1
|
Lu C, Yang Y, Zhang M, Li J, Song H, Zhao H, Mou Y, Li Y, Song X. Establishment of an in situ model to explore the tumor immune microenvironment in head and neck squamous cell carcinoma. Head Neck 2024; 46:1310-1321. [PMID: 38436502 DOI: 10.1002/hed.27707] [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: 08/22/2023] [Revised: 01/16/2024] [Accepted: 02/11/2024] [Indexed: 03/05/2024] Open
Abstract
OBJECTIVE Establish an in situ model for investigating HNSCC, focusing on tumor growth, metastasis, and the immune microenvironment. METHODS Generated a monoclonal SCCVII-ZsGreen cell line through lentiviral transfection. Selected monoclonal lines with growth rates similar to the original SCCVII for in vivo tumorigenesis. Monitored tumor development and metastasis through fluorescence in vivo imaging. Employed immunohistochemistry to assess immune cell distribution in the tumor microenvironment. RESULTS SCCVII-ZsGreen exhibited comparable proliferation and in vivo tumorigenicity to SCCVII. In situ tumor formation on day 10, with cervical metastasis in C57BL/6 mice by day 16. No significant fluorescence signals in organs like liver and lungs, while SCCVII-ZsGreen presence confirmed in cervical lymph node metastases. Immunohistochemistry revealed CD4+ T, CD8+ T, B, and dendritic cells distribution, with minimal macrophages. CONCLUSION Our model is a valuable tool for studying HNSCC occurrence, metastasis, and immune microenvironment. It allows dynamic observation of tumor development, aids preclinical drug experiments, and facilitates exploration of the tumor immune contexture.
Collapse
Affiliation(s)
- Congxian Lu
- Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Qingdao, Shandong, China
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai, Shandong, China
- Yantai Key Laboratory of Otorhinolaryngologic Diseases, Yantai, Shandong, China
| | - Yuteng Yang
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai, Shandong, China
- Yantai Key Laboratory of Otorhinolaryngologic Diseases, Yantai, Shandong, China
- The 2nd Medical College of Binzhou Medical University, Yantai, Shandong, China
| | - Mingjun Zhang
- Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Qingdao, Shandong, China
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai, Shandong, China
- Yantai Key Laboratory of Otorhinolaryngologic Diseases, Yantai, Shandong, China
| | - Jiaxuan Li
- Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Qingdao, Shandong, China
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai, Shandong, China
- Yantai Key Laboratory of Otorhinolaryngologic Diseases, Yantai, Shandong, China
| | - Hao Song
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai, Shandong, China
- Yantai Key Laboratory of Otorhinolaryngologic Diseases, Yantai, Shandong, China
- The 2nd Medical College of Binzhou Medical University, Yantai, Shandong, China
| | - Hongfei Zhao
- Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Qingdao, Shandong, China
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai, Shandong, China
- Yantai Key Laboratory of Otorhinolaryngologic Diseases, Yantai, Shandong, China
| | - Yakui Mou
- Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Qingdao, Shandong, China
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai, Shandong, China
- Yantai Key Laboratory of Otorhinolaryngologic Diseases, Yantai, Shandong, China
| | - Yumei Li
- Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Qingdao, Shandong, China
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai, Shandong, China
- Yantai Key Laboratory of Otorhinolaryngologic Diseases, Yantai, Shandong, China
| | - Xicheng Song
- Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Qingdao, Shandong, China
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai, Shandong, China
- Yantai Key Laboratory of Otorhinolaryngologic Diseases, Yantai, Shandong, China
| |
Collapse
|
2
|
Lainšček D, Golob-Urbanc A, Orehek S. In Vivo Bioluminescence and Fluorescence Imaging: Optical Tool for Cancer Research. Methods Mol Biol 2024; 2773:105-123. [PMID: 38236541 DOI: 10.1007/978-1-0716-3714-2_11] [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: 01/19/2024]
Abstract
In vivo whole-body imaging, using optical tools based on bioluminescence and fluorescence detection, offers tremendous opportunities to specifically determine the spatiotemporal resolution of cancer cells within the tested animals. This enables the study of many aspects of cancer biology, including cell proliferation, trafficking, and invasions. The antitumor therapeutic properties of various tested compounds (e.g., CD19 CAR-T cells, used for cancer immunotherapy) can be monitored within the same animal at different time points, significantly reducing the number of animals used in the study as indicated in this method.
Collapse
Affiliation(s)
- Duško Lainšček
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, Ljubljana, Slovenia.
- EN-FIST Centre of Excellence, Ljubljana, Slovenia.
| | - Anja Golob-Urbanc
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, Ljubljana, Slovenia
| | - Sara Orehek
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, Ljubljana, Slovenia
- Graduate School of Biomedicine, University of Ljubljana, Ljubljana, Slovenia
| |
Collapse
|
3
|
Almagro J, Messal HA. Volume imaging to interrogate cancer cell-tumor microenvironment interactions in space and time. Front Immunol 2023; 14:1176594. [PMID: 37261345 PMCID: PMC10228654 DOI: 10.3389/fimmu.2023.1176594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 04/26/2023] [Indexed: 06/02/2023] Open
Abstract
Volume imaging visualizes the three-dimensional (3D) complexity of tumors to unravel the dynamic crosstalk between cancer cells and the heterogeneous landscape of the tumor microenvironment (TME). Tissue clearing and intravital microscopy (IVM) constitute rapidly progressing technologies to study the architectural context of such interactions. Tissue clearing enables high-resolution imaging of large samples, allowing for the characterization of entire tumors and even organs and organisms with tumors. With IVM, the dynamic engagement between cancer cells and the TME can be visualized in 3D over time, allowing for acquisition of 4D data. Together, tissue clearing and IVM have been critical in the examination of cancer-TME interactions and have drastically advanced our knowledge in fundamental cancer research and clinical oncology. This review provides an overview of the current technical repertoire of fluorescence volume imaging technologies to study cancer and the TME, and discusses how their recent applications have been utilized to advance our fundamental understanding of tumor architecture, stromal and immune infiltration, vascularization and innervation, and to explore avenues for immunotherapy and optimized chemotherapy delivery.
Collapse
Affiliation(s)
- Jorge Almagro
- Robin Chemers Neustein Laboratory of Mammalian Cell Biology and Development, The Rockefeller University, New York, NY, United States
| | - Hendrik A. Messal
- Division of Molecular Pathology, Oncode Institute, The Netherlands Cancer Institute, Plesmanlaan, Amsterdam, Netherlands
| |
Collapse
|
4
|
Bioorthogonal surface-enhanced Raman scattering flower-like nanoprobe with embedded standards for accurate cancer cell imaging. Anal Chim Acta 2023; 1246:340895. [PMID: 36764777 DOI: 10.1016/j.aca.2023.340895] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 12/08/2022] [Accepted: 01/24/2023] [Indexed: 01/26/2023]
Abstract
Developing precise and effective strategies for cancer identification and imaging is attractive due to their importance for early cancer detection, prognosis, and subsequent treatment. Herein, we reported a novel bioorthogonal surface-enhanced Raman scattering (SERS) nanoprobe for accurate cancer cell imaging. A novel core-molecule-shell nanoflower (Au@4-MBN@Au) with rich electromagnetic hot spots and enhanced Raman scattering was first synthesized by optimizing the embedded concentrations of 4-mercaptobenzonitrile (4-MBN). Then, Au@4-MBN@Au was further modified with FA-PEG-SH molecules to acquire the bioorthogonal SERS nanoprobe Au@4-MBN@Au-PEG-FA. The SERS nanoprobe illustrated a robust and stable nitrile stretching vibration Raman signal (2223 cm-1) in the cellular silent region, ensuring high sensitivity and ultra-accuracy SERS imaging of cancer cells. Furthermore, cell imaging results demonstrated Au@4-MBN@Au-PEG-FA could recognize FR-positive HeLa cells with high selectivity due to the high affinity between folate receptor and folic acid. More notably, Au@4-MBN@Au-PEG-FA has been applied to identify FR-positive Hela cells from co-cultured cancer cells with similar morphology by SERS imaging for the first time. With improved signal-to-background ratio, high selectivity, and excellent stability, we anticipate the SERS nanoprobe Au@4-MBN@Au-PEG-FA could be applied for FR-related cancer theranostics and clinical detection in the future.
Collapse
|
5
|
Zaw Thin M, Moore C, Snoeks T, Kalber T, Downward J, Behrens A. Micro-CT acquisition and image processing to track and characterize pulmonary nodules in mice. Nat Protoc 2023; 18:990-1015. [PMID: 36494493 DOI: 10.1038/s41596-022-00769-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 08/09/2022] [Indexed: 12/14/2022]
Abstract
X-ray computed tomography is a reliable technique for the detection and longitudinal monitoring of pulmonary nodules. In preclinical stages of diagnostic or therapeutic development, the miniaturized versions of the clinical computed tomography scanners are ideally suited for carrying out translationally-relevant research in conditions that closely mimic those found in the clinic. In this Protocol, we provide image acquisition parameters optimized for low radiation dose, high-resolution and high-throughput computed tomography imaging using three commercially available micro-computed tomography scanners, together with a detailed description of the image analysis tools required to identify a variety of lung tumor types, characterized by specific radiological features. For each animal, image acquisition takes 4-8 min, and data analysis typically requires 10-30 min. Researchers with basic training in animal handling, medical imaging and software analysis should be able to implement this protocol across a wide range of lung cancer models in mice for investigating the molecular mechanisms driving lung cancer development and the assessment of diagnostic and therapeutic agents.
Collapse
Affiliation(s)
- May Zaw Thin
- Cancer Stem Cell Laboratory, Institute of Cancer Research, London, UK. .,Adult Stem Cell Laboratory, The Francis Crick Institute, London, UK.
| | - Christopher Moore
- Oncogene Biology Laboratory, The Francis Crick Institute, London, UK
| | - Thomas Snoeks
- Imaging Research Facility, The Francis Crick Institute, London, UK
| | - Tammy Kalber
- Centre for Advanced Biomedical Imaging (CABI), University College London, London, UK
| | - Julian Downward
- Oncogene Biology Laboratory, The Francis Crick Institute, London, UK. .,Lung Cancer Group, Division of Molecular Pathology, Institute of Cancer Research, London, UK.
| | - Axel Behrens
- Cancer Stem Cell Laboratory, Institute of Cancer Research, London, UK.,Adult Stem Cell Laboratory, The Francis Crick Institute, London, UK.,Department of Surgery and Cancer, Imperial College London, London, UK.,Cancer Research UK Convergence Science Centre, Imperial College London, London, UK
| |
Collapse
|
6
|
Human cancer cells generate spontaneous calcium transients and intercellular waves that modulate tumor growth. Biomaterials 2022; 290:121823. [DOI: 10.1016/j.biomaterials.2022.121823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Accepted: 09/24/2022] [Indexed: 11/02/2022]
|
7
|
|
8
|
Sengar P, Chauhan K, Hirata GA. Progress on carbon dots and hydroxyapatite based biocompatible luminescent nanomaterials for cancer theranostics. Transl Oncol 2022; 24:101482. [PMID: 35841822 PMCID: PMC9293661 DOI: 10.1016/j.tranon.2022.101482] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 06/07/2022] [Accepted: 07/06/2022] [Indexed: 11/17/2022] Open
Abstract
Biocompatible carbon dots (CDs) and nanohydroxyapatite (nHA) have attracted much attention for the development of optical imaging probes. This review discusses the development of CD and nHA based nanomaterials as multifunctional agents for cancer theranostics. The effect of synthesis strategies and doping on photoluminescent properties along with tuning of emission in biological window has been briefly reviewed. The cancer targeting strategies, biocompatibility and biodistribution of CDs and nHA based luminescent probes is discussed. A summary of current challenges and future perspectives is provided.
Despite the significant advancement in cancer diagnosis and therapy, a huge burden remains. Consequently, much research has been diverted on the development of multifunctional nanomaterials for improvement in conventional diagnosis and therapy. Luminescent nanomaterials offer a versatile platform for the development of such materials as their intrinsic photoluminescence (PL) property offers convergence of diagnosis as well as therapy at the same time. However, the clinical translation of nanomaterials faces various challenges, including biocompatibility and cost-effective scale up production. Thus, luminescent materials with facile synthesis approach along with intrinsic biocompatibility and anticancerous activity hold significant importance. As a result, carbon dots (CDs) and nanohydroxyapatite (nHA) have attracted much attention for the development of optical imaging probes. CDs are the newest members of the carbonaceous nanomaterials family that possess intrinsic luminescent and therapeutic properties, making them a promising candidate for cancer theranostic. Additionally, nHA is an excellent bioactive material due to its compositional similarity to the human bone matrix. The nHA crystal can efficiently host rare-earth elements to attain luminescent property, which can further be implemented for cancer theranostic applications. Herein, the development of CDs and nHA based nanomaterials as multifunctional agents for cancer has been briefly discussed. The emphasis has been given to different synthesis strategies leading to different morphologies and tunable PL spectra, followed by their diverse applications as biocompatible theranostic agents. Finally, the review has been summarized with the current challenges and future perspectives.
Collapse
Affiliation(s)
- Prakhar Sengar
- Centro de Nanociencias y Nanotecnología, Universidad Nacional Autónoma de México Ensenada, Baja California C.P. 22860, México
| | - Kanchan Chauhan
- Centro de Nanociencias y Nanotecnología, Universidad Nacional Autónoma de México Ensenada, Baja California C.P. 22860, México
| | - Gustavo A Hirata
- Centro de Nanociencias y Nanotecnología, Universidad Nacional Autónoma de México Ensenada, Baja California C.P. 22860, México.
| |
Collapse
|
9
|
Radzevičiūtė E, Malyško-Ptašinskė V, Novickij J, Novickij V, Girkontaitė I. Transfection by Electroporation of Cancer and Primary Cells Using Nanosecond and Microsecond Electric Fields. Pharmaceutics 2022; 14:1239. [PMID: 35745814 PMCID: PMC9230780 DOI: 10.3390/pharmaceutics14061239] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 06/08/2022] [Accepted: 06/09/2022] [Indexed: 12/18/2022] Open
Abstract
Gene transfer into primary immune cells as well as into cell lines is essential for scientific and therapeutical applications. One of the methods used for gene transfer is electroporation (EP). EP is a method where a pulsed electric field (PEF) causes a highly transient permeability of the targeted cell membrane. In this work, we present the electrotransfection of CHO-K1, 4T1 cell lines, and primary murine DCs with detectable protein-encoding plasmids in the sub-microsecond range. Microsecond (µs)- and nanosecond (ns)-range pulsed electric field transfection protocols were used. The efficiency of electrotransfection was evaluated using green fluorescent protein (GFP)-encoding plasmids (4.7 kbp; p-EGFP-N1) and plasmids expressing a firefly luciferase and red fluorescent protein (tdTomato) (8.5 kbp; pcDNA3.1(+)/Luc2 = tdT)). It was shown that the used nsPEFs protocol (7 kV/cm × 300 ns × 100, 1 MHz) ensured a better transfection efficiency than µsPEFs (1.2 kV/cm × 100 µs × 8, 1 Hz). Plasmid size and concentration had a strong impact on the cell transfection efficiency too. We also showed that there were no significant differences in transfection efficiency between immature and mature DCs. Finally, the nsPEF protocols were successfully applied for the stable transfection of the CHO-K1 cell line with the linearized pcDNA3.1(+)/Luc2 = tdT plasmid. The results of the study are applicable in gene therapy and DNA vaccination studies for the derivation of optimal electrotransfection conditions.
Collapse
Affiliation(s)
- Eivina Radzevičiūtė
- State Research Institute Centre for Innovative Medicine, Department of Immunology, 08406 Vilnius, Lithuania;
| | - Veronika Malyško-Ptašinskė
- Faculty of Electronics, Vilnius Gediminas Technical University, 03227 Vilnius, Lithuania; (V.M.-P.); (J.N.)
| | - Jurij Novickij
- Faculty of Electronics, Vilnius Gediminas Technical University, 03227 Vilnius, Lithuania; (V.M.-P.); (J.N.)
| | - Vitalij Novickij
- Faculty of Electronics, Vilnius Gediminas Technical University, 03227 Vilnius, Lithuania; (V.M.-P.); (J.N.)
| | - Irutė Girkontaitė
- State Research Institute Centre for Innovative Medicine, Department of Immunology, 08406 Vilnius, Lithuania;
| |
Collapse
|
10
|
Drzał A, Jasiński K, Gonet M, Kowolik E, Bartel Ż, Elas M. MRI and US imaging reveal evolution of spatial heterogeneity of murine tumor vasculature. Magn Reson Imaging 2022; 92:33-44. [DOI: 10.1016/j.mri.2022.06.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 05/25/2022] [Accepted: 06/02/2022] [Indexed: 11/15/2022]
|
11
|
Tazawa H, Shigeyasu K, Noma K, Kagawa S, Sakurai F, Mizuguchi H, Kobayashi H, Imamura T, Fujiwara T. Tumor‐targeted fluorescence labeling systems for cancer diagnosis and treatment. Cancer Sci 2022; 113:1919-1929. [PMID: 35398956 PMCID: PMC9207361 DOI: 10.1111/cas.15369] [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: 09/23/2021] [Revised: 03/25/2022] [Accepted: 04/02/2022] [Indexed: 11/28/2022] Open
Abstract
Conventional imaging techniques are available for clinical identification of tumor sites. However, detecting metastatic tumor cells that are spreading from primary tumor sites using conventional imaging techniques remains difficult. In contrast, fluorescence‐based labeling systems are useful tools for detecting tumor cells at the single‐cell level in cancer research. The ability to detect fluorescent‐labeled tumor cells enables investigations of the biodistribution of tumor cells for the diagnosis and treatment of cancer. For example, the presence of fluorescent tumor cells in the peripheral blood of cancer patients is a predictive biomarker for early diagnosis of distant metastasis. The elimination of fluorescent tumor cells without damaging normal tissues is ideal for minimally invasive treatment of cancer. To capture fluorescent tumor cells within normal tissues, however, tumor‐specific activated target molecules are needed. This review focuses on recent advances in tumor‐targeted fluorescence labeling systems, in which indirect reporter labeling using tumor‐specific promoters is applied to fluorescence labeling of tumor cells for the diagnosis and treatment of cancer. Telomerase promoter‐dependent fluorescence labeling using replication‐competent viral vectors produces fluorescent proteins that can be used to detect and eliminate telomerase‐positive tumor cells. Tissue‐specific promoter‐dependent fluorescence labeling enables identification of specific tumor cells. Vimentin promoter‐dependent fluorescence labeling is a useful tool for identifying tumor cells that undergo epithelial–mesenchymal transition (EMT). The evaluation of tumor cells undergoing EMT is important for accurately assessing metastatic potential. Thus, tumor‐targeted fluorescence labeling systems represent novel platforms that enable the capture of tumor cells for the diagnosis and treatment of cancer.
Collapse
Affiliation(s)
- Hiroshi Tazawa
- Department of Gastroenterological Surgery Okayama University Graduate School of Medicine Dentistry and Pharmaceutical Sciences Okayama Japan
- Center for Innovative Clinical Medicine Okayama University Hospital Okayama Japan
| | - Kunitoshi Shigeyasu
- Department of Gastroenterological Surgery Okayama University Graduate School of Medicine Dentistry and Pharmaceutical Sciences Okayama Japan
| | - Kazuhiro Noma
- Department of Gastroenterological Surgery Okayama University Graduate School of Medicine Dentistry and Pharmaceutical Sciences Okayama Japan
| | - Shunsuke Kagawa
- Department of Gastroenterological Surgery Okayama University Graduate School of Medicine Dentistry and Pharmaceutical Sciences Okayama Japan
- Minimally Invasive Therapy Center Okayama University Hospital Okayama Japan
| | - Fuminori Sakurai
- Laboratory of Biochemistry and Molecular Biology Graduate School of Pharmaceutical Sciences Osaka University Osaka Japan
| | - Hiroyuki Mizuguchi
- Laboratory of Biochemistry and Molecular Biology Graduate School of Pharmaceutical Sciences Osaka University Osaka Japan
| | - Hisataka Kobayashi
- Molecular Imaging Branch Center for Cancer Research National Cancer Institute National Institutes of Health Bethesda MD USA
| | - Takeshi Imamura
- Department of Molecular Medicine for Pathogenesis Ehime University Graduate School of Medicine Ehime Japan
| | - Toshiyoshi Fujiwara
- Department of Gastroenterological Surgery Okayama University Graduate School of Medicine Dentistry and Pharmaceutical Sciences Okayama Japan
| |
Collapse
|
12
|
Zheng F, Huang X, Ding J, Bi A, Wang S, Chen F, Zeng W. NIR-I Dye-Based Probe: A New Window for Bimodal Tumor Theranostics. Front Chem 2022; 10:859948. [PMID: 35402374 PMCID: PMC8984032 DOI: 10.3389/fchem.2022.859948] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Accepted: 02/25/2022] [Indexed: 11/13/2022] Open
Abstract
Near-infrared (NIR, 650-1700 nm) bioimaging has emerged as a powerful strategy in tumor diagnosis. In particular, NIR-I fluorescence imaging (650-950 nm) has drawn more attention, benefiting from the high quantum yield and good biocompatibility. Since their biomedical applications are slightly limited by their relatively low penetration depth, NIR-I fluorescence imaging probes have been under extensive development in recent years. This review summarizes the particular application of the NIR-I fluorescent dye-contained bimodal probes, with emphasis on related nanoprobes. These probes have enabled us to overcome the drawbacks of individual imaging modalities as well as achieve synergistic imaging. Meanwhile, the application of these NIR-I fluorescence-based bimodal probes for cancer theranostics is highlighted.
Collapse
Affiliation(s)
- Fan Zheng
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
- Hunan Key Laboratory of Diagnostic and Therapeutic Drug Research for Chronic Diseases, Changsha, China
| | - Xueyan Huang
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
- Hunan Key Laboratory of Diagnostic and Therapeutic Drug Research for Chronic Diseases, Changsha, China
| | - Jipeng Ding
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
- Hunan Key Laboratory of Diagnostic and Therapeutic Drug Research for Chronic Diseases, Changsha, China
| | - Anyao Bi
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
- Hunan Key Laboratory of Diagnostic and Therapeutic Drug Research for Chronic Diseases, Changsha, China
| | - Shifen Wang
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
- Hunan Key Laboratory of Diagnostic and Therapeutic Drug Research for Chronic Diseases, Changsha, China
| | - Fei Chen
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
- Hunan Key Laboratory of Diagnostic and Therapeutic Drug Research for Chronic Diseases, Changsha, China
| | - Wenbin Zeng
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
- Hunan Key Laboratory of Diagnostic and Therapeutic Drug Research for Chronic Diseases, Changsha, China
| |
Collapse
|
13
|
Knier NN, Pellizzari S, Zhou J, Foster PJ, Parsyan A. Preclinical Models of Brain Metastases in Breast Cancer. Biomedicines 2022; 10:biomedicines10030667. [PMID: 35327469 PMCID: PMC8945440 DOI: 10.3390/biomedicines10030667] [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: 02/23/2022] [Revised: 03/08/2022] [Accepted: 03/08/2022] [Indexed: 02/05/2023] Open
Abstract
Breast cancer remains a leading cause of mortality among women worldwide. Brain metastases confer extremely poor prognosis due to a lack of understanding of their specific biology, unique physiologic and anatomic features of the brain, and limited treatment strategies. A major roadblock in advancing the treatment of breast cancer brain metastases (BCBM) is the scarcity of representative experimental preclinical models. Current models are predominantly based on the use of animal xenograft models with immortalized breast cancer cell lines that poorly capture the disease’s heterogeneity. Recent years have witnessed the development of patient-derived in vitro and in vivo breast cancer culturing systems that more closely recapitulate the biology from individual patients. These advances led to the development of modern patient-tissue-based experimental models for BCBM. The success of preclinical models is also based on the imaging technologies used to detect metastases. Advances in animal brain imaging, including cellular MRI and multimodality imaging, allow sensitive and specific detection of brain metastases and monitoring treatment responses. These imaging technologies, together with novel translational breast cancer models based on patient-derived cancer tissues, represent a unique opportunity to advance our understanding of brain metastases biology and develop novel treatment approaches. This review discusses the state-of-the-art knowledge in preclinical models of this disease.
Collapse
Affiliation(s)
- Natasha N. Knier
- Department of Medical Biophysics, Western University, London, ON N6A 5C1, Canada; (N.N.K.); (P.J.F.)
- Imaging Laboratories, Robarts Research Institute, London, ON N6A 5B7, Canada
| | - Sierra Pellizzari
- Department of Anatomy and Cell Biology, Western University, London, ON N6A 3K7, Canada;
| | - Jiangbing Zhou
- Department of Neurosurgery, Yale University, New Haven, CT 06510, USA;
| | - Paula J. Foster
- Department of Medical Biophysics, Western University, London, ON N6A 5C1, Canada; (N.N.K.); (P.J.F.)
- Imaging Laboratories, Robarts Research Institute, London, ON N6A 5B7, Canada
| | - Armen Parsyan
- Department of Anatomy and Cell Biology, Western University, London, ON N6A 3K7, Canada;
- London Regional Cancer Program, London Health Science Centre, London, ON N6A 5W9, Canada
- Department of Oncology, Western University, London, ON N6A 4L6, Canada
- Department of Surgery, Western University, London, ON N6A 3K7, Canada
- Correspondence: ; Tel.: +1-519-646-4831; Fax: +1-519-646-6327
| |
Collapse
|
14
|
Komatsu S, Terui K, Nakata M, Shibata R, Oita S, Kawaguchi Y, Yoshizawa H, Hirokawa T, Nakatani E, Hishiki T. Combined Use of Three-Dimensional Construction and Indocyanine Green-Fluorescent Imaging for Resection of Multiple Lung Metastases in Hepatoblastoma. CHILDREN 2022; 9:children9030376. [PMID: 35327748 PMCID: PMC8947451 DOI: 10.3390/children9030376] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 02/28/2022] [Accepted: 03/06/2022] [Indexed: 11/26/2022]
Abstract
It is essential to accurately and safely resect all tumors during surgery for multiple lung metastases. Here, we report a case of hepatoblastoma (HB) with multiple pulmonary nodules that ultimately underwent complete resection using combined three-dimensional image reconstruction and indocyanine green (ICG) fluorescence guidance. A 1-year-old boy was diagnosed with HB and multiple lung metastases. After intensive chemotherapy, complete resection with subsegmentectomy (S5 + 6) and partial resection (S3, S8) were performed. More than 100 pulmonary nodules, which remained visible on computed tomography (CT) despite additional postoperative chemotherapy, were subjected to pulmonary resection. We used the SYNAPSE VINCENT software (Fujifilm Medical, Tokyo, Japan) to obtain three-dimensional images of the nodules. We numbered each nodule, and 33 lesions of the right lung were resected by multiple wedge resections through a right thoracotomy, with the aid of palpation and ICG fluorescence guidance. One month after the right metastasectomy, resection of 64 lesions in the left lung was performed via left thoracotomy. Postoperative CT showed complete clearance of the lung lesions, and the patient remained disease-free for 15 months after the treatment. This case study confirms that the combination of three-dimensional localization and ICG fluorescence guidance allows for accurate and safe resection of nearly 100 lung metastases.
Collapse
|
15
|
Takahashi K, Tanabe R, Ehata S, Kubota SI, Morishita Y, Ueda HR, Miyazono K. Visualization of the cancer cell cycle by tissue-clearing technology using the Fucci reporter system. Cancer Sci 2021; 112:3796-3809. [PMID: 34145937 PMCID: PMC8409402 DOI: 10.1111/cas.15034] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 06/16/2021] [Accepted: 06/17/2021] [Indexed: 02/06/2023] Open
Abstract
Tissue-clearing technology is an emerging imaging technique currently utilized not only in neuroscience research but also in cancer research. In our previous reports, tissue-clearing methods were used for the detection of metastatic tumors. Here, we showed that the cell cycles of primary and metastatic tumors were visualized by tissue-clearing methods using a reporter system. First, we established cancer cell lines stably expressing fluorescent ubiquitination-based cell cycle indicator (Fucci) reporter with widely used cancer cell lines A549 and 4T1. Fluorescence patterns of the Fucci reporter were investigated in various tumor inoculation models in mice. Interestingly, fluorescence patterns of the Fucci reporter of tumor colonies were different between various organs, and even among colonies in the same organs. The effects of antitumor drugs were also evaluated using these Fucci reporter cells. Of the three antitumor drugs studied, 5-fluorouracil treatment on 4T1-Fucci cells resulted in characteristic fluorescent patterns by the induction of G2 /M arrest both in vitro and in vivo. Thus, the combination of a tissue-clearing method with the Fucci reporter is useful for analyzing the mechanisms of cancer metastasis and drug resistance.
Collapse
Affiliation(s)
- Kei Takahashi
- Department of Molecular PathologyGraduate School of MedicineThe University of TokyoTokyoJapan
| | - Ryo Tanabe
- Department of Molecular PathologyGraduate School of MedicineThe University of TokyoTokyoJapan
| | - Shogo Ehata
- Department of Molecular PathologyGraduate School of MedicineThe University of TokyoTokyoJapan
- Environmental Science CenterThe University of TokyoTokyoJapan
| | - Shimpei I. Kubota
- Department of Molecular PathologyGraduate School of MedicineThe University of TokyoTokyoJapan
| | - Yasuyuki Morishita
- Department of Molecular PathologyGraduate School of MedicineThe University of TokyoTokyoJapan
| | - Hiroki R. Ueda
- Department of Systems PharmacologyGraduate School of MedicineThe University of TokyoTokyoJapan
- Laboratory for Synthetic BiologyRIKEN Center for Biosystems Dynamics ResearchSuitaJapan
| | - Kohei Miyazono
- Department of Molecular PathologyGraduate School of MedicineThe University of TokyoTokyoJapan
| |
Collapse
|
16
|
Wagalgave SM, Birajdar SS, Malegaonkar JN, Bhosale SV. Patented AIE materials for biomedical applications. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2021; 185:199-223. [PMID: 34782106 DOI: 10.1016/bs.pmbts.2021.07.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
In recent years aggregation induced emission (AIE) concept has attracted researcher's interest worldwide. Several organic building blocks are developed as AIE materials. This chapter discusses the patented AIE material and their utilization related in biological, medicinal and biotechnology fields. It is demonstrated that AIE chromophores such as tetraphenylethylene (TPE) as well as other AIE building blocks became important fluorescent emissive bioactive materials. Such emissive materials are widely employed as bioprobes for the detection of mitochondria, cellular imaging and tracking, protein carrier detection of S-phase DNA, detection of d-glucose, visualization of cancer treatment, drug screening, image-guided therapy, bacterial imaging, photodynamic therapy and drug screening. Such AIE materials upon imaging in cellular environment displays significant enhancement of fluorescence emission. Such patented AIE chromophores has a great potential for bioimaging and biomedical applications. In this chapter we compile some patented representative examples to explore their bioimaging/medicinal imaging applications since lot of new inventions are reported every day.
Collapse
Affiliation(s)
- Sopan M Wagalgave
- Polymers and Functional Materials Division, CSIR-Indian Institute of Chemical Technology, Hyderabad, Telangana, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, India
| | - Shailesh S Birajdar
- Polymers and Functional Materials Division, CSIR-Indian Institute of Chemical Technology, Hyderabad, Telangana, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, India
| | - Jotiram N Malegaonkar
- Polymers and Functional Materials Division, CSIR-Indian Institute of Chemical Technology, Hyderabad, Telangana, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, India
| | - Sidhanath Vishwanath Bhosale
- Polymers and Functional Materials Division, CSIR-Indian Institute of Chemical Technology, Hyderabad, Telangana, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, India.
| |
Collapse
|
17
|
|
18
|
Moody AS, Dayton PA, Zamboni WC. Imaging methods to evaluate tumor microenvironment factors affecting nanoparticle drug delivery and antitumor response. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2021; 4:382-413. [PMID: 34796317 PMCID: PMC8597952 DOI: 10.20517/cdr.2020.94] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 01/07/2021] [Accepted: 01/28/2021] [Indexed: 11/24/2022]
Abstract
Standard small molecule and nanoparticulate chemotherapies are used for cancer treatment; however, their effectiveness remains highly variable. One reason for this variable response is hypothesized to be due to nonspecific drug distribution and heterogeneity of the tumor microenvironment, which affect tumor delivery of the agents. Nanoparticle drugs have many theoretical advantages, but due to variability in tumor microenvironment (TME) factors, the overall drug delivery to tumors and associated antitumor response are low. The nanotechnology field would greatly benefit from a thorough analysis of the TME factors that create these physiological barriers to tumor delivery and treatment in preclinical models and in patients. Thus, there is a need to develop methods that can be used to reveal the content of the TME, determine how these TME factors affect drug delivery, and modulate TME factors to increase the tumor delivery and efficacy of nanoparticles. In this review, we will discuss TME factors involved in drug delivery, and how biomedical imaging tools can be used to evaluate tumor barriers and predict drug delivery to tumors and antitumor response.
Collapse
Affiliation(s)
- Amber S. Moody
- UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC 27599, USA
- UNC Lineberger Comprehensive Cancer Center, Chapel Hill, NC 27599, USA
- Carolina Institute for Nanomedicine, Chapel Hill, NC 27599, USA
- Joint Department of Biomedical Engineering, University of North Carolina and North Carolina State University, Chapel Hill, NC 27599, USA
| | - Paul A. Dayton
- UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC 27599, USA
- UNC Lineberger Comprehensive Cancer Center, Chapel Hill, NC 27599, USA
- Joint Department of Biomedical Engineering, University of North Carolina and North Carolina State University, Chapel Hill, NC 27599, USA
| | - William C. Zamboni
- UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC 27599, USA
- UNC Lineberger Comprehensive Cancer Center, Chapel Hill, NC 27599, USA
- Carolina Institute for Nanomedicine, Chapel Hill, NC 27599, USA
| |
Collapse
|
19
|
Defensor EB, Lim MA, Schaevitz LR. Biomonitoring and Digital Data Technology as an Opportunity for Enhancing Animal Study Translation. ILAR J 2021; 62:223-231. [PMID: 34097730 DOI: 10.1093/ilar/ilab018] [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: 08/20/2019] [Accepted: 03/17/2021] [Indexed: 02/01/2023] Open
Abstract
The failure of animal studies to translate to effective clinical therapeutics has driven efforts to identify underlying cause and develop solutions that improve the reproducibility and translatability of preclinical research. Common issues revolve around study design, analysis, and reporting as well as standardization between preclinical and clinical endpoints. To address these needs, recent advancements in digital technology, including biomonitoring of digital biomarkers, development of software systems and database technologies, as well as application of artificial intelligence to preclinical datasets can be used to increase the translational relevance of preclinical animal research. In this review, we will describe how a number of innovative digital technologies are being applied to overcome recurring challenges in study design, execution, and data sharing as well as improving scientific outcome measures. Examples of how these technologies are applied to specific therapeutic areas are provided. Digital technologies can enhance the quality of preclinical research and encourage scientific collaboration, thus accelerating the development of novel therapeutics.
Collapse
|
20
|
Monterosso ME, Futrega K, Lott WB, Vela I, Williams ED, Doran MR. Using the Microwell-mesh to culture microtissues in vitro and as a carrier to implant microtissues in vivo into mice. Sci Rep 2021; 11:5118. [PMID: 33664329 PMCID: PMC7933425 DOI: 10.1038/s41598-021-84154-4] [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: 08/28/2020] [Accepted: 02/03/2021] [Indexed: 11/09/2022] Open
Abstract
Prostate cancer (PCa) patient-derived xenografts (PDXs) are commonly propagated by serial transplantation of "pieces" of tumour in mice, but the cellular composition of pieces is not standardised. Herein, we optimised a microwell platform, the Microwell-mesh, to aggregate precise numbers of cells into arrays of microtissues, and then implanted the Microwell-mesh into NOD-scid IL2γ-/- (NSG) mice to study microtissue growth. First, mesh pore size was optimised using microtissues assembled from bone marrow-derived stromal cells, with mesh opening dimensions of 100×100 μm achieving superior microtissue vascularisation relative to mesh with 36×36 μm mesh openings. The optimised Microwell-mesh was used to assemble and implant PCa cell microtissue arrays (hereafter microtissues formed from cancer cells are referred to as microtumours) into mice. PCa cells were enriched from three different PDX lines, LuCaP35, LuCaP141, and BM18. 3D microtumours showed greater in vitro viability than 2D cultures, but neither proliferated. Microtumours were successfully established in mice 81% (57 of 70), 67% (4 of 6), 76% (19 of 25) for LuCaP35, LuCaP141, and BM18 PCa cells, respectively. Microtumour growth was tracked using live animal imaging for size or bioluminescence signal. If augmented with further imaging advances and cell bar coding, this microtumour model could enable greater resolution of PCa PDX drug response, and lead to the more efficient use of animals. The concept of microtissue assembly in the Microwell-mesh, and implantation in vivo may also have utility in implantation of islets, hair follicles or other organ-specific cells that self-assemble into 3D structures, providing an important bridge between in vitro assembly of mini-organs and in vivo implantation.
Collapse
Affiliation(s)
- Melissa E Monterosso
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, Australia.,Translational Research Institute, Brisbane, Australia
| | - Kathryn Futrega
- Translational Research Institute, Brisbane, Australia.,Centre for Biomedical Technologies (CBT), School of Mechanical, Medical, and Process Engineering (MMPE), Science and Engineering Faculty (SEF), Queensland University of Technology, Brisbane, Australia
| | - William B Lott
- Translational Research Institute, Brisbane, Australia.,Centre for Biomedical Technologies (CBT), School of Mechanical, Medical, and Process Engineering (MMPE), Science and Engineering Faculty (SEF), Queensland University of Technology, Brisbane, Australia
| | - Ian Vela
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, Australia.,Translational Research Institute, Brisbane, Australia.,Australian Prostate Cancer Research Centre - Queensland (APCRC-Q) and Queensland Bladder Cancer initiative (QBCI), Brisbane, Australia.,Department of Urology, Princess Alexandra Hospital, Brisbane, Australia
| | - Elizabeth D Williams
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, Australia.,Translational Research Institute, Brisbane, Australia.,Australian Prostate Cancer Research Centre - Queensland (APCRC-Q) and Queensland Bladder Cancer initiative (QBCI), Brisbane, Australia
| | - Michael R Doran
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, Australia. .,Translational Research Institute, Brisbane, Australia. .,Centre for Biomedical Technologies (CBT), School of Mechanical, Medical, and Process Engineering (MMPE), Science and Engineering Faculty (SEF), Queensland University of Technology, Brisbane, Australia. .,Australian Prostate Cancer Research Centre - Queensland (APCRC-Q) and Queensland Bladder Cancer initiative (QBCI), Brisbane, Australia. .,Mater Research Institute - University of Queensland (UQ), Translational Research Institute (TRI), Brisbane, Australia.
| |
Collapse
|
21
|
Prasad S, Chandra A, Cavo M, Parasido E, Fricke S, Lee Y, D'Amone E, Gigli G, Albanese C, Rodriguez O, Del Mercato LL. Optical and magnetic resonance imaging approaches for investigating the tumour microenvironment: state-of-the-art review and future trends. NANOTECHNOLOGY 2021; 32:062001. [PMID: 33065554 DOI: 10.1088/1361-6528/abc208] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
The tumour microenvironment (TME) strongly influences tumorigenesis and metastasis. Two of the most characterized properties of the TME are acidosis and hypoxia, both of which are considered hallmarks of tumours as well as critical factors in response to anticancer treatments. Currently, various imaging approaches exist to measure acidosis and hypoxia in the TME, including magnetic resonance imaging (MRI), positron emission tomography and optical imaging. In this review, we will focus on the latest fluorescent-based methods for optical sensing of cell metabolism and MRI as diagnostic imaging tools applied both in vitro and in vivo. The primary emphasis will be on describing the current and future uses of systems that can measure intra- and extra-cellular pH and oxygen changes at high spatial and temporal resolution. In addition, the suitability of these approaches for mapping tumour heterogeneity, and assessing response or failure to therapeutics will also be covered.
Collapse
Affiliation(s)
- Saumya Prasad
- Institute of Nanotechnology, National Research Council (CNR-NANOTEC), c/o Campus Ecotekne, via Monteroni, 73100, Lecce, Italy
| | - Anil Chandra
- Institute of Nanotechnology, National Research Council (CNR-NANOTEC), c/o Campus Ecotekne, via Monteroni, 73100, Lecce, Italy
| | - Marta Cavo
- Institute of Nanotechnology, National Research Council (CNR-NANOTEC), c/o Campus Ecotekne, via Monteroni, 73100, Lecce, Italy
| | - Erika Parasido
- Department of Oncology, Georgetown University Medical Center, Washington, DC, United States of America
- Center for Translational Imaging, Georgetown University Medical Center, Washington, DC, United States of America
| | - Stanley Fricke
- Department of Oncology, Georgetown University Medical Center, Washington, DC, United States of America
- Center for Translational Imaging, Georgetown University Medical Center, Washington, DC, United States of America
- Department of Radiology, Georgetown University Medical Center, Washington, DC, United States of America
| | - Yichien Lee
- Department of Oncology, Georgetown University Medical Center, Washington, DC, United States of America
| | - Eliana D'Amone
- Institute of Nanotechnology, National Research Council (CNR-NANOTEC), c/o Campus Ecotekne, via Monteroni, 73100, Lecce, Italy
| | - Giuseppe Gigli
- Institute of Nanotechnology, National Research Council (CNR-NANOTEC), c/o Campus Ecotekne, via Monteroni, 73100, Lecce, Italy
- Department of Mathematics and Physics 'Ennio De Giorgi', University of Salento, via Arnesano, 73100, Lecce, Italy
| | - Chris Albanese
- Department of Oncology, Georgetown University Medical Center, Washington, DC, United States of America
- Center for Translational Imaging, Georgetown University Medical Center, Washington, DC, United States of America
- Department of Radiology, Georgetown University Medical Center, Washington, DC, United States of America
| | - Olga Rodriguez
- Department of Oncology, Georgetown University Medical Center, Washington, DC, United States of America
- Center for Translational Imaging, Georgetown University Medical Center, Washington, DC, United States of America
| | - Loretta L Del Mercato
- Institute of Nanotechnology, National Research Council (CNR-NANOTEC), c/o Campus Ecotekne, via Monteroni, 73100, Lecce, Italy
| |
Collapse
|
22
|
Wu S, Lin X. Trials in developing a nanoscale material for extravascular contrast-enhanced ultrasound targeting hepatocellular carcinoma. PeerJ 2020; 8:e10403. [PMID: 33354418 PMCID: PMC7727372 DOI: 10.7717/peerj.10403] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 10/31/2020] [Indexed: 12/27/2022] Open
Abstract
Background Medical imaging is an important approach for the diagnosis of hepatocellular carcinoma (HCC), a common life threaten disease, however, the diagnostic efficiency is still not optimal. Developing a novel method to improve diagnosis is necessary. The aim of this project was to formulate a material that can combine with GPC3 of HCC for targeted enhanced ultrasound. Methods A material of sulfur hexafluoride (SF6) filled liposome microbubbles and conjugated with synthesized peptide (LSPMbs) was prepared and assessed in vitro and vivo. Liposome microbubbles were made of DPPC, DPPG, DSPE-PEG2000,and SF6, using thin film method to form shell, followed filling SF6, and conjugating peptide. A carbodiimide method was used for covalent conjugation of peptide to LSMbs. Results The prepared LSPMbs appeared round shaped, with size of 380.9 ± 176.5 nm, and Zeta potential of −51.4 ± 10.4mV. LSPMbs showed high affinity to Huh-7 cells in vitro, presented good enhanced ultrasound effects, did not show cytotoxicity, and did not exhibit targeted fluorescence and enhanced ultrasound in animal xenograft tumors. Conclusion Extravascular contrast-enhanced ultrasound targeted GPC3 on HCC may not be realized, and the reason may be that targeted contrast agents of microbubbles are hard to access and accumulate in the tumor stroma and matrix.
Collapse
Affiliation(s)
- Size Wu
- Department of Ultrasound, The First Affiliated Hospital of Hainan Medical University, Haikou, Hainan Province, China
| | - Xiyuan Lin
- Department of Emergency Medicine, The First Affiliated Hospital of Hainan Medical University, Haikou, Hainan Province, China
| |
Collapse
|
23
|
Zhang Y, Du Y, Zhuo Y, Qiu L. Functional Nucleic Acid-Based Live-Cell Fluorescence Imaging. Front Chem 2020; 8:598013. [PMID: 33363111 PMCID: PMC7759623 DOI: 10.3389/fchem.2020.598013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Accepted: 11/16/2020] [Indexed: 12/01/2022] Open
Abstract
Cell is the structural and functional unit of organism. It serves as a key research object in various biological processes, such as growth, ontogeny, metabolism and stress. Studying the spatiotemporal distribution and functional activity of specific biological molecules in living cells is crucial for exploring the mechanism governing life. It also facilitates the elucidation of pathogenesis, clinical prevention and disease theranostics. In recent years, the fluorescence imaging technique has been greatly exploited for live-cell imaging. However, the development of molecular probes has lagged far behind. Functional nucleic acids (FNAs), for example, aptamer and DNAzyme, possess special chemical and/or biological functions, hence severing as promising molecular tools for cellular imaging. The current mini review focuses on the applications of FNAs in live-cell fluorescence imaging.
Collapse
Affiliation(s)
- Yutong Zhang
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, China
| | - Yulin Du
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, China
| | - Yuting Zhuo
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, China
- NHC Key Laboratory of Birth Defect for Research and Prevention, Hunan Provincial Maternal and Child Health Care Hospital, Changsha, China
| | - Liping Qiu
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, China
- NHC Key Laboratory of Birth Defect for Research and Prevention, Hunan Provincial Maternal and Child Health Care Hospital, Changsha, China
| |
Collapse
|
24
|
Kostyuk AI, Panova AS, Kokova AD, Kotova DA, Maltsev DI, Podgorny OV, Belousov VV, Bilan DS. In Vivo Imaging with Genetically Encoded Redox Biosensors. Int J Mol Sci 2020; 21:E8164. [PMID: 33142884 PMCID: PMC7662651 DOI: 10.3390/ijms21218164] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 10/28/2020] [Accepted: 10/29/2020] [Indexed: 12/13/2022] Open
Abstract
Redox reactions are of high fundamental and practical interest since they are involved in both normal physiology and the pathogenesis of various diseases. However, this area of research has always been a relatively problematic field in the context of analytical approaches, mostly because of the unstable nature of the compounds that are measured. Genetically encoded sensors allow for the registration of highly reactive molecules in real-time mode and, therefore, they began a new era in redox biology. Their strongest points manifest most brightly in in vivo experiments and pave the way for the non-invasive investigation of biochemical pathways that proceed in organisms from different systematic groups. In the first part of the review, we briefly describe the redox sensors that were used in vivo as well as summarize the model systems to which they were applied. Next, we thoroughly discuss the biological results obtained in these studies in regard to animals, plants, as well as unicellular eukaryotes and prokaryotes. We hope that this work reflects the amazing power of this technology and can serve as a useful guide for biologists and chemists who work in the field of redox processes.
Collapse
Affiliation(s)
- Alexander I. Kostyuk
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, 117997 Moscow, Russia; (A.I.K.); (A.S.P.); (A.D.K.); (D.A.K.); (D.I.M.); (O.V.P.); (V.V.B.)
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, 117997 Moscow, Russia
| | - Anastasiya S. Panova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, 117997 Moscow, Russia; (A.I.K.); (A.S.P.); (A.D.K.); (D.A.K.); (D.I.M.); (O.V.P.); (V.V.B.)
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, 117997 Moscow, Russia
| | - Aleksandra D. Kokova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, 117997 Moscow, Russia; (A.I.K.); (A.S.P.); (A.D.K.); (D.A.K.); (D.I.M.); (O.V.P.); (V.V.B.)
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, 117997 Moscow, Russia
| | - Daria A. Kotova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, 117997 Moscow, Russia; (A.I.K.); (A.S.P.); (A.D.K.); (D.A.K.); (D.I.M.); (O.V.P.); (V.V.B.)
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, 117997 Moscow, Russia
| | - Dmitry I. Maltsev
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, 117997 Moscow, Russia; (A.I.K.); (A.S.P.); (A.D.K.); (D.A.K.); (D.I.M.); (O.V.P.); (V.V.B.)
- Federal Center for Cerebrovascular Pathology and Stroke, 117997 Moscow, Russia
| | - Oleg V. Podgorny
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, 117997 Moscow, Russia; (A.I.K.); (A.S.P.); (A.D.K.); (D.A.K.); (D.I.M.); (O.V.P.); (V.V.B.)
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, 117997 Moscow, Russia
| | - Vsevolod V. Belousov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, 117997 Moscow, Russia; (A.I.K.); (A.S.P.); (A.D.K.); (D.A.K.); (D.I.M.); (O.V.P.); (V.V.B.)
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, 117997 Moscow, Russia
- Federal Center for Cerebrovascular Pathology and Stroke, 117997 Moscow, Russia
- Institute for Cardiovascular Physiology, Georg August University Göttingen, D-37073 Göttingen, Germany
| | - Dmitry S. Bilan
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, 117997 Moscow, Russia; (A.I.K.); (A.S.P.); (A.D.K.); (D.A.K.); (D.I.M.); (O.V.P.); (V.V.B.)
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, 117997 Moscow, Russia
| |
Collapse
|
25
|
Wang Y, Hu Y, Pan K, Li H, Shang S, Wang Y, Tang G, Han X. In-vivo imaging revealed antigen-directed gingival B10 infiltration in experimental periodontitis. Biochim Biophys Acta Mol Basis Dis 2020; 1867:165991. [PMID: 33080346 DOI: 10.1016/j.bbadis.2020.165991] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 09/29/2020] [Accepted: 10/14/2020] [Indexed: 12/15/2022]
Abstract
Our previous study demonstrated that IL-10 secreting B (B10) cells alleviate inflammation and bone loss in experimental periodontitis. The purpose of this study is to determine whether antigen-specificity is required for the local infiltration of B10 cells. Experimental periodontitis was induced in the recipient mice by placement of silk ligature with or without the presence of live Porphyromonas gingivalis (P. gingivalis). Donor mice were pre-immunized by intraperitoneal (IP) injection of formalin-fixed P. gingivalis, or PBS as non-immunized control. Spleen B cells were purified and treated with LPS and CpG for 48 h to expand the B10 population in vitro. Fluorescence-labelled B10 cells were transferred into the recipient mice by tail vein injection and were tracked on day 0, 3, 5 and 10 using IVIS Spectrum in vivo imaging system. The number of B10 cells and P. gingivalis-binding B cells were significantly increased after in vitro treatment of LPS and CpG. On day 5, the fluorescence intensity in gingival tissues was the highest in mice transferred with B10 cells from pre-immunized donor mice. Gingival expression of IL-6, TNF-α, RANKL/OPG ratio and periodontal bone loss in recipient mice were significantly reduced, and the expression of IL-10 and the number of CD19+ B cells were significantly increased after pre-immunized B10 cell transfer in the presence of antigen, compared to those with non-immunized B10 cell transfer or no antigen presence. This study suggests that antigen specificity dictate the local infiltration of B10 cells into periodontal tissue and these antigen-specific B10 cells promote anti-inflammatory responses.
Collapse
Affiliation(s)
- Yufeng Wang
- Department of Oral Medicine, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, National Clinical Research Center of Stomatology, Shanghai 200011, China; Department of Immunology and Infectious Diseases, The Forsyth Institute, Harvard School of Dental Medicine Affiliate, Cambridge, MA 02142, United States
| | - Yang Hu
- Department of Immunology and Infectious Diseases, The Forsyth Institute, Harvard School of Dental Medicine Affiliate, Cambridge, MA 02142, United States; Department of Oral Medicine, Infection and Immunity, Harvard School of Dental Medicine, Boston, MA 02115, United States
| | - Keqing Pan
- Department of Immunology and Infectious Diseases, The Forsyth Institute, Harvard School of Dental Medicine Affiliate, Cambridge, MA 02142, United States; Department of Stomatology, the affiliated hospital of Qingdao University, Qingdao, Shandong 266003, China
| | - Hao Li
- Department of Immunology and Infectious Diseases, The Forsyth Institute, Harvard School of Dental Medicine Affiliate, Cambridge, MA 02142, United States; Department of Prosthodontics, the Affiliated Hospital of Stomatology, Guangxi Medical University, Nanning 530021, China
| | - Shu Shang
- Shanghai University of Medicine and Health Sciences, Shanghai 201318, China
| | - Yuhua Wang
- Department of Oral Medicine, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, National Clinical Research Center of Stomatology, Shanghai 200011, China
| | - Guoyao Tang
- Department of Oral Medicine, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, National Clinical Research Center of Stomatology, Shanghai 200011, China
| | - Xiaozhe Han
- Department of Immunology and Infectious Diseases, The Forsyth Institute, Harvard School of Dental Medicine Affiliate, Cambridge, MA 02142, United States; Department of Oral Medicine, Infection and Immunity, Harvard School of Dental Medicine, Boston, MA 02115, United States.
| |
Collapse
|
26
|
Matsuda M, Terai K. Experimental pathology by intravital microscopy and genetically encoded fluorescent biosensors. Pathol Int 2020; 70:379-390. [PMID: 32270554 PMCID: PMC7383902 DOI: 10.1111/pin.12925] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 03/13/2020] [Accepted: 03/16/2020] [Indexed: 01/03/2023]
Abstract
The invention of two‐photon excitation microscopes widens the potential application of intravital microscopy (IVM) to the broad field of experimental pathology. Moreover, the recent development of fluorescent protein‐based, genetically encoded biosensors provides an ideal tool to visualize the cell function in live animals. We start from a brief review of IVM with two‐photon excitation microscopes and genetically encoded biosensors based on the principle of Förster resonance energy transfer (FRET). Then, we describe how IVM using biosensors has revealed the pathogenesis of several disease models.
Collapse
Affiliation(s)
- Michiyuki Matsuda
- Department of Pathology and Biology of Diseases, Graduate School of Medicine, Kyoto University, Kyoto, Japan.,Research Center for Dynamic Living Systems, Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| | - Kenta Terai
- Research Center for Dynamic Living Systems, Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| |
Collapse
|
27
|
Shi K, Liu X, Du G, Cai X, Zhan Y. In vivo antitumour activity of Britanin against gastric cancer through nuclear factor-κB-mediated immune response. ACTA ACUST UNITED AC 2020; 72:607-618. [PMID: 31943207 DOI: 10.1111/jphp.13230] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2019] [Accepted: 12/17/2019] [Indexed: 12/14/2022]
Abstract
OBJECTIVES Britanin was explored for the antitumour effect on gastric cancer, which is a sesquiterpene lactone (SL) extracted from Inula japonica. METHODS In the present study, cell viability assays were performed to evaluate the antiproliferation effect of Britanin on gastric cancer cells. Tumour development in BGC-823 cell-bearing nude mice was monitored in real-time after Britanin treatment via a bioluminescent imaging method. Western blotting analysis and enzyme-linked immunosorbent assays detected proteins associated with the nuclear factor (NF)-κB signalling pathway. KEY FINDINGS Britanin can suppress the proliferation of gastric cancer cells in vitro and the growth of tumours in vivo. In the treatment group, decreased levels of p65 and phosphorylated (p)-p65 were observed. This indicated that NF-κB plays an important role in the antitumour effect of Britanin. Furthermore, considering the additional role of NF-κB in the immune system, the levels of the downstream molecules interleukin (IL)-2 and the cytokine IL-10 were subsequently determined in vivo. An increase in the IL-2 level and a decrease in the IL-10 level indicated that Britanin elicited an enhanced immune response. CONCLUSIONS Britanin may be a promising candidate for gastric cancer chemotherapy, and its anticancer effect likely depends on an NF-κB-mediated immune response.
Collapse
Affiliation(s)
- Keru Shi
- Engineering Research Center of Molecular & Neuroimaging, Ministry of Education, School of Life Science and Technology, Xidian University, Xi'an, China
| | - Xinyue Liu
- Engineering Research Center of Molecular & Neuroimaging, Ministry of Education, School of Life Science and Technology, Xidian University, Xi'an, China
| | - Getao Du
- Engineering Research Center of Molecular & Neuroimaging, Ministry of Education, School of Life Science and Technology, Xidian University, Xi'an, China
| | - Xiaoxia Cai
- Engineering Research Center of Molecular & Neuroimaging, Ministry of Education, School of Life Science and Technology, Xidian University, Xi'an, China
| | - Yonghua Zhan
- Engineering Research Center of Molecular & Neuroimaging, Ministry of Education, School of Life Science and Technology, Xidian University, Xi'an, China
| |
Collapse
|
28
|
Singh N, Kumar P, Riaz U. Applications of near infrared and surface enhanced Raman scattering techniques in tumor imaging: A short review. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2019; 222:117279. [PMID: 31234091 DOI: 10.1016/j.saa.2019.117279] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 06/08/2019] [Accepted: 06/15/2019] [Indexed: 06/09/2023]
Abstract
Imaging technologies play a vital role in clinical oncology and have undergone massive growth over the past few decades. Research in the field of tumor imaging and biomedical diagnostics requires early detection of physiological alterations so as to provide curative treatment in real time. The objective of this review is to provide an insight about near infrared fluorescence (NIRF) and surface enhanced Raman scattering (SERS) imaging techniques that can be used to expand their capabilities for the early detection and diagnosis of cancer cells. Basic setup, principle and working of the instruments has been provided and common NIRF imaging agents as well as SERS tags are also discussed besides the analytical advantages/disadvantages of these techniques. This review can help researchers working in the field of molecular imaging to design cost effective fluorophores and SERS tags to overcome the limitations of both NIRF as well as SERS imaging technologies.
Collapse
Affiliation(s)
- Neetika Singh
- Materials Research Laboratory, Department of Chemistry, Jamia Millia Islamia, New Delhi 110025, India; Advanced Instrumentation Research Facility, Jawaharlal Nehru University, New Delhi 110067, India
| | - Prabhat Kumar
- Materials Research Laboratory, Department of Chemistry, Jamia Millia Islamia, New Delhi 110025, India; Advanced Instrumentation Research Facility, Jawaharlal Nehru University, New Delhi 110067, India
| | - Ufana Riaz
- Materials Research Laboratory, Department of Chemistry, Jamia Millia Islamia, New Delhi 110025, India; Advanced Instrumentation Research Facility, Jawaharlal Nehru University, New Delhi 110067, India.
| |
Collapse
|
29
|
König TT, Goedeke J, Muensterer OJ. Multiphoton microscopy in surgical oncology- a systematic review and guide for clinical translatability. Surg Oncol 2019; 31:119-131. [PMID: 31654957 DOI: 10.1016/j.suronc.2019.10.011] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Revised: 10/02/2019] [Accepted: 10/13/2019] [Indexed: 02/07/2023]
Abstract
BACKGROUND Multiphoton microscopy (MPM) facilitates three-dimensional, high-resolution functional imaging of unlabeled tissues in vivo and ex vivo. This systematic review discusses the diagnostic value, advantages and challenges in the practical use of MPM in surgical oncology. METHOD AND FINDINGS A Medline search was conducted in April 2019. Fifty-three original research papers investigating MPM compared to standard histology in human patients with solid tumors were identified. A qualitative synopsis and meta-analysis of 14 blinded studies was performed. Risk of bias and applicability were evaluated. MPM can image fresh, frozen or fixed tissues up to a depth 1000 μm in the z-plane. Best results including functional imaging and virtual histochemistry are obtained by in vivo imaging or scanning fresh tissue immediately after excision. Two-photon excited fluorescence by natural fluorophores of the cytoplasm and second harmonic generation signals by fluorophores of the extracellular matrix can be scanned simultaneously, providing high resolution optical histochemistry comparable to standard histology. Functional parameters like fluorescence lifetime imaging or optical redox ratio provide additional objective information. A major concern is inability to visualize the nucleus. However, in a subpopulation analysis of 440 specimens, MPM yielded a sensitivity of 94%, specificity of 96% and accuracy of 95% for the detection of malignant tissue. CONCLUSION MPM is a promising emerging technique in surgical oncology. Ex vivo imaging has high sensitivity, specificity and accuracy for the detection of tumor cells. For broad clinical application in vivo, technical challenges need to be resolved.
Collapse
Affiliation(s)
| | - Jan Goedeke
- Universitätsmedizin Mainz, Department of Pediatric Surgery, Mainz, Germany
| | | |
Collapse
|
30
|
Nomura N, Nishihara R, Nakajima T, Kim SB, Iwasawa N, Hiruta Y, Nishiyama S, Sato M, Citterio D, Suzuki K. Biothiol-Activatable Bioluminescent Coelenterazine Derivative for Molecular Imaging in Vitro and in Vivo. Anal Chem 2019; 91:9546-9553. [PMID: 31291724 DOI: 10.1021/acs.analchem.9b00694] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
There is a high demand for sensitive biothiol probes targeting cysteine, glutathione, and homocysteine. These biothiols are known as playing essential roles to maintain homeostasis and work as indicators of many diseases. This work presents a bioluminescent probe (named AMCM) to detect biothiols in live mammalian cells and in vivo with a limit of detection of 0.11 μM for cysteine in solution and high selectivity for biothiols, making it suitable for real-time biothiol detection in biological systems. Upon application to live cells, AMCM showed low cytotoxicity and sensitively reported bioluminescence in response to changes of biothiol levels. Furthermore, a bioluminescence resonance energy transfer system consisting of AMCM combined with the near-infrared fluorescent protein iRFP713 was applied to in vivo imaging, with emitted tissue-permeable luminescence in living mice. In summary, this work demonstrates that AMCM is of high practical value for the detection of biothiols in living cells and for deep tissue imaging in living animals.
Collapse
Affiliation(s)
| | | | - Takahiro Nakajima
- Graduate School of Arts and Sciences , The University of Tokyo , 3-8-1 Komaba , Meguro , Tokyo , Japan
| | - Sung Bae Kim
- National Institute of Advanced Industrial Science and Technology , 1-1-1 Umezono , Tsukuba , Ibaraki 305-8560 , Japan
| | | | | | | | - Moritoshi Sato
- Graduate School of Arts and Sciences , The University of Tokyo , 3-8-1 Komaba , Meguro , Tokyo , Japan
| | | | | |
Collapse
|
31
|
Cho N, Shokeen M. Changing landscape of optical imaging in skeletal metastases. J Bone Oncol 2019; 17:100249. [PMID: 31316892 PMCID: PMC6611980 DOI: 10.1016/j.jbo.2019.100249] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Revised: 06/25/2019] [Accepted: 06/26/2019] [Indexed: 02/08/2023] Open
Abstract
Optical imaging is an emerging strategy for in vitro and in vivo visualization of the molecular mechanisms of cancer over time. An increasing number of optical imaging contrast agents and techniques have been developed in recent years specifically for bone research and skeletal metastases. Visualizing molecular processes in relation to bone remodeling in metastasized cancers provides valuable information for understanding disease mechanisms and monitoring expression of primary molecular targets and therapeutic efficacy. This review is intended to provide an overview of tumor-specific and non-specific contrast agents in the first near-infrared window (NIR-I) window from 650 nm to 950 nm that can be used to study functional and structural aspects of skeletal remodeling of cancer in preclinical animal models. Near-infrared (NIR) optical imaging techniques, specifically NIR spectroscopy and photoacoustic imaging, and their use in skeletal metastases will also be discussed. Perspectives on the promises and challenges facing this exciting field are then given.
Collapse
Affiliation(s)
- Nicholas Cho
- Department of Radiology, Washington University School of Medicine, 4515 McKinley Ave, St. Louis, MO 63110, United States.,Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO 63110, United States
| | - Monica Shokeen
- Department of Radiology, Washington University School of Medicine, 4515 McKinley Ave, St. Louis, MO 63110, United States.,Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO 63110, United States.,Alvin J. Siteman Cancer Center at Washington University School of Medicine and Barnes Jewish Hospital, St. Louis, MO 63110, United States
| |
Collapse
|
32
|
Taki S, Ardestani MS. Novel nanosized AS1411-chitosan-BODIPY conjugate for molecular fluorescent imaging. Int J Nanomedicine 2019; 14:3543-3555. [PMID: 31190811 PMCID: PMC6526928 DOI: 10.2147/ijn.s202561] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Accepted: 04/17/2019] [Indexed: 11/23/2022] Open
Abstract
Background: In recent years, non-invasive imaging technologies for early cancer detection have drawn worldwide attention. In this study, an antinucleolin aptamer, AS1411, was successfully conjugated to BODIPY-labeled chitosan and studied on T47D and HEK-293 cell lines. Methods: After conjugation of the aptamer to chitosan nanoparticles and purification, its structure was confirmed using atomic force microscopy (AFM), electrophoretic light scattering (ELS) and dynamic light scattering (DLS). Results of AFM, DLS and ELS of both conjugation and chitosan were compared for confirmation of conjugation. Conjugates were mixed with BODIPY FL fluorescent dye, purified and lyophilized. The labeled conjugate was characterized using Fourier-transform infrared spectroscopy, ultraviolet-visible spectroscopy, ELS and DLS. In vitro cellular uptake and cytotoxic effects of BODIPY-labeled chitosan-AS1411 aptamer conjugates were evaluated using the XTT assay on T47D and HEK-293 cells and flow cytometry on T47D cells. Results: The data showed that uptake of BODIPY-labeled chitosan-AS1411 aptamer conjugate was satisfactory. Moreover, there was no statistically significant cytotoxicity of the conjugate on either cell line. Conclusion: The outcomes confirmed the potential application of this new targeted imaging agent as a novel cancer diagnostic agent for molecular imaging.
Collapse
Affiliation(s)
- Setareh Taki
- Department of Radiopharmacy, International Campus, School of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Mehdi Shafiee Ardestani
- Department of Radiopharmacy, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
33
|
Yao C, Singh SC, ElKabbash M, Zhang J, Lu H, Guo C. Quasi-rhombus metasurfaces as multimode interference couplers for controlling the propagation of modes in dielectric-loaded waveguides. OPTICS LETTERS 2019; 44:1654-1657. [PMID: 30933114 DOI: 10.1364/ol.44.001654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Accepted: 02/21/2019] [Indexed: 06/09/2023]
Abstract
Metasurfaces can control the propagation of free space and guided modes by imparting a phase gradient and modifying the mode propagation properties. Here we propose a design to control optical signals in a dielectric-loaded waveguide using quasi-rhombus gradient plasmonic metasurface structure. The metasurface acts as a multimode interference coupler that can focus, route, and split the propagating field in UV-visible spectral range. The ability to gain full control on waveguided mode with minimal footprint can significantly impact miniaturization of optical devices and photonic integrated circuits.
Collapse
|
34
|
Shanmuganathan R, Edison TNJI, LewisOscar F, Kumar P, Shanmugam S, Pugazhendhi A. Chitosan nanopolymers: An overview of drug delivery against cancer. Int J Biol Macromol 2019; 130:727-736. [PMID: 30771392 DOI: 10.1016/j.ijbiomac.2019.02.060] [Citation(s) in RCA: 128] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2018] [Revised: 01/31/2019] [Accepted: 02/11/2019] [Indexed: 01/26/2023]
Abstract
Cancer is becoming a major reason for death troll worldwide due to the difficulty in finding an efficient, cost effective and target specific method of treatment or diagnosis. The variety of cancer therapy used in the present scenario have painful side effects, low effectiveness and high cost, which are some major drawbacks of the available therapies. Apart from the conventional cancer therapy, nanotechnology has grown extremely towards treating cancer. Nanotechnology is a promising area of science focusing on developing target specific drug delivery system for carrying small or large active molecules to diagnose and treat cancer cells. In the field of nanoscience, Chitosan nanopolymers (ChNPs) are been emerging as a potential carrier due to their biodegradability and biocompatibility. The easy modification and versatility in administration route of ChNPs has attracted attention of researchers towards loading chemicals, proteins and gene drugs for target specific therapy of cancer cells. Therefore, the present review deals with the growing concern towards cancer therapy, introduction of ChNPs, mode of action and other strategies employed by researchers till date towards cancer treatment and diagnosis ChNPs.
Collapse
Affiliation(s)
| | | | | | - Ponnuchamy Kumar
- Food Chemistry and Molecular Cancer Biology Lab, Department of Animal Health and Management, Alagappa University, Karaikudi 630 003, India
| | | | - Arivalagan Pugazhendhi
- Innovative Green Product Synthesis and Renewable Environment Development Research Group, Faculty of Environment and Labour Safety, Ton Duc Thang University, Ho Chi Minh City, Viet Nam.
| |
Collapse
|
35
|
ERK Activity Imaging During Migration of Living Cells In Vitro and In Vivo. Int J Mol Sci 2019; 20:ijms20030679. [PMID: 30764494 PMCID: PMC6387119 DOI: 10.3390/ijms20030679] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 01/31/2019] [Accepted: 02/01/2019] [Indexed: 12/12/2022] Open
Abstract
Extracellular signal-regulated kinase (ERK) is a major downstream factor of the EGFR-RAS-RAF signalling pathway, and thus the role of ERK in cell growth has been widely examined. The development of biosensors based on fluorescent proteins has enabled us to measure ERK activities in living cells, both after growth factor stimulation and in its absence. Long-term imaging unexpectedly revealed the oscillative activation of ERK in an epithelial sheet or a cyst in vitro. Studies using transgenic mice expressing the ERK biosensor have revealed inhomogeneous ERK activities among various cell species. In vivo Förster (or fluorescence) resonance energy transfer (FRET) imaging shed light on a novel role of ERK in cell migration. Neutrophils and epithelial cells in various organs such as intestine, skin, lung and bladder showed spatio-temporally different cell dynamics and ERK activities. Experiments using inhibitors confirmed that ERK activities are required for various pathological responses, including epithelial repair after injuries, inflammation, and niche formation of cancer metastasis. In conclusion, biosensors for ERK will be powerful and valuable tools to investigate the roles of ERK in situ.
Collapse
|
36
|
Tsubakihara Y, Moustakas A. Epithelial-Mesenchymal Transition and Metastasis under the Control of Transforming Growth Factor β. Int J Mol Sci 2018; 19:ijms19113672. [PMID: 30463358 PMCID: PMC6274739 DOI: 10.3390/ijms19113672] [Citation(s) in RCA: 117] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Revised: 11/12/2018] [Accepted: 11/14/2018] [Indexed: 02/08/2023] Open
Abstract
Metastasis of tumor cells from primary sites of malignancy to neighboring stromal tissue or distant localities entails in several instances, but not in every case, the epithelial-mesenchymal transition (EMT). EMT weakens the strong adhesion forces between differentiated epithelial cells so that carcinoma cells can achieve solitary or collective motility, which makes the EMT an intuitive mechanism for the initiation of tumor metastasis. EMT initiates after primary oncogenic events lead to secondary secretion of cytokines. The interaction between tumor-secreted cytokines and oncogenic stimuli facilitates EMT progression. A classic case of this mechanism is the cooperation between oncogenic Ras and the transforming growth factor β (TGFβ). The power of TGFβ to mediate EMT during metastasis depends on versatile signaling crosstalk and on the regulation of successive waves of expression of many other cytokines and the progressive remodeling of the extracellular matrix that facilitates motility through basement membranes. Since metastasis involves many organs in the body, whereas EMT affects carcinoma cell differentiation locally, it has frequently been debated whether EMT truly contributes to metastasis. Despite controversies, studies of circulating tumor cells, studies of acquired chemoresistance by metastatic cells, and several (but not all) metastatic animal models, support a link between EMT and metastasis, with TGFβ, often being a common denominator in this link. This article aims at discussing mechanistic cases where TGFβ signaling and EMT facilitate tumor cell dissemination.
Collapse
Affiliation(s)
- Yutaro Tsubakihara
- Department of Medical Biochemistry and Microbiology, Science for Life Laboratory, Uppsala University, Box 582, SE-751 23 Uppsala, Sweden.
- Ludwig Institute for Cancer Research, Biomedical Center, Uppsala University, Box 595, SE-751 24 Uppsala, Sweden.
| | - Aristidis Moustakas
- Department of Medical Biochemistry and Microbiology, Science for Life Laboratory, Uppsala University, Box 582, SE-751 23 Uppsala, Sweden.
- Ludwig Institute for Cancer Research, Biomedical Center, Uppsala University, Box 595, SE-751 24 Uppsala, Sweden.
| |
Collapse
|
37
|
Imamura T, Saitou T, Kawakami R. In vivo optical imaging of cancer cell function and tumor microenvironment. Cancer Sci 2018; 109:912-918. [PMID: 29465804 PMCID: PMC5891206 DOI: 10.1111/cas.13544] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2017] [Revised: 02/12/2018] [Accepted: 02/14/2018] [Indexed: 02/04/2023] Open
Abstract
In vivo optical imaging using fluorescence and bioluminescence is superior to other methods in terms of spatiotemporal resolution and specificity, and represents a new technology for comprehensively studying living organisms in a less invasive way. Nowadays, it is an indispensable technology for studying many aspects of cancer biology, including dynamic invasion and metastasis. In observations of fluorescence or bioluminescence signals in a living body, various problems were caused by optical characteristics such as absorption and scattering and, therefore, observation of deep tissue was difficult. Recent developments in techniques for observation of the deep tissues of living animals overcame this difficulty by improving bioluminescent proteins, fluorescent proteins, and fluorescent dyes, as well as detection technologies such as two‐photon excitation microscopy. In the present review, we introduce these technological developments and in vivo application of bioluminescence and fluorescence imaging, and discuss future perspectives on the use of in vivo optical imaging technology in cancer research.
Collapse
Affiliation(s)
- Takeshi Imamura
- Department of Molecular Medicine for Pathogenesis, Ehime University Graduate School of Medicine, Toon, Japan.,Translational Research Center, Ehime University Hospital, Toon, Japan
| | - Takashi Saitou
- Department of Molecular Medicine for Pathogenesis, Ehime University Graduate School of Medicine, Toon, Japan.,Translational Research Center, Ehime University Hospital, Toon, Japan
| | - Ryosuke Kawakami
- Department of Molecular Medicine for Pathogenesis, Ehime University Graduate School of Medicine, Toon, Japan
| |
Collapse
|