Arnesano, Italy
| | - Francesca Puppo
- University of California San Diego, Departments of Neurosciences, La Jolla, California, United States
| | - Bernardo L. Sabatini
- Harvard Medical School, Howard Hughes Medical Institute, Department of Neurobiology, Boston, Massachusetts, United States
| | - Sanaz Sadegh
- University of California San Diego, Departments of Neurosciences, La Jolla, California, United States
| | - Sava Sakadzic
- Massachusetts General Hospital, Harvard Medical School, Athinoula A. Martinos Center for Biomedical Imaging, Charlestown, Massachusetts, United States
| | - Shy Shoham
- New York University Grossman School of Medicine, Tech4Health and Neuroscience Institutes, New York, New York, United States
| | - Sanaya N. Shroff
- Boston University, Department of Biomedical Engineering, Boston, Massachusetts, United States
| | - R. Angus Silver
- University College London, Department of Neuroscience, Physiology and Pharmacology, London, United Kingdom
| | - Ruth R. Sims
- Sorbonne University, INSERM, CNRS, Institut de la Vision, Paris, France
| | - Spencer L. Smith
- University of California Santa Barbara, Department of Electrical and Computer Engineering, Santa Barbara, California, United States
| | - Vivek J. Srinivasan
- New York University Langone Health, Departments of Ophthalmology and Radiology, New York, New York, United States
| | - Martin Thunemann
- Boston University, Department of Biomedical Engineering, Boston, Massachusetts, United States
| | - Lei Tian
- Boston University, Departments of Electrical Engineering and Biomedical Engineering, Boston, Massachusetts, United States
| | - Lin Tian
- University of California Davis, Department of Biochemistry and Molecular Medicine, Davis, California, United States
| | - Thomas Troxler
- University of Pennsylvania, Perelman School of Medicine, Department of Biochemistry and Biophysics, Philadelphia, Pennsylvania, United States
- University of Pennsylvania, School of Arts and Sciences, Department of Chemistry, Philadelphia, Pennsylvania, United States
| | - Antoine Valera
- University College London, Department of Neuroscience, Physiology and Pharmacology, London, United Kingdom
| | - Alipasha Vaziri
- Rockefeller University, Laboratory of Neurotechnology and Biophysics, New York, New York, United States
- The Rockefeller University, The Kavli Neural Systems Institute, New York, New York, United States
| | - Sergei A. Vinogradov
- University of Pennsylvania, Perelman School of Medicine, Department of Biochemistry and Biophysics, Philadelphia, Pennsylvania, United States
- University of Pennsylvania, School of Arts and Sciences, Department of Chemistry, Philadelphia, Pennsylvania, United States
| | - Flavia Vitale
- Center for Neuroengineering and Therapeutics, Departments of Neurology, Bioengineering, Physical Medicine and Rehabilitation, Philadelphia, Pennsylvania, United States
| | - Lihong V. Wang
- California Institute of Technology, Andrew and Peggy Cherng Department of Medical Engineering, Department of Electrical Engineering, Pasadena, California, United States
| | - Hana Uhlířová
- Institute of Scientific Instruments of the Czech Academy of Sciences, Brno, Czech Republic
| | - Chris Xu
- Cornell University, School of Applied and Engineering Physics, Ithaca, New York, United States
| | - Changhuei Yang
- California Institute of Technology, Departments of Electrical Engineering, Bioengineering and Medical Engineering, Pasadena, California, United States
| | - Mu-Han Yang
- University of California San Diego, Department of Electrical and Computer Engineering, La Jolla, California, United States
| | - Gary Yellen
- Harvard Medical School, Department of Neurobiology, Boston, Massachusetts, United States
| | - Ofer Yizhar
- Weizmann Institute of Science, Department of Brain Sciences, Rehovot, Israel
| | - Yongxin Zhao
- Carnegie Mellon University, Department of Biological Sciences, Pittsburgh, Pennsylvania, United States
| |
Collapse
|
32
|
Zhang X, Nakajima T, Mizoi K, Tsushima Y, Ogihara T. Imaging modalities for monitoring acute therapeutic effects after near-infrared photoimmunotherapy in vivo. JOURNAL OF BIOPHOTONICS 2022; 15:e202100266. [PMID: 34783185 DOI: 10.1002/jbio.202100266] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 10/11/2021] [Accepted: 10/17/2021] [Indexed: 06/13/2023]
Abstract
Near-infrared photoimmunotherapy (NIR-PIT) induces immediate cell death after irradiation with near-infrared (NIR) light. Acute therapeutic effects caused by NIR-PIT before the change of tumor size is essential to be monitored by imaging modalities. We summarized and compared the imaging modalities for evaluating acute therapeutic effects after NIR-PIT, and aimed to provide a better understanding of advantages and disadvantages of each modality for evaluation in clinical applications. Fluorescence imaging and fluorescence lifetime, with high resolution, remains high accumulation of fluorescence dyes in the normal organs. High resolution and noninvasiveness are the major advantages of magnetic resonance imaging, while 18 F-fluorodeoxyglucose positron emission tomography provides information about the glucose metabolism. Optical coherence tomography provided more information about the blood vessels. Thus, all of the imaging modalities play an important role in evaluating acute therapeutic effects after NIR-PIT. Clinicians should choose suitable modality according to specific purpose and conditions in clinical application.
Collapse
Affiliation(s)
- Xieyi Zhang
- Laboratory of Biopharmaceutics, Department of Pharmacology, Faculty of Pharmacy, Takasaki University of Health and Welfare, Takasaki, Gunma, Japan
| | - Takahito Nakajima
- Department of Diagnostic and Interventional Radiology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
- Department of Diagnostic Radiology and Nuclear Medicine, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
| | - Kenta Mizoi
- Laboratory of Biopharmaceutics, Department of Pharmacology, Faculty of Pharmacy, Takasaki University of Health and Welfare, Takasaki, Gunma, Japan
| | - Yoshito Tsushima
- Department of Diagnostic Radiology and Nuclear Medicine, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
- Research Program for Diagnostic and Molecular Imaging, Division of Integrated Oncology Research, Gunma University Initiative for Advanced Research (GIAR), Maebashi, Gunma, Japan
| | - Takuo Ogihara
- Laboratory of Biopharmaceutics, Department of Pharmacology, Faculty of Pharmacy, Takasaki University of Health and Welfare, Takasaki, Gunma, Japan
- Laboratory of Clinical Pharmacokinetics, Graduate School of Pharmaceutical Sciences, Takasaki University of Health and Welfare, Takasaki, Gunma, Japan
| |
Collapse
|
33
|
Advancing Tumor Microenvironment Research by Combining Organs-on-Chips and Biosensors. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1379:171-203. [DOI: 10.1007/978-3-031-04039-9_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
34
|
Lahmann I, Zhang Y, Baum K, Wolf J, Birchmeier C. An oscillatory network controlling self-renewal of skeletal muscle stem cells. Exp Cell Res 2021; 409:112933. [PMID: 34793773 DOI: 10.1016/j.yexcr.2021.112933] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 10/04/2021] [Accepted: 11/14/2021] [Indexed: 12/11/2022]
Abstract
The balance between proliferation and differentiation of muscle stem cells is tightly controlled, ensuring the maintenance of a cellular pool needed for muscle growth and repair. Muscle stem cells can proliferate, they can generate differentiating cells, or they self-renew to produce new stem cells. Notch signaling plays a crucial role in this process. Recent studies revealed that expression of the Notch effector HES1 oscillates in activated muscle stem cells. The oscillatory expression of HES1 periodically represses transcription from the genes encoding the myogenic transcription factor MYOD and the Notch ligand DLL1, thereby driving MYOD and DLL1 oscillations. This oscillatory network allows muscle progenitor cells and activated muscle stem cells to remain in a proliferative and 'undecided' state, in which they can either differentiate or self-renew. When HES1 is downregulated, MYOD oscillations become unstable and are replaced by sustained expression, which drives the cells into terminal differentiation. During development and regeneration, proliferating stem cells contact each other and the stability of the oscillatory expression depends on regular DLL1 inputs provided by neighboring cells. In such communities of cells that receive and provide Notch signals, the appropriate timing of DLL1 inputs is important, as sustained DLL1 cannot replace oscillatory DLL1. Thus, in cell communities, DLL1 oscillations ensure the appropriate balance between self-renewal and differentiation. In summary, oscillations in myogenic cells are an important example of dynamic gene expression determining cell fate.
Collapse
Affiliation(s)
- Ines Lahmann
- Neurowissenschaftliches Forschungszentrum, NeuroCure Cluster of Excellence, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany; Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Developmental Biology/Signal Transduction Group, 13125, Berlin, Germany
| | - Yao Zhang
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Developmental Biology/Signal Transduction Group, 13125, Berlin, Germany
| | - Katharina Baum
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Mathematical Modelling of Cellular Processes, 13125, Berlin, Germany; New address: Hasso Plattner Institute, Digital Engineering Faculty, University of Potsdam, 14482, Potsdam, Germany
| | - Jana Wolf
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Mathematical Modelling of Cellular Processes, 13125, Berlin, Germany; Free University Berlin, Department of Mathematics and Computer Science, Arnimallee 14, 14195, Berlin, Germany
| | - Carmen Birchmeier
- Neurowissenschaftliches Forschungszentrum, NeuroCure Cluster of Excellence, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany; Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Developmental Biology/Signal Transduction Group, 13125, Berlin, Germany.
| |
Collapse
|
35
|
Moreira Teixeira L, Mezzanotte L. New bioimaging avenues for organs‐on‐chips by integration of bioluminescence. VIEW 2021. [DOI: 10.1002/viw.20200177] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Affiliation(s)
- Liliana Moreira Teixeira
- Department of Developmental Bioengineering Technical Medical Centre University of Twente Enschede The Netherlands
| | - Laura Mezzanotte
- Department of Radiology and Nuclear Medicine Erasmus Medical Center Rotterdam The Netherlands
- Department of Molecular Genetics Erasmus Medical Center Rotterdam The Netherlands
| |
Collapse
|
36
|
Wang FI, Ding G, Ng GS, Dixon SJ, Chidiac P. Luciferase-based GloSensor™ cAMP assay: Temperature optimization and application to cell-based kinetic studies. Methods 2021; 203:249-258. [PMID: 34737032 DOI: 10.1016/j.ymeth.2021.10.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 10/27/2021] [Accepted: 10/28/2021] [Indexed: 01/13/2023] Open
Abstract
G protein-coupled receptors (GPCRs) are an important receptor superfamily and common therapeutic targets. The second messenger cyclic adenosine monophosphate (cAMP) is a key mediator in many GPCR signaling pathways. Monitoring intracellular cAMP levels can help identify orthosteric agonists and antagonists, as well as allosteric modulators. In this regard, luminescence-based biosensors have revolutionized our ability to monitor GPCR signaling kinetics. The GloSensor™ cAMP assay enables real-time monitoring of signaling downstream of many GPCRs. However, it is crucial to optimize assay conditions such as temperature. As well, it has not been reported whether the effects of temperature on biosensor activity are reversible. Here, we describe the temperature sensitivity and reversibility of the GloSensor™ cAMP assay, and which GloSensor™ version is optimal for measuring cytosolic cAMP. We also present a detailed protocol for monitoring cAMP levels in live cells expressing endogenous or exogenous GPCRs. Temperature optimization studies were carried out using HEK293H cells transiently transfected with the adenosine receptor A2a and the GloSensor™ plasmid (pGloSensor-20F or -22F). We found that preincubation and luminescence reading at room temperature were optimal as compared to higher temperatures. As well, the GloSensor-22F biosensor had a superior signal-to-background ratio and the effect of temperature on biosensor activity was reversible. However, thermal instability of the biosensor may pose a problem for in vivo studies. Nevertheless, the GloSensor™ cAMP assay can be applied to analyze signaling by a wide range of GPCRs for drug discovery purposes.
Collapse
Affiliation(s)
- Fang I Wang
- Department of Physiology and Pharmacology, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, Canada
| | - Gucci Ding
- Department of Physiology and Pharmacology, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, Canada
| | - Garmen S Ng
- Department of Physiology and Pharmacology, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, Canada
| | - S Jeffrey Dixon
- Department of Physiology and Pharmacology, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, Canada
| | - Peter Chidiac
- Department of Physiology and Pharmacology, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, Canada.
| |
Collapse
|
37
|
Berglund K, Stern MA, Gross RE. Bioluminescence-Optogenetics. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1293:281-293. [PMID: 33398820 DOI: 10.1007/978-981-15-8763-4_17] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/16/2023]
Abstract
In this chapter, we introduce a relatively new, emerging method for molecular neuromodulation-bioluminescence-optogenetics. Bioluminescence-optogenetics is mediated by luminopsin fusion proteins-light-sensing opsins fused to light-emitting luciferases. We describe their structures and working mechanisms and discuss their unique benefits over conventional optogenetics and chemogenetics. We also summarize applications of bioluminescence-optogenetics in various neurological disease models in rodents.
Collapse
Affiliation(s)
- Ken Berglund
- Department of Neurosurgery, Emory University School of Medicine, Atlanta, GA, USA.
| | - Matthew A Stern
- Department of Neurosurgery, Emory University School of Medicine, Atlanta, GA, USA
| | - Robert E Gross
- Department of Neurosurgery, Emory University School of Medicine, Atlanta, GA, USA
| |
Collapse
|
38
|
Li Y, Tang Y, Yang GY. Therapeutic application of exosomes in ischaemic stroke. Stroke Vasc Neurol 2021; 6:483-495. [PMID: 33431513 PMCID: PMC8485240 DOI: 10.1136/svn-2020-000419] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 08/28/2020] [Accepted: 09/18/2020] [Indexed: 02/07/2023] Open
Abstract
Ischaemic stroke is a leading cause of long-term disability in the world, with limited effective treatments. Increasing evidence demonstrates that exosomes are involved in ischaemic pathology and exhibit restorative therapeutic effects by mediating cell–cell communication. The potential of exosome therapy for ischaemic stroke has been actively investigated in the past decade. In this review, we mainly discuss the current knowledge of therapeutic applications of exosomes from different cell types, different exosomal administration routes, and current advances of exosome tracking and targeting in ischaemic stroke. We also briefly summarised the pathology of ischaemic stroke, exosome biogenesis, exosome profile changes after stroke as well as registered clinical trials of exosome-based therapy.
Collapse
Affiliation(s)
- Yongfang Li
- Department of Neurology, Ruijin Hospital, School of medcine, Shanghai Jiao Tong University, Shanghai, China
| | - Yaohui Tang
- Neuroscience and Neuroengineering Center, Medx Research Institute, Shanghai Jiao Tong University School of Biomedical Engineering, Shanghai, China
| | - Guo-Yuan Yang
- Department of Neurology, Ruijin Hospital, School of medcine, Shanghai Jiao Tong University, Shanghai, China .,Neuroscience and Neuroengineering Center, Medx Research Institute, Shanghai Jiao Tong University School of Biomedical Engineering, Shanghai, China
| |
Collapse
|
39
|
Ham SJ, Choi Y, Kim KW, Cho YC, Woo DC, Lee CW, Jang YJ, Kim PN. Tract ablation after radiofrequency ablation to prevent viable tumor cell adhesion to the needle electrode. Int J Hyperthermia 2020; 37:1287-1292. [PMID: 33198552 DOI: 10.1080/02656736.2020.1846794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
Abstract
PURPOSE To evaluate whether the additive needle tract ablation (TA) can reduce adherent cells on the needle tract after radiofrequency ablation (RFA) in a preclinical HCC mouse model. METHODS Hep3B-Luc cells were engrafted in the Balb/c-nude mice. Nineteen mice were randomly assigned into three groups: the needle only group (needle placement only without performing RFA), the RFA only group (needle placement with active RFA treatment), and the RFA-TA group (needle placement with active RFA treatment and additive tract ablation). The 17-gauge needle with a 10-mm active tip was used. After RFA and TA, the viability of adherent tumor cells on the RFA needle was evaluated with bioluminescence imaging (BLI) and live-cell counting. RESULTS We observed that RFA-TA group had the lowest BLI values compared with other groups (needle only group, 11.2 ± 6.4 million; RFA only group, 13.6 ± 9.1 million; RFA-TA group, 1.11 ± 0.8 million, p = 0.001). Live cell counting with acridine orange/propidium iodide staining also confirmed that the counted viable cell numbers in RFA-TA group were lowest compared to the other groups (needle only group, 14.8 ± 4.5; RFA only group, 643.8 ± 131.9; RFA-TA group, 1.5 ± 0.9, p < 0.001). CONCLUSIONS The additive tract ablation can significantly reduce the number of viable tumor cells adherent to the RFA needle, which can prevent needle tract seeding after RFA procedure.
Collapse
Affiliation(s)
- Su Jung Ham
- Department of Radiology and Research Institute of Radiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Yoonseok Choi
- Medical Research Institute, Gangneung Asan Hospital, University of Ulsan College of Medicine, Gangneung-si, Republic of Korea
| | - Kyung Won Kim
- Department of Radiology and Research Institute of Radiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Young Chul Cho
- Department of Radiology and Research Institute of Radiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Dong-Cheol Woo
- Department of Convergence Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.,Convergence Medicine Research Center, Asan Medical Center, Asan Institute for Life Sciences, Seoul, Republic of Korea
| | - Choong Wook Lee
- Department of Radiology and Research Institute of Radiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Yun-Jin Jang
- Department of Radiology and Research Institute of Radiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Pyo Nyun Kim
- Department of Radiology and Research Institute of Radiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
40
|
Hall MS, Decker JT, Shea LD. Towards systems tissue engineering: Elucidating the dynamics, spatial coordination, and individual cells driving emergent behaviors. Biomaterials 2020; 255:120189. [PMID: 32569865 PMCID: PMC7396312 DOI: 10.1016/j.biomaterials.2020.120189] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Revised: 04/20/2020] [Accepted: 06/09/2020] [Indexed: 12/11/2022]
Abstract
Biomaterial systems have enabled the in vitro production of complex, emergent tissue behaviors that were not possible with conventional two-dimensional culture systems, allowing for analysis of both normal development and disease processes. We propose that the path towards developing the design parameters for biomaterial systems lies with identifying the molecular drivers of emergent behavior through leveraging technological advances in systems biology, including single cell omics, genetic engineering, and high content imaging. This growing research opportunity at the intersection of the fields of tissue engineering and systems biology - systems tissue engineering - can uniquely interrogate the mechanisms by which complex tissue behaviors emerge with the potential to capture the contribution of i) dynamic regulation of tissue development and dysregulation, ii) single cell heterogeneity and the function of rare cell types, and iii) the spatial distribution and structure of individual cells and cell types within a tissue. By leveraging advances in both biological and materials data science, systems tissue engineering can facilitate the identification of biomaterial design parameters that will accelerate basic science discovery and translation.
Collapse
Affiliation(s)
- Matthew S Hall
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Joseph T Decker
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Lonnie D Shea
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
41
|
Trimaglio G, Tilkin-Mariamé AF, Feliu V, Lauzéral-Vizcaino F, Tosolini M, Valle C, Ayyoub M, Neyrolles O, Vergnolle N, Rombouts Y, Devaud C. Colon-specific immune microenvironment regulates cancer progression versus rejection. Oncoimmunology 2020; 9:1790125. [PMID: 32923152 PMCID: PMC7458593 DOI: 10.1080/2162402x.2020.1790125] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Immunotherapies have achieved clinical benefit in many types of cancer but remain limited to a subset of patients in colorectal cancer (CRC). Resistance to immunotherapy can be attributed in part to tissue-specific factors constraining antitumor immunity. Thus, a better understanding of how the colon microenvironment shapes the immune response to CRC is needed to identify mechanisms of resistance to immunotherapies and guide the development of novel therapeutics. In an orthotopic mouse model of MC38-CRC, tumor progression was monitored by bioluminescence imaging and the immune signatures were assessed at a transcriptional level using NanoString and at a cellular level by flow cytometry. Despite initial tumor growth in all mice, only 25% to 35% of mice developed a progressive lethal CRC while the remaining animals spontaneously rejected their solid tumor. No tumor rejection was observed in the absence of adaptive immunity, nor when MC38 cells were injected in non-orthotopic locations, subcutaneously or into the liver. We observed that progressive CRC tumors exhibited a protumor immune response, characterized by a regulatory T-lymphocyte pattern, discernible shortly post-tumor implantation, as well as suppressive myeloid cells. In contrast, tumor-rejecting mice presented an early inflammatory response and an antitumor microenvironment enriched in CD8+ T cells. Taken together, our data demonstrate the role of the colon microenvironment in regulating the balance between anti or protumor immune responses. While emphasizing the relevance of the CRC orthotopic model, they set the basis for exploring the impact of the identified signatures in colon cancer response to immunotherapy.
Collapse
Affiliation(s)
- Giulia Trimaglio
- Institut De Pharmacologie Et De Biologie Structurale (IPBS), Université De Toulouse, CNRS, UPS, Toulouse, France
| | | | - Virginie Feliu
- Centre De Recherches En Cancérologie De Toulouse (CRCT), INSERM U1037, Toulouse, France.,Immune Monitoring Core Facility, Institut Universitaire Du Cancer (IUCT)- Oncopôle, Toulouse, France
| | - Françoise Lauzéral-Vizcaino
- Immune Monitoring Core Facility, Institut Universitaire Du Cancer (IUCT)- Oncopôle, Toulouse, France.,Université Toulouse III Paul Sabatier, Toulouse, France
| | - Marie Tosolini
- Centre De Recherches En Cancérologie De Toulouse (CRCT), INSERM U1037, Toulouse, France
| | - Carine Valle
- Centre De Recherches En Cancérologie De Toulouse (CRCT), INSERM U1037, Toulouse, France
| | - Maha Ayyoub
- Centre De Recherches En Cancérologie De Toulouse (CRCT), INSERM U1037, Toulouse, France.,Immune Monitoring Core Facility, Institut Universitaire Du Cancer (IUCT)- Oncopôle, Toulouse, France.,Université Toulouse III Paul Sabatier, Toulouse, France
| | - Olivier Neyrolles
- Institut De Pharmacologie Et De Biologie Structurale (IPBS), Université De Toulouse, CNRS, UPS, Toulouse, France
| | - Nathalie Vergnolle
- INSERM (U1220), INRA, ENVT, UPS, Institut De Recherche En Santé Digestive (IRSD), Toulouse, France
| | - Yoann Rombouts
- Institut De Pharmacologie Et De Biologie Structurale (IPBS), Université De Toulouse, CNRS, UPS, Toulouse, France
| | - Christel Devaud
- INSERM (U1220), INRA, ENVT, UPS, Institut De Recherche En Santé Digestive (IRSD), Toulouse, France
| |
Collapse
|
42
|
Li C, Yao L, Gong J, Pang H, Shan Q, Wang Z, Lu J, Wang Z. Efficacy of Gefitinib Combined with 125I Radioactive Particles in the Treatment of Transplanted Lung Cancer Tumors in Nude Mice. Cardiovasc Intervent Radiol 2020. [PMID: 32607617 DOI: 10.1007/s00270-020-02550-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
OBJECTIVE To investigate the efficacy of gefitinib combined with iodine-125 (125I) radioactive particles in the treatment of transplanted tumors of the lung cancer cell line A549 in nude mice. MATERIALS AND METHODS Twenty-four nude mice were inoculated with A549-luc human lung adenocarcinoma cells stably expressing luciferase. The tumor size was approximately 8-10 mm after 20 days. The mice were randomly divided into four groups: a control group (n = 6), an 125I particles group (n = 6), a gefitinib group (n = 6) and a gefitinib combined 125I particles implantation group (n = 6). Tumor growth was observed, and changes in tumor size were continuously measured. Bioluminescence imaging was used to detect the bioluminescence activity of human lung adenocarcinoma A549-luc cells containing the luciferase reporter gene in vivo. After 35 days, the nude mice were sacrificed, and a tumor growth curve was drawn. RESULTS Before treatment, the tumor volumes of the four groups were not significantly different. The tumor volume difference was statistically significant in the four groups (control group, 125I radioactive particles, gefitinib group and combined drug group) at 5 weeks after treatment (F = 10.305, P < 0.05). The tumor size in the gefitinib combined with 125I particles group was significantly smaller than that in the gefitinib, 125I particles and control groups and significantly smaller than that before treatment. There was no significant difference in the bioluminescence signal intensity between the four groups before treatment. The numbers of biofluorescence photons difference were statistically significant in the four groups (F = 28.975, P < 0.05). The bioluminescence signal intensity in the gefitinib combined with 125I particles group was significantly lower than that in the 125I particles, gefitinib and control groups and significantly lower than that before treatment. CONCLUSION Gefitinib combined with 125I radioactive particles brachytherapy can significantly inhibit tumor growth.
Collapse
Affiliation(s)
- Chaojie Li
- Department of Radiology, Ruijin Hospital/Luwan Branch, Shanghai Jiao Tong University School of Medicine, No.197, Ruijin 2nd Road, Huangpu District, Shanghai, 200020, China
| | - Linyan Yao
- Department of Radiology, Ruijin Hospital/Luwan Branch, Shanghai Jiao Tong University School of Medicine, No.197, Ruijin 2nd Road, Huangpu District, Shanghai, 200020, China
| | - Ju Gong
- Department of Radiology, Ruijin Hospital/Luwan Branch, Shanghai Jiao Tong University School of Medicine, No.197, Ruijin 2nd Road, Huangpu District, Shanghai, 200020, China
| | - Haopeng Pang
- Department of Radiology, Ruijin Hospital/Luwan Branch, Shanghai Jiao Tong University School of Medicine, No.197, Ruijin 2nd Road, Huangpu District, Shanghai, 200020, China
| | - Qungang Shan
- Department of Radiology, Ruijin Hospital/Luwan Branch, Shanghai Jiao Tong University School of Medicine, No.197, Ruijin 2nd Road, Huangpu District, Shanghai, 200020, China
| | - Ziyin Wang
- Department of Radiology, Ruijin Hospital/Luwan Branch, Shanghai Jiao Tong University School of Medicine, No.197, Ruijin 2nd Road, Huangpu District, Shanghai, 200020, China
| | - Jian Lu
- Department of Radiology, Ruijin Hospital/Luwan Branch, Shanghai Jiao Tong University School of Medicine, No.197, Ruijin 2nd Road, Huangpu District, Shanghai, 200020, China.
| | - Zhongmin Wang
- Department of Radiology, Ruijin Hospital/Luwan Branch, Shanghai Jiao Tong University School of Medicine, No.197, Ruijin 2nd Road, Huangpu District, Shanghai, 200020, China.
| |
Collapse
|
43
|
Maier J, Elmenofi S, Taschauer A, Anton M, Sami H, Ogris M. Luminescent and fluorescent triple reporter plasmid constructs for Wnt, Hedgehog and Notch pathway. PLoS One 2019; 14:e0226570. [PMID: 31860685 PMCID: PMC6924688 DOI: 10.1371/journal.pone.0226570] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 11/29/2019] [Indexed: 12/11/2022] Open
Abstract
Tracking the activity of signalling pathways is a fundamental method for basic science, as well as in cancer- and pharmaceutical research. The developmental pathways Wnt, Hedgehog and Notch are frequently deregulated in cancers and represent a valuable target for the discovery of novel anticancer compounds. Here we present reporter systems for tracking activity of these pathways by using specific promoter elements driving the expression of either sensitive luciferases or fluorescent proteins. A high level of sensitivity was obtained using the luciferase reporter genes for firefly (FLuc), secreted Gaussia (GLuc) and synthetic NanoLuc (NLuc). As fluorescent reporter proteins, mTurqouise2, tdTomato and iRFP720 were chosen. Specificity of pathway activity was validated by co-transfection with pathway activating genes, showing significant response to induction. In addition, multi-gene plasmids were cloned, allowing the detection of all three pathways by one vector. By using the multi-gene vector 3P-Luc (wnt-NLuc, hedgehog-FLuc, Notch-GLuc), we could unambiguously demonstrate the crosstalk between pathways, while excluding cross reactivity of luciferase substrates. First studies with synthetic compounds confirmed the applicability of the system for future drug screening approaches.
Collapse
Affiliation(s)
- Julia Maier
- Laboratory of MacroMolecular Cancer Therapeutics (MMCT), Center of Pharmaceutical Sciences, Department of Pharmaceutical Chemistry, University of Vienna, Althanstrasse, Vienna, Austria
| | - Salma Elmenofi
- Laboratory of MacroMolecular Cancer Therapeutics (MMCT), Center of Pharmaceutical Sciences, Department of Pharmaceutical Chemistry, University of Vienna, Althanstrasse, Vienna, Austria
| | - Alexander Taschauer
- Laboratory of MacroMolecular Cancer Therapeutics (MMCT), Center of Pharmaceutical Sciences, Department of Pharmaceutical Chemistry, University of Vienna, Althanstrasse, Vienna, Austria
| | - Martina Anton
- Institutes of Molecular Immunology and Experimental Oncology, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Haider Sami
- Laboratory of MacroMolecular Cancer Therapeutics (MMCT), Center of Pharmaceutical Sciences, Department of Pharmaceutical Chemistry, University of Vienna, Althanstrasse, Vienna, Austria
- * E-mail: (MO); (HS)
| | - Manfred Ogris
- Laboratory of MacroMolecular Cancer Therapeutics (MMCT), Center of Pharmaceutical Sciences, Department of Pharmaceutical Chemistry, University of Vienna, Althanstrasse, Vienna, Austria
- * E-mail: (MO); (HS)
| |
Collapse
|
44
|
Dou L, Matz EL, Gu X, Shu F, Paxton J, Song J, Yoo J, Atala A, Jackson J, Zhang Y. Non-Invasive Cell Tracking with Brighter and Red-Transferred Luciferase for Potential Application in Stem Cell Therapy. Cell Transplant 2019; 28:1542-1551. [PMID: 31684762 PMCID: PMC6923553 DOI: 10.1177/0963689719885078] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
This study investigated the safety of a novel cell-labeling technology with mKATE and
Renilla reniformis luciferase (mKATE-renLUC) and assessed the efficacy
on tracking implanted human placental stromal cells (PSC) in an erectile dysfunction (ED)
animal model. Human PSC were labeled with mKATE-renLUC using a lentivirus. Cell viability,
apoptosis, proliferation, migration, surface marker expression and differentiation
potential of the labeled PSC were evaluated and compared with non-labeled PSC. The
paracrine profile of labeled cells was examined using an angiogenesis protein array. The
brightness and duration of labeled cells with different densities were evaluated. An ED
rat model was established and labeled PSC were injected into cavernosal tissue of the
penis. The migration and distribution of transplanted PSC were monitored using an IVIS
imaging system in real time. Implanted PSC were identified in isolated tissues via
detection of mKATE fluorescence. The cell viability, morphology, proliferation, migration,
surface marker expression and differentiation potential of mKATE-renLUC-labeled PSC were
similar to those of non-labeled cells in vitro (no statistical difference
p>0.05). Similar expressions of trophic factors were found between
labeled and non-labeled PSC. The migration and distribution of PSC expressing renLUC were
tracked in vivo using IVIS imaging system. mKATE-positive PSC were detected in penile,
kidney, prostate and hepatic tissues using histological methods. This labeling technology
provides a safe and effective cell-tracking approach with a brighter fluorophore and
codon-optimized luciferase.
Collapse
Affiliation(s)
- Lei Dou
- Stomatological Hospital of Chongqing Medical University, Chongqing, China.,Wake Forest Institute for Regenerative Medicine, Winston-Salem, NC, USA
| | - Ethan L Matz
- Wake Forest Institute for Regenerative Medicine, Winston-Salem, NC, USA
| | - Xin Gu
- Wake Forest Institute for Regenerative Medicine, Winston-Salem, NC, USA
| | - Fangpeng Shu
- Wake Forest Institute for Regenerative Medicine, Winston-Salem, NC, USA
| | - Jennifer Paxton
- Wake Forest Institute for Regenerative Medicine, Winston-Salem, NC, USA
| | - Jinlin Song
- Stomatological Hospital of Chongqing Medical University, Chongqing, China
| | - James Yoo
- Wake Forest Institute for Regenerative Medicine, Winston-Salem, NC, USA
| | - Anthony Atala
- Wake Forest Institute for Regenerative Medicine, Winston-Salem, NC, USA
| | - John Jackson
- Wake Forest Institute for Regenerative Medicine, Winston-Salem, NC, USA
| | - Yuanyuan Zhang
- Wake Forest Institute for Regenerative Medicine, Winston-Salem, NC, USA
| |
Collapse
|
45
|
Coix Seed Extract Enhances the Anti-Pancreatic Cancer Efficacy of Gemcitabine through Regulating ABCB1- and ABCG2-Mediated Drug Efflux: A Bioluminescent Pharmacokinetic and Pharmacodynamic Study. Int J Mol Sci 2019; 20:ijms20215250. [PMID: 31652737 PMCID: PMC6862065 DOI: 10.3390/ijms20215250] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 10/18/2019] [Accepted: 10/21/2019] [Indexed: 01/29/2023] Open
Abstract
A deep insight into the function and kinetics of ATP-binding cassette (ABC) transporters may aid in the development of pharmaceutics that can minimize the particular facet of chemo-resistance. We utilized bioluminescence imaging to monitor the ABC transporter mediated intracellular drug efflux function. We also investigated the potential association between the intracellular bioluminescent pharmacokinetic profiles and the anti-tumor efficacy of the coix seed extract and gemcitabine against pancreatic cancer cells in vitro and in vivo. The bioluminescent pharmacokinetic parameters and pharmacodynamic index (IC50 and TGI) were determined. The expression levels ABCB1 and ABCG2 were assessed. Results showed that coix seed extract could synergistically enhance the anti-cancer efficacy of gemcitabine (p < 0.05). Meanwhile coix seed extract alone or in combination with gemcitabine could significantly increase the AUCluc while decreasing the Kluc (p < 0.01). Western blot and immunohistochemistry assay demonstrated that coix seed extract could significantly mitigate gemcitabine-induced upregulation of ABCB1 and ABCG2 protein. The Pearson correlation analysis demonstrated that the bioluminescent pharmacokinetic parameters and pharmacodynamic index have strong association in vitro and in vivo. In conclusion coix seed extract could augment the efficacy of gemcitabine therapy in pancreatic cancer cells may at least partly due to the alteration of ABC transporter-mediated drug efflux function.
Collapse
|
46
|
Bhuckory S, Kays JC, Dennis AM. In Vivo Biosensing Using Resonance Energy Transfer. BIOSENSORS 2019; 9:E76. [PMID: 31163706 PMCID: PMC6628364 DOI: 10.3390/bios9020076] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Revised: 05/20/2019] [Accepted: 05/27/2019] [Indexed: 01/05/2023]
Abstract
Solution-phase and intracellular biosensing has substantially enhanced our understanding of molecular processes foundational to biology and pathology. Optical methods are favored because of the low cost of probes and instrumentation. While chromatographic methods are helpful, fluorescent biosensing further increases sensitivity and can be more effective in complex media. Resonance energy transfer (RET)-based sensors have been developed to use fluorescence, bioluminescence, or chemiluminescence (FRET, BRET, or CRET, respectively) as an energy donor, yielding changes in emission spectra, lifetime, or intensity in response to a molecular or environmental change. These methods hold great promise for expanding our understanding of molecular processes not just in solution and in vitro studies, but also in vivo, generating information about complex activities in a natural, organismal setting. In this review, we focus on dyes, fluorescent proteins, and nanoparticles used as energy transfer-based optical transducers in vivo in mice; there are examples of optical sensing using FRET, BRET, and in this mammalian model system. After a description of the energy transfer mechanisms and their contribution to in vivo imaging, we give a short perspective of RET-based in vivo sensors and the importance of imaging in the infrared for reduced tissue autofluorescence and improved sensitivity.
Collapse
Affiliation(s)
- Shashi Bhuckory
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA.
| | - Joshua C Kays
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA.
| | - Allison M Dennis
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA.
- Division of Materials Science and Engineering, Boston University, Boston, MA 02215, USA.
| |
Collapse
|
47
|
Yan Y, Shi P, Song W, Bi S. Chemiluminescence and Bioluminescence Imaging for Biosensing and Therapy: In Vitro and In Vivo Perspectives. Theranostics 2019; 9:4047-4065. [PMID: 31281531 PMCID: PMC6592176 DOI: 10.7150/thno.33228] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Accepted: 04/25/2019] [Indexed: 12/11/2022] Open
Abstract
Chemiluminescence (CL) and bioluminescence (BL) imaging technologies, which require no external light source so as to avoid the photobleaching, background interference and autoluminescence, have become powerful tools in biochemical analysis and biomedical science with the development of advanced imaging equipment. CL imaging technology has been widely applied to high-throughput detection of a variety of analytes because of its high sensitivity, high efficiency and high signal-to-noise ratio (SNR). Using luciferase and fluorescent proteins as reporters, various BL imaging systems have been developed innovatively for real-time monitoring of diverse molecules in vivo based on the reaction between luciferin and the substrate. Meanwhile, the kinetics of protein interactions even in deep tissues has been studied by BL imaging. In this review, we summarize in vitro and in vivo applications of CL and BL imaging for biosensing and therapy. We first focus on in vitro CL imaging from the view of improving the sensitivity. Then, in vivo CL applications in cells and tissues based on different CL systems are demonstrated. Subsequently, the recent in vitro and in vivo applications of BL imaging are summarized. Finally, we provide the insight into the development trends and future perspectives of CL and BL imaging technologies.
Collapse
|
48
|
Berglund K, Fernandez AM, Gutekunst CAN, Hochgeschwender U, Gross RE. Step-function luminopsins for bimodal prolonged neuromodulation. J Neurosci Res 2019; 98:422-436. [PMID: 30957296 DOI: 10.1002/jnr.24424] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Revised: 02/26/2019] [Accepted: 03/14/2019] [Indexed: 11/09/2022]
Abstract
Although molecular tools for controlling neuronal activity by light have vastly expanded, there are still unmet needs which require development and refinement. For example, light delivery into the brain is still a major practical challenge that hinders potential translation of optogenetics in human patients. In addition, it would be advantageous to manipulate neuronal activity acutely and precisely as well as chronically and non-invasively, using the same genetic construct in animal models. We have previously addressed these challenges by employing bioluminescence and have created a new line of opto-chemogenetic probes termed luminopsins by fusing light-sensing opsins with light-emitting luciferases. In this report, we incorporated Chlamydomonas channelrhodopsin 2 with step-function mutations as the opsin moiety in the new luminopsin fusion protein termed step-function luminopsin (SFLMO). Bioluminescence-induced photocurrent lasted longer than the bioluminescence signal due to very slow deactivation of the mutated channel. In addition, bioluminescence was able to activate most of the channels on the cell surface due to the extremely high light sensitivity of the channel. This efficient channel activation was partly mediated by radiationless bioluminescence resonance energy transfer due to the proximity of luciferase and opsin. To test the utility of SFLMOs in vivo, we transduced the substantia nigra unilaterally via a viral vector in male rats. Injection of the luciferase substrate as well as conventional photostimulation via fiber optics elicited circling behaviors. Thus, SFLMOs expand the current approaches for manipulation of neuronal activity in the brain and add more versatility and practicality to optogenetics in freely behaving animals.
Collapse
Affiliation(s)
- Ken Berglund
- Department of Neurosurgery, Emory University School of Medicine, Atlanta, Georgia
| | | | | | - Ute Hochgeschwender
- Neuroscience Program and College of Medicine, Central Michigan University, Mt Pleasant, Michigan
| | - Robert E Gross
- Department of Neurosurgery, Emory University School of Medicine, Atlanta, Georgia
| |
Collapse
|
49
|
Kim CK, Cho KF, Kim MW, Ting AY. Luciferase-LOV BRET enables versatile and specific transcriptional readout of cellular protein-protein interactions. eLife 2019; 8:43826. [PMID: 30942168 PMCID: PMC6447360 DOI: 10.7554/elife.43826] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Accepted: 03/16/2019] [Indexed: 12/21/2022] Open
Abstract
Technologies that convert transient protein-protein interactions (PPIs) into stable expression of a reporter gene are useful for genetic selections, high-throughput screening, and multiplexing with omics technologies. We previously reported SPARK (Kim et al., 2017), a transcription factor that is activated by the coincidence of blue light and a PPI. Here, we report an improved, second-generation SPARK2 that incorporates a luciferase moiety to control the light-sensitive LOV domain. SPARK2 can be temporally gated by either external light or addition of a small-molecule luciferin, which causes luciferase to open LOV via proximity-dependent BRET. Furthermore, the nested 'AND' gate design of SPARK2-in which both protease recruitment to the membrane-anchored transcription factor and LOV domain opening are regulated by the PPI of interest-yields a lower-background system and improved PPI specificity. We apply SPARK2 to high-throughput screening for GPCR agonists and for the detection of trans-cellular contacts, all with versatile transcriptional readout.
Collapse
Affiliation(s)
- Christina K Kim
- Department of Genetics, Stanford University, Stanford, United States
| | - Kelvin F Cho
- Cancer Biology Program, Stanford University, Stanford, United States
| | - Min Woo Kim
- Department of Genetics, Stanford University, Stanford, United States
| | - Alice Y Ting
- Department of Genetics, Stanford University, Stanford, United States.,Department of Biology, Stanford University, Stanford, United States.,Chan Zuckerberg Biohub, San Francisco, United States
| |
Collapse
|
50
|
Yeh HW, Wu T, Chen M, Ai HW. Identification of Factors Complicating Bioluminescence Imaging. Biochemistry 2019; 58:1689-1697. [PMID: 30810040 DOI: 10.1021/acs.biochem.8b01303] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
In vivo bioluminescence imaging (BLI) has become a standard, non-invasive imaging modality for following gene expression or the fate of proteins and cells in living animals. Currently, bioluminescent reporters used in laboratories are mostly derivatives of two major luciferase families: ATP-dependent insect luciferases and ATP-independent marine luciferases. Inconsistent results of experiments using different bioluminescent reporters, such as Akaluc and Antareas2, have been reported. Herein, we re-examined the inconsistency in several experimental settings and identified the factors, such as ATP dependency, stability in serum, and molecular sizes of luciferases, that contributed to the observed differences. We expect this study will make the research community aware of these factors and facilitate more accurate interpretation of BLI data by considering the nature of each bioluminescent reporter.
Collapse
Affiliation(s)
- Hsien-Wei Yeh
- Center for Membrane and Cell Physiology, Department of Molecular Physiology and Biological Physics, Department of Chemistry, and UVA Cancer Center , University of Virginia , 1340 Jefferson Park Avenue , Charlottesville , Virginia 22908 , United States
| | - Tianchen Wu
- Center for Membrane and Cell Physiology, Department of Molecular Physiology and Biological Physics, Department of Chemistry, and UVA Cancer Center , University of Virginia , 1340 Jefferson Park Avenue , Charlottesville , Virginia 22908 , United States
| | - Minghai Chen
- Center for Membrane and Cell Physiology, Department of Molecular Physiology and Biological Physics, Department of Chemistry, and UVA Cancer Center , University of Virginia , 1340 Jefferson Park Avenue , Charlottesville , Virginia 22908 , United States
| | - Hui-Wang Ai
- Center for Membrane and Cell Physiology, Department of Molecular Physiology and Biological Physics, Department of Chemistry, and UVA Cancer Center , University of Virginia , 1340 Jefferson Park Avenue , Charlottesville , Virginia 22908 , United States
| |
Collapse
|