1
|
Chakraborty A, Kamat SS. Lysophosphatidylserine: A Signaling Lipid with Implications in Human Diseases. Chem Rev 2024; 124:5470-5504. [PMID: 38607675 DOI: 10.1021/acs.chemrev.3c00701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/14/2024]
Abstract
Lysophosphatidylserine (lyso-PS) has emerged as yet another important signaling lysophospholipid in mammals, and deregulation in its metabolism has been directly linked to an array of human autoimmune and neurological disorders. It has an indispensable role in several biological processes in humans, and therefore, cellular concentrations of lyso-PS are tightly regulated to ensure optimal signaling and functioning in physiological settings. Given its biological importance, the past two decades have seen an explosion in the available literature toward our understanding of diverse aspects of lyso-PS metabolism and signaling and its association with human diseases. In this Review, we aim to comprehensively summarize different aspects of lyso-PS, such as its structure, biodistribution, chemical synthesis, and SAR studies with some synthetic analogs. From a biochemical perspective, we provide an exhaustive coverage of the diverse biological activities modulated by lyso-PSs, such as its metabolism and the receptors that respond to them in humans. We also briefly discuss the human diseases associated with aberrant lyso-PS metabolism and signaling and posit some future directions that may advance our understanding of lyso-PS-mediated mammalian physiology.
Collapse
Affiliation(s)
- Arnab Chakraborty
- Department of Biology, Indian Institute of Science Education and Research, Dr. Homi Bhabha Road, Pashan, Pune 411008, Maharashtra, India
| | - Siddhesh S Kamat
- Department of Biology, Indian Institute of Science Education and Research, Dr. Homi Bhabha Road, Pashan, Pune 411008, Maharashtra, India
| |
Collapse
|
2
|
Martynowycz MW, Andreev K, Mor A, Gidalevitz D. Cancer-Associated Gangliosides as a Therapeutic Target for Host Defense Peptide Mimics. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2023; 39:12541-12549. [PMID: 37647566 DOI: 10.1021/acs.langmuir.3c01008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/01/2023]
Abstract
Aberrant levels of glycolipids expressed on cellular surfaces are characteristic of different types of cancers. The oligomer of acylated lysine (OAK) mimicking antimicrobial peptides displays in vitro activity against human and murine melanoma cell lines with upregulated GD3 and GM3 gangliosides. Herein, we demonstrate the capability of OAK to intercalate into the sialo-oligosaccharides of DPPC/GD3 and DPPC/GM3 lipid monolayers using X-ray scattering. The lack of insertion into monolayers containing phosphatidylserine suggests that the mechanism of action by OAKs against glycosylated lipid membranes is not merely driven by charge effects. The fluorescence microscopy data demonstrates the membrane-lytic activity of OAK. Understanding the molecular basis for selectivity toward GD3 and GM3 gangliosides by antimicrobial lipopeptides will contribute to the development of novel therapies to cure melanoma and other malignancies.
Collapse
Affiliation(s)
- Michael W Martynowycz
- Department of Physics, Center for Molecular Study of Condensed Soft Matter (μCoSM), Pritzker Institute of Biomedical Science and Engineering, Illinois Institute of Technology, 10 W 35th Street, Chicago, Illinois 60616, United States
| | - Konstantin Andreev
- Department of Physics, Center for Molecular Study of Condensed Soft Matter (μCoSM), Pritzker Institute of Biomedical Science and Engineering, Illinois Institute of Technology, 10 W 35th Street, Chicago, Illinois 60616, United States
| | - Amram Mor
- Department of Biotechnology and Food Engineering, Technion─Israel Institute of Technology, Technion City, Haifa 32000, Israel
| | - David Gidalevitz
- Department of Physics, Center for Molecular Study of Condensed Soft Matter (μCoSM), Pritzker Institute of Biomedical Science and Engineering, Illinois Institute of Technology, 10 W 35th Street, Chicago, Illinois 60616, United States
| |
Collapse
|
3
|
Ho Shon I, Hogg PJ. Imaging of cell death in malignancy: Targeting pathways or phenotypes? Nucl Med Biol 2023; 124-125:108380. [PMID: 37598518 DOI: 10.1016/j.nucmedbio.2023.108380] [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: 06/19/2023] [Revised: 08/06/2023] [Accepted: 08/10/2023] [Indexed: 08/22/2023]
Abstract
Cell death is fundamental in health and disease and resisting cell death is a hallmark of cancer. Treatment of malignancy aims to cause cancer cell death, however current clinical imaging of treatment response does not specifically image cancer cell death but assesses this indirectly either by changes in tumor size (using x-ray computed tomography) or metabolic activity (using 2-[18F]fluoro-2-deoxy-glucose positron emission tomography). The ability to directly image tumor cell death soon after commencement of therapy would enable personalised response adapted approaches to cancer treatment that is presently not possible with current imaging, which is in many circumstances neither sufficiently accurate nor timely. Several cell death pathways have now been identified and characterised that present multiple potential targets for imaging cell death including externalisation of phosphatidylserine and phosphatidylethanolamine, caspase activation and La autoantigen redistribution. However, targeting one specific cell death pathway carries the risk of not detecting cell death by other pathways and it is now understood that cancer treatment induces cell death by different and sometimes multiple pathways. An alternative approach is targeting the cell death phenotype that is "agnostic" of the death pathway. Cell death phenotypes that have been targeted for cell death imaging include loss of plasma membrane integrity and dissipation of the mitochondrial membrane potential. Targeting the cell death phenotype may have the advantage of being a more sensitive and generalisable approach to cancer cell death imaging. This review describes and summarises the approaches and radiopharmaceuticals investigated for imaging cell death by targeting cell death pathways or cell death phenotype.
Collapse
Affiliation(s)
- Ivan Ho Shon
- Department of Nuclear Medicine and PET, Prince of Wales Hospital, Sydney, Australia; School of Clinical Medicine, UNSW Medicine & Health, Randwick Clinical Campus, UNSW Sydney, Australia.
| | - Philip J Hogg
- The Centenary Institute, University of Sydney, Sydney, Australia
| |
Collapse
|
4
|
Dirvelyte E, Bujanauskiene D, Jankaityte E, Daugelaviciene N, Kisieliute U, Nagula I, Budvytyte R, Neniskyte U. Genetically encoded phosphatidylserine biosensor for in vitro, ex vivo and in vivo labelling. Cell Mol Biol Lett 2023; 28:59. [PMID: 37501184 PMCID: PMC10373266 DOI: 10.1186/s11658-023-00472-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Accepted: 06/27/2023] [Indexed: 07/29/2023] Open
Abstract
BACKGROUND The dynamics of phosphatidylserine in the plasma membrane is a tightly regulated feature of eukaryotic cells. Phosphatidylserine (PS) is found preferentially in the inner leaflet of the plasma membrane. Disruption of this asymmetry leads to the exposure of phosphatidylserine on the cell surface and is associated with cell death, synaptic pruning, blood clotting and other cellular processes. Due to the role of phosphatidylserine in widespread cellular functions, an efficient phosphatidylserine probe is needed to study them. Currently, a few different phosphatidylserine labelling tools are available; however, these labels have unfavourable signal-to-noise ratios and are difficult to use in tissues due to limited permeability. Their application in living tissue requires injection procedures that damage the tissue and release damage-associated molecular patterns, which in turn stimulates phosphatidylserine exposure. METHODS For this reason, we developed a novel genetically encoded phosphatidylserine probe based on the C2 domain of the lactadherin (MFG-E8) protein, suitable for labelling exposed phosphatidylserine in various research models. We tested the C2 probe specificity to phosphatidylserine on hybrid bilayer lipid membranes by observing surface plasmon resonance angle shift. Then, we analysed purified fused C2 proteins on different cell culture lines or engineered AAVs encoding C2 probes on tissue cultures after apoptosis induction. For in vivo experiments, neurotropic AAVs were intravenously injected into perinatal mice, and after 2 weeks, brain slices were collected to observe C2-SNAP expression. RESULTS The biophysical analysis revealed the high specificity of the C2 probe for phosphatidylserine. The fused recombinant C2 proteins were suitable for labelling phosphatidylserine on the surface of apoptotic cells in various cell lines. We engineered AAVs and validated them in organotypic brain tissue cultures for non-invasive delivery of the genetically encoded C2 probe and showed that these probes were expressed in the brain in vivo after intravenous AAV delivery to mice. CONCLUSIONS We have demonstrated that the developed genetically encoded PS biosensor can be utilised in a variety of assays as a two-component system of C2 and C2m2 fusion proteins. This system allows for precise quantification and PS visualisation at directly specified threshold levels, enabling the evaluation of PS exposure in both physiological and cell death processes.
Collapse
Affiliation(s)
- Eimina Dirvelyte
- VU LSC-EMBL Partnership Institute for Genome Editing Technologies, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Daina Bujanauskiene
- VU LSC-EMBL Partnership Institute for Genome Editing Technologies, Life Sciences Center, Vilnius University, Vilnius, Lithuania
- Institute of Bioscience, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Evelina Jankaityte
- VU LSC-EMBL Partnership Institute for Genome Editing Technologies, Life Sciences Center, Vilnius University, Vilnius, Lithuania
- Institute of Biochemistry, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Neringa Daugelaviciene
- VU LSC-EMBL Partnership Institute for Genome Editing Technologies, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Ugne Kisieliute
- Institute of Bioscience, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Igor Nagula
- VU LSC-EMBL Partnership Institute for Genome Editing Technologies, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Rima Budvytyte
- VU LSC-EMBL Partnership Institute for Genome Editing Technologies, Life Sciences Center, Vilnius University, Vilnius, Lithuania
- Institute of Biochemistry, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Urte Neniskyte
- VU LSC-EMBL Partnership Institute for Genome Editing Technologies, Life Sciences Center, Vilnius University, Vilnius, Lithuania.
- Institute of Bioscience, Life Sciences Center, Vilnius University, Vilnius, Lithuania.
| |
Collapse
|
5
|
Kang Y, Zhai X, Lu S, Vuletic I, Wang L, Zhou K, Peng Z, Ren Q, Xie Z. A Hybrid Imaging Platform(CT/PET/FMI) for Evaluating Tumor Necrosis and Apoptosis in Real-Time. Front Oncol 2022; 12:772392. [PMID: 35814447 PMCID: PMC9257022 DOI: 10.3389/fonc.2022.772392] [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: 09/08/2021] [Accepted: 05/19/2022] [Indexed: 11/13/2022] Open
Abstract
Multimodality imaging is an advanced imaging tool for monitoring tumor behavior and therapy in vivo. In this study, we have developed a novel hybrid tri-modality system that includes two molecular imaging methods: positron emission computed tomography (PET) and fluorescence molecular imaging (FMI) and the anatomic imaging modality X-ray computed tomography (CT). The following paper describes the system development. Also, its imaging performance was tested in vitro (phantom) and in vivo, in Balb/c nude mice bearing a head and neck tumor xenograft treated with novel gene therapy [a new approach to the delivery of recombinant bacterial gene (IL-24-expressing strain)]. Using the tri-modality imaging system, we simultaneously monitored the therapeutic effect, including the apoptotic and necrotic induction within the tumor in vivo. The apoptotic induction was examined in real-time using an 18F-ML-10 tracer; the cell death was detected using ICG. A CT was used to evaluate the anatomical situation. An increased tumor inhibition (including tumor growth and tumor cell apoptosis) was observed in the treatment group compared to the control groups, which further confirmed the therapeutic effect of a new IL-24-expressing strain gene therapy on the tumor in vivo. By being able to offer concurrent morphological and functional information, our system is able to characterize malignant tissues more accurately. Therefore, this new tri-modality system (PET/CT/FMI) is an effective imaging tool for simultaneously investigating and monitoring tumor progression and therapy outcomes in vivo.
Collapse
Affiliation(s)
- Yulin Kang
- Institute of Environmental Information, Chinese Research Academy of Environmental Sciences, Beijing, China
- Department of Biomedical Engineering, College of Engineering, Peking University, Beijing, China
- *Correspondence: Qiushi Ren, ; Zhaoheng Xie, ; Yulin Kang,
| | - Xiaohui Zhai
- Department of Biomedical Engineering, College of Engineering, Peking University, Beijing, China
| | - Sifen Lu
- Precision Medicine Key Laboratory of Sichuan Province and Precision Medicine Center, West China Hospital, Sichuan University, Chengdu, China
| | - Ivan Vuletic
- Department of Biomedical Engineering, College of Engineering, Peking University, Beijing, China
| | - Lin Wang
- Department of Biomedical Engineering, College of Engineering, Peking University, Beijing, China
| | - Kun Zhou
- Department of Biomedical Engineering, College of Engineering, Peking University, Beijing, China
| | - Zhiqiang Peng
- State Key Laboratory of Proteomics, National Centre for Protein Sciences, Beijing Institute of Lifeomics, Bejing, China
| | - Qiushi Ren
- Department of Biomedical Engineering, College of Engineering, Peking University, Beijing, China
- *Correspondence: Qiushi Ren, ; Zhaoheng Xie, ; Yulin Kang,
| | - Zhaoheng Xie
- Department of Biomedical Engineering, College of Engineering, Peking University, Beijing, China
- *Correspondence: Qiushi Ren, ; Zhaoheng Xie, ; Yulin Kang,
| |
Collapse
|
6
|
Van de Wiele C, Maes A. Gamma camera imaging of apoptosis. Nucl Med Mol Imaging 2022. [DOI: 10.1016/b978-0-12-822960-6.00212-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
|
7
|
Jouberton E, Schmitt S, Maisonial-Besset A, Chautard E, Penault-Llorca F, Cachin F. Interest and Limits of [18F]ML-10 PET Imaging for Early Detection of Response to Conventional Chemotherapy. Front Oncol 2021; 11:789769. [PMID: 34988022 PMCID: PMC8722713 DOI: 10.3389/fonc.2021.789769] [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: 10/19/2021] [Accepted: 11/29/2021] [Indexed: 11/25/2022] Open
Abstract
One of the current challenges in oncology is to develop imaging tools to early detect the response to conventional chemotherapy and adjust treatment strategies when necessary. Several studies evaluating PET imaging with 2-deoxy-2-[18F]fluoro-D-glucose ([18F]FDG) as a predictive tool of therapeutic response highlighted its insufficient specificity and sensitivity. The [18F]FDG uptake reflects only tumor metabolic activity and not treatment-induced cell death, which seems to be relevant for therapeutic evaluation. Therefore, to evaluate this parameter in vivo, several cell death radiotracers have been developed in the last years. However, few of them have reached the clinical trials. This systematic review focuses on the use of [18F]ML-10 (2-(5-[18F]fluoropentyl)-2-methylmalonic acid) as radiotracer of apoptosis and especially as a measure of tumor response to treatment. A comprehensive literature review concerning the preclinical and clinical investigations conducted with [18F]ML-10 was performed. The abilities and applications of this radiotracer as well as its clinical relevance and limitations were discussed. Most studies highlighted a good ability of the radiotracer to target apoptotic cells. However, the increase in apoptosis during treatment did not correlate with the radiotracer tumoral uptake, even using more advanced image analysis (voxel-based analysis). [18F]ML-10 PET imaging does not meet current clinical expectations for early detection of the therapeutic response to conventional chemotherapy. This review has pointed out the challenges of applying various apoptosis imaging strategies in clinical trials, the current methodologies available for image analysis and the future of molecular imaging to assess this therapeutic response.
Collapse
Affiliation(s)
- Elodie Jouberton
- Service de Médecine Nucléaire, Centre Jean PERRIN, Clermont-Ferrand, France
- Imagerie Moléculaire et Stratégies Théranostiques, UMR1240, Université Clermont Auvergne, INSERM, Clermont-Ferrand, France
- *Correspondence: Elodie Jouberton,
| | - Sébastien Schmitt
- Imagerie Moléculaire et Stratégies Théranostiques, UMR1240, Université Clermont Auvergne, INSERM, Clermont-Ferrand, France
| | - Aurélie Maisonial-Besset
- Imagerie Moléculaire et Stratégies Théranostiques, UMR1240, Université Clermont Auvergne, INSERM, Clermont-Ferrand, France
| | - Emmanuel Chautard
- Imagerie Moléculaire et Stratégies Théranostiques, UMR1240, Université Clermont Auvergne, INSERM, Clermont-Ferrand, France
- Service de Pathologie, Centre Jean PERRIN, Clermont-Ferrand, France
| | - Frédérique Penault-Llorca
- Imagerie Moléculaire et Stratégies Théranostiques, UMR1240, Université Clermont Auvergne, INSERM, Clermont-Ferrand, France
- Service de Pathologie, Centre Jean PERRIN, Clermont-Ferrand, France
| | - Florent Cachin
- Service de Médecine Nucléaire, Centre Jean PERRIN, Clermont-Ferrand, France
- Imagerie Moléculaire et Stratégies Théranostiques, UMR1240, Université Clermont Auvergne, INSERM, Clermont-Ferrand, France
| |
Collapse
|
8
|
Li J, Song M, Wen H, Zhang Y, Li Y, Lyu L, Wang X, Qi X. Gonadal lipidomics profile of an ovoviviparity teleost, black rockfish, during gonadal development. FISH PHYSIOLOGY AND BIOCHEMISTRY 2021; 47:811-828. [PMID: 33694040 DOI: 10.1007/s10695-021-00936-7] [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: 10/15/2020] [Accepted: 02/10/2021] [Indexed: 06/12/2023]
Abstract
In order to study the variation of gonad lipidomics during reproductive cycle, black rockfish was employed as the research model in the present study. Using histology, lipidomics, and qPCR, the profile of gonad lipidomics and the expression levels of related genes during different developmental stages were detected and analyzed to show the potential regulatory network of lipid metabolism. Based on Ultra High-Performance Liquid Tandem Chromatography Quadrupole Time of Flight Mass Spectrometry (UHPLC-QTOFMS), four significant differential glycerophospholipid metabolic pathways including phosphatidylcholine (PC), phosphatidylethanolamine (PE), phosphatidylserine (PS), and phosphatidic acid (PA) were enriched by KEGG. Pathway-related enzyme-coding genes, including phosphatidylserine decarboxylase (pisd), phosphatidylserine synthase (ptdss1, ptdss2), and phospholipase D (pld1, pld2) were identified from the whole genome data and confirmed by cloning. The expression profiles of these genes were tested by qPCR in the tissues and gonads in developmental stages, and we found that pisd, pld, and ptdss genes were all downregulated through the developmental process in the brain of male, and the latter two genes were upregulated in the liver and testis at stage IV, which were the opposite trend observed in the female. Thus, our findings would be helpful in further understanding the substance metabolism and regulation during gonad development in ovoviviparity teleosts.
Collapse
Affiliation(s)
- Jianshuang Li
- Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao, People's Republic of China
| | - Min Song
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, People's Republic of China
| | - Haishen Wen
- Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao, People's Republic of China
| | - Ying Zhang
- Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao, People's Republic of China
| | - Yun Li
- Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao, People's Republic of China
| | - Likang Lyu
- Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao, People's Republic of China
| | - Xiaojie Wang
- Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao, People's Republic of China
| | - Xin Qi
- Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao, People's Republic of China.
| |
Collapse
|
9
|
So IS, Kang JH, Hong JW, Sung S, Hasan AF, Sa KH, Han SW, Kim IS, Kang YM. A novel apoptosis probe, cyclic ApoPep-1, for in vivo imaging with multimodal applications in chronic inflammatory arthritis. Apoptosis 2021; 26:209-218. [PMID: 33655467 DOI: 10.1007/s10495-021-01659-z] [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] [Accepted: 02/04/2021] [Indexed: 11/26/2022]
Abstract
Apoptosis plays an essential role in the pathophysiologic processes of rheumatoid arthritis. A molecular probe that allows spatiotemporal observation of apoptosis in vitro, in vivo, and ex vivo concomitantly would be useful to monitoring or predicting pathophysiologic stages. In this study we investigated whether cyclic apoptosis-targeting peptide-1 (CApoPep-1) can be used as an apoptosis imaging probe in inflammatory arthritis. We tested the utility of CApoPep-1 for detecting apoptotic immune cells in vitro and ex vivo using flow cytometry and immunofluorescence. The feasibility of visualizing and quantifying apoptosis using this probe was evaluated in a murine collagen-induced arthritis (CIA) model, especially after treatment. CApoPep-1 peptide may successfully replace Annexin V for in vitro and terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) assay for ex vivo in the measurement of apoptotic cells, thus function as a sensitive probe enough to be used clinically. In vivo imaging in CIA mice revealed that CApoPep-1 had 42.9 times higher fluorescence intensity than Annexin V for apoptosis quantification. Furthermore, it may be used as an imaging probe for early detection of apoptotic response in situ after treatment. The CApoPep-1 signal was mostly co-localized with the TUNEL signal (69.6% of TUNEL+ cells) in defined cell populations in joint tissues of CIA mice. These results demonstrate that CApoPep-1 is sufficiently sensitive to be used as an apoptosis imaging probe for multipurpose applications which could detect the same target across in vitro, in vivo, to ex vivo in inflammatory arthritis.
Collapse
Affiliation(s)
- In-Seop So
- Department of Internal Medicine (Rheumatology), Kyungpook National University School of Medicine, 680 Gukchaebosang-ro, Junggu, Daegu, 41944, Republic of Korea
- Cell and Matrix Research Institute, Kyungpook National University, Daegu, Republic of Korea
| | - Jin Hee Kang
- Department of Internal Medicine (Rheumatology), Kyungpook National University School of Medicine, 680 Gukchaebosang-ro, Junggu, Daegu, 41944, Republic of Korea
- Cell and Matrix Research Institute, Kyungpook National University, Daegu, Republic of Korea
- Department of Biochemistry and Cell Biology, Kyungpook National University School of Medicine, Daegu, Republic of Korea
| | - Jung Wan Hong
- Department of Internal Medicine (Rheumatology), Kyungpook National University School of Medicine, 680 Gukchaebosang-ro, Junggu, Daegu, 41944, Republic of Korea
- Cell and Matrix Research Institute, Kyungpook National University, Daegu, Republic of Korea
| | - Shijin Sung
- Department of Internal Medicine (Rheumatology), Kyungpook National University School of Medicine, 680 Gukchaebosang-ro, Junggu, Daegu, 41944, Republic of Korea
- Cell and Matrix Research Institute, Kyungpook National University, Daegu, Republic of Korea
- Department of Biochemistry and Cell Biology, Kyungpook National University School of Medicine, Daegu, Republic of Korea
| | - Al Faruque Hasan
- Department of Internal Medicine (Rheumatology), Kyungpook National University School of Medicine, 680 Gukchaebosang-ro, Junggu, Daegu, 41944, Republic of Korea
- Cell and Matrix Research Institute, Kyungpook National University, Daegu, Republic of Korea
- Department of Biochemistry and Cell Biology, Kyungpook National University School of Medicine, Daegu, Republic of Korea
| | - Keum Hee Sa
- Department of Internal Medicine (Rheumatology), Kyungpook National University School of Medicine, 680 Gukchaebosang-ro, Junggu, Daegu, 41944, Republic of Korea
- Cell and Matrix Research Institute, Kyungpook National University, Daegu, Republic of Korea
- Department of Biochemistry and Cell Biology, Kyungpook National University School of Medicine, Daegu, Republic of Korea
| | - Seung Woo Han
- Department of Internal Medicine (Rheumatology), Kyungpook National University School of Medicine, 680 Gukchaebosang-ro, Junggu, Daegu, 41944, Republic of Korea
| | - In San Kim
- Cell and Matrix Research Institute, Kyungpook National University, Daegu, Republic of Korea
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology, Seoul, Republic of Korea
| | - Young Mo Kang
- Department of Internal Medicine (Rheumatology), Kyungpook National University School of Medicine, 680 Gukchaebosang-ro, Junggu, Daegu, 41944, Republic of Korea.
- Cell and Matrix Research Institute, Kyungpook National University, Daegu, Republic of Korea.
- Department of Biochemistry and Cell Biology, Kyungpook National University School of Medicine, Daegu, Republic of Korea.
| |
Collapse
|
10
|
Chaudhry F, Kawai H, Johnson KW, Narula N, Shekhar A, Chaudhry F, Nakahara T, Tanimoto T, Kim D, Adapoe MKMY, Blankenberg FG, Mattis JA, Pak KY, Levy PD, Ozaki Y, Arbustini E, Strauss HW, Petrov A, Fuster V, Narula J. Molecular Imaging of Apoptosis in Atherosclerosis by Targeting Cell Membrane Phospholipid Asymmetry. J Am Coll Cardiol 2021; 76:1862-1874. [PMID: 33059832 DOI: 10.1016/j.jacc.2020.08.047] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 08/20/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND Apoptosis in atherosclerotic lesions contributes to plaque vulnerability by lipid core enlargement and fibrous cap attenuation. Apoptosis is associated with exteriorization of phosphatidylserine (PS) and phosphatidylethanolamine (PE) on the cell membrane. Although PS-avid radiolabeled annexin-V has been employed for molecular imaging of high-risk plaques, PE-targeted imaging in atherosclerosis has not been studied. OBJECTIVES This study sought to evaluate the feasibility of molecular imaging with PE-avid radiolabeled duramycin in experimental atherosclerotic lesions in a rabbit model and compare duramycin targeting with radiolabeled annexin-V. METHODS Of the 27 rabbits, 21 were fed high-cholesterol, high-fat diet for 16 weeks. Nine of the 21 rabbits received 99mTc-duramycin (test group), 6 received 99mTc-linear duramycin (duramycin without PE-binding capability, negative radiotracer control group), and 6 received 99mTc-annexin-V for radionuclide imaging. The remaining normal chow-fed 6 animals (disease control group) received 99mTc-duramycin. In vivo microSPECT/microCT imaging was performed, and the aortas were explanted for ex vivo imaging and for histological characterization of atherosclerosis. RESULTS A significantly higher duramycin uptake was observed in the test group compared with that of disease control and negative radiotracer control animals; duramycin uptake was also significantly higher than the annexin-V uptake. Quantitative duramycin uptake, represented as the square root of percent injected dose per cm (√ID/cm) of abdominal aorta was >2-fold higher in atherosclerotic lesions in test group (0.08 ± 0.01%) than in comparable regions of disease control animals (0.039 ± 0.0061%, p = 3.70·10-8). Mean annexin uptake (0.060 ± 0.010%) was significantly lower than duramycin (p = 0.001). Duramycin uptake corresponded to the lesion severity and macrophage burden. The radiation burden to the kidneys was substantially lower with duramycin (0.49% ID/g) than annexin (5.48% ID/g; p = 4.00·10-4). CONCLUSIONS Radiolabeled duramycin localizes in lipid-rich areas with high concentration of apoptotic macrophages in the experimental atherosclerosis model. Duramycin uptake in atherosclerotic lesions was significantly greater than annexin-V uptake and produced significantly lower radiation burden to nontarget organs.
Collapse
Affiliation(s)
- Farhan Chaudhry
- Icahn School of Medicine at Mount Sinai, New York, New York; Wayne State University School of Medicine, Detroit, Michigan
| | - Hideki Kawai
- Icahn School of Medicine at Mount Sinai, New York, New York; Department of Cardiology, Fujita Health University, Toyoake, Aichi, Japan
| | - Kipp W Johnson
- Icahn School of Medicine at Mount Sinai, New York, New York
| | - Navneet Narula
- New York University Langone Medical Center, New York, New York
| | - Aditya Shekhar
- Icahn School of Medicine at Mount Sinai, New York, New York
| | | | | | | | - Dongbin Kim
- Icahn School of Medicine at Mount Sinai, New York, New York
| | | | | | - Jeffrey A Mattis
- Molecular Targeting Technologies, Inc., West Chester, Pennsylvania
| | - Koon Y Pak
- Molecular Targeting Technologies, Inc., West Chester, Pennsylvania
| | - Phillip D Levy
- Wayne State University School of Medicine, Detroit, Michigan
| | - Yukio Ozaki
- Department of Cardiology, Fujita Health University, Toyoake, Aichi, Japan
| | | | - H William Strauss
- Icahn School of Medicine at Mount Sinai, New York, New York; Memorial Sloan Kettering Cancer Center, New York, New York
| | - Artiom Petrov
- Icahn School of Medicine at Mount Sinai, New York, New York.
| | - Valentin Fuster
- Icahn School of Medicine at Mount Sinai, New York, New York; Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
| | - Jagat Narula
- Icahn School of Medicine at Mount Sinai, New York, New York
| |
Collapse
|
11
|
Lin G, Mu Q, Revia R, Stephen Z, Jeon M, Zhang M. A highly selective iron oxide-based imaging nanoparticle for long-term monitoring of drug-induced tumor cell apoptosis. Biomater Sci 2021; 9:471-481. [PMID: 32662460 PMCID: PMC7855362 DOI: 10.1039/d0bm00518e] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
The ability to visualize and quantify apoptosis in vivo is critical to monitoring the disease response to treatment and providing prognostic information. However, the application of current apoptosis labeling probes faces significant challenges including nonspecific tissue uptake, inefficient apoptotic cell labeling and short monitoring windows. Here we report a highly specific apoptosis labeling nanoparticle (NP) probe with Pisum sativum agglutinin (PSA) as a tumor targeting ligand for prolonged in vivo apoptosis imaging. The NP (namely, IONP-Neu-PSA) consists of a magnetic iron oxide core (IONP) conjugated with PSA, and a reporter fluorophore. IONP-Neu-PSA demonstrated minimal cytotoxicity and high labeling specificity towards apoptotic cells in vitro. When applied in vivo, IONP-Neu-PSA tracks apoptotic tumors for a prolonged period of two weeks under near-IR imaging with low background noise. Moreover, IONP-Neu-PSA possesses T2 contrast enhancing properties that can potentially enable apoptosis detection by magnetic resonance imaging (MRI). The high specificity for apoptotic cells, sustained fluorescence signals, and non-invasive imaging capability exhibited by IONP-Neu-PSA make it a versatile tool for cancer treatment monitoring and pathological research.
Collapse
Affiliation(s)
- Guanyou Lin
- Department of Materials Sciences and Engineering, University of Washington, Seattle, Washington 98195, USA.
| | - Qingxin Mu
- Department of Pharmaceutics, University of Washington, Seattle, Washington 98195, USA
| | - Richard Revia
- Department of Materials Sciences and Engineering, University of Washington, Seattle, Washington 98195, USA.
| | - Zachary Stephen
- Department of Materials Sciences and Engineering, University of Washington, Seattle, Washington 98195, USA.
| | - Mike Jeon
- Department of Materials Sciences and Engineering, University of Washington, Seattle, Washington 98195, USA.
| | - Miqin Zhang
- Department of Materials Sciences and Engineering, University of Washington, Seattle, Washington 98195, USA. and Department of Neurological Surgery, University of Washington, Seattle, Washington 98195, USA
| |
Collapse
|
12
|
Qi G, Wang J, Ma K, Zhang Y, Zhang J, Xu W, Jin Y. Label-Free Single-Particle Surface-Enhanced Raman Spectroscopy Detection of Phosphatidylserine Externalization on Cell Membranes with Multifunctional Micron-Nano Composite Probes. Anal Chem 2021; 93:2183-2190. [DOI: 10.1021/acs.analchem.0c04038] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Guohua Qi
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, Jilin, P. R. China
| | - Jiafeng Wang
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, Jilin, P. R. China
- Department of Endodontics, School and Hospital of Stomatology, Jilin University, Changchun 130021, Jilin, P.R. China
| | - Kongshuo Ma
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, Jilin, P. R. China
- University of Science and Technology of China, Hefei 230026, P. R. China
| | - Ying Zhang
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, Jilin, P. R. China
- University of Science and Technology of China, Hefei 230026, P. R. China
| | - Jie Zhang
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, Jilin, P. R. China
| | - Weiqing Xu
- State Key Laboratory of Supramolecular Structure and Materials, Jilin University, 2699 Qianjin Avenue, Changchun 130012, P. R. China
| | - Yongdong Jin
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, Jilin, P. R. China
- University of Science and Technology of China, Hefei 230026, P. R. China
| |
Collapse
|
13
|
Li Y, Bouza M, Wu C, Guo H, Huang D, Doron G, Temenoff JS, Stecenko AA, Wang ZL, Fernández FM. Sub-nanoliter metabolomics via mass spectrometry to characterize volume-limited samples. Nat Commun 2020; 11:5625. [PMID: 33159052 PMCID: PMC7648103 DOI: 10.1038/s41467-020-19444-y] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 09/28/2020] [Indexed: 01/18/2023] Open
Abstract
The human metabolome provides a window into the mechanisms and biomarkers of various diseases. However, because of limited availability, many sample types are still difficult to study by metabolomic analyses. Here, we present a mass spectrometry (MS)-based metabolomics strategy that only consumes sub-nanoliter sample volumes. The approach consists of combining a customized metabolomics workflow with a pulsed MS ion generation method, known as triboelectric nanogenerator inductive nanoelectrospray ionization (TENGi nanoESI) MS. Samples tested with this approach include exhaled breath condensate collected from cystic fibrosis patients as well as in vitro-cultured human mesenchymal stromal cells. Both test samples are only available in minimum amounts. Experiments show that picoliter-volume spray pulses suffice to generate high-quality spectral fingerprints, which increase the information density produced per unit sample volume. This TENGi nanoESI strategy has the potential to fill in the gap in metabolomics where liquid chromatography-MS-based analyses cannot be applied. Our method opens up avenues for future investigations into understanding metabolic changes caused by diseases or external stimuli.
Collapse
Affiliation(s)
- Yafeng Li
- School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Marcos Bouza
- School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Changsheng Wu
- School of Materials Science and Engineering, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Hengyu Guo
- School of Materials Science and Engineering, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Danning Huang
- School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Gilad Doron
- W.H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, 30332, USA
| | - Johnna S Temenoff
- W.H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, 30332, USA.,Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Arlene A Stecenko
- Emory + Children's Center for Cystic Fibrosis and Airways Disease Research and Department of Pediatrics, Emory University School of Medicine and Children's Healthcare of Atlanta, Atlanta, GA, 30322, USA
| | - Zhong Lin Wang
- School of Materials Science and Engineering, Georgia Institute of Technology, Atlanta, GA, 30332, USA.,Beijing Institute of Nanoenergy and Nanosystems, Chinese Academy of Sciences, Beijing, 100083, China
| | - Facundo M Fernández
- School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, GA, 30332, USA. .,Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, 30332, USA.
| |
Collapse
|
14
|
Jin KT, Yao JY, Ying XJ, Lin Y, Chen YF. Nanomedicine and Early Cancer Diagnosis: Molecular Imaging using Fluorescence Nanoparticles. Curr Top Med Chem 2020; 20:2737-2761. [PMID: 32962614 DOI: 10.2174/1568026620666200922112640] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 10/15/2020] [Accepted: 10/30/2020] [Indexed: 12/15/2022]
Abstract
Incorporating nanotechnology into fluorescent imaging and magnetic resonance imaging (MRI) has shown promising potential for accurate diagnosis of cancer at an earlier stage than the conventional imaging modalities. Molecular imaging (MI) aims to quantitatively characterize, visualize, and measure the biological processes or living cells at molecular and genetic levels. MI modalities have been exploited in different applications including noninvasive determination and visualization of diseased tissues, cell trafficking visualization, early detection, treatment response monitoring, and in vivo visualization of living cells. High-affinity molecular probe and imaging modality to detect the probe are the two main requirements of MI. Recent advances in nanotechnology and allied modalities have facilitated the use of nanoparticles (NPs) as MI probes. Within the extensive group of NPs, fluorescent NPs play a prominent role in optical molecular imaging. The fluorescent NPs used in molecular and cellular imaging can be categorized into three main groups including quantum dots (QDs), upconversion, and dyedoped NPs. Fluorescent NPs have great potential in targeted theranostics including cancer imaging, immunoassay- based cells, proteins and bacteria detections, imaging-guided surgery, and therapy. Fluorescent NPs have shown promising potentials for drug and gene delivery, detection of the chromosomal abnormalities, labeling of DNA, and visualizing DNA replication dynamics. Multifunctional NPs have been successfully used in a single theranostic modality integrating diagnosis and therapy. The unique characteristics of multifunctional NPs make them potential theranostic agents that can be utilized concurrently for diagnosis and therapy. This review provides the state of the art of the applications of nanotechnologies in early cancer diagnosis focusing on fluorescent NPs, their synthesis methods, and perspectives in clinical theranostics.
Collapse
Affiliation(s)
- Ke-Tao Jin
- Department of Colorectal Surgery, Jinhua Hosptial, Zhejiang University School of Medicine, Jinhua, Zhejiang 321000, P.R. China
| | - Jia-Yu Yao
- Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Zhejiang Provincial People's Hospital (People's Hospital of Hangzhou Medical College), Hangzhou 310014, P.R. China,Clinical Research Institute, Zhejiang Provincial People's Hospital (People's Hospital Hangzhou Medical College), Hangzhou 310014, P.R. China
| | - Xiao-Jiang Ying
- Department of Colorectal Surgery Shaoxing People's Hospital (Shaoxing Hospital, Zhejiang University School of Medicine), Shaoxing, 312000, Zhejiang Province, P.R. China
| | - Yan Lin
- Department of Gastroenterology, Zhejiang Provincial People’s Hospital (People’s Hospital of Hangzhou Medical College), Hangzhou 310014, Zhejiang Province, P.R China
| | - Yun-Fang Chen
- Department of Stomatology, Zhejiang Provincial People’s Hospital (People’s Hospital of Hangzhou Medical College), Hangzhou 310014, P.R. China
| |
Collapse
|
15
|
Hosseini ES, Nikkhah M, Hamidieh AA, Fearnhead HO, Concordet JP, Hosseinkhani S. The Lumiptosome, an engineered luminescent form of the apoptosome can report cell death by using the same Apaf-1 dependent pathway. J Cell Sci 2020; 133:133/10/jcs242636. [DOI: 10.1242/jcs.242636] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Accepted: 02/28/2020] [Indexed: 01/09/2023] Open
Abstract
ABSTRACT
Detection of the apoptosis signature becomes central in understanding cell death modes. We present here a whole-cell biosensor that detects Apaf-1 association and apoptosome formation using a split-luciferase complementary assay. Fusion of N-terminal (Nluc) and C-terminal (Cluc)-fragments of firefly luciferase to the N-terminus of human Apaf-1 was performed in HEK293 cells by using CRISPR-Cas9 technology. This resulted in a luminescent form of the apoptosome that we named ‘Lumiptosome’. During Apaf-1 gene editing, a high number of knock-in events were observed without selection, suggesting that the Apaf-1 locus is important for the integration of exogenous transgenes. Since activation of caspase-9 is directly dependent on the apoptosome formation, measured reconstitution of luciferase activity should result from the cooperative association of Nluc-Apaf-1 and Cluc-Apaf-1. Time-response measurements also confirmed that formation of the apoptosome occurs prior to activation of caspase-3. Additionally, overexpression of the Bcl2 apoptosis regulator in transgenic and normal HEK293 cells confirmed that formation of the Lumiptosome depends on release of cytochrome c. Thus, HEK293 cells that stably express the Lumiptosome can be utilized to screen pro- and anti-apoptotic drugs, and to examine Apaf-1-dependent cellular pathways.
Collapse
Affiliation(s)
- Elaheh Sadat Hosseini
- Department of Nanobiotechnology, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
- Department of Nanobiotechnology, Pharmacology and Therapeutics, School of Medicine, NUI, Galway, Ireland
- Department of Nanobiotechnology, Museum National d'Histoire Naturelle, Laboratoire Structure et Instabilité des Génomes - INSERM U1154 - CNRS 7196; Laboratoire de Biophysique, Sorbonne Universités, Paris, F-75231, France
| | - Maryam Nikkhah
- Department of Nanobiotechnology, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Amir Ali Hamidieh
- Pediatric Cell Therapy Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Howard O. Fearnhead
- Department of Nanobiotechnology, Pharmacology and Therapeutics, School of Medicine, NUI, Galway, Ireland
| | - Jean-Paul Concordet
- Department of Nanobiotechnology, Museum National d'Histoire Naturelle, Laboratoire Structure et Instabilité des Génomes - INSERM U1154 - CNRS 7196; Laboratoire de Biophysique, Sorbonne Universités, Paris, F-75231, France
| | - Saman Hosseinkhani
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran 14115-175, Iran
| |
Collapse
|
16
|
Abstract
One major characteristic of programmed cell death (apoptosis) results in the increased expression of phosphatidylserine (PS) on the outer membrane of dying cells. Consequently, PS represents an excellent target for non-invasive imaging of apoptosis by single-photon emission computed tomography (SPECT) and positron emission tomography (PET). Annexin V is a 36 kDa protein which binds with high affinity to PS in the presence of Ca2+ ions. This makes radiolabeled annexins valuable apoptosis imaging agents for clinical and biomedical research applications for monitoring apoptosis in vivo. However, the use of radiolabeled annexin V for in vivo imaging of cell death has been met with a variety of challenges which have prevented its translation into the clinic. These difficulties include: complicated and time-consuming radiolabeling procedures, sub-optimal biodistribution, inadequate pharmacokinetics leading to poor tumour-to-blood contrast ratios, reliance upon Ca2+ concentrations in vivo, low tumor tissue penetration, and an incomplete understanding of what constitutes the best imaging protocol following induction of apoptosis. Therefore, new concepts and improved strategies for the development of PS-binding radiotracers are needed. Radiolabeled PS-binding peptides and various Zn(II) complexes as phosphate chemosensors offer an innovative strategy for radionuclide-based molecular imaging of apoptosis with PET and SPECT. Radiolabeled peptides and Zn(II) complexes provide several advantages over annexin V including better pharmacokinetics due to their smaller size, better availability, simpler synthesis and radiolabeling strategies as well as facilitated tissue penetration due to their smaller size and faster blood clearance profile allowing for optimized image contrast. In addition, peptides can be structurally modified to improve metabolic stability along with other pharmacokinetic and pharmacodynamic properties. The present review will summarize the current status of radiolabeled annexins, peptides and Zn(II) complexes developed as radiotracers for imaging apoptosis through targeting PS utilizing PET and SPECT imaging.
Collapse
|
17
|
Brand C, Greve B, Bölling T, Eich HT, Willich N, Harrach S, Hintelmann H, Lenz G, Mesters RM, Kessler T, Schliemann C, Berdel WE, Schwöppe C. Radiation synergizes with antitumor activity of CD13-targeted tissue factor in a HT1080 xenograft model of human soft tissue sarcoma. PLoS One 2020; 15:e0229271. [PMID: 32084238 PMCID: PMC7034830 DOI: 10.1371/journal.pone.0229271] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2019] [Accepted: 02/03/2020] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Truncated tissue factor (tTF) retargeted by NGR-peptides to aminopeptidase N (CD13) in tumor vasculature is effective in experimental tumor therapy. tTF-NGR induces tumor growth inhibition in a variety of human tumor xenografts of different histology. To improve on the therapeutic efficacy we have combined tTF-NGR with radiotherapy. METHODS Serum-stimulated human umbilical vein endothelial cells (HUVEC) and human HT1080 sarcoma cells were irradiated in vitro, and upregulated early-apoptotic phosphatidylserine (PS) on the cell surface was measured by standard flow cytometry. Increase of cellular procoagulant function in relation to irradiation and PS cell surface concentration was measured in a tTF-NGR-dependent Factor X activation assay. In vivo experiments with CD-1 athymic mice bearing human HT1080 sarcoma xenotransplants were performed to test the systemic therapeutic effects of tTF-NGR on tumor growth alone or in combination with regional tumor ionizing radiotherapy. RESULTS As shown by flow cytometry with HUVEC and HT1080 sarcoma cells in vitro, irradiation with 4 and 6 Gy in the process of apoptosis induced upregulation of PS presence on the outer surface of both cell types. Proapoptotic HUVEC and HT1080 cells both showed significantly higher procoagulant efficacy on the basis of equimolar concentrations of tTF-NGR as measured by FX activation. This effect can be reverted by masking of PS with Annexin V. HT1080 human sarcoma xenografted tumors showed shrinkage induced by combined regional radiotherapy and systemic tTF-NGR as compared to growth inhibition achieved by either of the treatment modalities alone. CONCLUSIONS Irradiation renders tumor and tumor vascular cells procoagulant by PS upregulation on their outer surface and radiotherapy can significantly improve the therapeutic antitumor efficacy of tTF-NGR in the xenograft model used. This synergistic effect will influence design of future clinical combination studies.
Collapse
Affiliation(s)
- Caroline Brand
- Department of Medicine A, Hematology, Oncology, University Hospital Muenster, Muenster, Germany
| | - Burkhard Greve
- Department of Radiation Therapy and Radiation-Oncology, University Hospital Muenster, Muenster, Germany
| | - Tobias Bölling
- Department of Radiation Therapy and Radiation-Oncology, University Hospital Muenster, Muenster, Germany
| | - Hans T. Eich
- Department of Radiation Therapy and Radiation-Oncology, University Hospital Muenster, Muenster, Germany
| | - Normann Willich
- Department of Radiation Therapy and Radiation-Oncology, University Hospital Muenster, Muenster, Germany
| | - Saliha Harrach
- Department of Medicine A, Hematology, Oncology, University Hospital Muenster, Muenster, Germany
| | - Heike Hintelmann
- Department of Medicine A, Hematology, Oncology, University Hospital Muenster, Muenster, Germany
| | - Georg Lenz
- Department of Medicine A, Hematology, Oncology, University Hospital Muenster, Muenster, Germany
| | - Rolf M. Mesters
- Department of Medicine A, Hematology, Oncology, University Hospital Muenster, Muenster, Germany
| | - Torsten Kessler
- Department of Medicine A, Hematology, Oncology, University Hospital Muenster, Muenster, Germany
| | - Christoph Schliemann
- Department of Medicine A, Hematology, Oncology, University Hospital Muenster, Muenster, Germany
| | - Wolfgang E. Berdel
- Department of Medicine A, Hematology, Oncology, University Hospital Muenster, Muenster, Germany
- * E-mail: (CSch); (WEB)
| | - Christian Schwöppe
- Department of Medicine A, Hematology, Oncology, University Hospital Muenster, Muenster, Germany
- * E-mail: (CSch); (WEB)
| |
Collapse
|
18
|
Chi X, Nguyen D, Pemberton JM, Osterlund EJ, Liu Q, Brahmbhatt H, Zhang Z, Lin J, Leber B, Andrews DW. The carboxyl-terminal sequence of bim enables bax activation and killing of unprimed cells. eLife 2020; 9:44525. [PMID: 31976859 PMCID: PMC6980855 DOI: 10.7554/elife.44525] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Accepted: 01/03/2020] [Indexed: 12/12/2022] Open
Abstract
The Bcl-2 family BH3 protein Bim promotes apoptosis at mitochondria by activating the pore-forming proteins Bax and Bak and by inhibiting the anti-apoptotic proteins Bcl-XL, Bcl-2 and Mcl-1. Bim binds to these proteins via its BH3 domain and to the mitochondrial membrane by a carboxyl-terminal sequence (CTS). In cells killed by Bim, the expression of a Bim mutant in which the CTS was deleted (BimL-dCTS) triggered apoptosis that correlated with inhibition of anti-apoptotic proteins being sufficient to permeabilize mitochondria isolated from the same cells. Detailed analysis of the molecular mechanism demonstrated that BimL-dCTS inhibited Bcl-XL but did not activate Bax. Examination of additional point mutants unexpectedly revealed that the CTS of Bim directly interacts with Bax, is required for physiological concentrations of Bim to activate Bax and that different residues in the CTS enable Bax activation and binding to membranes.
Collapse
Affiliation(s)
- Xiaoke Chi
- Department of Chemistry and Chemical Biology, McMaster University, Hamilton, Canada.,Biological Sciences, Sunnybrook Research Institute, Toronto, Canada
| | - Dang Nguyen
- Biological Sciences, Sunnybrook Research Institute, Toronto, Canada.,Department of Medical Biophysics, University of Toronto, Ontario, Canada
| | - James M Pemberton
- Biological Sciences, Sunnybrook Research Institute, Toronto, Canada.,Department of Medical Biophysics, University of Toronto, Ontario, Canada
| | - Elizabeth J Osterlund
- Biological Sciences, Sunnybrook Research Institute, Toronto, Canada.,Department of Biochemistry, University of Toronto, Toronto, Canada
| | - Qian Liu
- Biological Sciences, Sunnybrook Research Institute, Toronto, Canada
| | - Hetal Brahmbhatt
- Department of Chemistry and Chemical Biology, McMaster University, Hamilton, Canada.,Biological Sciences, Sunnybrook Research Institute, Toronto, Canada.,Department of Medicine, McMaster University, Hamilton, Canada
| | - Zhi Zhang
- Department of Biochemistry, University of Oklahoma Health Sciences Center, Oklahoma City, United States.,Molecular Biology and Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, United States
| | - Jialing Lin
- Department of Biochemistry, University of Oklahoma Health Sciences Center, Oklahoma City, United States.,Molecular Biology and Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, United States
| | - Brian Leber
- Department of Medicine, McMaster University, Hamilton, Canada
| | - David W Andrews
- Biological Sciences, Sunnybrook Research Institute, Toronto, Canada.,Department of Medical Biophysics, University of Toronto, Ontario, Canada.,Department of Biochemistry, University of Toronto, Toronto, Canada
| |
Collapse
|
19
|
Liu G, Jin BK, Ma C, Chen Z, Zhu JJ. Potential-Resolved Electrochemiluminescence Nanoprobes for Visual Apoptosis Evaluation at Single-Cell Level. Anal Chem 2019; 91:6363-6370. [PMID: 30964659 DOI: 10.1021/acs.analchem.9b01401] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
In this work, a potential-resolved electrochemiluminescence (ECL) method is developed and used for the apoptosis diagnosis at the single-cell level. The apoptosis of cells usually induces the decreasing expression of epidermal growth factor receptor (EGFR) and promotes phosphatidylserine (PS) eversion on the cell membrane. Here, Au@L012 and g-C3N4 as ECL probes are functionalized with epidermal growth factor (EGF) and peptide (PSBP) to recognize the EGFR and PS on the cell surface, respectively, showing two well-separated ECL signals during a potential scanning. Experimental results reveal that the relative ECL change of g-C3N4 and Au@L012 correlates with the degree of apoptosis, which provides an accurate way to investigate apoptosis without interference that solely changes EGFR or PS. With a homemade ECL microscopy, we simultaneously evaluate the EGFR and PS expression of abundant individual cells and, therefore, achieve the visualization analysis of the apoptosis rate for normal and cancer cell samples. This strategy contributes to visually studying tumor markers and pushing the application of ECL imaging for the disease diagnosis at the single-cell level.
Collapse
Affiliation(s)
- Gen Liu
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering , Nanjing University , Nanjing , 210023 , China.,College of Chemistry and Chemical Engineering , Anhui University , Hefei , Anhui 230601 , China
| | - Bao-Kang Jin
- College of Chemistry and Chemical Engineering , Anhui University , Hefei , Anhui 230601 , China
| | - Cheng Ma
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering , Nanjing University , Nanjing , 210023 , China
| | - Zixuan Chen
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering , Nanjing University , Nanjing , 210023 , China
| | - Jun-Jie Zhu
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering , Nanjing University , Nanjing , 210023 , China
| |
Collapse
|
20
|
Khoshbakht S, Beiki D, Geramifar P, Kobarfard F, Sabzevari O, Amini M, Bolourchian N, Shamshirian D, Shahhosseini S. Design, Synthesis, Radiolabeling, and Biologic Evaluation of Three 18F-FDG-Radiolabeled Targeting Peptides for the Imaging of Apoptosis. Cancer Biother Radiopharm 2019; 34:271-279. [PMID: 30835137 DOI: 10.1089/cbr.2018.2709] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Background: Early detection of apoptosis is very important for therapy and follow-up treatment in various pathologic conditions. Annexin V interacts strongly and specifically with phosphatidylserine, specific biomarkers of apoptosis with some limitations. Small peptides are suitable alternatives to annexin V. A reliable and noninvasive in vivo technique for the detection of apoptosis is in great demand. Based on our previous studies, three new peptide analogs of LIKKPF (Leu-Ile-Lys-Lys-Pro-Phe) as apoptosis imaging agents were developed. Materials and Methods: Aoa-LIKKP-Cl-F, Aoe-LIKKP-Pyr-F, and Aoe-LIKKP-Nap-F were synthesized, functionalized with aminooxy, and radiolabeled with 18F-FDG. Their biologic properties were evaluated in vitro using apoptotic Jurkat cells. 18F-FDG-Aoe-LIKKP-Pyr-F peptide was injected into normal and apoptotic mice models for biodistribution and in vivo positron emission tomography/computed tomography imaging studies. Results: 18F-FDG-Aoe-LIKKP-Pyr-F peptide showed higher affinity for apoptotic cells. The localization of peptide in apoptotic liver mice was confirmed in biodistribution and imaging studies. Conclusion: The results showed that Aoe-LIKKP-Pyr-F peptide is an auspicious agent for molecular imaging of apoptosis.
Collapse
Affiliation(s)
- Sepideh Khoshbakht
- 1 Shohada-E-Tajrish Hospital, School of Medicine, Shahid Behesti University of Medical Sciences, Tehran, Iran
| | - Davood Beiki
- 2 Research Center for Nuclear Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Parham Geramifar
- 2 Research Center for Nuclear Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Farzad Kobarfard
- 3 Department of Pharmaceutical Chemistry and Radiopharmacy, School of Pharmacy, Phytochemistry Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Omid Sabzevari
- 4 Department of Toxicology and Pharmacology, Faculty of Pharmacy, Toxicology and Poisoning Research Centre, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohsen Amini
- 5 Department of Medicinal Chemistry, Faculty of Pharmacy, Drug Design and Development Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Noushin Bolourchian
- 6 Department of Pharmaceutics, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Danial Shamshirian
- 7 PET/CT Center, Masih Daneshvari Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Soraya Shahhosseini
- 8 Department of Pharmaceutical Chemistry and Radiopharmacy, School of Pharmacy, Protein Technology Research Center, Shahid Behesti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
21
|
Miao T, Floreani RA, Liu G, Chen X. Nanotheranostics-Based Imaging for Cancer Treatment Monitoring. Bioanalysis 2019. [DOI: 10.1007/978-3-030-01775-0_16] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
|
22
|
Kawai H, Chaudhry F, Shekhar A, Petrov A, Nakahara T, Tanimoto T, Kim D, Chen J, Lebeche D, Blankenberg FG, Pak KY, Kolodgie FD, Virmani R, Sengupta P, Narula N, Hajjar RJ, Strauss HW, Narula J. Molecular Imaging of Apoptosis in Ischemia Reperfusion Injury With Radiolabeled Duramycin Targeting Phosphatidylethanolamine. JACC Cardiovasc Imaging 2018; 11:1823-1833. [DOI: 10.1016/j.jcmg.2017.11.037] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Revised: 11/28/2017] [Accepted: 11/29/2017] [Indexed: 12/21/2022]
|
23
|
Johnson SE, Ugolkov A, Haney CR, Bondarenko G, Li L, Waters EA, Bergan R, Tran A, O'Halloran TV, Mazar A, Zhao M. Whole-body Imaging of Cell Death Provides a Systemic, Minimally Invasive, Dynamic, and Near-real Time Indicator for Chemotherapeutic Drug Toxicity. Clin Cancer Res 2018; 25:1331-1342. [PMID: 30420445 DOI: 10.1158/1078-0432.ccr-18-1846] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Revised: 10/09/2018] [Accepted: 11/07/2018] [Indexed: 12/31/2022]
Abstract
PURPOSE Response to toxicity in chemotherapies varies considerably from tissue to tissue and from patient to patient. An ability to monitor the tissue damage done by chemotherapy may have a profound impact on treatment and prognosis allowing for a proactive management in understanding and mitigating such events. For the first time, we investigated the feasibility of using whole-body imaging to map chemotherapeutic drug-induced toxicity on an individual basis. EXPERIMENTAL DESIGN In a preclinical proof-of-concept, rats were treated with a single clinical dose of cyclophosphamide, methotrexate, or cisplatin. In vivo whole-body imaging data were acquired using 99mTc-duramycin, which identifies dead and dying cells as an unambiguous marker for tissue injury in susceptible organs. Imaging results were cross-validated using quantitative ex vivo measurements and histopathology and compared with standard blood and serum panels for toxicology. RESULTS The in vivo whole-body imaging data detected widespread changes, where spatially heterogeneous toxic effects were identified across different tissues, within substructures of organs, as well as among different individuals. The signal changes were consistent with established toxicity profiles of these chemotherapeutic drugs. Apart from generating a map of susceptible tissues, this in vivo imaging approach was more sensitive compared with conventional blood and serum markers used in toxicology. Also, repeated imaging during the acute period after drug treatment captured different kinetics of tissue injury among susceptible organs in males and females. CONCLUSIONS This novel and highly translational imaging approach shows promise in optimizing therapeutic decisions by detecting and managing drug toxicity on a personalized basis.Toxicity to normal tissues is a significant limitation in chemotherapies. This work demonstrated an in vivo imaging-based approach for characterizing toxicity-induced tissue injury in a systemic, dynamic, and near-real time fashion. This novel approach shows promise in optimizing therapeutic decisions by monitoring drug toxicity on a personalized basis.
Collapse
Affiliation(s)
- Steven E Johnson
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Andrey Ugolkov
- Division of Hematology/Oncology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois.,Chemistry of Life Processes Institute, Northwestern University, Evanston, Illinois
| | - Chad R Haney
- Center for Advanced Molecular Imaging, Chemistry of Life Processes Institute, Northwestern University, Evanston, Illinois
| | - Gennadiy Bondarenko
- Division of Hematology/Oncology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois.,Chemistry of Life Processes Institute, Northwestern University, Evanston, Illinois
| | - Lin Li
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Emily A Waters
- Center for Advanced Molecular Imaging, Chemistry of Life Processes Institute, Northwestern University, Evanston, Illinois
| | - Raymond Bergan
- Division of Hematology/Oncology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Andy Tran
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Thomas V O'Halloran
- Chemistry of Life Processes Institute, Northwestern University, Evanston, Illinois.,Department of Chemistry, Northwestern University, Evanston, Illinois
| | - Andrew Mazar
- Chemistry of Life Processes Institute, Northwestern University, Evanston, Illinois. .,Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Ming Zhao
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois. .,Chemistry of Life Processes Institute, Northwestern University, Evanston, Illinois
| |
Collapse
|
24
|
Celentano A, McCullough M, Cirillo N. Glucocorticoids reduce chemotherapeutic effectiveness on OSCC cells via glucose-dependent mechanisms. J Cell Physiol 2018; 234:2013-2020. [PMID: 30240006 DOI: 10.1002/jcp.27227] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Accepted: 07/17/2018] [Indexed: 01/12/2023]
Abstract
Synthetic corticosteroids are routinely administered during the treatment of several diseases, including malignancies. However, recent evidence suggests that corticosteroids may have tumor-promoting effects, particularly in epithelial neoplasms. Our aim was to assess the role of the recently characterized cancer-associated glucocorticoid (GC) system in the resistance to chemotherapy of oral malignant keratinocytes. Human malignant oral keratinocyte cell lines H314/H357/H400/BICR16/BICR56 were tested with: two chemotherapeutic agents, doxorubicin (DOXO) and 5-fluorouracil (5-FU), as well as hydrocortisone (HC), adrenocorticotropic hormone (ACTH), 5-pregnen-3-beta-ol-20-one-16-alfa-carbonitrile (PCN), and two glucose uptake inhibitors, Fasentin and WZB. Both DOXO and 5-FU induced apoptosis in a dose-dependent and time-dependent manner. HC administration (100 nM) reduced the effectiveness of both chemotherapeutic agents to a variable extent in all 5 oral squamous cell carcinoma cell lines. ACTH also reduced the effectiveness of DOXO on 2 cell lines tested (H357 and BICR56). The glucose uptake inhibitors Fasentin and WZB were able to partially block the increased resistance to the cytotoxic drugs induced by HC. In summary, we have demonstrated, for the first time, the importance of cortisol on oral cancer cells ability to proliferate and combat the effectiveness of chemotherapeutic agents. This effect appears to be glucose dependent.
Collapse
Affiliation(s)
- Antonio Celentano
- Melbourne Dental School, The University of Melbourne, Melbourne, Victoria, Australia
| | - Michael McCullough
- Melbourne Dental School, The University of Melbourne, Melbourne, Victoria, Australia
| | - Nicola Cirillo
- Melbourne Dental School, The University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
25
|
Yang E, Al-Mugheiry TS, Normando EM, Cordeiro MF. Real-Time Imaging of Retinal Cell Apoptosis by Confocal Scanning Laser Ophthalmoscopy and Its Role in Glaucoma. Front Neurol 2018; 9:338. [PMID: 29867744 PMCID: PMC5962659 DOI: 10.3389/fneur.2018.00338] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Accepted: 04/27/2018] [Indexed: 12/31/2022] Open
Abstract
Glaucoma is one of the leading causes of irreversible blindness in the world. It is characterized by the progressive loss of retinal ganglion cells (RGCs), mainly through the process of apoptosis. Glaucoma patients often come to clinical attention when irreversible loss of visual function has been already established; therefore, early recognition of RGC apoptosis is inordinately important in disease prevention. The novel technology called Detection of Apoptosing Retinal Cells (DARC) allows real-time in vivo quantification of apoptosing cells through the use of a fluorescent biomarker and a confocal scanning ophthalmoscope. A recent Phase I clinical trial has evaluated the safety of DARC and its ability to detect retinal apoptosis in glaucoma patients and healthy volunteers. Results suggest that DARC may have potential in the early detection of glaucoma, which could help alleviate the medical, social, and economic burden associated with this blinding condition.
Collapse
Affiliation(s)
- Elizabeth Yang
- The Western Eye Hospital, Imperial College Healthcare NHS Trust, London, United Kingdom.,The Imperial College Ophthalmic Research Group (ICORG), Imperial College London, London, United Kingdom
| | - Toby S Al-Mugheiry
- Queen Elizabeth Hospital, King's Lynn NHS Foundation Trust, Norfolk, United Kingdom
| | - Eduardo M Normando
- The Western Eye Hospital, Imperial College Healthcare NHS Trust, London, United Kingdom.,The Imperial College Ophthalmic Research Group (ICORG), Imperial College London, London, United Kingdom.,Insitute of Ophthalmology, University College London, London, United Kingdom
| | - Maria F Cordeiro
- The Western Eye Hospital, Imperial College Healthcare NHS Trust, London, United Kingdom.,The Imperial College Ophthalmic Research Group (ICORG), Imperial College London, London, United Kingdom.,Insitute of Ophthalmology, University College London, London, United Kingdom
| |
Collapse
|
26
|
Rybczynska AA, Boersma HH, de Jong S, Gietema JA, Noordzij W, Dierckx RAJO, Elsinga PH, van Waarde A. Avenues to molecular imaging of dying cells: Focus on cancer. Med Res Rev 2018. [PMID: 29528513 PMCID: PMC6220832 DOI: 10.1002/med.21495] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Successful treatment of cancer patients requires balancing of the dose, timing, and type of therapeutic regimen. Detection of increased cell death may serve as a predictor of the eventual therapeutic success. Imaging of cell death may thus lead to early identification of treatment responders and nonresponders, and to “patient‐tailored therapy.” Cell death in organs and tissues of the human body can be visualized, using positron emission tomography or single‐photon emission computed tomography, although unsolved problems remain concerning target selection, tracer pharmacokinetics, target‐to‐nontarget ratio, and spatial and temporal resolution of the scans. Phosphatidylserine exposure by dying cells has been the most extensively studied imaging target. However, visualization of this process with radiolabeled Annexin A5 has not become routine in the clinical setting. Classification of death modes is no longer based only on cell morphology but also on biochemistry, and apoptosis is no longer found to be the preponderant mechanism of cell death after antitumor therapy, as was earlier believed. These conceptual changes have affected radiochemical efforts. Novel probes targeting changes in membrane permeability, cytoplasmic pH, mitochondrial membrane potential, or caspase activation have recently been explored. In this review, we discuss molecular changes in tumors which can be targeted to visualize cell death and we propose promising biomarkers for future exploration.
Collapse
Affiliation(s)
- Anna A Rybczynska
- Molecular Imaging Center, Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands.,Department of Genetics, University of Groningen, Groningen, the Netherlands
| | - Hendrikus H Boersma
- Molecular Imaging Center, Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands.,Department of Clinical Pharmacy & Pharmacology, University of Groningen, Groningen, the Netherlands
| | - Steven de Jong
- Department of Medical Oncology, University of Groningen, Groningen, the Netherlands
| | - Jourik A Gietema
- Department of Medical Oncology, University of Groningen, Groningen, the Netherlands
| | - Walter Noordzij
- Molecular Imaging Center, Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Rudi A J O Dierckx
- Molecular Imaging Center, Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands.,Department of Nuclear Medicine, Ghent University, Ghent, Belgium
| | - Philip H Elsinga
- Molecular Imaging Center, Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Aren van Waarde
- Molecular Imaging Center, Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| |
Collapse
|
27
|
Burvenich IJG, Parakh S, Parslow AC, Lee ST, Gan HK, Scott AM. Receptor Occupancy Imaging Studies in Oncology Drug Development. AAPS JOURNAL 2018. [DOI: 10.1208/s12248-018-0203-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
28
|
Shekhar A, Heeger P, Reutelingsperger C, Arbustini E, Narula N, Hofstra L, Bax JJ, Narula J. Targeted Imaging for Cell Death in Cardiovascular Disorders. JACC Cardiovasc Imaging 2018; 11:476-493. [DOI: 10.1016/j.jcmg.2017.11.018] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Revised: 11/19/2017] [Accepted: 11/27/2017] [Indexed: 01/30/2023]
|
29
|
Stucke-Ring J, Ronnacker J, Brand C, Höltke C, Schliemann C, Kessler T, Schmidt LH, Harrach S, Mantke V, Hintelmann H, Hartmann W, Wardelmann E, Lenz G, Wünsch B, Müller-Tidow C, Mesters RM, Schwöppe C, Berdel WE. Combinatorial effects of doxorubicin and retargeted tissue factor by intratumoral entrapment of doxorubicin and proapoptotic increase of tumor vascular infarction. Oncotarget 2018; 7:82458-82472. [PMID: 27738341 PMCID: PMC5347705 DOI: 10.18632/oncotarget.12559] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Accepted: 10/04/2016] [Indexed: 12/14/2022] Open
Abstract
Truncated tissue factor (tTF), retargeted to tumor vasculature by GNGRAHA peptide (tTF-NGR), and doxorubicin have therapeutic activity against a variety of tumors. We report on combination experiments of both drugs using different schedules. We have tested fluorescence- and HPLC-based intratumoral pharmacokinetics of doxorubicin, flow cytometry for cellular phosphatidylserine (PS) expression, and tumor xenograft studies for showing in vivo apoptosis, proliferation decrease, and tumor shrinkage upon combination therapy with doxorubicin and induced tumor vascular infarction. tTF-NGR given before doxorubicin inhibits the uptake of the drug into human fibrosarcoma xenografts in vivo. Reverse sequence does not influence the uptake of doxorubicin into tumor, but significantly inhibits the late wash-out phase, thus entrapping doxorubicin in tumor tissue by vascular occlusion. Incubation of endothelial and tumor cells with doxorubicin in vitro increases PS concentrations in the outer layer of the cell membrane as a sign of early apoptosis. Cells expressing increased PS concentrations show comparatively higher procoagulatory efficacy on the basis of equimolar tTF-NGR present in the Factor X assay. Experiments using human M21 melanoma and HT1080 fibrosarcoma xenografts in athymic nude mice indeed show a combinatorial tumor growth inhibition applying doxorubicin and tTF-NGR in sequence over single drug treatment. Combination of cytotoxic drugs such as doxorubicin with tTF-NGR-induced tumor vessel infarction can improve pharmacodynamics of the drugs by new mechanisms, entrapping a cytotoxic molecule inside tumor tissue and reciprocally improving procoagulatory activity of tTF-NGR in the tumor vasculature via apoptosis induction in tumor endothelial and tumor cells.
Collapse
Affiliation(s)
- Janine Stucke-Ring
- Department of Medicine A (Hematology, Hemostaseology, Oncology and Pneumology), University Hospital of Muenster, Muenster, Germany
| | - Julian Ronnacker
- Department of Medicine A (Hematology, Hemostaseology, Oncology and Pneumology), University Hospital of Muenster, Muenster, Germany
| | - Caroline Brand
- Department of Medicine A (Hematology, Hemostaseology, Oncology and Pneumology), University Hospital of Muenster, Muenster, Germany
| | - Carsten Höltke
- Department of Clinical Radiology, University Hospital of Muenster, Muenster, Germany
| | - Christoph Schliemann
- Department of Medicine A (Hematology, Hemostaseology, Oncology and Pneumology), University Hospital of Muenster, Muenster, Germany
| | - Torsten Kessler
- Department of Medicine A (Hematology, Hemostaseology, Oncology and Pneumology), University Hospital of Muenster, Muenster, Germany
| | - Lars Henning Schmidt
- Department of Medicine A (Hematology, Hemostaseology, Oncology and Pneumology), University Hospital of Muenster, Muenster, Germany
| | - Saliha Harrach
- Department of Medicine A (Hematology, Hemostaseology, Oncology and Pneumology), University Hospital of Muenster, Muenster, Germany
| | - Verena Mantke
- Department of Medicine A (Hematology, Hemostaseology, Oncology and Pneumology), University Hospital of Muenster, Muenster, Germany
| | - Heike Hintelmann
- Department of Medicine A (Hematology, Hemostaseology, Oncology and Pneumology), University Hospital of Muenster, Muenster, Germany
| | - Wolfgang Hartmann
- Gerhard-Domagk Institute for Pathology, University Hospital of Muenster, Muenster, Germany
| | - Eva Wardelmann
- Gerhard-Domagk Institute for Pathology, University Hospital of Muenster, Muenster, Germany
| | - Georg Lenz
- Department of Medicine A (Hematology, Hemostaseology, Oncology and Pneumology), University Hospital of Muenster, Muenster, Germany
| | - Bernhard Wünsch
- Department of Pharmaceutical Chemistry, Westfalian Wilhelms-University, Muenster, Germany
| | - Carsten Müller-Tidow
- Department of Hematology and Oncology, University Hospital Halle, Halle, Germany
| | - Rolf M Mesters
- Department of Medicine A (Hematology, Hemostaseology, Oncology and Pneumology), University Hospital of Muenster, Muenster, Germany
| | - Christian Schwöppe
- Department of Medicine A (Hematology, Hemostaseology, Oncology and Pneumology), University Hospital of Muenster, Muenster, Germany
| | - Wolfgang E Berdel
- Department of Medicine A (Hematology, Hemostaseology, Oncology and Pneumology), University Hospital of Muenster, Muenster, Germany
| |
Collapse
|
30
|
Delvaeye T, Wyffels L, Deleye S, Lemeire K, Gonçalves A, Decrock E, Staelens S, Leybaert L, Vandenabeele P, Krysko DV. Noninvasive Whole-Body Imaging of Phosphatidylethanolamine as a Cell Death Marker Using 99mTc-Duramycin During TNF-Induced SIRS. J Nucl Med 2018; 59:1140-1145. [PMID: 29419481 DOI: 10.2967/jnumed.117.205815] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Accepted: 01/16/2018] [Indexed: 01/30/2023] Open
Abstract
Systemic inflammatory response syndrome (SIRS) is an inflammatory state affecting the whole body. It is associated with the presence of pro- and antiinflammatory cytokines in serum, including tumor necrosis factor (TNF). TNF has multiple effects and leads to cytokine production, leukocyte infiltration, and blood pressure reduction and coagulation, thereby contributing to tissue damage and organ failure. A sterile mouse model of sepsis, TNF-induced SIRS, was used to visualize the temporal and spatial distribution of damage in susceptible tissues during SIRS. For this, a radiopharmaceutical agent, 99mTc-duramycin, that binds to exposed phosphatidylethanolamine on dying cells was longitudinally visualized using SPECT/CT imaging. Methods: C57BL/6J mice were challenged with intravenous injections of murine TNF or vehicle, and necrostatin-1 was used to interfere with cell death. Two hours after vehicle or TNF treatment, mice received 99mTc-duramycin intravenously (35.44 ± 3.80 MBq). Static whole-body 99mTc-duramycin SPECT/CT imaging was performed 2, 4, and 6 h after tracer injection. Tracer uptake in different organs was quantified by volume-of-interest analysis using PMOD software and expressed as SUVmean After the last scan, ex vivo biodistribution was performed to validate the SPECT imaging data. Lastly, terminal deoxynucleotidyl-transferase-mediated deoxyuridine triphosphate nick-end labeling (TUNEL) staining was performed to correlate the obtained results to cell death. Results: An increased 99mTc-duramycin uptake was detected in mice injected with TNF, when compared with control mice, in lungs (0.55 ± 0.1 vs. 0.34 ± 0.05), intestine (0.75 ± 0.13 vs. 0.56 ± 0.1), and liver (1.03 ± 0.14 vs. 0.64 ± 0.04) 4 h after TNF and remained significantly elevated until 8 h after TNF. The imaging results were consistent with ex vivo γ-counting results. Significantly increased levels of tissue damage were detected via TUNEL staining in the lungs and intestine of mice injected with TNF. Interestingly, necrostatin-1 pretreatment conferred protection against lethal SIRS and reduced the 99mTc-duramycin uptake in the lungs 8 h after TNF (SUV, 0.32 ± 0.1 vs. 0.51 ± 0.15). Conclusion: This study demonstrated that noninvasive 99mTc-duramycin SPECT imaging can be used to characterize temporal and spatial kinetics of injury and cell death in susceptible tissues during TNF-induced SIRS, making it useful for global, whole-body assessment of tissue damage during diseases associated with inflammation and injury.
Collapse
Affiliation(s)
- Tinneke Delvaeye
- VIB Center for Inflammation Research, Ghent, Belgium.,Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium.,Physiology Group, Department of Basic Medical Sciences, Ghent University, Ghent, Belgium
| | - Leonie Wyffels
- Molecular Imaging Center Antwerp, University of Antwerp, Wilrijk, Belgium
| | - Steven Deleye
- Molecular Imaging Center Antwerp, University of Antwerp, Wilrijk, Belgium
| | - Kelly Lemeire
- VIB Center for Inflammation Research, Ghent, Belgium.,Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Amanda Gonçalves
- VIB Center for Inflammation Research, Ghent, Belgium.,Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium.,VIB BioImaging Core, Ghent, Belgium; and
| | - Elke Decrock
- Physiology Group, Department of Basic Medical Sciences, Ghent University, Ghent, Belgium
| | - Steven Staelens
- Molecular Imaging Center Antwerp, University of Antwerp, Wilrijk, Belgium
| | - Luc Leybaert
- Physiology Group, Department of Basic Medical Sciences, Ghent University, Ghent, Belgium
| | - Peter Vandenabeele
- VIB Center for Inflammation Research, Ghent, Belgium.,Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Dmitri V Krysko
- Anatomy and Embryology Group, Department of Basic Medical Sciences, Ghent University, Ghent, Belgium
| |
Collapse
|
31
|
Johnson LL. Death, near death, and an antibiotic. J Nucl Cardiol 2018; 25:101-103. [PMID: 28900870 DOI: 10.1007/s12350-017-1053-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Accepted: 08/17/2017] [Indexed: 10/18/2022]
Affiliation(s)
- Lynne L Johnson
- Columbia University, 622 West 168th St, New York, NY, 10032, USA.
| |
Collapse
|
32
|
SPECT and PET radiopharmaceuticals for molecular imaging of apoptosis: from bench to clinic. Oncotarget 2017; 8:20476-20495. [PMID: 28108738 PMCID: PMC5386778 DOI: 10.18632/oncotarget.14730] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Accepted: 01/09/2017] [Indexed: 11/25/2022] Open
Abstract
Owing to the central role of apoptosis in many human diseases and the wide-spread application of apoptosis-based therapeutics, molecular imaging of apoptosis in clinical practice is of great interest for clinicians, and holds great promises. Based on the well-defined biochemical changes for apoptosis, a rich assortment of probes and approaches have been developed for molecular imaging of apoptosis with various imaging modalities. Among these imaging techniques, nuclear imaging (including single photon emission computed tomography and positron emission tomography) remains the premier clinical method owing to their high specificity and sensitivity. Therefore, the corresponding radiopharmaceuticals have been a major focus, and some of them like 99mTc-Annexin V, 18F-ML-10, 18F-CP18, and 18F-ICMT-11 are currently under clinical investigations in Phase I/II or Phase II/III clinical trials on a wide scope of diseases. In this review, we summarize these radiopharmaceuticals that have been widely used in clinical trials and elaborate them in terms of radiosynthesis, pharmacokinetics and dosimetry, and their applications in different clinical stages. We also explore the unique features required to qualify a desirable radiopharmaceutical for imaging apoptosis in clinical practice. Particularly, a perspective of the impact of these clinical efforts, namely, apoptosis imaging as predictive and prognostic markers, early-response indicators and surrogate endpoints, is also the highlight of this review.
Collapse
|
33
|
|
34
|
Baust JM, Vogel MJ, Van Buskirk R, Baust JG. A Molecular Basis of Cryopreservation Failure and its Modulation to Improve Cell Survival. Cell Transplant 2017. [DOI: 10.3727/000000001783986413] [Citation(s) in RCA: 116] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Affiliation(s)
- John M. Baust
- Institute of Biomedical Technology, State University of New York, Binghamton, NY 13902
| | - Martin J. Vogel
- Institute of Biomedical Technology, State University of New York, Binghamton, NY 13902
| | - Robert Van Buskirk
- Institute of Biomedical Technology, State University of New York, Binghamton, NY 13902
| | - John G. Baust
- Institute of Biomedical Technology, State University of New York, Binghamton, NY 13902
| |
Collapse
|
35
|
Phosphatidylethanolamine targeting for cell death imaging in early treatment response evaluation and disease diagnosis. Apoptosis 2017. [DOI: 10.1007/s10495-017-1384-0] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
36
|
Lee BY, Chon J, Kim HS, Lee JH, Yun DH, Yoo SD, Kim DH, Lee SA, Han YJ, Lee H, Kim JC, Soh Y, Chung JH, Kim SK, Park HJ. Association Between a Polymorphism in CASP3 and CASP9 Genes and Ischemic Stroke. Ann Rehabil Med 2017; 41:197-203. [PMID: 28503451 PMCID: PMC5426270 DOI: 10.5535/arm.2017.41.2.197] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Accepted: 08/29/2016] [Indexed: 12/23/2022] Open
Abstract
Objective To investigate whether the polymorphisms of CASP3 gene (rs4647602, intron A/C and rs1049216, UTR C/T) and CASP9 gene (rs1052576, Gln/Arg G/A and rs1052571, Ser/Val T/C) were associated with the development, and clinical severity of ischemic stroke and functional consequences after stroke. Methods Genomic DNA from 121 ischemic stroke patients and 201 healthy control subjects were extracted, and polymerase chain reaction products were sequenced. To investigate the association of polymorphisms and the development, and National Institutes of Health Stroke Scale (K-NIHSS), logistic regression models were analyzed. Results Polymorphism of the untranslational region of CASP3 (rs1049216, UTR C/T) has been associated with the development of ischemic stroke—in codominant1 model (odds ratio [OR], 0.51; 95% confidence interval [CI], 0.29–0.88; p=0.017), in dominant model (OR, 0.57; 95% CI, 0.34–0.97; p=0.034), and in the overdominant model (OR, 0.50; 95% CI, 0.29–0.87; p=0.011). A missense SNP of CASP9 gene (rs1052571, Ser/Val T/C) was associated with the development of ischemic stroke (OR, 1.93; 95% CI, 1.05–3.55; p=0.034 in recessive model). Conclusion These results indicate the possibility that CASP3 and CASP9 genes are markers for the development of ischemic stroke.
Collapse
Affiliation(s)
- Bae Youl Lee
- Department of Rehabilitation Medicine, Kyung Hee University Medical Center, Seoul, Korea
| | - Jinmann Chon
- Department of Rehabilitation Medicine, Kyung Hee University Medical Center, Seoul, Korea
| | - Hee-Sang Kim
- Department of Rehabilitation Medicine, Kyung Hee University Medical Center, Seoul, Korea
| | - Jong Ha Lee
- Department of Rehabilitation Medicine, Kyung Hee University Medical Center, Seoul, Korea
| | - Dong Hwan Yun
- Department of Rehabilitation Medicine, Kyung Hee University Medical Center, Seoul, Korea
| | - Seung Don Yoo
- Department of Rehabilitation Medicine, Kyung Hee University Medical Center, Seoul, Korea
| | - Dong Hwan Kim
- Department of Rehabilitation Medicine, Kyung Hee University Medical Center, Seoul, Korea
| | - Seung Ah Lee
- Department of Rehabilitation Medicine, Kyung Hee University Medical Center, Seoul, Korea
| | - Yoo Jin Han
- Department of Rehabilitation Medicine, Kyung Hee University Medical Center, Seoul, Korea
| | - Hyunseok Lee
- Department of Rehabilitation Medicine, Kyung Hee University Medical Center, Seoul, Korea
| | - Jin Chul Kim
- Department of Rehabilitation Medicine, Kyung Hee University Medical Center, Seoul, Korea
| | - Yunsoo Soh
- Department of Rehabilitation Medicine, Kyung Hee University Medical Center, Seoul, Korea
| | - Joo-Ho Chung
- Kohwang Medical Research Institute, Kyung Hee University, Seoul, Korea
| | - Su Kang Kim
- Kohwang Medical Research Institute, Kyung Hee University, Seoul, Korea
| | - Hae Jeong Park
- Kohwang Medical Research Institute, Kyung Hee University, Seoul, Korea
| |
Collapse
|
37
|
Preliminary biological evaluation of 18F-AlF-NOTA-MAL-Cys-Annexin V as a novel apoptosis imaging agent. Oncotarget 2017; 8:51086-51095. [PMID: 28881632 PMCID: PMC5584233 DOI: 10.18632/oncotarget.16994] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Accepted: 02/12/2017] [Indexed: 11/25/2022] Open
Abstract
A novel annexin V derivative (Cys-Annexin V) with a single cysteine residue at its C-terminal has been successfully labeled site-specifically with NOTA-maleimide aluminum [18F]fluoride complexation and evaluated it as a novel apoptosis agent in vitro and in vivo. The total synthesis time of 18F-AlF-NOTA-MAL-Cys-Annexin V from [18F]fluoride was about 65 min. The tracer was stable in vitro and it was excreted through renal in normal mice. The rate of the tracer bound to erythrocytes with exposed phosphatidylserine was 89.36±0.61% and this binding could be blocked by unlabeled Cys-Annexin V. In rats treated with cycloheximide, there were 6.23±0.23 times (n=4) increase in hepatic uptake of the tracer as compared to normal rats at 1h p.i. The uptake of the tracer in liver also could be blocked by co-injection of unlabeled Cys-Annexin V. These results indicated the favorable characterizations such as convenient synthesis and specific apoptotic cells targeting of18F-AlF-NOTA-MAL- Cys-Annexin V were suitable for its further investigation in clinical apoptosis imaging.
Collapse
|
38
|
Schumacher D, Lemke O, Helma J, Gerszonowicz L, Waller V, Stoschek T, Durkin PM, Budisa N, Leonhardt H, Keller BG, Hackenberger CPR. Broad substrate tolerance of tubulin tyrosine ligase enables one-step site-specific enzymatic protein labeling. Chem Sci 2017; 8:3471-3478. [PMID: 28507719 PMCID: PMC5418632 DOI: 10.1039/c7sc00574a] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Accepted: 03/18/2017] [Indexed: 12/31/2022] Open
Abstract
The broad substrate tolerance of tubulin tyrosine ligase enables its wide applicability for protein functionalization.
The broad substrate tolerance of tubulin tyrosine ligase is the basic rationale behind its wide applicability for chemoenzymatic protein functionalization. In this context, we report that the wild-type enzyme enables ligation of various unnatural amino acids that are substantially bigger than and structurally unrelated to the natural substrate, tyrosine, without the need for extensive protein engineering. This unusual substrate flexibility is due to the fact that the enzyme's catalytic pocket forms an extended cavity during ligation, as confirmed by docking experiments and all-atom molecular dynamics simulations. This feature enabled one-step C-terminal biotinylation and fluorescent coumarin labeling of various functional proteins as demonstrated with ubiquitin, an antigen binding nanobody, and the apoptosis marker Annexin V. Its broad substrate tolerance establishes tubulin tyrosine ligase as a powerful tool for in vitro enzyme-mediated protein modification with single functional amino acids in a specific structural context.
Collapse
Affiliation(s)
- Dominik Schumacher
- Department of Chemical-Biology , Leibniz-Institut für Molekulare Pharmakologie (FMP) , Robert-Rössle-Str. 10 , 13125 Berlin , Germany . .,Department of Chemistry , Humboldt Universität zu Berlin , Brook-Taylor-Strasse 2 , 12489 Berlin , Germany
| | - Oliver Lemke
- Department of Biology, Chemistry, Pharmacy , Freie Universität Berlin , Takustr. 3 , 14195 Berlin , Germany .
| | - Jonas Helma
- Department of Biology II , Ludwig Maximilians Universität München and Center for Integrated Protein Science Munich , Großhadenerstr. 2 , 82152 Martinsried , Germany
| | - Lena Gerszonowicz
- Department of Chemistry , Humboldt Universität zu Berlin , Brook-Taylor-Strasse 2 , 12489 Berlin , Germany
| | - Verena Waller
- Department of Biology II , Ludwig Maximilians Universität München and Center for Integrated Protein Science Munich , Großhadenerstr. 2 , 82152 Martinsried , Germany
| | - Tina Stoschek
- Department of Biology II , Ludwig Maximilians Universität München and Center for Integrated Protein Science Munich , Großhadenerstr. 2 , 82152 Martinsried , Germany
| | - Patrick M Durkin
- Department of Chemistry , TU Berlin , Müller-Breslau-Str. 10 , 10623 Berlin , Germany
| | - Nediljko Budisa
- Department of Chemistry , TU Berlin , Müller-Breslau-Str. 10 , 10623 Berlin , Germany
| | - Heinrich Leonhardt
- Department of Biology II , Ludwig Maximilians Universität München and Center for Integrated Protein Science Munich , Großhadenerstr. 2 , 82152 Martinsried , Germany
| | - Bettina G Keller
- Department of Biology, Chemistry, Pharmacy , Freie Universität Berlin , Takustr. 3 , 14195 Berlin , Germany .
| | - Christian P R Hackenberger
- Department of Chemical-Biology , Leibniz-Institut für Molekulare Pharmakologie (FMP) , Robert-Rössle-Str. 10 , 13125 Berlin , Germany . .,Department of Chemistry , Humboldt Universität zu Berlin , Brook-Taylor-Strasse 2 , 12489 Berlin , Germany
| |
Collapse
|
39
|
Synthesis, gallium labelling and characterization of P04087, a functionalized phosphatidylserine-binding peptide. EJNMMI Radiopharm Chem 2017. [PMID: 29527564 PMCID: PMC5835976 DOI: 10.1186/s41181-016-0021-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Background Radiolabeled phosphatidylserine (PS)-binding peptides represent an innovative strategy for molecular imaging of apoptosis and thrombus. The hexapeptide PGDLSR was described as a selective and high affinity ligand for PS. In this work, we synthesized and evaluated a gallium labelled-PGDLSR peptide as a potential and selective radiopharmaceutical for nuclear imaging. PGDLSR-β-alanine-NODAGA (P04087) was prepared using Fmoc-based synthesis and then chelated with cold gallium, 68Ga and 67Ga. The affinity of Ga-P04087 for PS was evaluated by a competitive binding assay using biotinylated AnnexinV. The in vitro stability of the radiotracer was checked at room temperature and after incubation in human serum at 37 °C with and without a metalloprotease inhibitor. The in vivo binding of 67Ga-P04087 to phosphatidylserine was evaluated in a rat model of infective endocarditis. Results PGDLSR was successfully prepared with a yield of 31%. P04087 was obtained with a yield of 28% and in high chemical purity (>95%). The radiochemical purities of 67Ga-P04087 and 68Ga-P04087 exceeded 98% in all cases. IC50 of P04087 and Ga-P04087 were in the same order of magnitude (10−7M). The radiolabelled product was stable for 24 h at room temperature, but was very rapidly degraded in human serum in the absence of a protease inhibitor, which had a stabilizing effect. No focal uptake could be detected visually in the cardiac area on SPECT images. On autoradiography however, a focal uptake of 67Ga-P04087 in the valve area was present and histological slices demonstrated localization of peptide binding at the peripheral layer of vegetations. Conclusion In spite of the preservation of the peptide affinity to the PS after its conjugation to the NODAGA chelator, and of the presence of 67Ga-P04087 uptake on autoradiography, the absence of detectable foci in vivo in the valve area may be attributed to both the low intensity of the signal and the presence of background activity originating from blood pool and surrounding tissues in the living animals. Further modifications are necessary to design a radiolabeled peptide with higher binding potencies to PS while possessing enhanced metabolic stability in vivo.
Collapse
|
40
|
Bower AJ, Marjanovic M, Zhao Y, Li J, Chaney EJ, Boppart SA. Label-free in vivo cellular-level detection and imaging of apoptosis. JOURNAL OF BIOPHOTONICS 2017; 10:143-150. [PMID: 27089867 PMCID: PMC5071126 DOI: 10.1002/jbio.201600003] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Revised: 03/07/2016] [Accepted: 03/23/2016] [Indexed: 05/18/2023]
Abstract
Cell death plays a critical role in health and homeostasis as well as in the pathogenesis and treatment of a broad spectrum of diseases and can be broadly divided into two main categories: apoptosis, or programmed cell death, and necrosis, or acute cell death. While these processes have been characterized extensively in vitro, label-free detection of apoptosis and necrosis at the cellular level in vivo has yet to be shown. In this study, for the first time, fluorescence lifetime imaging microscopy (FLIM) of intracellular reduced nicotinamide adenine dinucleotide (NADH) was utilized to assess the metabolic response of in vivo mouse epidermal keratinocytes following induction of apoptosis and necrosis. Results show significantly elevated levels of both the mean lifetime of NADH and the intracellular ratio of protein bound-to-free NADH in the apoptotic compared to the necrotic tissue. In addition, the longitudinal profiles of these two cell death processes show remarkable differences. By identifying and extracting these temporal metabolic signatures, apoptosis in single cells can be studied in native tissue environments within the living organism.
Collapse
Affiliation(s)
- Andrew J Bower
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL, USA
- Department of Electrical and Computer Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Marina Marjanovic
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL, USA
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Youbo Zhao
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Joanne Li
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL, USA
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Eric J Chaney
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Stephen A Boppart
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL, USA
- Department of Electrical and Computer Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, USA
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL, USA
- Department of Internal Medicine, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| |
Collapse
|
41
|
Ogawa K. Biocomplexes in radiochemistry. PHYSICAL SCIENCES REVIEWS 2016. [DOI: 10.1515/psr-2016-0005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
42
|
Shimizu Y, Kuge Y. Recent Advances in the Development of PET/SPECT Probes for Atherosclerosis Imaging. Nucl Med Mol Imaging 2016; 50:284-291. [PMID: 27994683 DOI: 10.1007/s13139-016-0418-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2016] [Accepted: 04/12/2016] [Indexed: 01/23/2023] Open
Abstract
The rupture of vulnerable atherosclerotic plaques and subsequent thrombus formation are the major causes of myocardial and cerebral infarction. Accordingly, the detection of vulnerable plaques is important for risk stratification and to provide appropriate treatment. Inflammation imaging using 2-deoxy-2-[18F]fluoro-D-glucose (18F-FDG) has been most extensively studied for detecting vulnerable atherosclerotic plaques. It is of great importance to develop PET/SPECT probes capable of specifically visualizing the biological molecules involved in atherosclerotic plaque formation and/or progression. In this article, we review recent advances in the development of PET/SPECT probes for visualizing atherosclerotic plaques and their application to therapy monitoring, mainly focusing on experimental studies.
Collapse
Affiliation(s)
- Yoichi Shimizu
- Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
| | - Yuji Kuge
- Central Institute of Isotope Science, Hokkaido University, Kita 15 Nishi 7, Kita-ku, Sapporo, 060-0815 Japan ; Hokkaido University Graduate School of Medicine, Sapporo, Japan
| |
Collapse
|
43
|
Thukkani AK, Shoghi KI, Zhou D, Xu J, Chu W, Novak E, Chen DL, Gropler RJ, Mach RH. PET imaging of in vivo caspase-3/7 activity following myocardial ischemia-reperfusion injury with the radiolabeled isatin sulfonamide analogue [(18)F]WC-4-116. AMERICAN JOURNAL OF NUCLEAR MEDICINE AND MOLECULAR IMAGING 2016; 6:110-119. [PMID: 27186438 PMCID: PMC4858607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Accepted: 02/09/2016] [Indexed: 06/05/2023]
Abstract
The utility of [(18)F]WC-4-116, a PET tracer for imaging caspase-3 activation, was evaluated in an animal model of myocardial apoptosis. [(18)F]WC-4-116 was injected into rats at 3 hours after a 30 min period of ischemia induced by temporary occlusion of the left anterior descending coronary artery in Sprague-Dawley rats. [(18)F]WC-4-116 uptake was quantified by 1) autoradiography, 2) microPET imaging studies, and 3) post-PET biodistribution studies. MicroPET imaging also assessed uptake of the non-caspase-3-targeted tracer [(18)F]ICMT-18 at 3 hours postischemia. Enzyme assays and Western blotting assessed caspase-3 activation in both at-risk and not-at-risk regions. Caspase-3 enzyme activity increased in the at-risk but not in the not-at-risk myocardium. Quantitative autoradiographic analysis of [(18)F]WC-4-116 demonstrated nearly 2-fold higher uptake in the ischemia-reperfusion (IR) versus sham animals. [(18)F]WC-4-116 microPET imaging studies demonstrated that the IR animals was similarly elevated in relation to sham. [(18)F]ICMT-18 uptake did not increase in at-risk myocardium despite evidence of caspase-3 activation. Biodistribution studies with [(18)F]WC-4-116 confirmed the microPET findings. These data indicate that the caspase-3-PET tracer [(18)F]WC-4-116 can noninvasively image in vivo caspase activity during myocardial apoptosis and may be useful for clinical imaging in humans.
Collapse
Affiliation(s)
- Arun K Thukkani
- Mallinckrodt Institute of Radiology, Washington University School of MedicineSt. Louis, MO 63110, USA
- Cardiovascular Division, Washington University School of MedicineSt Louis, MO 63110, USA
| | - Kooresh I Shoghi
- Mallinckrodt Institute of Radiology, Washington University School of MedicineSt. Louis, MO 63110, USA
- Department of Biomedical Engineering, Washington University School of MedicineSt. Louis, MO 63110, USA
| | - Dong Zhou
- Mallinckrodt Institute of Radiology, Washington University School of MedicineSt. Louis, MO 63110, USA
| | - Jinbin Xu
- Mallinckrodt Institute of Radiology, Washington University School of MedicineSt. Louis, MO 63110, USA
| | - Wenhua Chu
- Mallinckrodt Institute of Radiology, Washington University School of MedicineSt. Louis, MO 63110, USA
| | - Eric Novak
- Cardiovascular Division, Washington University School of MedicineSt Louis, MO 63110, USA
| | - Delphine L Chen
- Mallinckrodt Institute of Radiology, Washington University School of MedicineSt. Louis, MO 63110, USA
| | - Robert J Gropler
- Mallinckrodt Institute of Radiology, Washington University School of MedicineSt. Louis, MO 63110, USA
- Cardiovascular Division, Washington University School of MedicineSt Louis, MO 63110, USA
| | - Robert H Mach
- Mallinckrodt Institute of Radiology, Washington University School of MedicineSt. Louis, MO 63110, USA
- Department of Cell Biology & Physiology, Washington University School of MedicineSt. Louis, MO 63110, USA
- Department of Biochemistry & Molecular Biophysics, Washington University School of MedicineSt. Louis, MO 63110, USA
| |
Collapse
|
44
|
Khoshbakht S, Beiki D, Geramifar P, Kobarfard F, Sabzevari O, Amini M, Shahhosseini S. 18FDG-labeled LIKKPF: a PET tracer for apoptosis imaging. J Radioanal Nucl Chem 2016. [DOI: 10.1007/s10967-016-4793-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
|
45
|
Saito A, Mekawy MM, Sumiyoshi A, Riera JJ, Shimizu H, Kawashima R, Tominaga T. Noninvasive targeting delivery and in vivo magnetic resonance tracking method for live apoptotic cells in cerebral ischemia with functional Fe2O3 magnetic nanoparticles. J Nanobiotechnology 2016; 14:19. [PMID: 26969152 PMCID: PMC4788935 DOI: 10.1186/s12951-016-0173-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Accepted: 02/26/2016] [Indexed: 11/14/2022] Open
Abstract
Background Apoptotic neuronal death is known as programmed cell death. Inhibition of this progression might contribute to a new treatment strategy. However, methods for in vivo detection of live apoptotic cells are in need to be developed and established. Context and purpose The purpose of this study is to develop a new method for in vivo brain imaging for live apoptotic lesions using magnetic resonance imaging (MRI). We focused on the specific accumulation of our recently developed functional magnetic nanoparticles (FMNPs) into apoptotic cells using a rat cerebral ischemia model. Sulphorhodamine B, fluorescent dye was linked to valylalanylaspartic acid fluoromethyl ketone as a pan-caspase inhibitor to form SR-FLIVO. SR-FLIVO was bound with FMNPs to develop SR-FLIVO-FMNP probe. Ischemic rat brains were scanned by 7T MRI before and after intravenous injection of SR-FLIVO-FMNP and the distribution was evaluated by subtraction images of T2* colored mapping. SR-FLIVO, intracellular FMNPs, and T2* reduction area were histologically analyzed. The distribution of SR-FLIVO-FMNP was evaluated by subtracting the T2* signal images and was significantly correlated with the histological findings by TUNEL staining. Results Our experimental results revealed several findings where our newly developed probe SR-FLIVO-FMNP was intravenously administered into ischemic rats and FLIVO expression was tracked and found in apoptotic cells in rat brains after cerebral ischemia. A remarkable T2* reduction within the ischemic lesion was recorded using MRI based SR-FLIVO-FMNP probe as a contrasting agent due to the specific probe accumulation in apoptotic cells whereas, no observation of T2* reduction within the non-ischemic lesion due to no probe accumulation in non-apoptotic cells. Histological analysis based on the correlation between FLIVO and TUNEL staining showed that almost all FLIVO-positive cells were positive for TUNEL staining. These findings suggest the possibility for establishment of in vivo targeting delivery methods to live apoptotic cells based on conjugation of magnetic and fluorescent dual functional probes. Conclusion A newly developed probe SR-FLIVO-FMNP might be considered as a useful probe for in vivo apoptotic detection, and FMNPs might be a strong platform for noninvasive imaging and targeting delivery. Electronic supplementary material The online version of this article (doi:10.1186/s12951-016-0173-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Atsushi Saito
- Department of Neurosurgery, Aomori Prefectural Central Hospital, 2-1-1 Higashitsukurimichi, Aomori, 030-8553, Japan. .,Department of Neurosurgery, Graduate School of Medicine, Tohoku University, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8575, Japan.
| | - Moataz M Mekawy
- Department of Neurosurgery, Graduate School of Medicine, Tohoku University, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8575, Japan. .,National Institute for Materials Science, 1-Chome-2-1 Sengen, Tsukuba, Ibaraki Prefecture, 305-0047, Japan.
| | - Akira Sumiyoshi
- Department of Functional Brain Imaging, Institute of Development, Aging and Cancer, Tohoku University, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8575, Japan
| | - Jorge J Riera
- Department of Functional Brain Imaging, Institute of Development, Aging and Cancer, Tohoku University, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8575, Japan
| | - Hiroaki Shimizu
- Department of Neurosurgery, Graduate School of Medicine, Akita University, 1-1-1 Hondo, Akita, 010-8543, Japan
| | - Ryuta Kawashima
- Department of Functional Brain Imaging, Institute of Development, Aging and Cancer, Tohoku University, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8575, Japan
| | - Teiji Tominaga
- Department of Neurosurgery, Graduate School of Medicine, Tohoku University, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8575, Japan
| |
Collapse
|
46
|
Winetraub Y, SoRelle ED, Liba O, de la Zerda A. Quantitative contrast-enhanced optical coherence tomography. APPLIED PHYSICS LETTERS 2016; 108:023702. [PMID: 26869724 PMCID: PMC4714990 DOI: 10.1063/1.4939547] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2015] [Accepted: 12/22/2015] [Indexed: 05/18/2023]
Abstract
We have developed a model to accurately quantify the signals produced by exogenous scattering agents used for contrast-enhanced Optical Coherence Tomography (OCT). This model predicts distinct concentration-dependent signal trends that arise from the underlying physics of OCT detection. Accordingly, we show that real scattering particles can be described as simplified ideal scatterers with modified scattering intensity and concentration. The relation between OCT signal and particle concentration is approximately linear at concentrations lower than 0.8 particle per imaging voxel. However, at higher concentrations, interference effects cause signal to increase with a square root dependence on the number of particles within a voxel. Finally, high particle concentrations cause enough light attenuation to saturate the detected signal. Predictions were validated by comparison with measured OCT signals from gold nanorods (GNRs) prepared in water at concentrations ranging over five orders of magnitude (50 fM to 5 nM). In addition, we validated that our model accurately predicts the signal responses of GNRs in highly heterogeneous scattering environments including whole blood and living animals. By enabling particle quantification, this work provides a valuable tool for current and future contrast-enhanced in vivo OCT studies. More generally, the model described herein may inform the interpretation of detected signals in modalities that rely on coherence-based detection or are susceptible to interference effects.
Collapse
|
47
|
Khoshbakht S, Beiki D, Geramifar P, Kobarfard F, Sabzevari O, Amini M, Mehrnejad F, Shahhosseini S. Synthesis, Radiolabeling, and Biological Evaluation of Peptide LIKKPF Functionalized with HYNIC as Apoptosis Imaging Agent. IRANIAN JOURNAL OF PHARMACEUTICAL RESEARCH : IJPR 2016; 15:415-24. [PMID: 27642312 PMCID: PMC5018269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
A noninvasive method of detecting exposure of phosphatidylserine (PS) on the external surface of the plasma membrane such as nuclear imaging could assist the diagnosis and therapy of apoptosis related pathologies. The most studied imaging agent for apoptosis is Annexin V so far. Because of limitations of Annexin V other agents have been introduced such as small peptides and molecules. Radiopeptides that have affinity and bind to PS are good candidates for noninvasive imaging of apoptosis. The LIKKPF, introduced by Burtea et al, with nanomolar affinity for PS, was used as templete. The biological properties of LIKKPF radiolabeled with Tc-99 m was assessed in-vitro using apoptotic Jurkat cells and in-vivo using mouse model of liver apoptosis. The radiolabeled LIKKPF with (99m)Tc was stable in human serum at 37˚C for at least 2 h. Results showed that the radiolabeled LIKKPF has less affinity to PS compare to original phage peptide, but high enough for specific binding to apoptotic cells in-vitro and in-vivo. It is concluded that the less affinity of radiolabeled LIKKPF might be attributed to hydrophobicity of peptide. The future peptides should be more hydrophobic compare to LIKKPF.
Collapse
Affiliation(s)
- Sepideh Khoshbakht
- Department of Radiopharmacy, Faculty of Pharmacy, Tehran University of Medical Sciences.
| | - Davood Beiki
- Research Center for Nuclear Medicine, Tehran University of Medical Sciences.
| | - Parham Geramifar
- Research Center for Nuclear Medicine, Tehran University of Medical Sciences.
| | - Farzad Kobarfard
- Department of Pharmaceutical Chemistry, School of Pharmacy, Phytochemistry Research Center, Shahid Beheshti University of Medical Sciences.
| | - Omid Sabzevari
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, and Toxicology and Poisoning Research Center, Tehran University of Medical Sciences.
| | - Mohsen Amini
- Department of Medicinal Chemistry, and Drug Design and Development Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences.
| | - Faramarz Mehrnejad
- Department of Life Science Engineering, Faculty of New Sciences & Technologies, University of Tehran.
| | - Soraya Shahhosseini
- Department of Pharmaceutical Chemistry, School of Pharmacy, and PET/CT Unit, Ferdous Nuclear Medicine Center, Masih Daneshvari Hospital, Shahid Behesti University of Medical Sciences, Tehran, Iran.,
| |
Collapse
|
48
|
Tian K, Shibata-Germanos S, Pahlitzsch M, Cordeiro MF. Current perspective of neuroprotection and glaucoma. Clin Ophthalmol 2015; 9:2109-18. [PMID: 26635467 PMCID: PMC4646599 DOI: 10.2147/opth.s80445] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Glaucoma is the second leading cause of blindness worldwide and is most notably characterized by progressive optic nerve atrophy and advancing loss of retinal ganglion cells (RGCs). The main concomitant factor is the elevated intraocular pressure (IOP). Existing treatments are focused generally on lowering IOP. However, both RGC loss and optic nerve atrophy can independently occur with IOP at normal levels. In recent years, there has been substantial progress in the development of neuroprotective therapies for glaucoma in order to restore vital visual function. The present review intends to offer a brief insight into conventional glaucoma treatments and discuss exciting current developments of mostly preclinical data in novel neuroprotective strategies for glaucoma that include recent advances in noninvasive diagnostics going beyond IOP maintenance for an enhanced global view. Such strategies now target RGC loss and optic nerve damage, opening a critical therapeutic window for preventative monitoring and treatment.
Collapse
Affiliation(s)
- Kailin Tian
- Glaucoma and Retinal Neurodegeneration Research Group, UCL Institute of Ophthalmology, London, UK ; Eye Centre, Renmin Hospital of Wuhan University, Wuhan, People's Republic of China
| | - Shannon Shibata-Germanos
- Glaucoma and Retinal Neurodegeneration Research Group, UCL Institute of Ophthalmology, London, UK
| | - Milena Pahlitzsch
- Glaucoma and Retinal Neurodegeneration Research Group, UCL Institute of Ophthalmology, London, UK
| | - M Francesca Cordeiro
- Glaucoma and Retinal Neurodegeneration Research Group, UCL Institute of Ophthalmology, London, UK ; Western Eye Hospital, ICORG, Imperial College NHS Trust, London, UK
| |
Collapse
|
49
|
Clear KJ, Harmatys KM, Rice DR, Wolter WR, Suckow MA, Wang Y, Rusckowski M, Smith BD. Phenoxide-Bridged Zinc(II)-Bis(dipicolylamine) Probes for Molecular Imaging of Cell Death. Bioconjug Chem 2015; 27:363-75. [PMID: 26334386 DOI: 10.1021/acs.bioconjchem.5b00447] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Cell death is involved in many pathological conditions, and there is a need for clinical and preclinical imaging agents that can target and report cell death. One of the best known biomarkers of cell death is exposure of the anionic phospholipid phosphatidylserine (PS) on the surface of dead and dying cells. Synthetic zinc(II)-bis(dipicolylamine) (Zn2BDPA) coordination complexes are known to selectively recognize PS-rich membranes and act as cell death molecular imaging agents. However, there is a need to improve in vivo imaging performance by selectively increasing target affinity and decreasing off-target accumulation. This present study compared the cell death targeting ability of two new deep-red fluorescent probes containing phenoxide-bridged Zn2BDPA complexes. One probe was a bivalent version of the other and associated more strongly with PS-rich liposome membranes. However, the bivalent probe exhibited self-quenching on the membrane surface, so the monovalent version produced brighter micrographs of dead and dying cells in cell culture and also better fluorescence imaging contrast in two living animal models of cell death (rat implanted tumor with necrotic core and mouse thymus atrophy). An (111)In-labeled radiotracer version of the monovalent probe also exhibited selective cell death targeting ability in the mouse thymus atrophy model, with relatively high amounts detected in dead and dying tissue and low off-target accumulation in nonclearance organs. The in vivo biodistribution profile is the most favorable yet reported for a Zn2BDPA complex; thus, the monovalent phenoxide-bridged Zn2BDPA scaffold is a promising candidate for further development as a cell death imaging agent in living subjects.
Collapse
Affiliation(s)
- Kasey J Clear
- Department of Chemistry and Biochemistry, University of Notre Dame , 236 Nieuwland Science Hall, Notre Dame, Indiana 46556, United States
| | - Kara M Harmatys
- Department of Chemistry and Biochemistry, University of Notre Dame , 236 Nieuwland Science Hall, Notre Dame, Indiana 46556, United States
| | - Douglas R Rice
- Department of Chemistry and Biochemistry, University of Notre Dame , 236 Nieuwland Science Hall, Notre Dame, Indiana 46556, United States
| | - William R Wolter
- Freimann Life Science Center, University of Notre Dame , 400 Galvin Life Science, Notre Dame, Indiana 46556, United States
| | - Mark A Suckow
- Freimann Life Science Center, University of Notre Dame , 400 Galvin Life Science, Notre Dame, Indiana 46556, United States
| | - Yuzhen Wang
- Division of Nuclear Medicine, Department of Radiology, University of Massachusetts Medical School , Worcester, Massachusetts 01655, United States
| | - Mary Rusckowski
- Division of Nuclear Medicine, Department of Radiology, University of Massachusetts Medical School , Worcester, Massachusetts 01655, United States
| | - Bradley D Smith
- Department of Chemistry and Biochemistry, University of Notre Dame , 236 Nieuwland Science Hall, Notre Dame, Indiana 46556, United States
| |
Collapse
|
50
|
18F-Labeled wild-type annexin V: comparison of random and site-selective radiolabeling methods. Amino Acids 2015; 48:65-74. [DOI: 10.1007/s00726-015-2068-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Accepted: 08/03/2015] [Indexed: 10/23/2022]
|