1
|
Wu KL, Martinez-Paniagua M, Reichel K, Menon PS, Deo S, Roysam B, Varadarajan N. Automated detection of apoptotic bodies and cells in label-free time-lapse high-throughput video microscopy using deep convolutional neural networks. Bioinformatics 2023; 39:btad584. [PMID: 37773981 PMCID: PMC10563152 DOI: 10.1093/bioinformatics/btad584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 09/06/2023] [Accepted: 09/28/2023] [Indexed: 10/01/2023] Open
Abstract
MOTIVATION Reliable label-free methods are needed for detecting and profiling apoptotic events in time-lapse cell-cell interaction assays. Prior studies relied on fluorescent markers of apoptosis, e.g. Annexin-V, that provide an inconsistent and late indication of apoptotic onset for human melanoma cells. Our motivation is to improve the detection of apoptosis by directly detecting apoptotic bodies in a label-free manner. RESULTS Our trained ResNet50 network identified nanowells containing apoptotic bodies with 92% accuracy and predicted the onset of apoptosis with an error of one frame (5 min/frame). Our apoptotic body segmentation yielded an IoU accuracy of 75%, allowing associative identification of apoptotic cells. Our method detected apoptosis events, 70% of which were not detected by Annexin-V staining. AVAILABILITY AND IMPLEMENTATION Open-source code and sample data provided at https://github.com/kwu14victor/ApoBDproject.
Collapse
Affiliation(s)
- Kwan-Ling Wu
- William A. Brookshire Department of Chemical and Biomolecular Engineering, University of Houston, Houston, TX 77204, United States
| | - Melisa Martinez-Paniagua
- William A. Brookshire Department of Chemical and Biomolecular Engineering, University of Houston, Houston, TX 77204, United States
| | - Kate Reichel
- William A. Brookshire Department of Chemical and Biomolecular Engineering, University of Houston, Houston, TX 77204, United States
| | - Prashant S Menon
- William A. Brookshire Department of Chemical and Biomolecular Engineering, University of Houston, Houston, TX 77204, United States
| | - Shravani Deo
- William A. Brookshire Department of Chemical and Biomolecular Engineering, University of Houston, Houston, TX 77204, United States
| | - Badrinath Roysam
- Department of Electrical and Computer Engineering, University of Houston, Houston, TX 77204, United States
| | - Navin Varadarajan
- William A. Brookshire Department of Chemical and Biomolecular Engineering, University of Houston, Houston, TX 77204, United States
| |
Collapse
|
2
|
Safarpour A, Ebrahimi M, Fazeli SAS, Amoozegar MA. A phenol amine molecule from Salinivenus iranica acts as the inhibitor of cancer stem cells in breast cancer cell lines. Sci Rep 2023; 13:12669. [PMID: 37542193 PMCID: PMC10403564 DOI: 10.1038/s41598-023-39736-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Accepted: 07/30/2023] [Indexed: 08/06/2023] Open
Abstract
In recent years, the anticancer properties of metabolites from halophilic microorganisms have received a lot of attention. Twenty-nine halophilic bacterial strains were selected from a culture collection to test the effects of their supernatant metabolites on stem cell-like properties of six human cancer cell lines. Human fibroblasts were used as normal control. Sphere and colony formation assay were done to assess the stem cell-like properties. invasion and migration assay, and tumor development in mice model were done to assess the anti-tumorigenesis effect in vitro and in vivo. The metabolites from Salinivenus iranica demonstrated the most potent cytotoxic effect on breast cancer cell lines (IC50 = 100 µg/mL) among all strains, with no effect on normal cells. In MDA-MB-231 cells, the supernatant metabolites enhanced both early and late apoptosis (approximately 9.5% and 48.8%, respectively) and decreased the sphere and colony formation ability of breast cancer cells. Furthermore, after intratumor injection of metabolites, tumors developed in the mice models reduced dramatically, associated with increased pro-apoptotic caspase-3 expression. The purified cytotoxic molecule, a phenol amine with a molecular weight of 1961.73 Dalton (IC50 = 1 µg/mL), downregulated pluripotency gene SRY-Box Transcription Factor 2 (SOX-2) expression in breast cancer cells which is associated with resistance to conventional anticancer treatment. In conclusion, we suggested that the phenol amine molecule from Salinivenus iranica could be a potential anti-breast cancer component.
Collapse
Affiliation(s)
- Atefeh Safarpour
- Extremophiles Laboratory, Department of Microbiology, School of Biology and Center of Excellence in Phylogeny of Living Organisms, College of Science, University of Tehran, Tehran, Iran
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, 19395-4644, Iran
| | - Marzieh Ebrahimi
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, 19395-4644, Iran.
| | - Seyed Abolhassan Shahzadeh Fazeli
- Department of Molecular and Cellular Biology, Faculty of Basic Sciences and Advanced Technologies in Biology, University of Science and Culture, ACECR, Tehran, Iran
| | - Mohammad Ali Amoozegar
- Extremophiles Laboratory, Department of Microbiology, School of Biology and Center of Excellence in Phylogeny of Living Organisms, College of Science, University of Tehran, Tehran, Iran.
| |
Collapse
|
3
|
Barth ND, Mendive‐Tapia L, Subiros‐Funosas R, Ghashghaei O, Lavilla R, Maiorino L, He X, Dransfield I, Egeblad M, Vendrell M. A Bivalent Activatable Fluorescent Probe for Screening and Intravital Imaging of Chemotherapy-Induced Cancer Cell Death. ANGEWANDTE CHEMIE (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 134:e202113020. [PMID: 38505298 PMCID: PMC10947113 DOI: 10.1002/ange.202113020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Indexed: 11/11/2022]
Abstract
The detection and quantification of apoptotic cells is a key process in cancer research, particularly during the screening of anticancer therapeutics and in mechanistic studies using preclinical models. Intravital optical imaging enables high-resolution visualisation of cellular events in live organisms; however, there are few fluorescent probes that can reliably provide functional readouts in situ without interference from tissue autofluorescence. We report the design and optimisation of the fluorogenic probe Apotracker Red for real-time detection of cancer cell death. The strong fluorogenic behaviour, high selectivity, and excellent stability of Apotracker Red make it a reliable optical reporter for the characterisation of the effects of anticancer drugs in cells in vitro and for direct imaging of chemotherapy-induced apoptosis in vivo in mouse models of breast cancer.
Collapse
Affiliation(s)
- Nicole D. Barth
- Centre for Inflammation ResearchThe University of EdinburghUK
| | | | | | - Ouldouz Ghashghaei
- Laboratory of Medicinal ChemistryFaculty of Pharmacy and Institute of Biomedicine (IBUB)University of BarcelonaSpain
| | - Rodolfo Lavilla
- Laboratory of Medicinal ChemistryFaculty of Pharmacy and Institute of Biomedicine (IBUB)University of BarcelonaSpain
| | - Laura Maiorino
- Cold Spring Harbor LaboratoryCold Spring HarborNY11724USA
| | - Xue‐Yan He
- Cold Spring Harbor LaboratoryCold Spring HarborNY11724USA
| | - Ian Dransfield
- Centre for Inflammation ResearchThe University of EdinburghUK
| | - Mikala Egeblad
- Cold Spring Harbor LaboratoryCold Spring HarborNY11724USA
| | - Marc Vendrell
- Centre for Inflammation ResearchThe University of EdinburghUK
| |
Collapse
|
4
|
Barth ND, Mendive‐Tapia L, Subiros‐Funosas R, Ghashghaei O, Lavilla R, Maiorino L, He X, Dransfield I, Egeblad M, Vendrell M. A Bivalent Activatable Fluorescent Probe for Screening and Intravital Imaging of Chemotherapy-Induced Cancer Cell Death. Angew Chem Int Ed Engl 2022; 61:e202113020. [PMID: 34762762 PMCID: PMC8991960 DOI: 10.1002/anie.202113020] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Indexed: 11/21/2022]
Abstract
The detection and quantification of apoptotic cells is a key process in cancer research, particularly during the screening of anticancer therapeutics and in mechanistic studies using preclinical models. Intravital optical imaging enables high-resolution visualisation of cellular events in live organisms; however, there are few fluorescent probes that can reliably provide functional readouts in situ without interference from tissue autofluorescence. We report the design and optimisation of the fluorogenic probe Apotracker Red for real-time detection of cancer cell death. The strong fluorogenic behaviour, high selectivity, and excellent stability of Apotracker Red make it a reliable optical reporter for the characterisation of the effects of anticancer drugs in cells in vitro and for direct imaging of chemotherapy-induced apoptosis in vivo in mouse models of breast cancer.
Collapse
Affiliation(s)
- Nicole D. Barth
- Centre for Inflammation ResearchThe University of EdinburghUK
| | | | | | - Ouldouz Ghashghaei
- Laboratory of Medicinal ChemistryFaculty of Pharmacy and Institute of Biomedicine (IBUB)University of BarcelonaSpain
| | - Rodolfo Lavilla
- Laboratory of Medicinal ChemistryFaculty of Pharmacy and Institute of Biomedicine (IBUB)University of BarcelonaSpain
| | - Laura Maiorino
- Cold Spring Harbor LaboratoryCold Spring HarborNY11724USA
| | - Xue‐Yan He
- Cold Spring Harbor LaboratoryCold Spring HarborNY11724USA
| | - Ian Dransfield
- Centre for Inflammation ResearchThe University of EdinburghUK
| | - Mikala Egeblad
- Cold Spring Harbor LaboratoryCold Spring HarborNY11724USA
| | - Marc Vendrell
- Centre for Inflammation ResearchThe University of EdinburghUK
| |
Collapse
|
5
|
Kundu R, Chandra A, Datta A. Fluorescent Chemical Tools for Tracking Anionic Phospholipids. Isr J Chem 2021. [DOI: 10.1002/ijch.202100003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
- Rajasree Kundu
- Department of Chemical Sciences Tata Institute of Fundamental Research 1 Homi Bhabha Road, Colaba Mumbai 400005 India
| | - Amitava Chandra
- Department of Chemical Sciences Tata Institute of Fundamental Research 1 Homi Bhabha Road, Colaba Mumbai 400005 India
| | - Ankona Datta
- Department of Chemical Sciences Tata Institute of Fundamental Research 1 Homi Bhabha Road, Colaba Mumbai 400005 India
| |
Collapse
|
6
|
Cytotoxicity test for the use of freeze-dried amniotic membranes against viability, proliferation, and apoptosis on brain cell culture: An in vitro study. INTERDISCIPLINARY NEUROSURGERY 2021. [DOI: 10.1016/j.inat.2020.100947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
|
7
|
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
|
8
|
Zhang D, Jin Q, Jiang C, Gao M, Ni Y, Zhang J. Imaging Cell Death: Focus on Early Evaluation of Tumor Response to Therapy. Bioconjug Chem 2020; 31:1025-1051. [PMID: 32150392 DOI: 10.1021/acs.bioconjchem.0c00119] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Cell death plays a prominent role in the treatment of cancer, because most anticancer therapies act by the induction of cell death including apoptosis, necrosis, and other pathways of cell death. Imaging cell death helps to identify treatment responders from nonresponders and thus enables patient-tailored therapy, which will increase the likelihood of treatment response and ultimately lead to improved patient survival. By taking advantage of molecular probes that specifically target the biomarkers/biochemical processes of cell death, cell death imaging can be successfully achieved. In recent years, with the increased understanding of the molecular mechanism of cell death, a variety of well-defined biomarkers/biochemical processes of cell death have been identified. By targeting these established cell death biomarkers/biochemical processes, a set of molecular imaging probes have been developed and evaluated for early monitoring treatment response in tumors. In this review, we mainly present the recent advances in identifying useful biomarkers/biochemical processes for both apoptosis and necrosis imaging and in developing molecular imaging probes targeting these biomarkers/biochemical processes, with a focus on their application in early evaluation of tumor response to therapy.
Collapse
Affiliation(s)
- Dongjian Zhang
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, P.R. China.,Laboratories of Translational Medicine, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, P.R. China
| | - Qiaomei Jin
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, P.R. China.,Laboratories of Translational Medicine, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, P.R. China
| | - Cuihua Jiang
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, P.R. China.,Laboratories of Translational Medicine, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, P.R. China
| | - Meng Gao
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, P.R. China.,Laboratories of Translational Medicine, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, P.R. China
| | - Yicheng Ni
- Theragnostic Laboratory, Campus Gasthuisberg, KU Leuven, Leuven 3000, Belgium
| | - Jian Zhang
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, P.R. China.,Laboratories of Translational Medicine, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, P.R. China
| |
Collapse
|
9
|
Rajavenkatesh K, Padmaja M, Janani I, Aishwarya S, Purna Sai K, Thennarasu S. Design and synthesis of a novel peptide for selective detection of cancer cells. Chem Biol Drug Des 2020; 95:610-623. [PMID: 32147880 DOI: 10.1111/cbdd.13675] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Revised: 01/28/2020] [Accepted: 02/23/2020] [Indexed: 01/01/2023]
Abstract
Using a minimalist approach, an 11-residue peptide (Peptide 1) tagged with rhodamine fluorophore was designed and synthesized for selective detection of cancer cells. Peptide 1 contains RGD and NGR motifs to bind, respectively, integrins and aminopeptidase CD13, which are over expressed in cancer cells. Surface tension measurements revealed that peptide 1 possess surface-active property owing to the overall hydrophobicity and cationic nature of the peptide. Peptide 1 displays cancer cell-selective binding at ≤5.0 µM concentrations, while peptide 2 (randomized sequence of 1) shows non-selective binding to normal and cancer cells. Fluorescence microscopy and FACS analysis demonstrated the intracellular localization of peptide 1 in three different cancer cell lines, confirming the role of RGD and NGR motifs. Cytotoxicity assay exhibited the viability of normal and cancer cells up to 100 µM concentrations of peptide 1. Steady-state fluorescence measurements disclosed the preferential interactions of the peptide 1 with anionic POPC/POPG bilayers rather than with zwitterionic POPC lipid bilayers. Circular dichroism studies showed minimal changes in the secondary structure of peptide 1 upon binding with the anionic lipid bilayers. Peptide 1 is largely unordered, non-toxic, and useful for identification of cancer cells. Peptide 1 provides a template for designing drug-loaded peptides for targeted delivery into cancer cells.
Collapse
Affiliation(s)
| | - Murali Padmaja
- Biological Materials Laboratory, CSIR-Central Leather Research Institute, Chennai, India
| | - Indrakumar Janani
- Biological Materials Laboratory, CSIR-Central Leather Research Institute, Chennai, India
| | - Satish Aishwarya
- Biological Materials Laboratory, CSIR-Central Leather Research Institute, Chennai, India
| | - Korrapati Purna Sai
- Biological Materials Laboratory, CSIR-Central Leather Research Institute, Chennai, India
| | - Sathiah Thennarasu
- Organic and Bioorganic Chemistry Laboratory, CSIR-Central Leather Research Institute, Chennai, India
| |
Collapse
|
10
|
Cho HJ, Park SJ, Lee YS, Kim S. Theranostic iRGD peptide containing cisplatin prodrug: Dual-cargo tumor penetration for improved imaging and therapy. J Control Release 2019; 300:73-80. [PMID: 30831135 DOI: 10.1016/j.jconrel.2019.02.043] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Revised: 02/07/2019] [Accepted: 02/28/2019] [Indexed: 01/30/2023]
Abstract
In theranostics, peptide-based platforms have widely been exploited owing to their unique biological functions and chemical versatilities. As a tumor-homing ligand, internalizing RGD peptide (iRGD), composed of a tumor-targeting sequence (RGD) and a cell-penetrating C-end Rule (CendR) motif, is known to facilitate the tumor-specific delivery of payloads that are covalently conjugated on its N-terminal fragment or co-administered without any covalent linkages. However, theranostic uses of the iRGD-based platform remain in its infancy with its full potential unexplored; for instance, detailed mechanism of iRGD fragmentation during internalization, strategies for the tumor-specific release of payloads from iRGD and the role of the C-terminal iRGD fragment in delivery have yet to be revealed. In this study, we designed a dual-channel fluorescent cyclic iRGD (TAMRA-iRGDC-Cy5.5) to track each of the N- and C-terminal fragments separately during the tumor internalization process. It turned out that both fragments undergo translocation into cancer cells together and are localized within endosomal-lysosomal compartments. The resulting co-internalization of both iRGD fragments allowed us to develop a new theranostic peptide platform (Cy5.5-iRGDC-Pt(IV)) by conjugating a fluorescent dye and a cisplatin prodrug on each terminus of cyclic iRGD for simultaneous cancer-targeted imaging and therapy. Compared to a control peptide having a non-iRGD sequence, the Cy5.5-iRGDC-Pt(IV) did not only provide a better tumor imaging contrast but also induced tumor-specific apoptosis leading to efficacious tumor suppression. Besides the outstanding cancer imaging and therapeutic performance, the Cy5.5-iRGDC-Pt(IV) revealed negligible systemic toxicity, holding potential to be applied for theranostic uses.
Collapse
Affiliation(s)
- Hong-Jun Cho
- Center for Theragnosis, Korea Institute of Science and Technology, 5 Hwarang-ro 14-gil, Seongbuk-gu, Seoul 02792, Republic of Korea
| | - Sung-Jun Park
- Center for Theragnosis, Korea Institute of Science and Technology, 5 Hwarang-ro 14-gil, Seongbuk-gu, Seoul 02792, Republic of Korea; School of Chemical and Biological Engineering, Seoul National University, Seoul 08826, Republic of Korea
| | - Yoon-Sik Lee
- School of Chemical and Biological Engineering, Seoul National University, Seoul 08826, Republic of Korea
| | - Sehoon Kim
- Center for Theragnosis, Korea Institute of Science and Technology, 5 Hwarang-ro 14-gil, Seongbuk-gu, Seoul 02792, Republic of Korea; KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Republic of Korea; Division of Bio-Medical Science & Technology, KIST School, Korea University of Science and Technology (UST), Seoul 02792, Republic of Korea.
| |
Collapse
|
11
|
Bae SM, Park SJ, Choi M, Song M, Cho YE, Do EJ, Ryu YM, Park S, Lee BH, Lee SW, Hwang SW, Park SH, Yang DH, Ye BD, Byeon JS, Yang SK, Joo J, Kim SY, Myung SJ. PSP1, a Phosphatidylserine-Recognizing Peptide, Is Useful for Visualizing Radiation-Induced Apoptosis in Colorectal Cancer In Vitro and In Vivo. Transl Oncol 2018; 11:1044-1052. [PMID: 29982102 PMCID: PMC6034579 DOI: 10.1016/j.tranon.2018.06.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Revised: 06/12/2018] [Accepted: 06/15/2018] [Indexed: 02/07/2023] Open
Abstract
Accurate and timely visualization of apoptotic status in response to radiation is necessary for deciding whether to continue radiation or change to another mode of treatment. This is especially critical in patients with colorectal cancer, which requires a delicate combination of surgery, radiation, and chemotherapy in order to achieve optimal outcome. In this study, we investigated the potential of phosphatidylserine-recognizing peptide 1 (PSP1) as an apoptosis-targeting probe, which identifies phosphatidylserine on cell surfaces. We first screened colon cancer cell lines for their sensitivity to radiation and selected two cell lines: HCT116 and HT29. Cell binding assay using fluorescence-activated cell sorting and optical imaging showed that HCT116 cells had better binding to PSP1 than HT29 cells. Thus, mouse xenograft model using HCT116 cells was generated and was topically irradiated with either single or fractionated dose of radiation followed by systemic administration of PSP1 for subsequent molecular optical imaging. We confirmed that the PSP1 probe was selectively bound to apoptosis-induced tumor in a radiation dose-dependent manner. We also observed that fractionated radiation regimen, which is recently being used in clinical situation, was more effective in inducing tumor apoptosis than corresponding single-dose radiation treatment. We then evaluated the correlation between tumor targeting of PSP1 and suppression effect of tumor development and found that tumor volume and fluorescence intensity were correlated before (correlation coefficient r2 = 0.534) and after (r2 = 0.848) radiation therapy. Our study shows that PSP1 peptide is an efficient index probe for deciding "go or no-go" for radiation therapy in colorectal cancer.
Collapse
Affiliation(s)
- Sang Mun Bae
- Asan Institute for Life Science, Asan Medical Center, University of Ulsan College of medicine, Seoul, Republic of Korea
| | - Soo Jung Park
- Asan Institute for Life Science, Asan Medical Center, University of Ulsan College of medicine, Seoul, Republic of Korea; Department of Internal Medicine and Institute of Gastroenterology, Yonsei University College of medicine, Seoul, Republic of Korea
| | - Myoungeun Choi
- Department of Dermatology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Miyeoun Song
- Department of Life and Nanopharmaceutical Science, Kyung Hee University, Seoul, Republic of Korea
| | - Young Eun Cho
- Asan Institute for Life Science, Asan Medical Center, University of Ulsan College of medicine, Seoul, Republic of Korea
| | - Eun-Ju Do
- Asan Institute for Life Science, Asan Medical Center, University of Ulsan College of medicine, Seoul, Republic of Korea
| | - Yeon-Mi Ryu
- Asan Institute for Life Science, Asan Medical Center, University of Ulsan College of medicine, Seoul, Republic of Korea
| | - Sunha Park
- Asan Institute for Life Science, Asan Medical Center, University of Ulsan College of medicine, Seoul, Republic of Korea
| | - Byung-Heon Lee
- Department of Biochemistry and Cell Biology, Kyungpook National University School of Medicine, Daegu, Republic of Korea
| | - Sang-Wook Lee
- Department of Radiation Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Sung Wook Hwang
- Department of Gastroenterology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Sang Hyoung Park
- Department of Gastroenterology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Dong-Hoon Yang
- Department of Gastroenterology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Byong Duk Ye
- Department of Gastroenterology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Jeong-Sik Byeon
- Department of Gastroenterology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Suk-Kyun Yang
- Department of Gastroenterology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Jinmyoung Joo
- Biomedical Engineering Research Center, Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea; Department of Convergence Medicine, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Sang-Yeob Kim
- Asan Institute for Life Science, Asan Medical Center, University of Ulsan College of medicine, Seoul, Republic of Korea; Department of Convergence Medicine, University of Ulsan College of Medicine, Seoul, Republic of Korea.
| | - Seung-Jae Myung
- Asan Institute for Life Science, Asan Medical Center, University of Ulsan College of medicine, Seoul, Republic of Korea; Department of Gastroenterology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea; Department of Convergence Medicine, University of Ulsan College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
12
|
Kang JS. Fluorescence Detection of Cell Death in Liver of Mice Treated with Thioacetamide. Toxicol Res 2018; 34:1-6. [PMID: 29371995 PMCID: PMC5776913 DOI: 10.5487/tr.2018.34.1.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Revised: 12/12/2017] [Accepted: 12/13/2017] [Indexed: 11/20/2022] Open
Abstract
The purpose of this study was to detect cell death in the liver of mice treated with thioacetamide (TAA) using fluorescence bioimaging and compare this outcome with that using conventional histopathological examination. At 6 weeks of age, 24 mice were randomly divided into three groups: group 1 (G1), control group; group 2 (G2), fluorescence probe control group; group 3 (G3), TAA-treated group. G3 mice were treated with TAA. Twenty-two hours after TAA treatment, G2 and G3 mice were treated with Annexin-Vivo 750. Fluorescence in vivo bioimaging was performed by fluorescence molecular tomography at two hours after Annexin-Vivo 750 treatment, and fluorescence ex vivo bioimaging of the liver was performed. Liver damage was validated by histopathological examination. In vivo bioimaging showed that the fluorescence intensity was increased in the right upper part of G3 mice compared with that in G2 mice, whereas G1 mice showed no signal. Additionally ex vivo bioimaging showed that the fluorescence intensity was significantly increased in the livers of G3 mice compared with those in G1 or G2 mice (p < 0.05). Histopathological examination of the liver showed no cell death in G1 and G2 mice. However, in G3 mice, there was destruction of hepatocytes and increased cell death. Terminal deoxynucleotidyl transferase dUTP nick end labeling staining confirmed many cell death features in the liver of G3 mice, whereas no pathological findings were observed in the liver of G1 and G2 mice. Taken together, fluorescence bioimaging in this study showed the detection of cell death and made it possible to quantify the level of cell death in male mice. The outcome was correlated with conventional biomedical examination. As it was difficult to differentiate histological location by fluorescent bioimaging, it is necessary to develop specific fluorescent dyes for monitoring hepatic disease progression and to exploit new bioimaging techniques without dye-labeling.
Collapse
Affiliation(s)
- Jin Seok Kang
- Department of Biomedical Laboratory Science, Namseoul University, Cheonan, Korea.,Molecular Diagnostics Research Institute, Namseoul University, Cheonan, Korea
| |
Collapse
|
13
|
Abbady AQ, Twair A, Ali B, Murad H. Characterization of Annexin V Fusion with the Superfolder GFP in Liposomes Binding and Apoptosis Detection. Front Physiol 2017; 8:317. [PMID: 28579961 PMCID: PMC5437369 DOI: 10.3389/fphys.2017.00317] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Accepted: 05/02/2017] [Indexed: 12/29/2022] Open
Abstract
Programed cell death is a critical and unavoidable part of life. One of the most widely used markers for dying cells, by apoptosis or pyroptosis, is the redistribution of phosphatidylserine (PS) from the inner to the outer plasma membrane leaflet. Annexin V protein is a sensitive and specific probe to mark this event because of its high affinity to the exposed PS. Beyond that, annexin V can bind to any PS-containing phospholipid bilayer of almost all tiny forms of membranous vesicles like blood platelets, exosomes, or even nanostructured liposomes. In this work, recombinant human annexin V was produced as a fusion with a highly fluorescent superfolder derivative of the green fluorescent protein (sfGFP) in Escherichia coli. The fusion protein(sfGFP-ANXV, 64 kDa), annexin V (ANXV, 40 kDa), and sfGFP (27 kDa) were separately produced after cloning their encoding genes in pRSET plasmid, and all proteins were expressed in a soluble form, then purified in high yields because of their N-terminal 6× His tag (~150 mg of pure protein per 1 L culture). Superiority of this fluorescent fusion protein over fluorescein-conjugated annexin V was demonstrated in binding to phospholipids (and their liposomes), prepared from natural sources (soya bean and egg yolk) that have different content of PS, by using different methods including ELISA, dot-blotting, surface plasmon resonance, and flow cytometry. We also applied fluorescent annexin V in the detection of apoptotic cells by flow cytometry and fluorescent microscopy. Interestingly, sfGFP-ANXV fusion was more sensitive to early apoptotic stressed HeLa cells than fluorescein-conjugated-ANXV. This highly expressed and functional sfGFP-ANXV fusion protein provides a promising ready-to-use molecular tool for quantifying liposomes (or similarly exosomes) and detecting apoptosis in cells.
Collapse
Affiliation(s)
- Abdul Qader Abbady
- Department of Molecular Biology and Biotechnology, Atomic Energy Commission of SyriaDamascus, Syria
| | - Aya Twair
- Department of Molecular Biology and Biotechnology, Atomic Energy Commission of SyriaDamascus, Syria.,Department of Animal Biology, Faculty of Sciences, Damascus UniversityDamascus, Syria
| | - Bouthaina Ali
- Department of Molecular Biology and Biotechnology, Atomic Energy Commission of SyriaDamascus, Syria
| | - Hossam Murad
- Department of Molecular Biology and Biotechnology, Atomic Energy Commission of SyriaDamascus, Syria
| |
Collapse
|
14
|
Perreault A, Richter S, Bergman C, Wuest M, Wuest F. Targeting Phosphatidylserine with a 64Cu-Labeled Peptide for Molecular Imaging of Apoptosis. Mol Pharm 2016; 13:3564-3577. [DOI: 10.1021/acs.molpharmaceut.6b00666] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Amanda Perreault
- Department of Oncology, Cross
Cancer Institute, University of Alberta, 11560 University Avenue, Edmonton, Alberta T6G 2X4, Canada
| | - Susan Richter
- Department of Oncology, Cross
Cancer Institute, University of Alberta, 11560 University Avenue, Edmonton, Alberta T6G 2X4, Canada
| | - Cody Bergman
- Department of Oncology, Cross
Cancer Institute, University of Alberta, 11560 University Avenue, Edmonton, Alberta T6G 2X4, Canada
| | - Melinda Wuest
- Department of Oncology, Cross
Cancer Institute, University of Alberta, 11560 University Avenue, Edmonton, Alberta T6G 2X4, Canada
| | - Frank Wuest
- Department of Oncology, Cross
Cancer Institute, University of Alberta, 11560 University Avenue, Edmonton, Alberta T6G 2X4, Canada
| |
Collapse
|
15
|
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
|