101
|
Favreau PF, He J, Gil DA, Deming DA, Huisken J, Skala MC. Label-free redox imaging of patient-derived organoids using selective plane illumination microscopy. BIOMEDICAL OPTICS EXPRESS 2020; 11:2591-2606. [PMID: 32499946 PMCID: PMC7249841 DOI: 10.1364/boe.389164] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 04/07/2020] [Accepted: 04/09/2020] [Indexed: 05/04/2023]
Abstract
High-throughput drug screening of patient-derived organoids offers an attractive platform to determine cancer treatment efficacy. Here, selective plane illumination microscopy (SPIM) was used to determine treatment response in organoids with endogenous fluorescence from the metabolic coenzymes NAD(P)H and FAD. Rapid 3-D autofluorescence imaging of colorectal cancer organoids was achieved. A quantitative image analysis approach was developed to segment each organoid and quantify changes in endogenous fluorescence caused by treatment. Quantitative analysis of SPIM volumes confirmed the sensitivity of patient-derived organoids to standard therapies. This proof-of-principle study demonstrates that SPIM is a powerful tool for high-throughput screening of organoid treatment response.
Collapse
Affiliation(s)
| | - Jiaye He
- Morgridge Institute for Research, Madison, WI 53715, USA
- Max Planck Institute for Molecular Cell Biology and Genetics, 01307 Dresden, Saxony, Germany
- Department of Integrative Biology, University of Wisconsin–Madison, Madison, WI 53715, USA
| | - Daniel A. Gil
- Morgridge Institute for Research, Madison, WI 53715, USA
- Department of Biomedical Engineering, University of Wisconsin–Madison, Madison, WI 53715, USA
| | - Dustin A. Deming
- University of Wisconsin Carbone Cancer Center, Madison, WI 53715, USA
- Division of Hematology and Oncology, Department of Medicine, University of Wisconsin–Madison, Madison, WI 53715, USA
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin–Madison, Madison, WI 53715, USA
- William S Middleton Memorial Veterans Hospital, Madison, WI 53715, USA
| | - Jan Huisken
- Morgridge Institute for Research, Madison, WI 53715, USA
- Department of Integrative Biology, University of Wisconsin–Madison, Madison, WI 53715, USA
| | - Melissa C. Skala
- Morgridge Institute for Research, Madison, WI 53715, USA
- Department of Biomedical Engineering, University of Wisconsin–Madison, Madison, WI 53715, USA
| |
Collapse
|
102
|
Pouli D, Thieu HT, Genega EM, Baecher-Lind L, House M, Bond B, Roncari DM, Evans ML, Rius-Diaz F, Munger K, Georgakoudi I. Label-free, High-Resolution Optical Metabolic Imaging of Human Cervical Precancers Reveals Potential for Intraepithelial Neoplasia Diagnosis. CELL REPORTS MEDICINE 2020; 1. [PMID: 32577625 PMCID: PMC7311071 DOI: 10.1016/j.xcrm.2020.100017] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
While metabolic changes are considered a cancer hallmark, their assessment has not been incorporated in the detection of early or precancers, when treatment is most effective. Here, we demonstrate that metabolic changes are detected in freshly excised human cervical precancerous tissues using label-free, non-destructive imaging of the entire epithelium. The images rely on two-photon excited fluorescence from two metabolic co-enzymes, NAD(P)H and FAD, and have micron-level resolution, enabling sensitive assessments of the redox ratio and mitochondrial fragmentation, which yield metrics of metabolic function and heterogeneity. Simultaneous characterization of morphological features, such as the depth-dependent variation of the nuclear:cytoplasmic ratio, is demonstrated. Multi-parametric analysis combining several metabolic metrics with morphological ones enhances significantly the diagnostic accuracy of identifying high-grade squamous intraepithelial lesions. Our results motivate the translation of such functional metabolic imaging to in vivo studies, which may enable improved identification of cervical lesions, and other precancers, at the bedside.
Collapse
Affiliation(s)
- Dimitra Pouli
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, MA 02155, USA.,Present address: Department of Pathology, Beth Israel Deaconess Medical Center and Harvard Medical School, 330 Brookline Avenue, Boston, MA 02115, USA
| | - Hong-Thao Thieu
- Department of Obstetrics and Gynecology, Tufts University School of Medicine, Tufts Medical Center, 800 Washington Street, Boston, MA 02111, USA
| | - Elizabeth M Genega
- Department of Pathology and Laboratory Medicine, Tufts University School of Medicine, Tufts Medical Center, 800 Washington Street, Boston, MA 02111, USA
| | - Laura Baecher-Lind
- Department of Obstetrics and Gynecology, Tufts University School of Medicine, Tufts Medical Center, 800 Washington Street, Boston, MA 02111, USA
| | - Michael House
- Department of Obstetrics and Gynecology, Tufts University School of Medicine, Tufts Medical Center, 800 Washington Street, Boston, MA 02111, USA
| | - Brian Bond
- Department of Obstetrics and Gynecology, Tufts University School of Medicine, Tufts Medical Center, 800 Washington Street, Boston, MA 02111, USA.,Present address: Department of Obstetrics and Gynecology, University of Massachusetts School of Medicine, 55 Lake Avenue North, Worcester, MA 01655, USA
| | - Danielle M Roncari
- Department of Obstetrics and Gynecology, Tufts University School of Medicine, Tufts Medical Center, 800 Washington Street, Boston, MA 02111, USA
| | - Megan L Evans
- Department of Obstetrics and Gynecology, Tufts University School of Medicine, Tufts Medical Center, 800 Washington Street, Boston, MA 02111, USA
| | - Francisca Rius-Diaz
- Department of Preventive Medicine and Public Health, Faculty of Medicine, University of Málaga, 32 Louis Pasteur Boulevard, 29071 Málaga, Spain
| | - Karl Munger
- Department of Developmental, Molecular, and Chemical Biology, Tufts University School of Medicine, 136 Harrison Avenue, Boston, MA 02111, USA
| | - Irene Georgakoudi
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, MA 02155, USA.,Lead Contact
| |
Collapse
|
103
|
Datta R, Heaster TM, Sharick JT, Gillette AA, Skala MC. Fluorescence lifetime imaging microscopy: fundamentals and advances in instrumentation, analysis, and applications. JOURNAL OF BIOMEDICAL OPTICS 2020; 25:1-43. [PMID: 32406215 PMCID: PMC7219965 DOI: 10.1117/1.jbo.25.7.071203] [Citation(s) in RCA: 399] [Impact Index Per Article: 79.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Accepted: 04/24/2020] [Indexed: 05/18/2023]
Abstract
SIGNIFICANCE Fluorescence lifetime imaging microscopy (FLIM) is a powerful technique to distinguish the unique molecular environment of fluorophores. FLIM measures the time a fluorophore remains in an excited state before emitting a photon, and detects molecular variations of fluorophores that are not apparent with spectral techniques alone. FLIM is sensitive to multiple biomedical processes including disease progression and drug efficacy. AIM We provide an overview of FLIM principles, instrumentation, and analysis while highlighting the latest developments and biological applications. APPROACH This review covers FLIM principles and theory, including advantages over intensity-based fluorescence measurements. Fundamentals of FLIM instrumentation in time- and frequency-domains are summarized, along with recent developments. Image segmentation and analysis strategies that quantify spatial and molecular features of cellular heterogeneity are reviewed. Finally, representative applications are provided including high-resolution FLIM of cell- and organelle-level molecular changes, use of exogenous and endogenous fluorophores, and imaging protein-protein interactions with Förster resonance energy transfer (FRET). Advantages and limitations of FLIM are also discussed. CONCLUSIONS FLIM is advantageous for probing molecular environments of fluorophores to inform on fluorophore behavior that cannot be elucidated with intensity measurements alone. Development of FLIM technologies, analysis, and applications will further advance biological research and clinical assessments.
Collapse
Affiliation(s)
- Rupsa Datta
- Morgridge Institute for Research, Madison, Wisconsin, United States
| | - Tiffany M. Heaster
- Morgridge Institute for Research, Madison, Wisconsin, United States
- University of Wisconsin, Department of Biomedical Engineering, Madison, Wisconsin, United States
| | - Joe T. Sharick
- Morgridge Institute for Research, Madison, Wisconsin, United States
| | - Amani A. Gillette
- Morgridge Institute for Research, Madison, Wisconsin, United States
- University of Wisconsin, Department of Biomedical Engineering, Madison, Wisconsin, United States
| | - Melissa C. Skala
- Morgridge Institute for Research, Madison, Wisconsin, United States
- University of Wisconsin, Department of Biomedical Engineering, Madison, Wisconsin, United States
| |
Collapse
|
104
|
Awasthi K, Chang FL, Hsieh PY, Hsu HY, Ohta N. Characterization of endogenous fluorescence in nonsmall lung cancerous cells: A comparison with nonmalignant lung normal cells. JOURNAL OF BIOPHOTONICS 2020; 13:e201960210. [PMID: 32067342 DOI: 10.1002/jbio.201960210] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 02/11/2020] [Accepted: 02/13/2020] [Indexed: 06/10/2023]
Abstract
Monitoring fluorescence properties of endogenous fluorophores such as nicotinamide adenine dinucleotide (NADH) and flavin adenine dinucleotide (FAD) in normal and cancerous cells provide substantial information noninvasively on biochemical and biophysical aspects of metabolic dysfunction of cancerous cells. Time-resolved spectral profiles and fluorescence lifetime images of NADH and FAD were obtained in human lung nonsmall carcinomas (H661 and A549) and normal lung cells (MRC-5). Both fluorophores show the fast and slowly decaying emission components upon pulsed excitation, and fluorescence spectra of NADH and FAD show blue- and red-shifts, respectively, during their decay. All identified lifetime components of NADH and FAD were found to be shorter in cancerous cells than in normal cells, no matter how they were measured under different extra-cellular conditions (cells suspended in cuvette and cells attached on glass substrate), indicating that the changes in metabolism likely altered the subcellular milieu and potentially also affected the interaction of NADH and FAD with enzymes to which these cofactors were bound. The intensity ratio of NADH and FAD of cancerous cells was also shown to be larger than that of normal cells.
Collapse
Affiliation(s)
- Kamlesh Awasthi
- Department of Applied Chemistry and Institute of Molecular Science, National Chiao Tung University, Hsinchu, Taiwan
- Center for Emergent Functional Matter Science, National Chiao Tung University, Hsinchu, Taiwan
| | - Feng-Lin Chang
- Department of Applied Chemistry and Institute of Molecular Science, National Chiao Tung University, Hsinchu, Taiwan
| | - Pei-Ying Hsieh
- Center for Emergent Functional Matter Science, National Chiao Tung University, Hsinchu, Taiwan
| | - Hsin-Yun Hsu
- Department of Applied Chemistry and Institute of Molecular Science, National Chiao Tung University, Hsinchu, Taiwan
- Center for Emergent Functional Matter Science, National Chiao Tung University, Hsinchu, Taiwan
| | - Nobuhiro Ohta
- Department of Applied Chemistry and Institute of Molecular Science, National Chiao Tung University, Hsinchu, Taiwan
- Center for Emergent Functional Matter Science, National Chiao Tung University, Hsinchu, Taiwan
| |
Collapse
|
105
|
Human Colon Organoids and Other Laboratory Strategies to Enhance Patient Treatment Selection. Curr Treat Options Oncol 2020; 21:35. [PMID: 32328818 DOI: 10.1007/s11864-020-00737-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
OPINION STATEMENT Though many advancements in personalized medicine have been made, better methods are still needed to predict treatment benefit for patients with colorectal cancer. Patient-derived cancer organoids (PDCOs) are a major advance towards true personalization of treatment strategies. A growing body of literature is demonstrating the feasibility of PDCOs as an accurate and high-throughput preclinical tool for patient treatment selection. Many studies demonstrate that these cultures are readily generated and represent the tumors they were derived from phenotypically and based on their mutation profile. This includes maintenance of the driver muatations giving the cancer cells a selective growth advantage, and also heterogeneity, including molecular and metabolic heterogeneity. Additionally, PDCOs are now being utilized to develop patient biospecimen repositories, perform high to moderate-throughput drug screening, and to potentially predict treatment response for individual patients that are undergoing anti-cancer treatments. In order to develop PDCOs as a true clinical tool, further studies are required to determine the reproducibility and accuracy of these models to predict patient response.
Collapse
|
106
|
Yang L, Liu B, Chen H, Gao R, Huang K, Guo Q, Li F, Chen W, He J. Progress in the application of organoids to breast cancer research. J Cell Mol Med 2020; 24:5420-5427. [PMID: 32283573 PMCID: PMC7214171 DOI: 10.1111/jcmm.15216] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2019] [Revised: 02/13/2020] [Accepted: 03/06/2020] [Indexed: 12/17/2022] Open
Abstract
Breast cancer is the most common cancer diagnosed in women. Breast cancer research is currently based mainly on animal models and traditional cell culture. However, the inherent species gap between humans and animals, as well as differences in organization between organs and cells, limits research advances. The breast cancer organoid can reproduce many of the key features of human breast cancer, thereby providing a new platform for investigating the mechanisms underlying the development, progression, metastasis and drug resistance of breast cancer. The application of organoid technology can also promote drug discovery and the design of individualized treatment strategies. Here, we discuss the latest advances in the use of organoid technology for breast cancer research.
Collapse
Affiliation(s)
- Liping Yang
- Department of Breast Surgery, Peking University Shenzhen Hospital, Shenzhen, China.,Department of Breast Surgery, Shenzhen Second People's Hospital, the First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Baoer Liu
- Department of Breast Surgery, Peking University Shenzhen Hospital, Shenzhen, China.,Department of Breast Surgery, Shenzhen Second People's Hospital, the First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Haodong Chen
- Department of Breast Surgery, Peking University Shenzhen Hospital, Shenzhen, China
| | - Rui Gao
- Department of Breast Surgery, Peking University Shenzhen Hospital, Shenzhen, China
| | - Kanghua Huang
- Department of Breast Surgery, Peking University Shenzhen Hospital, Shenzhen, China
| | - Qiuyi Guo
- Department of Breast Surgery, Peking University Shenzhen Hospital, Shenzhen, China
| | - Feng Li
- Department of Breast Surgery, Peking University Shenzhen Hospital, Shenzhen, China
| | - Weicai Chen
- Department of Breast Surgery, Shenzhen Second People's Hospital, the First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Jinsong He
- Department of Breast Surgery, Peking University Shenzhen Hospital, Shenzhen, China
| |
Collapse
|
107
|
Lin M, Gao M, Pandalai PK, Cavnar MJ, Kim J. An Organotypic Microcosm for the Pancreatic Tumor Microenvironment. Cancers (Basel) 2020; 12:E811. [PMID: 32231028 PMCID: PMC7225919 DOI: 10.3390/cancers12040811] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 03/24/2020] [Accepted: 03/26/2020] [Indexed: 12/16/2022] Open
Abstract
Pancreatic duct adenocarcinoma (PDAC) is projected to become the second leading cause of cancer-related deaths in the next few years. Unfortunately, the development of novel therapies for PDAC has been challenged by a uniquely complex tumor microenvironment. The development of in vitro cancer organoids in recent years has demonstrated potential to increase therapies for patients with PDAC. Organoids have been established from PDAC murine and human tissues and they are representative of the primary tumor. Further, organoids have been shown beneficial in studies of molecular mechanisms and drug sensitivity testing. This review will cover the use of organoids to study PDAC development, invasiveness, and therapeutic resistance in the context of the tumor microenvironment, which is characterized by a dense desmoplastic reaction, hindered immune activity, and pro-tumor metabolic signaling. We describe investigations utilizing organoids to characterize the tumor microenvironment and also describe their limitations. Overall, organoids have great potential to serve as a versatile model of drug response and may be used to increase available therapies and improve survival for patients with PDAC.
Collapse
Affiliation(s)
| | | | | | | | - Joseph Kim
- Department of Surgery, University of Kentucky, Lexington, KY 40536, USA; (M.L.); (M.G.); (P.K.P.); (M.J.C.)
| |
Collapse
|
108
|
Jones JD, Ramser HE, Woessner AE, Veves A, Quinn KP. Quantifying Age-Related Changes in Skin Wound Metabolism Using In Vivo Multiphoton Microscopy. Adv Wound Care (New Rochelle) 2020; 9:90-102. [PMID: 31993251 PMCID: PMC6985773 DOI: 10.1089/wound.2019.1030] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Accepted: 07/28/2019] [Indexed: 12/27/2022] Open
Abstract
Objective: The elderly are at high risk for developing chronic skin wounds, but the effects of intrinsic aging on skin healing are difficult to isolate due to common comorbidities like diabetes. Our objective is to use multiphoton microscopy (MPM) to find endogenous, noninvasive biomarkers to differentiate changes in skin wound healing metabolism between young and aged mice in vivo. Approach: We utilized MPM to monitor skin metabolism at the edge of full-thickness, excisional wounds in 24- and 4-month-old mice of both sexes for 10 days. MPM can assess quantitative biomarkers of cellular metabolism in vivo by utilizing autofluorescence from the cofactors nicotinamide adenine dinucleotide (NADH) and flavin adenine dinucleotide (FAD). Results: An optical redox ratio of FAD/(NADH+FAD) autofluorescence and NADH fluorescence lifetime imaging revealed dynamic changes in keratinocyte function during healing. Aged female mice demonstrated an attenuation of keratinocyte proliferation during wound healing detectable optically through a higher redox ratio and longer NADH fluorescence lifetime. By measuring the correlation between NADH lifetime and the optical redox ratio at each day, we also demonstrate sensitivity to the proliferative phase of wound healing. Innovation: Label-free MPM was used to longitudinally monitor individual wounds in vivo, which revealed age-dependent differences in wound metabolism. Conclusion: These results indicate in vivo MPM can provide quantitative biomarkers of age-related delays in healing, which can be used in the future to provide patient-specific wound care.
Collapse
Affiliation(s)
- Jake D. Jones
- Department of Biomedical Engineering, University of Arkansas, Fayetteville, Arkansas
| | - Hallie E. Ramser
- Department of Biomedical Engineering, University of Arkansas, Fayetteville, Arkansas
| | - Alan E. Woessner
- Department of Biomedical Engineering, University of Arkansas, Fayetteville, Arkansas
| | - Aristidis Veves
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Kyle P. Quinn
- Department of Biomedical Engineering, University of Arkansas, Fayetteville, Arkansas
| |
Collapse
|
109
|
Favreau PF, Deal JA, Harris B, Weber DS, Rich TC, Leavesley SJ. Label-free spectroscopic tissue characterization using fluorescence excitation-scanning spectral imaging. JOURNAL OF BIOPHOTONICS 2020; 13:e201900183. [PMID: 31566889 PMCID: PMC8491137 DOI: 10.1002/jbio.201900183] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Revised: 09/09/2019] [Accepted: 09/15/2019] [Indexed: 05/04/2023]
Abstract
Spectral imaging approaches provide new possibilities for measuring and discriminating fluorescent molecules in living cells and tissues. These approaches often employ tunable filters and robust image processing algorithms to identify many fluorescent labels in a single image set. Here, we present results from a novel spectral imaging technology that scans the fluorescence excitation spectrum, demonstrating that excitation-scanning hyperspectral image data can discriminate among tissue types and estimate the molecular composition of tissues. This approach allows fast, accurate quantification of many fluorescent species from multivariate image data without the need of exogenous labels or dyes. We evaluated the ability of the excitation-scanning approach to identify endogenous fluorescence signatures in multiple unlabeled tissue types. Signatures were screened using multi-pass principal component analysis. Endmember extraction techniques revealed conserved autofluorescent signatures across multiple tissue types. We further examined the ability to detect known molecular signatures by constructing spectral libraries of common endogenous fluorophores and applying multiple spectral analysis techniques on test images from lung, liver and kidney. Spectral deconvolution revealed structure-specific morphologic contrast generated from pure molecule signatures. These results demonstrate that excitation-scanning spectral imaging, coupled with spectral imaging processing techniques, provides an approach for discriminating among tissue types and assessing the molecular composition of tissues. Additionally, excitation scanning offers the ability to rapidly screen molecular markers across a range of tissues without using fluorescent labels. This approach lays the groundwork for translation of excitation-scanning technologies to clinical imaging platforms.
Collapse
Affiliation(s)
- Peter F Favreau
- Department of Biochemistry, University of Wisconsin-Madison, Madison, Wisconsin
| | - Joshua A Deal
- Department of Chemical and Biomolecular Engineering, Center for Lung Biology, University of South Alabama, Mobile, Alabama
| | - Bradley Harris
- Department of Medical Sciences, University of South Alabama, Mobile, Alabama
| | - David S Weber
- Department of Physiology, University of South Alabama, Mobile, Alabama
| | - Thomas C Rich
- Department of Pharmacology, Center for Lung Biology, University of South Alabama, Mobile, Alabama
| | - Silas J Leavesley
- Department of Chemical and Biomolecular Engineering, Center for Lung Biology, University of South Alabama, Mobile, Alabama
- Department of Pharmacology, Center for Lung Biology, University of South Alabama, Mobile, Alabama
| |
Collapse
|
110
|
Cao R, Wallrabe H, Periasamy A. Multiphoton FLIM imaging of NAD(P)H and FAD with one excitation wavelength. JOURNAL OF BIOMEDICAL OPTICS 2020; 25:1-16. [PMID: 31920048 PMCID: PMC6951488 DOI: 10.1117/1.jbo.25.1.014510] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Accepted: 12/12/2019] [Indexed: 05/07/2023]
Abstract
Two-photon fluorescence lifetime imaging microscopy (FLIM) is widely used to capture autofluorescence signals from cellular components to investigate dynamic physiological changes in live cells and tissues. Among these intrinsic fluorophores, nicotinamide adenine dinucleotide (phosphate) (NAD(P)H) and flavin adenine dinucleotide (FAD)-essential coenzymes in cellular respiration-have been used as intrinsic fluorescent biomarkers for metabolic states in cancer and other pathologies. Traditional FLIM imaging for NAD(P)H, FAD, and in particular fluorescence lifetime redox ratio (FLIRR) requires a sequential multiwavelength excitation to avoid spectral bleed-through (SBT). This sequential imaging complicates image acquisition, may introduce motion artifacts, and reduce temporal resolution. Testing several two-photon excitation wavelengths in combination with optimized emission filters, we have proved a FLIM imaging protocol, allowing simultaneous image acquisition with a single 800-nm wavelength excitation for NADH and FAD with negligible SBT. As a first step, standard NADH and FAD single and mixed solutions were tested that mimic biological sample conditions. After these optimization steps, the assay was applied to two prostate cancer live cell lines: African-American (AA) and Caucasian-American (LNCaP), used in our previous publications. FLIRR result shows that, in cells, the 800-nm two-photon excitation wavelength is suitable for NADH and FAD FLIM imaging with negligible SBT. While NAD(P)H signals are decreased, sufficient photons are present for accurate lifetime fitting and FAD signals are measurably increased at lower laser power, compared with the common 890-nm excitation conditions. This single wavelength excitation allows a simplification of NADH and FAD FLIM imaging data analysis, decreasing the total imaging time. It also avoids motion artifacts and increases temporal resolution. This simplified assay will also make it more suitable to be applied in a clinical setting.
Collapse
Affiliation(s)
- Ruofan Cao
- University of Virginia, WM Keck Center for Cellular Imaging, Department of Biology, Charlottesville, Virginia, United States
| | - Horst Wallrabe
- University of Virginia, WM Keck Center for Cellular Imaging, Department of Biology, Charlottesville, Virginia, United States
| | - Ammasi Periasamy
- University of Virginia, WM Keck Center for Cellular Imaging, Department of Biology, Charlottesville, Virginia, United States
- University of Virginia, Department of Biomedical Engineering, Charlottesville, Virginia, United States
- Address all correspondence to Ammasi Periasamy, E-mail:
| |
Collapse
|
111
|
Smith JT, Yao R, Sinsuebphon N, Rudkouskaya A, Un N, Mazurkiewicz J, Barroso M, Yan P, Intes X. Fast fit-free analysis of fluorescence lifetime imaging via deep learning. Proc Natl Acad Sci U S A 2019; 116:24019-24030. [PMID: 31719196 PMCID: PMC6883809 DOI: 10.1073/pnas.1912707116] [Citation(s) in RCA: 81] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Fluorescence lifetime imaging (FLI) provides unique quantitative information in biomedical and molecular biology studies but relies on complex data-fitting techniques to derive the quantities of interest. Herein, we propose a fit-free approach in FLI image formation that is based on deep learning (DL) to quantify fluorescence decays simultaneously over a whole image and at fast speeds. We report on a deep neural network (DNN) architecture, named fluorescence lifetime imaging network (FLI-Net) that is designed and trained for different classes of experiments, including visible FLI and near-infrared (NIR) FLI microscopy (FLIM) and NIR gated macroscopy FLI (MFLI). FLI-Net outputs quantitatively the spatially resolved lifetime-based parameters that are typically employed in the field. We validate the utility of the FLI-Net framework by performing quantitative microscopic and preclinical lifetime-based studies across the visible and NIR spectra, as well as across the 2 main data acquisition technologies. These results demonstrate that FLI-Net is well suited to accurately quantify complex fluorescence lifetimes in cells and, in real time, in intact animals without any parameter settings. Hence, FLI-Net paves the way to reproducible and quantitative lifetime studies at unprecedented speeds, for improved dissemination and impact of FLI in many important biomedical applications ranging from fundamental discoveries in molecular and cellular biology to clinical translation.
Collapse
Affiliation(s)
- Jason T Smith
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY 12180;
| | - Ruoyang Yao
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY 12180
| | - Nattawut Sinsuebphon
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY 12180
| | - Alena Rudkouskaya
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY 12208
| | - Nathan Un
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY 12180
| | - Joseph Mazurkiewicz
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, Albany, NY 12208
| | - Margarida Barroso
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY 12208
| | - Pingkun Yan
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY 12180
| | - Xavier Intes
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY 12180;
| |
Collapse
|
112
|
Lin H, Fan T, Sui J, Wang G, Chen J, Zhuo S, Zhang H. Recent advances in multiphoton microscopy combined with nanomaterials in the field of disease evolution and clinical applications to liver cancer. NANOSCALE 2019; 11:19619-19635. [PMID: 31599299 DOI: 10.1039/c9nr04902a] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Multiphoton microscopy (MPM) is expected to become a powerful clinical tool, with its unique advantages of being label-free, high resolution, deep imaging depth, low light photobleaching and low phototoxicity. Nanomaterials, with excellent physical and chemical properties, are biocompatible and easy to prepare and functionalize. The addition of nanomaterials exactly compensates for some defects of MPM, such as the weak endogenous signal strength, limited imaging materials, insufficient imaging depth and lack of therapeutic effects. Therefore, combining MPM with nanomaterials is a promising biomedical imaging method. Here, we mainly review the principle of MPM and its application in liver cancer, especially in disease evolution and clinical applications, including monitoring tumor progression, diagnosing tumor occurrence, detecting tumor metastasis, and evaluating cancer therapy response. Then, we introduce the latest advances in the combination of MPM with nanomaterials, including the MPM imaging of gold nanoparticles (AuNPs) and carbon dots (CDs). Finally, we also propose the main challenges and future research directions of MPM technology in HCC.
Collapse
Affiliation(s)
- Hongxin Lin
- Fujian Normal University, Key Laboratory of OptoElectronic Science and Technology for Medicine of Ministry of Education, Fujian Provincial Key Laboratory of Photonics Technology, Fuzhou, 350007, China.
| | - Taojian Fan
- Shenzhen Engineering Laboratory of Phosphorene and Optoelectronics and Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, Shenzhen University, Shenzhen, 518060, China.
| | - Jian Sui
- Department of Gastrointestinal surgery, Fujian Provincial Hospital, Fuzhou, 350000, China
| | - Guangxing Wang
- Fujian Normal University, Key Laboratory of OptoElectronic Science and Technology for Medicine of Ministry of Education, Fujian Provincial Key Laboratory of Photonics Technology, Fuzhou, 350007, China.
| | - Jianxin Chen
- Fujian Normal University, Key Laboratory of OptoElectronic Science and Technology for Medicine of Ministry of Education, Fujian Provincial Key Laboratory of Photonics Technology, Fuzhou, 350007, China.
| | - Shuangmu Zhuo
- Fujian Normal University, Key Laboratory of OptoElectronic Science and Technology for Medicine of Ministry of Education, Fujian Provincial Key Laboratory of Photonics Technology, Fuzhou, 350007, China.
| | - Han Zhang
- Shenzhen Engineering Laboratory of Phosphorene and Optoelectronics and Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, Shenzhen University, Shenzhen, 518060, China.
| |
Collapse
|
113
|
Wang S, Chacko JV, Sagar AK, Eliceiri KW, Yuan M. Nonparametric empirical Bayesian framework for fluorescence-lifetime imaging microscopy. BIOMEDICAL OPTICS EXPRESS 2019; 10:5497-5517. [PMID: 31799027 PMCID: PMC6865096 DOI: 10.1364/boe.10.005497] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 08/31/2019] [Accepted: 09/29/2019] [Indexed: 05/02/2023]
Abstract
Fluorescence lifetime imaging microscopy (FLIM) is a powerful imaging tool used to study the molecular environment of flurophores. In time domain FLIM, extracting lifetime from fluorophores signals entails fitting data to a decaying exponential distribution function. However, most existing techniques for this purpose need large amounts of photons at each pixel and a long computation time, thus making it difficult to obtain reliable inference in applications requiring either short acquisition or minimal computation time. In this work, we introduce a new nonparametric empirical Bayesian framework for FLIM data analysis (NEB-FLIM), leading to both improved pixel-wise lifetime estimation and a more robust and computationally efficient integral property inference. This framework is developed based on a newly proposed hierarchical statistical model for FLIM data and adopts a novel nonparametric maximum likelihood estimator to estimate the prior distribution. To demonstrate the merit of the proposed framework, we applied it on both simulated and real biological datasets and compared it with previous classical methods on these datasets.
Collapse
Affiliation(s)
- Shulei Wang
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jenu V Chacko
- Laboratory for Optical and Computational Instrumentation, University of Wisconsin at Madison, Madison, WI 53706, USA
| | - Abdul K Sagar
- Laboratory for Optical and Computational Instrumentation, University of Wisconsin at Madison, Madison, WI 53706, USA
| | - Kevin W Eliceiri
- Laboratory for Optical and Computational Instrumentation, University of Wisconsin at Madison, Madison, WI 53706, USA
- Morgridge Institute for Research, Madison, WI 53706, USA
| | - Ming Yuan
- Department of Statistics, Columbia University, New York, NY 10027, USA
| |
Collapse
|
114
|
Broekgaarden M, Anbil S, Bulin AL, Obaid G, Mai Z, Baglo Y, Rizvi I, Hasan T. Modulation of redox metabolism negates cancer-associated fibroblasts-induced treatment resistance in a heterotypic 3D culture platform of pancreatic cancer. Biomaterials 2019; 222:119421. [PMID: 31494503 PMCID: PMC6934357 DOI: 10.1016/j.biomaterials.2019.119421] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 08/07/2019] [Accepted: 08/11/2019] [Indexed: 12/18/2022]
Abstract
The complex interplay between cancer cells and their microenvironment remains a major challenge in the design and optimization of treatment strategies for pancreatic ductal adenocarcinoma (PDAC). Recent investigations have demonstrated that mechanistically distinct combination therapies hold promise for treatment of PDAC, but effective clinical translation requires more accurate models that account for the abundant tumor-stroma and its influence on cancer growth, metabolism and treatment insensitivity. In this study, a modular 3D culture model that comprised PDAC cells and patient-derived cancer-associated fibroblasts (CAFs) was developed to assess the effects of PDAC-CAF interactions on treatment efficacies. Using newly-developed high-throughput imaging and image analysis tools, it was found that CAFs imparted a notable and statistically significant resistance to oxaliplatin chemotherapy and benzoporphyrin derivative-mediated photodynamic therapy, which associated with increased levels of basal oxidative metabolism. Increased treatment resistance and redox states were similarly observed in an orthotopic xenograft model of PDAC in which cancer cells and CAFs were co-implanted in mice. Combination therapies of oxaliplatin and PDT with the mitochondrial complex I inhibitor metformin overcame CAF-induced treatment resistance. The findings underscore that heterotypic microtumor culture models recapitulate metabolic alterations stemming from tumor-stroma interactions. The presented infrastructure can be adapted with disease-specific cell types and is compatible with patient-derived tissues to enable personalized screening and optimization of new metabolism-targeted treatment regimens for pancreatic cancer.
Collapse
Affiliation(s)
- Mans Broekgaarden
- Wellman Center for Photomedicine, Department of Dermatology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Sriram Anbil
- Wellman Center for Photomedicine, Department of Dermatology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA; The University of Texas School of Medicine, San Antonio, TX, USA
| | - Anne-Laure Bulin
- Wellman Center for Photomedicine, Department of Dermatology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Girgis Obaid
- Wellman Center for Photomedicine, Department of Dermatology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Zhiming Mai
- Wellman Center for Photomedicine, Department of Dermatology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Yan Baglo
- Wellman Center for Photomedicine, Department of Dermatology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Imran Rizvi
- Wellman Center for Photomedicine, Department of Dermatology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Tayyaba Hasan
- Wellman Center for Photomedicine, Department of Dermatology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA; Division of Health Sciences and Technology, Harvard-Massachusetts Institute of Technology, Boston, MA, USA.
| |
Collapse
|
115
|
Heaster TM, Landman BA, Skala MC. Quantitative Spatial Analysis of Metabolic Heterogeneity Across in vivo and in vitro Tumor Models. Front Oncol 2019; 9:1144. [PMID: 31737571 PMCID: PMC6839277 DOI: 10.3389/fonc.2019.01144] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Accepted: 10/15/2019] [Indexed: 12/12/2022] Open
Abstract
Metabolic preferences of tumor cells vary within a single tumor, contributing to tumor heterogeneity, drug resistance, and patient relapse. However, the relationship between tumor treatment response and metabolically distinct tumor cell populations is not well-understood. Here, a quantitative approach was developed to characterize spatial patterns of metabolic heterogeneity in tumor cell populations within in vivo xenografts and 3D in vitro cultures (i.e., organoids) of head and neck cancer. Label-free images of cell metabolism were acquired using two-photon fluorescence lifetime microscopy of the metabolic co-enzymes NAD(P)H and FAD. Previous studies have shown that NAD(P)H mean fluorescence lifetimes can identify metabolically distinct cells with varying drug response. Thus, density-based clustering of the NAD(P)H mean fluorescence lifetime was used to identify metabolic sub-populations of cells, then assessed in control, cetuximab-, cisplatin-, and combination-treated xenografts 13 days post-treatment and organoids 24 h post-treatment. Proximity analysis of these metabolically distinct cells was designed to quantify differences in spatial patterns between treatment groups and between xenografts and organoids. Multivariate spatial autocorrelation and principal components analyses of all autofluorescence intensity and lifetime variables were developed to further improve separation between cell sub-populations. Spatial principal components analysis and Z-score calculations of autofluorescence and spatial distribution variables also visualized differences between models. This analysis captures spatial distributions of tumor cell sub-populations influenced by treatment conditions and model-specific environments. Overall, this novel spatial analysis could provide new insights into tumor growth, treatment resistance, and more effective drug treatments across a range of microscopic imaging modalities (e.g., immunofluorescence, imaging mass spectrometry).
Collapse
Affiliation(s)
- Tiffany M. Heaster
- Department of Biomedical Engineering, University of Wisconsin—Madison, Madison, WI, United States
- Morgridge Institute for Research, Madison, WI, United States
| | - Bennett A. Landman
- Department of Electrical Engineering, Computer Engineering, and Computer Science, Vanderbilt University, Nashville, TN, United States
| | - Melissa C. Skala
- Department of Biomedical Engineering, University of Wisconsin—Madison, Madison, WI, United States
- Morgridge Institute for Research, Madison, WI, United States
| |
Collapse
|
116
|
Chacko JV, Eliceiri KW. NAD(P)H fluorescence lifetime measurements in fixed biological tissues. Methods Appl Fluoresc 2019; 7:044005. [PMID: 31553966 DOI: 10.1088/2050-6120/ab47e5] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Autofluorescence based fluorescence lifetime imaging microscopy (AF-FLIM) techniques have come a long way from early studies on cancer characterization and have now been widely employed in several cellular and animal studies covering a wide range of diseases. The majority of research in autofluorescence imaging (AFI) study metabolic fluxes in live biological samples. However, tissues from clinical or scientific studies are often chemically fixed for preservation and stabilization of tissue morphology. Fixation is particularly crucial for enzymatic, functional, or histopathology studies. Interpretations of metabolic imaging such as optical redox intensity imaging and AF-FLIM, have often been viewed as potentially unreliable in a fixed sample due to lack of studies in this field. In this study, we carefully evaluate the possibility of extracting microenvironment information in fixed tissues using reduced nicotinamide adenine dinucleotide (phosphate) (NAD(P)H) endogenous fluorescence. The ability to distinguish changes such as metabolism and pH using intrinsic fluorescence in fixed tissues has great pathological value. In this work, we show that the lifetime based metabolic contrast in a sample is preserved after chemical fixation. The fluorescence lifetime of a sample increases with an additive fixative like formaldehyde; however, the fixed tissues retain metabolic signatures even after fixation. This study presents an opportunity to successfully image archived unstained histopathology tissues, and generate useful AF-FLIM signatures. We demonstrate the capability to draw metabolic interpretations in fixed tissues even after long periods of storage.
Collapse
Affiliation(s)
- Jenu V Chacko
- Laboratory for Optical and Computational Instrumentation, U. Wisconsin at Madison, Madison WI, United States of America
| | | |
Collapse
|
117
|
Tang H, Song C, Ye F, Gao G, Ou X, Zhang L, Xie X, Xie X. miR-200c suppresses stemness and increases cellular sensitivity to trastuzumab in HER2+ breast cancer. J Cell Mol Med 2019; 23:8114-8127. [PMID: 31599500 PMCID: PMC6850933 DOI: 10.1111/jcmm.14681] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2019] [Revised: 08/14/2019] [Accepted: 08/28/2019] [Indexed: 12/22/2022] Open
Abstract
Resistance to trastuzumab remains a major obstacle in HER2‐overexpressing breast cancer treatment. miR‐200c is important for many functions in cancer stem cells (CSCs), including tumour recurrence, metastasis and resistance. We hypothesized that miR‐200c contributes to trastuzumab resistance and stemness maintenance in HER2‐overexpressing breast cancer. In this study, we used HER2‐positive SKBR3, HER2‐negative MCF‐7, and their CD44+CD24− phenotype mammospheres SKBR3‐S and MCF‐7‐S to verify. Our results demonstrated that miR‐200c was weakly expressed in breast cancer cell lines and cell line stem cells. Overexpression of miR‐200c resulted in a significant reduction in the number of tumour spheres formed and the population of CD44+CD24− phenotype mammospheres in SKBR3‐S. Combining miR‐200c with trastuzumab can significantly reduce proliferation and increase apoptosis of SKBR3 and SKBR3‐S. Overexpression of miR‐200c also eliminated its downstream target genes. These genes were highly expressed and positively related in breast cancer patients. Overexpression of miR‐200c also improved the malignant progression of SKBR3‐S and SKBR3 in vivo. miR‐200c plays an important role in the maintenance of the CSC‐like phenotype and increases drug sensitivity to trastuzumab in HER2+ cells and stem cells.
Collapse
Affiliation(s)
- Hailin Tang
- State Key Laboratory of Oncology in South China, Department of Breast Oncology, Collaborative Innovation Center of Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Cailu Song
- State Key Laboratory of Oncology in South China, Department of Breast Oncology, Collaborative Innovation Center of Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Feng Ye
- State Key Laboratory of Oncology in South China, Department of Breast Oncology, Collaborative Innovation Center of Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Guanfeng Gao
- State Key Laboratory of Oncology in South China, Department of Breast Oncology, Collaborative Innovation Center of Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Xueqi Ou
- State Key Laboratory of Oncology in South China, Department of Breast Oncology, Collaborative Innovation Center of Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Lijuan Zhang
- State Key Laboratory of Oncology in South China, Department of Breast Oncology, Collaborative Innovation Center of Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Xinhua Xie
- State Key Laboratory of Oncology in South China, Department of Breast Oncology, Collaborative Innovation Center of Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Xiaoming Xie
- State Key Laboratory of Oncology in South China, Department of Breast Oncology, Collaborative Innovation Center of Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| |
Collapse
|
118
|
Hutfilz A, Sonntag SR, Lewke B, Theisen-Kunde D, Grisanti S, Brinkmann R, Miura Y. Fluorescence Lifetime Imaging Ophthalmoscopy of the Retinal Pigment Epithelium During Wound Healing After Laser Irradiation. Transl Vis Sci Technol 2019; 8:12. [PMID: 31588376 PMCID: PMC6748347 DOI: 10.1167/tvst.8.5.12] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Accepted: 06/19/2019] [Indexed: 12/22/2022] Open
Abstract
Purpose To investigate the change in fluorescence lifetime of retinal pigment epithelium (RPE) after laser irradiation by using an organ culture model. Methods Porcine RPE-choroid-sclera explants were irradiated with selective retina treatment laser (wavelength: 527 nm, beam diameter: 200 μm, energy: 80–150 μJ). At 24 and 72 hours after irradiation, the mean fluorescence lifetime (τm) was measured with fluorescence lifetime imaging ophthalmoscopy (FLIO) (excitation wavelength: 473 nm, emission: short spectral channel: 498-560 nm, long spectral channel: 560–720 nm). For every laser spot, central damaged zone (zone 1: 120 × 120 μm), area including wound rim (280 × 280 μm except zone 1), and environmental zone (440 × 440 μm except zone 1 and 2) were analyzed. Peripheral zone at a distance from laser spots longer than 2000 μm was examined for comparison. Cell viability was evaluated with calcein-acetoxymethyl ester and morphology with fluorescence microscopy for filamentous-actin. Results The RPE defect after selective retina treatment was mostly closed within 72 hours. FLIO clearly demarcated the irradiated region, with prolonged τm at the center of the defect decreasing with eccentricity. In short spectral channel, but not in long spectral channel, τm in the environmental zone after 72 hours was still significantly longer than in the peripheral zone. Conclusions FLIO may clearly demarcate the RPE defect, demonstrate its closure, and, moreover, indicate the induced metabolic changes of surrounding cells during wound healing. Translational Relevance This ex vivo study showed that FLIO may be used to evaluate the extent and quality of restoration of the damaged RPE and to detect its metabolic change in human fundus noninvasively.
Collapse
Affiliation(s)
- Alessa Hutfilz
- Institute of Biomedical Optics, University of Lübeck, Lübeck, Germany.,Medical Laser Center Lübeck, Lübeck, Germany
| | - Svenja Rebecca Sonntag
- Department of Ophthalmology, University Hospital Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
| | - Britta Lewke
- Institute of Biomedical Optics, University of Lübeck, Lübeck, Germany.,Medical Laser Center Lübeck, Lübeck, Germany
| | | | - Salvatore Grisanti
- Department of Ophthalmology, University Hospital Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
| | - Ralf Brinkmann
- Institute of Biomedical Optics, University of Lübeck, Lübeck, Germany.,Medical Laser Center Lübeck, Lübeck, Germany
| | - Yoko Miura
- Institute of Biomedical Optics, University of Lübeck, Lübeck, Germany.,Department of Ophthalmology, University Hospital Schleswig-Holstein, Campus Lübeck, Lübeck, Germany.,Medical Laser Center Lübeck, Lübeck, Germany
| |
Collapse
|
119
|
Broekgaarden M, Bulin AL, Frederick J, Mai Z, Hasan T. Tracking Photodynamic- and Chemotherapy-Induced Redox-State Perturbations in 3D Culture Models of Pancreatic Cancer: A Tool for Identifying Therapy-Induced Metabolic Changes. J Clin Med 2019; 8:jcm8091399. [PMID: 31500115 PMCID: PMC6788194 DOI: 10.3390/jcm8091399] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 08/22/2019] [Accepted: 09/03/2019] [Indexed: 12/25/2022] Open
Abstract
The metabolic plasticity of cancer cells is considered a highly advantageous phenotype that is crucial for disease progression and acquisition of treatment resistance. A better understanding of cancer metabolism and its adaptability after treatments is vital to develop more effective therapies. To screen novel therapies and combination regimens, three-dimensional (3D) culture models of cancers are attractive platforms as they recapitulate key features of cancer. By applying non-perturbative intensity-based redox imaging combined with high-throughput image analysis, we demonstrated metabolic heterogeneity in various 3D culture models of pancreatic cancer. Photodynamic therapy and oxaliplatin chemotherapy, two cancer treatments with relevance to pancreatic cancer, induced perturbations in redox state in 3D microtumor cultures of pancreatic cancer. In an orthotopic mouse model of pancreatic cancer, a similar disruption in redox homeostasis was observed on ex vivo slices following photodynamic therapy in vivo. Taken together, redox imaging on cancer tissues combined with high-throughput analysis can elucidate dynamic spatiotemporal changes in metabolism following treatment, which will benefit the design of new metabolism-targeted therapeutic approaches.
Collapse
Affiliation(s)
- Mans Broekgaarden
- Wellman Center for Photomedicine, Department of Dermatology, Harvard Medical School and Massachusetts General Hospital, 40 Blossom Street, Boston, MA 02114, USA.
| | - Anne-Laure Bulin
- Wellman Center for Photomedicine, Department of Dermatology, Harvard Medical School and Massachusetts General Hospital, 40 Blossom Street, Boston, MA 02114, USA.
| | - Jane Frederick
- Wellman Center for Photomedicine, Department of Dermatology, Harvard Medical School and Massachusetts General Hospital, 40 Blossom Street, Boston, MA 02114, USA.
| | - Zhiming Mai
- Wellman Center for Photomedicine, Department of Dermatology, Harvard Medical School and Massachusetts General Hospital, 40 Blossom Street, Boston, MA 02114, USA.
| | - Tayyaba Hasan
- Wellman Center for Photomedicine, Department of Dermatology, Harvard Medical School and Massachusetts General Hospital, 40 Blossom Street, Boston, MA 02114, USA.
| |
Collapse
|
120
|
Liu Y, Xu J. High-resolution microscopy for imaging cancer pathobiology. CURRENT PATHOBIOLOGY REPORTS 2019; 7:85-96. [PMID: 32953251 PMCID: PMC7500261 DOI: 10.1007/s40139-019-00201-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
PURPOSE OF REVIEW Light microscopy plays an essential role in clinical diagnosis and understanding the pathogenesis of cancer. Conventional bright-field microscope is used to visualize abnormality in tissue architecture and nuclear morphology, but often suffers from many limitations. This review focuses on the potential of new imaging techniques to improve basic and clinical research in pathobiology. RECENT FINDINGS Light microscopy has significantly expanded its ability in resolution, imaging volume, speed and contrast. It now allows 3D high-resolution volumetric imaging of tissue architecture from large tissue and molecular structures at nanometer resolution. SUMMARY Pathologists and researchers now have access to various imaging tools to study cancer pathobiology in both breadth and depth. Although clinical adoption of a new technique is slow, the new imaging tools will provide significant new insights and open new avenues for improving early cancer detection, personalized risk assessment and identifying the best treatment strategies.
Collapse
Affiliation(s)
- Yang Liu
- Biomedical Optical Imaging Laboratory, Departments of Medicine and Bioengineering, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Jianquan Xu
- Biomedical Optical Imaging Laboratory, Departments of Medicine and Bioengineering, University of Pittsburgh, Pittsburgh, PA 15213, USA
| |
Collapse
|
121
|
Shcheslavskiy VI, Shirmanova MV, Jelzow A, Becker W. Multiparametric Time-Correlated Single Photon Counting Luminescence Microscopy. BIOCHEMISTRY (MOSCOW) 2019; 84:S51-S68. [PMID: 31213195 DOI: 10.1134/s0006297919140049] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Classic time-correlated single photon counting (TCSPC) technique involves detection of single photons of a periodic optical signal, registration of the photon arrival time in respect to the reference pulse, and construction of photon distribution with regard to the detection times. This technique achieves extremely high time resolution and near-ideal detection efficiency. Modern TCSPC is multi-dimensional, i.e., in addition to the photon arrival time relative to the excitation pulse, spatial coordinates within the image area, wavelength, time from the start of the experiment, and many other parameters are determined for each photon. Hence, the multi-dimensional TCSPC allows generation of photon distributions over these parameters. This review describes both classic and multi-dimensional types of TCSPC microscopy and their application for fluorescence lifetime imaging in different areas of biological studies.
Collapse
Affiliation(s)
- V I Shcheslavskiy
- Becker&Hickl GmbH, Berlin, 12277, Germany. .,Privolzhskiy Medical Research University, Nizhny Novgorod, 603005, Russia
| | - M V Shirmanova
- Privolzhskiy Medical Research University, Nizhny Novgorod, 603005, Russia
| | - A Jelzow
- Becker&Hickl GmbH, Berlin, 12277, Germany
| | - W Becker
- Becker&Hickl GmbH, Berlin, 12277, Germany
| |
Collapse
|
122
|
Pasch CA, Favreau PF, Yueh AE, Babiarz CP, Gillette AA, Sharick JT, Karim MR, Nickel KP, DeZeeuw AK, Sprackling CM, Emmerich PB, DeStefanis RA, Pitera RT, Payne SN, Korkos DP, Clipson L, Walsh CM, Miller D, Carchman EH, Burkard ME, Lemmon KK, Matkowskyj KA, Newton MA, Ong IM, Bassetti MF, Kimple RJ, Skala MC, Deming DA. Patient-Derived Cancer Organoid Cultures to Predict Sensitivity to Chemotherapy and Radiation. Clin Cancer Res 2019; 25:5376-5387. [PMID: 31175091 DOI: 10.1158/1078-0432.ccr-18-3590] [Citation(s) in RCA: 158] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Revised: 03/08/2019] [Accepted: 06/03/2019] [Indexed: 11/16/2022]
Abstract
PURPOSE Cancer treatment is limited by inaccurate predictors of patient-specific therapeutic response. Therefore, some patients are exposed to unnecessary side effects and delays in starting effective therapy. A clinical tool that predicts treatment sensitivity for individual patients is needed. EXPERIMENTAL DESIGN Patient-derived cancer organoids were derived across multiple histologies. The histologic characteristics, mutation profile, clonal structure, and response to chemotherapy and radiation were assessed using bright-field and optical metabolic imaging on spheroid and single-cell levels, respectively. RESULTS We demonstrate that patient-derived cancer organoids represent the cancers from which they were derived, including key histologic and molecular features. These cultures were generated from numerous cancers, various biopsy sample types, and in different clinical settings. Next-generation sequencing reveals the presence of subclonal populations within the organoid cultures. These cultures allow for the detection of clonal heterogeneity with a greater sensitivity than bulk tumor sequencing. Optical metabolic imaging of these organoids provides cell-level quantification of treatment response and tumor heterogeneity allowing for resolution of therapeutic differences between patient samples. Using this technology, we prospectively predict treatment response for a patient with metastatic colorectal cancer. CONCLUSIONS These studies add to the literature demonstrating feasibility to grow clinical patient-derived organotypic cultures for treatment effectiveness testing. Together, these culture methods and response assessment techniques hold great promise to predict treatment sensitivity for patients with cancer undergoing chemotherapy and/or radiation.
Collapse
Affiliation(s)
- Cheri A Pasch
- University of Wisconsin Carbone Cancer Center, Madison, Wisconsin
| | | | - Alexander E Yueh
- Division of Hematology and Oncology, Department of Medicine, University of Wisconsin-Madison, Madison, Wisconsin
| | - Christopher P Babiarz
- Division of Hematology and Oncology, Department of Medicine, University of Wisconsin-Madison, Madison, Wisconsin
| | - Amani A Gillette
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, Wisconsin
| | - Joe T Sharick
- Morgridge Institute for Research, Madison, Wisconsin
| | | | - Kwangok P Nickel
- University of Wisconsin Carbone Cancer Center, Madison, Wisconsin.,Department of Human Oncology, University of Wisconsin-Madison, Madison, Wisconsin
| | - Alyssa K DeZeeuw
- Division of Hematology and Oncology, Department of Medicine, University of Wisconsin-Madison, Madison, Wisconsin
| | | | - Philip B Emmerich
- Division of Hematology and Oncology, Department of Medicine, University of Wisconsin-Madison, Madison, Wisconsin
| | - Rebecca A DeStefanis
- Division of Hematology and Oncology, Department of Medicine, University of Wisconsin-Madison, Madison, Wisconsin
| | - Rosabella T Pitera
- Division of Hematology and Oncology, Department of Medicine, University of Wisconsin-Madison, Madison, Wisconsin
| | - Susan N Payne
- University of Wisconsin Carbone Cancer Center, Madison, Wisconsin
| | - Demetra P Korkos
- Division of Hematology and Oncology, Department of Medicine, University of Wisconsin-Madison, Madison, Wisconsin
| | - Linda Clipson
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin-Madison, Madison, Wisconsin
| | | | - Devon Miller
- Division of Hematology and Oncology, Department of Medicine, University of Wisconsin-Madison, Madison, Wisconsin
| | - Evie H Carchman
- University of Wisconsin Carbone Cancer Center, Madison, Wisconsin.,Department of Surgery, University of Wisconsin-Madison, Madison, Wisconsin
| | - Mark E Burkard
- University of Wisconsin Carbone Cancer Center, Madison, Wisconsin.,Division of Hematology and Oncology, Department of Medicine, University of Wisconsin-Madison, Madison, Wisconsin
| | - Kayla K Lemmon
- University of Wisconsin Carbone Cancer Center, Madison, Wisconsin
| | - Kristina A Matkowskyj
- University of Wisconsin Carbone Cancer Center, Madison, Wisconsin.,Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, Wisconsin.,William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin
| | - Michael A Newton
- Departments of Statistics and of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, Wisconsin
| | - Irene M Ong
- University of Wisconsin Carbone Cancer Center, Madison, Wisconsin.,Departments of Statistics and of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, Wisconsin
| | - Michael F Bassetti
- University of Wisconsin Carbone Cancer Center, Madison, Wisconsin.,Department of Human Oncology, University of Wisconsin-Madison, Madison, Wisconsin
| | - Randall J Kimple
- University of Wisconsin Carbone Cancer Center, Madison, Wisconsin.,Department of Human Oncology, University of Wisconsin-Madison, Madison, Wisconsin
| | - Melissa C Skala
- University of Wisconsin Carbone Cancer Center, Madison, Wisconsin.,Morgridge Institute for Research, Madison, Wisconsin.,Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, Wisconsin
| | - Dustin A Deming
- University of Wisconsin Carbone Cancer Center, Madison, Wisconsin. .,Division of Hematology and Oncology, Department of Medicine, University of Wisconsin-Madison, Madison, Wisconsin.,McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin-Madison, Madison, Wisconsin
| |
Collapse
|
123
|
Xu HN, Zhao H, Chellappa K, Davis JG, Nioka S, Baur JA, Li LZ. Optical Redox Imaging of Fixed Unstained Muscle Slides Reveals Useful Biological Information. Mol Imaging Biol 2019; 21:417-425. [PMID: 30977079 PMCID: PMC6581512 DOI: 10.1007/s11307-019-01348-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
PURPOSE Optical redox imaging (ORI) technique images cellular autofluorescence of nicotinamide adenine dinucleotide (NADH) and oxidized flavoproteins (Fp containing FAD, i.e., flavin adenine dinucleotide). ORI has found wide applications in the study of cellular energetics and metabolism and may potentially assist in disease diagnosis and prognosis. Fixed tissues have been reported to exhibit autofluorescence with similar spectral characteristics to those of NADH and Fp. However, few studies report on quantitative ORI of formalin-fixed paraffin-embedded (FFPE) unstained tissue slides for disease biomarkers. We investigate whether ORI of FFPE unstained skeletal muscle slides may provide relevant quantitative biological information. PROCEDURES Living mouse muscle fibers and frozen and FFPE mouse muscle slides were subjected to ORI. Living mouse muscle fibers were imaged ex vivo before and after paraformaldehyde fixation. FFPE muscle slides of three mouse groups (young, mid-age, and muscle-specific overexpression of nicotinamide phosphoribosyltransferase (Nampt) transgenic mid-age) were imaged and compared to detect age-related redox differences. RESULTS We observed that living muscle fiber and frozen and FFPE slides all had strong autofluorescence signals in the NADH and Fp channels. Paraformaldehyde fixation resulted in a significant increase in the redox ratio Fp/(NADH + Fp) of muscle fibers. Quantitative image analysis on FFPE unstained slides showed that mid-age gastrocnemius muscles had stronger NADH and Fp signals than young muscles. Gastrocnemius muscles from mid-age Nampt mice had lower NADH compared to age-matched controls, but had higher Fp than young controls. Soleus muscles had the same trend of change and appeared to be more oxidative than gastrocnemius muscles. Differential NADH and Fp signals were found between gastrocnemius and soleus muscles within both mid-aged control and Nampt groups. CONCLUSION Aging effect on redox status quantified by ORI of FFPE unstained muscle slides was reported for the first time. Quantitative information from ORI of FFPE unstained slides may be useful for biomedical applications.
Collapse
Affiliation(s)
- He N Xu
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Britton Chance Laboratory of Redox Imaging, Johnson Research Foundation, Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Institute of Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA.
| | - Huaqing Zhao
- Department of Clinical Sciences, Temple University School of Medicine, Philadelphia, PA, USA
| | - Karthikeyani Chellappa
- Institute for Diabetes, Obesity, and Metabolism and Department of Physiology, University of Pennsylvania, Philadelphia, PA, USA
| | - James G Davis
- Institute for Diabetes, Obesity, and Metabolism and Department of Physiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Shoko Nioka
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Britton Chance Laboratory of Redox Imaging, Johnson Research Foundation, Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Joseph A Baur
- Institute for Diabetes, Obesity, and Metabolism and Department of Physiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Lin Z Li
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Britton Chance Laboratory of Redox Imaging, Johnson Research Foundation, Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Institute of Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
124
|
Sanchez T, Zhang M, Needleman D, Seli E. Metabolic imaging via fluorescence lifetime imaging microscopy for egg and embryo assessment. Fertil Steril 2019; 111:212-218. [PMID: 30691624 DOI: 10.1016/j.fertnstert.2018.12.014] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Accepted: 12/14/2018] [Indexed: 01/07/2023]
Abstract
Current strategies for embryo assessment in the assisted reproductive technology laboratories rely primarily on morphologic parameters that have limited accuracy for determining embryo viability. Even with the addition of invasive diagnostic interventions such as preimplantation genetic testing for aneuploidy alone or in combination with mitochondrial DNA copy number assessment, at least one third of embryos fail to implant. Therefore, at a time when the clinical benefits of single ET are widely accepted, improving viability assessment of embryos is ever more important. Building on the previous work demonstrating the importance of metabolic state in oocytes and embryos, metabolic imaging via fluorescence lifetime imaging microscopy offers new and potentially useful diagnostic method by detecting natural fluorescence of FAD and NADH, the two electron transporters that play a central role in oxidative phosphorylation. Recent studies demonstrate that fluorescence lifetime imaging microscopy can detect oocyte and embryo metabolic function and dysfunction in a multitude of experimental models and provide encouraging evidence for use in scientific investigation and possibly for clinical application.
Collapse
Affiliation(s)
- Tim Sanchez
- Department of Molecular and Cellular Biology and Faculty of Arts and Sciences Center for Systems Biology and John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, Massachusetts
| | - Man Zhang
- Department of Obstetrics, Gynecology and Reproductive Science, Yale University, New Haven, Connecticut
| | - Dan Needleman
- Department of Molecular and Cellular Biology and Faculty of Arts and Sciences Center for Systems Biology and John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, Massachusetts
| | - Emre Seli
- Department of Obstetrics, Gynecology and Reproductive Science, Yale University, New Haven, Connecticut.
| |
Collapse
|
125
|
Silva SF, Domingues JP, Morgado AM. Can we use rapid lifetime determination for fast, fluorescence lifetime based, metabolic imaging? Precision and accuracy of double-exponential decay measurements with low total counts. PLoS One 2019; 14:e0216894. [PMID: 31086413 PMCID: PMC6516636 DOI: 10.1371/journal.pone.0216894] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 04/30/2019] [Indexed: 01/19/2023] Open
Abstract
Fluorescence lifetime imaging microscopy (FLIM) can assess cell’s metabolism through the fluorescence of the co-enzymes NADH and FAD, which exhibit a double-exponential decay, with components related to free and protein-bound conditions. In vivo real time clinical imaging applications demand fast acquisition. As photodamage limits excitation power, this is best achieved using wide-field techniques, like time-gated FLIM, and algorithms that require few images to calculate the decay parameters. The rapid lifetime determination (RLD) algorithm requires only four images to analyze a double-exponential decay. Using computational simulations, we evaluated the accuracy and precision of RLD when measuring endogenous fluorescence lifetimes and metabolic free to protein-bound ratios, for total counts per pixel (TC) lower than 104. The simulations were based on a time-gated FLIM instrument, accounting for its instrument response function, gain and noise. While the optimal acquisition setting depends on the values being measured, the accuracy of the free to protein-bound ratio α2/α1 is stable for low gains and gate separations larger than 1000 ps, while its precision is almost constant for gate separations between 1500 and 2500 ps. For the gate separations and free to protein-bound ratios considered, the accuracy error can be as high as 30% and the precision error can reach 60%. Precision errors lower than 10% cannot be obtained. The best performance occurs for low camera gains and gate separations near 1800 ps. When considering the narrow physiological ranges for the free to protein-bound ratio, the precision errors can be confined to an interval between 10% and 20%. RLD is a valid option when for real time FLIM. The simulations and methodology presented here can be applied to any time-gated FLIM instrument and are useful to obtain the accuracy and precision limits for RLD in the demanding conditions of TC lower than 104.
Collapse
Affiliation(s)
- Susana Figueiredo Silva
- CIBIT—Coimbra Institute for Biomedical Imaging and Translational Research/INCAS—Institute of Nuclear Sciences Applied to Health, University of Coimbra, Coimbra, Portugal
| | - José Paulo Domingues
- CIBIT—Coimbra Institute for Biomedical Imaging and Translational Research/INCAS—Institute of Nuclear Sciences Applied to Health, University of Coimbra, Coimbra, Portugal
- Department of Physics, Faculty of Sciences and Technology, University of Coimbra, Coimbra, Portugal
| | - António Miguel Morgado
- CIBIT—Coimbra Institute for Biomedical Imaging and Translational Research/INCAS—Institute of Nuclear Sciences Applied to Health, University of Coimbra, Coimbra, Portugal
- Department of Physics, Faculty of Sciences and Technology, University of Coimbra, Coimbra, Portugal
- * E-mail:
| |
Collapse
|
126
|
Sharick JT, Jeffery JJ, Karim MR, Walsh CM, Esbona K, Cook RS, Skala MC. Cellular Metabolic Heterogeneity In Vivo Is Recapitulated in Tumor Organoids. Neoplasia 2019; 21:615-626. [PMID: 31078067 PMCID: PMC6514366 DOI: 10.1016/j.neo.2019.04.004] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Revised: 04/09/2019] [Accepted: 04/10/2019] [Indexed: 12/14/2022] Open
Abstract
Heterogeneous populations within a tumor have varying metabolic profiles, which can muddle the interpretation of bulk tumor imaging studies of treatment response. Although methods to study tumor metabolism at the cellular level are emerging, these methods provide a single time point “snapshot” of tumor metabolism and require a significant time and animal burden while failing to capture the longitudinal metabolic response of a single tumor to treatment. Here, we investigated a novel method for longitudinal, single-cell tracking of metabolism across heterogeneous tumor cell populations using optical metabolic imaging (OMI), which measures autofluorescence of metabolic coenzymes as a report of metabolic activity. We also investigated whether in vivo cellular metabolic heterogeneity can be accurately captured using tumor-derived three-dimensional organoids in a genetically engineered mouse model of breast cancer. OMI measurements of response to paclitaxel and the phosphatidylinositol-3-kinase inhibitor XL147 in tumors and organoids taken at single cell resolution revealed parallel shifts in metaboltruic heterogeneity. Interestingly, these previously unappreciated heterogeneous metabolic responses in tumors and organoids could not be attributed to tumor cell fate or varying leukocyte content within the microenvironment, suggesting that heightened metabolic heterogeneity upon treatment is largely due to heterogeneous metabolic shifts within tumor cells. Together, these studies show that OMI revealed remarkable heterogeneity in response to treatment, which could provide a novel approach to predict the presence of potentially unresponsive tumor cell subpopulations lurking within a largely responsive bulk tumor population, which might otherwise be overlooked by traditional measurements.
Collapse
Affiliation(s)
- Joe T Sharick
- Department of Biomedical Engineering, Vanderbilt University, PMB 351631, 2301 Vanderbilt Place, Nashville, TN, 37235, USA; Morgridge Institute for Research, 330 N. Orchard Street, Madison, WI, 53715, USA
| | - Justin J Jeffery
- University of Wisconsin Carbone Cancer Center, 600 Highland Avenue, Madison, WI, 53792, USA
| | - Mohammad R Karim
- Morgridge Institute for Research, 330 N. Orchard Street, Madison, WI, 53715, USA
| | - Christine M Walsh
- Morgridge Institute for Research, 330 N. Orchard Street, Madison, WI, 53715, USA
| | - Karla Esbona
- Department of Pathology and Laboratory Medicine, University of Wisconsin School of Medicine and Public Health, 3170 UW Medical Foundation Centennial Building, 1685 Highland Avenue, Madison, WI, 53705, USA
| | - Rebecca S Cook
- Department of Biomedical Engineering, Vanderbilt University, PMB 351631, 2301 Vanderbilt Place, Nashville, TN, 37235, USA; Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, PMB 407935 U-3218, Medical Research Building III, Nashville, TN, 37240, USA;; Department of Cancer Biology, Vanderbilt University School of Medicine, Nashville, TN; Vanderbilt Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN
| | - Melissa C Skala
- Morgridge Institute for Research, 330 N. Orchard Street, Madison, WI, 53715, USA; Department of Biomedical Engineering, University of Wisconsin-Madison, Engineering Centers Building, 1550 Engineering Drive Room #2130, Madison, WI, 53706.
| |
Collapse
|
127
|
Kovaleva TF, Maksimova NS, Zhukov IY, Pershin VI, Mukhina IV, Gainullin MR. Cofilin: Molecular and Cellular Functions and Its Role in the Functioning of the Nervous System. NEUROCHEM J+ 2019. [DOI: 10.1134/s1819712419010124] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
128
|
Low dose photodynamic therapy harmonizes with radiation therapy to induce beneficial effects on pancreatic heterocellular spheroids. Oncotarget 2019; 10:2625-2643. [PMID: 31080554 PMCID: PMC6499000 DOI: 10.18632/oncotarget.26780] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Accepted: 02/21/2019] [Indexed: 12/14/2022] Open
Abstract
Photodynamic therapy (PDT) has seen long standing interest as a therapy for resistant cancers, but the main Achilles’ heel for its successful clinical exploitation is the use of poorly penetrating visible light. This limitation could be overcome by using radioluminescent nanoparticles, which can be excited during radiation therapy (RT) with penetrating X-rays. When infused in tumors, X-ray activated-nanoscintillators act as internal light sources and excite nearby photosensitizers. Recent studies demonstrated that it is realistic to achieve low dose PDT with current nanoscintillators. However, as the origin of enhanced RT efficacy with nanoscintillators may have varying origins, we aimed to answer the basic question: Is a combination of low-dose PDT beneficial to the RT efficacy in clinically relevant models of cancer? Pancreatic cancer (PanCa) remains a lethal disease for which RT is part of the palliative care and for which PDT demonstrated promising results in clinical trial. We thus evaluated the combination of low-dose PDT and RT delivered in absence of nanoscintillators on various heterocellular spheroid models that recapitulate the clinical heterogeneity of PanCa. Although therapeutic effects emerged at different timepoints in each model, the RT/PDT combination uniformly achieved favorable outcomes. With RT providing stunted tumor growth while PDT drove adjuvant apoptotic and necrotic cell death, the combination produced significantly smaller and less viable PanCa spheroids. In conclusion, the beneficial RT/PDT treatment outcomes encourage the further development of nanoscinitillators for X-ray-activated PDT. Assessment of such combination treatments should encompass multiparametric and temporally-spaced assessment of treatment effects in preclinical cancer models.
Collapse
|
129
|
Zhu C, Li M, Vincent T, Martin HL, Crouch BT, Martinez AF, Madonna MC, Palmer GM, Dewhirst MW, Ramanujam N. Simultaneous in vivo optical quantification of key metabolic and vascular endpoints reveals tumor metabolic diversity in murine breast tumor models. JOURNAL OF BIOPHOTONICS 2019; 12:e201800372. [PMID: 30565420 PMCID: PMC8744479 DOI: 10.1002/jbio.201800372] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Revised: 12/15/2018] [Accepted: 12/16/2018] [Indexed: 05/24/2023]
Abstract
Therapeutically exploiting vascular and metabolic endpoints becomes critical to translational cancer studies because altered vascularity and deregulated metabolism are two important cancer hallmarks. The metabolic and vascular phenotypes of three sibling breast tumor lines with different metastatic potential are investigated in vivo with a newly developed quantitative spectroscopy system. All tumor lines have different metabolic and vascular characteristics compared to normal tissues, and there are strong positive correlations between metabolic (glucose uptake and mitochondrial membrane potential) and vascular (oxygen saturations and hemoglobin concentrations) parameters for metastatic (4T1) tumors but not for micrometastatic (4T07) and nonmetastatic (67NR) tumors. A longitudinal study shows that both vascular and metabolic endpoints of 4T1 tumors increased up to a specific tumor size threshold beyond which these parameters decreased. The synchronous changes between metabolic and vascular parameters, along with the strong positive correlations between these endpoints suggest that 4T1 tumors rely on strong oxidative phosphorylation in addition to glycolysis. This study illustrates the great potential of our optical technique to provide valuable dynamic information about the interplay between the metabolic and vascular status of tumors, with important implications for translational cancer investigations.
Collapse
Affiliation(s)
- Caigang Zhu
- Department of Biomedical Engineering, Duke University, Durham, North Carolina
| | - Martin Li
- Department of Biomedical Engineering, Duke University, Durham, North Carolina
| | - Thomas Vincent
- Department of Biomedical Engineering, Duke University, Durham, North Carolina
| | - Hannah L Martin
- Department of Biomedical Engineering, Duke University, Durham, North Carolina
| | - Brian T Crouch
- Department of Biomedical Engineering, Duke University, Durham, North Carolina
| | - Amy F Martinez
- Department of Biomedical Engineering, Duke University, Durham, North Carolina
- Office of Research, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Megan C Madonna
- Department of Biomedical Engineering, Duke University, Durham, North Carolina
| | - Gregory M Palmer
- Department of Radiation Oncology, Duke University, Durham, North Carolina
| | - Mark W Dewhirst
- Department of Radiation Oncology, Duke University, Durham, North Carolina
| | - Nimmi Ramanujam
- Department of Biomedical Engineering, Duke University, Durham, North Carolina
| |
Collapse
|
130
|
Kolenc OI, Quinn KP. Evaluating Cell Metabolism Through Autofluorescence Imaging of NAD(P)H and FAD. Antioxid Redox Signal 2019; 30:875-889. [PMID: 29268621 PMCID: PMC6352511 DOI: 10.1089/ars.2017.7451] [Citation(s) in RCA: 176] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
SIGNIFICANCE Optical imaging using the endogenous fluorescence of metabolic cofactors has enabled nondestructive examination of dynamic changes in cell and tissue function both in vitro and in vivo. Quantifying NAD(P)H and FAD fluorescence through an optical redox ratio and fluorescence lifetime imaging (FLIM) provides sensitivity to the relative balance between oxidative phosphorylation and glucose catabolism. Since its introduction decades ago, the use of NAD(P)H imaging has expanded to include applications involving almost every major tissue type and a variety of pathologies. Recent Advances: This review focuses on the use of two-photon excited fluorescence and NAD(P)H fluorescence lifetime techniques in cancer, neuroscience, tissue engineering, and other biomedical applications over the last 5 years. In a variety of cancer models, NAD(P)H fluorescence intensity and lifetime measurements demonstrate a sensitivity to the Warburg effect, suggesting potential for early detection or high-throughput drug screening. The sensitivity to the biosynthetic demands of stem cell differentiation and tissue repair processes indicates the range of applications for this imaging technology may be broad. CRITICAL ISSUES As the number of applications for these fluorescence imaging techniques expand, identifying and characterizing additional intrinsic fluorophores and chromophores present in vivo will be vital to accurately measure and interpret metabolic outcomes. Understanding the full capabilities and limitations of FLIM will also be key to future advances. FUTURE DIRECTIONS Future work is needed to evaluate whether a combination of different biochemical and structural outcomes using these imaging techniques can provide complementary information regarding the utilization of specific metabolic pathways.
Collapse
Affiliation(s)
- Olivia I Kolenc
- Department of Biomedical Engineering, University of Arkansas, Fayetteville, Arkansas
| | - Kyle P Quinn
- Department of Biomedical Engineering, University of Arkansas, Fayetteville, Arkansas
| |
Collapse
|
131
|
Miura Y. Two-Photon Microscopy (TPM) and Fluorescence Lifetime Imaging Microscopy (FLIM) of Retinal Pigment Epithelium (RPE) of Mice In Vivo. Methods Mol Biol 2019; 1753:73-88. [PMID: 29564782 DOI: 10.1007/978-1-4939-7720-8_5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Retinal pigment epithelium (RPE), a monolayer of epithelial cells located between the neural retina and the choroid, plays a significant role in the maintenance of retinal function. Its in vivo imaging is still technically challenging in human eye. With the mouse eye, there is a possibility to look into the RPE through the sclera using two-photon microscopy (TPM). TPM is a two photon-excited nonlinear fluorescence microscopy that enables the observation of deep tissues up to several hundred micrometers. Since the simultaneous absorption of two photons occurs only at the focal plane, spatial resolution of the TPM is quite high, such that pinhole as used in a confocal microscope is not necessary. TPM enables observation of autofluorescence at the cellular level, and thus may provide new insights into the fluorescent molecules in/around RPE cells.The combination of TPM with fluorescence lifetime imaging microscopy (FLIM) may expand the breadth of information about cells and tissues. Fluorescence lifetime is a fluorophore-specific property, which is independent of fluorescence intensity and changes with the alteration of molecular environment. FLIM may have therefore the potentials to distinguish different fluorophores and to indicate the change in the environment of a fluorophore. Some energy metabolisms-related intracellular fluorophores, such as NADH (nicotinamide adenine dinucleotide) and FAD (flavin adenine dinucleotide), show characteristic fluorescence lifetimes that shift under different molecular environments, and thus their fluorescence lifetime have been used to indicate cell energy metabolic states. These nonlabeling imaging methods offer us the opportunity to engage in the study of the RPE in vivo as well as in vitro both in morphological as well as metabolic aspects.
Collapse
Affiliation(s)
- Yoko Miura
- Institute of Biomedical Optics, University of Lübeck, Lübeck, Germany. .,Department of Ophthalmology, University of Lübeck, Lübeck, Germany.
| |
Collapse
|
132
|
Gandhi N, Das GM. Metabolic Reprogramming in Breast Cancer and Its Therapeutic Implications. Cells 2019; 8:E89. [PMID: 30691108 PMCID: PMC6406734 DOI: 10.3390/cells8020089] [Citation(s) in RCA: 152] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Revised: 01/20/2019] [Accepted: 01/22/2019] [Indexed: 12/22/2022] Open
Abstract
Current standard-of-care (SOC) therapy for breast cancer includes targeted therapies such as endocrine therapy for estrogen receptor-alpha (ERα) positive; anti-HER2 monoclonal antibodies for human epidermal growth factor receptor-2 (HER2)-enriched; and general chemotherapy for triple negative breast cancer (TNBC) subtypes. These therapies frequently fail due to acquired or inherent resistance. Altered metabolism has been recognized as one of the major mechanisms underlying therapeutic resistance. There are several cues that dictate metabolic reprogramming that also account for the tumors' metabolic plasticity. For metabolic therapy to be efficacious there is a need to understand the metabolic underpinnings of the different subtypes of breast cancer as well as the role the SOC treatments play in targeting the metabolic phenotype. Understanding the mechanism will allow us to identify potential therapeutic vulnerabilities. There are some very interesting questions being tackled by researchers today as they pertain to altered metabolism in breast cancer. What are the metabolic differences between the different subtypes of breast cancer? Do cancer cells have a metabolic pathway preference based on the site and stage of metastasis? How do the cell-intrinsic and -extrinsic cues dictate the metabolic phenotype? How do the nucleus and mitochondria coordinately regulate metabolism? How does sensitivity or resistance to SOC affect metabolic reprogramming and vice-versa? This review addresses these issues along with the latest updates in the field of breast cancer metabolism.
Collapse
Affiliation(s)
- Nishant Gandhi
- Department of Pharmacology and Therapeutics, Center for Genetics & Pharmacology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA.
| | - Gokul M Das
- Department of Pharmacology and Therapeutics, Center for Genetics & Pharmacology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA.
| |
Collapse
|
133
|
In-vivo correlations between skin metabolic oscillations and vasomotion in wild-type mice and in a model of oxidative stress. Sci Rep 2019; 9:186. [PMID: 30655574 PMCID: PMC6336806 DOI: 10.1038/s41598-018-36970-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Accepted: 11/27/2018] [Indexed: 12/17/2022] Open
Abstract
Arterioles in the cutaneous microcirculation frequently display an oscillatory phenomenon defined vasomotion, consistent with periodic diameter variations in the micro-vessels associated with particular physiological or abnormal conditions. The cellular mechanisms underlying vasomotion and its physiological role have not been completely elucidated. Various mechanisms were demonstrated, based on cell Ca2+ oscillations determined by the activity of channels in the plasma membrane or sarcoplasmic reticulum of vascular cells. However, the possible engagement in vasomotion of cell metabolic oscillations of mitochondrial or glycolytic origin has been poorly explored. Metabolic oscillations associated with the production of ATP energy were previously described in cells, while limited studies have investigated these fluctuations in-vivo. Here, we characterised a low-frequency metabolic oscillator (MO-1) in skin from live wild-type and Nrf2−/− mice, by combination of fluorescence spectroscopy and wavelet transform processing technique. Furthermore, the relationships between metabolic and microvascular oscillators were examined during phenylephrine-induced vasoconstriction. We found a significant interaction between MO-1 and the endothelial EDHF vasomotor mechanism that was reduced in the presence of oxidative stress (Nrf2−/− mice). Our findings suggest indirectly that metabolic oscillations may be involved in the mechanisms underlying endothelium-mediated skin vasomotion, which might be altered in the presence of metabolic disturbance.
Collapse
|
134
|
Breast tumour organoids: promising models for the genomic and functional characterisation of breast cancer. Biochem Soc Trans 2019; 47:109-117. [PMID: 30626705 DOI: 10.1042/bst20180375] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Revised: 11/22/2018] [Accepted: 11/26/2018] [Indexed: 01/08/2023]
Abstract
Until recently, established cancer cell lines have been used extensively in breast cancer research, due largely to the difficulties associated with the manipulation and long-term maintenance in culture of primary tumour cells from patients. The recent development of organoid cultures has provided new opportunities to model and analyse patient samples, allowing the propagation of malignant cells under conditions that resemble the three-dimensional growth of breast tumours. They have proved efficacious in preserving the heterogeneity of primary samples and are emerging as a new model to further characterise the molecular features of breast cancer. Organoids formed from patient-derived cells are now in use for the evaluation of drug sensitivity and to validate disease-causing genomic variations. Here, the advantages and limitations of organoid cultures will be discussed and compared with the parallel development of other two- and three-dimensional culture strategies and with patient-derived xenografts. In particular, we will focus on the molecular characterisation of breast cancer organoids and provide some examples of how they have been used in functional studies.
Collapse
|
135
|
Chacko JV, Eliceiri KW. Autofluorescence lifetime imaging of cellular metabolism: Sensitivity toward cell density, pH, intracellular, and intercellular heterogeneity. Cytometry A 2019; 95:56-69. [PMID: 30296355 PMCID: PMC6329636 DOI: 10.1002/cyto.a.23603] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Revised: 08/14/2018] [Accepted: 08/20/2018] [Indexed: 12/14/2022]
Abstract
Autofluorescence imaging (AFI) has greatly accelerated in the last decade, way past its origins in detecting endogenous signals in biological tissues to identify differences between samples. There are many endogenous fluorescence sources of contrast but the most robust and widely utilized have been those associated with metabolism. The intrinsically fluorescent metabolic cofactors nicotinamide adenine dinucleotide (NAD+ /NADH) and flavin adenine dinucleotide (FAD/FADH2 ) have been utilized in a number of AFI applications including basic research, clinical, and pharmaceutical studies. Fluorescence lifetime imaging microscopy (FLIM) has emerged as one of the more powerful AFI tools for NADH and FAD characterization due to its unique ability to noninvasively detect metabolite bound and free states and quantitate cellular redox ratio. However, despite this widespread biological use, many standardization methods are still needed to extend FLIM-based AFI into a fully robust research and clinical diagnostic tools. FLIM is sensitive to a wide range of factors in the fluorophore microenvironment, and there are a number of analysis variables as well. To this end, there has been an emphasis on developing imaging standards and ways to make the image acquisition and analysis more consistent. However, biological conditions during FLIM-based AFI imaging are rarely considered as key sources of FLIM variability. Here, we present several experimental factors with supporting data of the cellular microenvironment such as confluency, pH, inter-/intracellular heterogeneity, and choice of cell line that need to be considered for accurate quantitative FLIM-based AFI measurement of cellular metabolism. © 2018 International Society for Advancement of Cytometry.
Collapse
Affiliation(s)
- Jenu V. Chacko
- Laboratory for Optical and Computational Instrumentation, University of Wisconsin at Madison, Madison WI, USA
| | - Kevin W. Eliceiri
- Laboratory for Optical and Computational Instrumentation, University of Wisconsin at Madison, Madison WI, USA
- Biomedical Engineering Department, University of Wisconsin at Madison, Madison WI, USA
- Morgridge Institute for Research, Madison WI, USA
| |
Collapse
|
136
|
Gil DA, Sharick JT, Mancha S, Gamm UA, Choma MA, Skala MC. Redox imaging and optical coherence tomography of the respiratory ciliated epithelium. JOURNAL OF BIOMEDICAL OPTICS 2019; 24:1-4. [PMID: 30701725 PMCID: PMC6985682 DOI: 10.1117/1.jbo.24.1.010501] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Accepted: 01/16/2019] [Indexed: 05/17/2023]
Abstract
Optical coherence tomography (OCT) is an emerging technology for in vivo airway and lung imaging. However, OCT lacks sensitivity to the metabolic changes caused by inflammation, which drives chronic respiratory diseases such as asthma and chronic obstructive pulmonary disorder. Redox imaging (RI) is a label-free technique that uses the autofluorescence of the metabolic coenzymes NAD(P)H and flavin adenine dinucleotide (FAD) to probe cellular metabolism and could provide complimentary information to OCT for airway and lung imaging. We demonstrate OCT and RI of respiratory ciliated epithelial function in ex vivo mouse tracheae. We applied RI to measure cellular metabolism via the redox ratio [intensity of NAD(P)H divided by FAD] and particle tracking velocimetry OCT to quantify cilia-driven fluid flow. To model mitochondrial dysfunction, a key aspect of the inflammatory process, cyanide was used to inhibit oxidative metabolism and reduce ciliary motility. Cyanide exposure over 20 min significantly increased the redox ratio and reversed cilia-driven fluid flow. We propose that RI provides complementary information to OCT to assess inflammation in the airway and lungs.
Collapse
Affiliation(s)
- Daniel A. Gil
- University of Wisconsin–Madison, Department of Biomedical Engineering, Madison, Wisconsin, United States
- Morgridge Institute for Research, Madison, Wisconsin, United States
| | - Joe T. Sharick
- Morgridge Institute for Research, Madison, Wisconsin, United States
- Vanderbilt University, Department of Biomedical Engineering, Nashville, Tennessee, United States
| | - Sophie Mancha
- University of Wisconsin–Madison, Department of Biomedical Engineering, Madison, Wisconsin, United States
- Morgridge Institute for Research, Madison, Wisconsin, United States
| | - Ute A. Gamm
- Yale University, Department of Diagnostic Radiology, New Haven, Connecticut, United States
| | - Michael A. Choma
- Yale University, Department of Diagnostic Radiology, New Haven, Connecticut, United States
- Yale University, Department of Biomedical Engineering, New Haven, Connecticut, United States
- Yale University, Department of Pediatrics, New Haven, Connecticut, United States
- Yale University, Department of Applied Physics, New Haven, Connecticut, United States
| | - Melissa C. Skala
- University of Wisconsin–Madison, Department of Biomedical Engineering, Madison, Wisconsin, United States
- Morgridge Institute for Research, Madison, Wisconsin, United States
- Address all correspondence to Melissa C. Skala, E-mail:
| |
Collapse
|
137
|
Nagle PW, Plukker JTM, Muijs CT, van Luijk P, Coppes RP. Patient-derived tumor organoids for prediction of cancer treatment response. Semin Cancer Biol 2018; 53:258-264. [DOI: 10.1016/j.semcancer.2018.06.005] [Citation(s) in RCA: 86] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Revised: 06/21/2018] [Accepted: 06/28/2018] [Indexed: 12/12/2022]
|
138
|
Jones JD, Ramser HE, Woessner AE, Quinn KP. In vivo multiphoton microscopy detects longitudinal metabolic changes associated with delayed skin wound healing. Commun Biol 2018; 1:198. [PMID: 30480099 PMCID: PMC6242983 DOI: 10.1038/s42003-018-0206-4] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 10/26/2018] [Indexed: 01/08/2023] Open
Abstract
Chronic wounds are difficult to diagnose and characterize due to a lack of quantitative biomarkers. Label-free multiphoton microscopy has emerged as a useful imaging modality capable of quantifying changes in cellular metabolism using an optical redox ratio of FAD/(NADH+FAD) autofluorescence. However, the utility of an optical redox ratio for long-term in vivo monitoring of tissue metabolism has not been robustly evaluated. In this study, we demonstrate how multiphoton microscopy can be used to monitor changes in the metabolism of individual full-thickness skin wounds in vivo. 3D optical redox ratio maps and NADH fluorescence lifetime images identify differences between diabetic and control mice during the re-epithelialization of wounds. These metabolic changes are associated with a transient increase in keratinocyte proliferation at the wound edge. Our study demonstrates that high-resolution, non-invasive autofluorescence imaging can be performed in vivo and that optical redox ratios can serve as quantitative optical biomarkers of impaired wound healing.
Collapse
Affiliation(s)
- Jake D Jones
- Department of Biomedical Engineering, University of Arkansas, 123 John A. White Jr. Engineering Hall, Fayetteville, AR, 72701, USA
| | - Hallie E Ramser
- Department of Biomedical Engineering, University of Arkansas, 123 John A. White Jr. Engineering Hall, Fayetteville, AR, 72701, USA
| | - Alan E Woessner
- Department of Biomedical Engineering, University of Arkansas, 123 John A. White Jr. Engineering Hall, Fayetteville, AR, 72701, USA
| | - Kyle P Quinn
- Department of Biomedical Engineering, University of Arkansas, 123 John A. White Jr. Engineering Hall, Fayetteville, AR, 72701, USA.
| |
Collapse
|
139
|
Carver GE, Locknar SA, Weaver DL, Stein JL, Stein GS. Real-time detection of breast cancer at the cellular level. J Cell Physiol 2018; 234:5413-5419. [PMID: 30362286 DOI: 10.1002/jcp.27451] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Accepted: 08/29/2018] [Indexed: 11/05/2022]
Abstract
Novel optoelectronic instrumentation has been developed for the multispectral imaging of autofluorescence emitted by metabolic fluorophores. The images resolve individual cells while spectra are collected for each pixel in the images. These datacubes are generated at a rate of 10 per second-fast enough for surgical guidance. The data is processed in real time to provide a single color-coded image to the surgeon. To date, the system has been applied to fresh, ex vivo, human surgical specimens and has distinguished breast cancer from benign tissue. The approach is applicable to in vivo measurements of surgical margins and needle-based optical biopsies. Ongoing work demonstrates that the system has great potential for translation to a hand-held probe with high sensitivity and specificity.
Collapse
Affiliation(s)
| | | | - Donald L Weaver
- Department of Pathology and Laboratory Medicine, University of Vermont Cancer Center, Burlington, Vermont
| | - Janet L Stein
- Department of Biochemistry, University of Vermont Cancer Center, Burlington, Vermont
| | - Gary S Stein
- Department of Pathology and Laboratory Medicine, University of Vermont Cancer Center, Burlington, Vermont
| |
Collapse
|
140
|
Bower AJ, Li J, Chaney EJ, Marjanovic M, Spillman DR, Boppart SA. High-speed imaging of transient metabolic dynamics using two-photon fluorescence lifetime imaging microscopy. OPTICA 2018; 5:1290-1296. [PMID: 30984802 PMCID: PMC6457362 DOI: 10.1364/optica.5.001290] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/02/2023]
Abstract
Two-photon fluorescence lifetime imaging microscopy (2P-FLIM) of autofluorescent metabolic coenzymes has been widely used to investigate energetic perturbations in living cells and tissues in a label-free manner with subcellular resolution. While the currently used state-of-the-art instruments are highly sensitive to local molecular changes associated with these metabolic processes, they are inherently slow and limit the study of dynamic metabolic environments. Here, a sustained video-rate 2P-FLIM imaging system is demonstrated for time-lapse lifetime imaging of reduced nicotinamide adenine dinucleotide, an autofluorescent metabolic coenzyme involved in both aerobic and anaerobic processes. This system is sufficiently sensitive to differences in metabolic activity between aggressive and nonaggressive cancer cell lines and is demonstrated for both wide field-of-view autofluorescence imaging as well as sustained video-rate image acquisition of metabolic dynamics following induction of apoptosis. The unique capabilities ofthis imaging platform provide a powerful technological advance to further explore rapid metabolic dynamics in living cells.
Collapse
Affiliation(s)
- Andrew J. Bower
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, 405 N. MathewsAve, Urbana, Illinois 61801, USA
- Department of Electrical and Computer Engineering, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA
| | - Joanne Li
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, 405 N. MathewsAve, Urbana, Illinois 61801, USA
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA
| | - Eric J. Chaney
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, 405 N. MathewsAve, Urbana, Illinois 61801, USA
| | - Marina Marjanovic
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, 405 N. MathewsAve, Urbana, Illinois 61801, USA
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA
- Carle-Illinois College of Medicine, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA
| | - Darold R. Spillman
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, 405 N. MathewsAve, Urbana, Illinois 61801, USA
| | - Stephen A. Boppart
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, 405 N. MathewsAve, Urbana, Illinois 61801, USA
- Department of Electrical and Computer Engineering, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA
- Carle-Illinois College of Medicine, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA
- Corresponding author:
| |
Collapse
|
141
|
Alturkistany F, Nichani K, Houston KD, Houston JP. Fluorescence lifetime shifts of NAD(P)H during apoptosis measured by time-resolved flow cytometry. Cytometry A 2018; 95:70-79. [PMID: 30369063 PMCID: PMC6587805 DOI: 10.1002/cyto.a.23606] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Revised: 08/01/2018] [Accepted: 08/20/2018] [Indexed: 12/16/2022]
Abstract
Autofluorescence from the intracellular metabolite, NAD(P)H, is a biomarker that is widely used and known to reliably screen and report metabolic activity as well as metabolic fluctuations within cells. As a ubiquitous endogenous fluorophore, NAD(P)H has a unique rate of fluorescence decay that is altered when bound to coenzymes. In this work we measure the shift in the fluorescence decay, or average fluorescence lifetime (1–3 ns), of NAD(P)H and correlate this shift to changes in metabolism that cells undergo during apoptosis. Our measurements are made with a flow cytometer designed specifically for fluorescence lifetime acquisition within the ultraviolet to violet spectrum. Our methods involved culture, treatment, and preparation of cells for cytometry and microscopy measurements. The evaluation we performed included observations and quantification of the changes in endogenous emission owing to the induction of apoptosis as well as changes in the decay kinetics of the emission measured by flow cytometry. Shifts in NAD(P)H fluorescence lifetime were observed as early as 15 min post‐treatment with an apoptosis inducing agent. Results also include a phasor analysis to evaluate free to bound ratios of NAD(P)H at different time points. We defined the free to bound ratios as the ratio of ‘short‐to‐long’ (S/L) fluorescence lifetime, where S/L was found to consistently decrease with an increase in apoptosis. With a quantitative framework such as phasor analysis, the short and long lifetime components of NAD(P)H can be used to map the cycling of free and bound NAD(P)H during the early‐to‐late stages of apoptosis. The combination of lifetime screening and phasor analyses provides the first step in high throughput metabolic profiling of single cells and can be leveraged for screening and sorting for a range of applications in biomedicine. © 2018 The Authors. Cytometry Part A published by Wiley Periodicals, Inc. on behalf of International Society for Advancement of Cytometry.
Collapse
Affiliation(s)
| | - Kapil Nichani
- Chemical & Materials Engineering, New Mexico State University, Las Cruces, New Mexico
| | - Kevin D Houston
- Chemistry & Biochemistry, New Mexico State University, Las Cruces, New Mexico.,Molecular Biology, New Mexico State University, Las Cruces, New Mexico
| | - Jessica P Houston
- Chemical & Materials Engineering, New Mexico State University, Las Cruces, New Mexico.,Molecular Biology, New Mexico State University, Las Cruces, New Mexico
| |
Collapse
|
142
|
Lagarto JL, Dyer BT, Talbot CB, Peters NS, French PMW, Lyon AR, Dunsby C. Characterization of NAD(P)H and FAD autofluorescence signatures in a Langendorff isolated-perfused rat heart model. BIOMEDICAL OPTICS EXPRESS 2018; 9:4961-4978. [PMID: 30319914 PMCID: PMC6179415 DOI: 10.1364/boe.9.004961] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Revised: 08/10/2018] [Accepted: 08/11/2018] [Indexed: 05/22/2023]
Abstract
Autofluorescence spectroscopy is a promising label-free approach to characterize biological samples with demonstrated potential to report structural and biochemical alterations in tissues in a number of clinical applications. We report a characterization of the ex vivo autofluorescence fingerprint of cardiac tissue, exploiting a Langendorff-perfused isolated rat heart model to induce physiological insults to the heart, with a view to understanding how metabolic alterations affect the autofluorescence signals. Changes in the autofluorescence intensity and lifetime signatures associated with reduced nicotinamide adenine dinucleotide (phosphate) (NAD(P)H) and flavin adenine dinucleotide (FAD) were characterized during oxygen- or glucose-depletion protocols. Results suggest that both NAD(P)H and FAD autofluorescence intensity and lifetime parameters are sensitive to changes in the metabolic state of the heart owing to oxygen deprivation. We also observed changes in NAD(P)H fluorescence intensity and FAD lifetime parameter on reperfusion of oxygen, which might provide information on reperfusion injury, and permanent tissue damage or changes to the tissue during recovery from oxygen deprivation. We found that changes in the autofluorescence signature following glucose-depletion are, in general, less pronounced, and most clearly visible in NAD(P)H related parameters. Overall, the results reported in this investigation can serve as baseline for future investigations of cardiac tissue involving autofluorescence measurements.
Collapse
Affiliation(s)
- João L Lagarto
- Photonics Group, Department of Physics, Imperial College London, Prince Consort Road, London SW7 2AZ, UK
- Authors contributed equally to this work
| | - Benjamin T Dyer
- National Heart and Lung Institute, Imperial College London, Du Cane Road, London W12 0NN, UK
- Authors contributed equally to this work
| | - Clifford B Talbot
- Photonics Group, Department of Physics, Imperial College London, Prince Consort Road, London SW7 2AZ, UK
| | - Nicholas S Peters
- National Heart and Lung Institute, Imperial College London, Du Cane Road, London W12 0NN, UK
| | - Paul M W French
- Photonics Group, Department of Physics, Imperial College London, Prince Consort Road, London SW7 2AZ, UK
| | - Alexander R Lyon
- National Heart and Lung Institute, Imperial College London, Du Cane Road, London W12 0NN, UK
- Authors contributed equally to this work
| | - Chris Dunsby
- Photonics Group, Department of Physics, Imperial College London, Prince Consort Road, London SW7 2AZ, UK
- Centre for Pathology, Imperial College London, Du Cane Road, London, W12 0NN, UK
- Authors contributed equally to this work
| |
Collapse
|
143
|
Kalinina S, Breymayer J, Reeß K, Lilge L, Mandel A, Rück A. Correlation of intracellular oxygen and cell metabolism by simultaneous PLIM of phosphorescent TLD1433 and FLIM of NAD(P)H. JOURNAL OF BIOPHOTONICS 2018; 11:e201800085. [PMID: 29877627 DOI: 10.1002/jbio.201800085] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Revised: 05/17/2018] [Accepted: 06/05/2018] [Indexed: 05/20/2023]
Abstract
During photodynamic therapy (PDT), disruption of cell respiration and metabolic changes could be one of the first events. Photophysical characteristics of the photosensitizer (PS) and its specific redox potential define consumption of molecular oxygen followed by generation of reactive oxygen species. The potential PS TLD1433 is based on transition metal Ru(II) and possess an oxygen-dependent luminescence. This enables the study of oxygen consumption by PS-phosphorescence lifetime imaging (PLIM) and simultaneously changes the cellular metabolic state by nicotinamide adenine dinucleotide (NAD(P)H)-fluorescence lifetime imaging (FLIM). Within this study, localization and cellular function of TLD1433 is investigated in bladder carcinoma cells using time-resolved and confocal laser scanning microscopy. Simultaneous FLIM/PLIM of NAD(P)H and TLD1433 during PDT correlated oxygen consumption, redox state and cellular energy metabolism. Our investigations aimed to provide a personalized protocol in theranostic PDT procedures and demonstrate the potential use of TLD1433 PDT also under hypoxic conditions, which are otherwise difficult to treat.
Collapse
Affiliation(s)
- Sviatlana Kalinina
- Core Facility Confocal and Multiphoton Microscopy, University of Ulm, Ulm, Germany
| | - Jasmin Breymayer
- Core Facility Confocal and Multiphoton Microscopy, University of Ulm, Ulm, Germany
| | - Kirsten Reeß
- Core Facility Confocal and Multiphoton Microscopy, University of Ulm, Ulm, Germany
| | - Lothar Lilge
- Department of Medical Biophysics, Princess Margaret Cancer Institute/University of Toronto, Toronto, ON, Canada
| | | | - Angelika Rück
- Core Facility Confocal and Multiphoton Microscopy, University of Ulm, Ulm, Germany
| |
Collapse
|
144
|
Schaefer PM, Kalinina S, Rueck A, von Arnim CAF, von Einem B. NADH Autofluorescence-A Marker on its Way to Boost Bioenergetic Research. Cytometry A 2018; 95:34-46. [PMID: 30211978 DOI: 10.1002/cyto.a.23597] [Citation(s) in RCA: 128] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 07/20/2018] [Accepted: 08/04/2018] [Indexed: 12/20/2022]
Abstract
More than 60 years ago, the idea was introduced that NADH autofluorescence could be used as a marker of cellular redox state and indirectly also of cellular energy metabolism. Fluorescence lifetime imaging microscopy of NADH autofluorescence offers a marker-free readout of the mitochondrial function of cells in their natural microenvironment and allows different pools of NADH to be distinguished within a cell. Despite its many advantages in terms of spatial resolution and in vivo applicability, this technique still requires improvement in order to be fully useful in bioenergetics research. In the present review, we give a summary of technical and biological challenges that have so far limited the spread of this powerful technology. To help overcome these challenges, we provide a comprehensible overview of biological applications of NADH imaging, along with a detailed summary of valid imaging approaches that may be used to tackle many biological questions. This review is meant to provide all scientists interested in bioenergetics with support on how to embed successfully NADH imaging in their research. © 2018 International Society for Advancement of Cytometry.
Collapse
Affiliation(s)
| | - Sviatlana Kalinina
- Core Facility Confocal and Multiphoton Microscopy, Ulm University, Ulm, Germany
| | - Angelika Rueck
- Core Facility Confocal and Multiphoton Microscopy, Ulm University, Ulm, Germany
| | - Christine A F von Arnim
- Department of Neurology, Ulm University, Ulm, Germany.,Clinic for Neurogeriatrics and Neurological Rehabilitation, University- and Rehabilitation Hospital Ulm, Ulm, Germany
| | | |
Collapse
|
145
|
Castagnoli L, Iorio E, Dugo M, Koschorke A, Faraci S, Canese R, Casalini P, Nanni P, Vernieri C, Di Nicola M, Morelli D, Tagliabue E, Pupa SM. Intratumor lactate levels reflect HER2 addiction status in HER2-positive breast cancer. J Cell Physiol 2018; 234:1768-1779. [PMID: 30132876 PMCID: PMC6282573 DOI: 10.1002/jcp.27049] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Accepted: 06/25/2018] [Indexed: 12/17/2022]
Abstract
Despite different molecular tumor profiles indicate that human epidermal growth factor receptor 2 (HER2) messenger RNA (mRNA) levels mirror HER2 addiction and trastuzumab benefit in HER2-positive breast cancer (BC), the identification of noninvasive clinical predictors of trastuzumab sensitivity remains an unmet clinical need. In the current study, we investigated whether intratumor lactate levels reflect HER2 addiction and, in turn, trastuzumab susceptibility. Accordingly, the gene expression profiles of transgenic murine BC cell lines expressing the human d16HER2 variant (HER2-addicted) or human full-length HER2 (WTHER2; HER2-nonaddicted) revealed a significant enrichment of glycolysis-related gene pathways in HER2-addicted cells. We studied the metabolic content of 22 human HER2-positive BC by quantitative nuclear magnetic resonance spectroscopy and found that those cases with higher lactate levels were characterized by higher HER2 transcript levels. Moreover, gene expression analyses of HER2-positive BC samples from a TCGA data set revealed a significant enrichment in glycolysis-related pathways in high/HER2-addicted tumors. These data were confirmed by metabolic analyses of human HER2-positive BC cell lines with high or low HER2 transcript levels, which revealed significantly more active glycolytic metabolism in high HER2 transcript than in low HER2 transcript cells. Overall, our results provide evidence for noninvasive intratumor lactate detection as a potential metabolic biomarker of HER2 addiction and trastuzumab response suggesting the possibility to use in vivo imaging to assess lactate levels and, in turn, select HER2-positive BC patients who are more likely to benefit from anti-HER2 treatments.
Collapse
Affiliation(s)
- Lorenzo Castagnoli
- Molecular Targeting Unit, Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Egidio Iorio
- Core Facilities, NMR Unit, Istituto Superiore di Sanità, Roma, Italy
| | - Matteo Dugo
- Functional Genomics and Bioinformatics Core Facility, Department of Applied Research and Technology Development, Fondazione IRCCS Istituto Nazionale deiTumori, Milan, Italy
| | - Ada Koschorke
- Molecular Targeting Unit, Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Simona Faraci
- Molecular Targeting Unit, Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Rossella Canese
- Core Facilities, NMR Unit, Istituto Superiore di Sanità, Roma, Italy
| | - Patrizia Casalini
- Molecular Targeting Unit, Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Patrizia Nanni
- Laboratory of Immunology and Biology of Metastasis, Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy
| | - Claudio Vernieri
- IFOM, FIRC Institute of Molecular Oncology, Milan, Italy.,Department of Medical Oncology and Hematology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Massimo Di Nicola
- Unit of Immunotherapy and Anticancer Innovative Therapeutics, Department of Medical Oncology and Hematology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Daniele Morelli
- Laboratory Medicine Unit, Department of Pathology and Laboratory Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Elda Tagliabue
- Molecular Targeting Unit, Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Serenella M Pupa
- Molecular Targeting Unit, Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| |
Collapse
|
146
|
Romero-Cordoba SL, Rodriguez-Cuevas S, Bautista-Pina V, Maffuz-Aziz A, D'Ippolito E, Cosentino G, Baroni S, Iorio MV, Hidalgo-Miranda A. Loss of function of miR-342-3p results in MCT1 over-expression and contributes to oncogenic metabolic reprogramming in triple negative breast cancer. Sci Rep 2018; 8:12252. [PMID: 30115973 PMCID: PMC6095912 DOI: 10.1038/s41598-018-29708-9] [Citation(s) in RCA: 83] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Accepted: 07/17/2018] [Indexed: 12/20/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is a heterogeneous and aggressive neoplasia lacking the expression of hormonal receptors and human epidermal growth factor receptor-2. Accumulating evidence has highlighted the importance of miRNAs dysregulation in the establishment of cancer programs, but the functional role of many miRNAs remains unclear. The description of miRNAs roles might provide novel strategies for treatment. In the present work, an integrated analysis of miRNA transcriptional landscape was performed (N = 132), identifying the significant down-modulation of miR-342-3p in TNBC, probably because of the aberrant activity of estrogen receptor, which serves as a transcription factor of the miRNA, as demonstrated by a siRNA-knockdown approach. The enhanced expression of miR-342-3p significantly decreased cell proliferation, viability and migration rates of diverse TN cells in vitro. Bioinformatic and functional analyses revealed that miR-342-3p directly targets the monocarboxylate transporter 1 (MCT1), which promotes lactate and glucose fluxes alteration, thus disrupting the metabolic homeostasis of tumor cells. Optical metabolic imaging assay defined a higher optical redox ratio in glycolytic cells overexpressing miR-342-3p. Furthermore, we found that hypoxic conditions and glucose starvation attenuate miR-342-3p expression, suggesting a crosstalk program between these metabolic factors. Consistently, miR-342-3p down-modulation is associated with an increased MCT1 expression level and glycolytic score in human triple negative tumors. Overall, we described for the first time the regulatory activity of miR-342-3p on relevant metabolic carcinogenic pathways in TN breast cancers.
Collapse
Affiliation(s)
- Sandra L Romero-Cordoba
- Cancer Genomics Laboratory, National Institute of Genomic Medicine, Mexico City, Mexico
- Start Up Unit, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | | | | | | | - Elvira D'Ippolito
- Start Up Unit, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Giulia Cosentino
- Start Up Unit, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Sara Baroni
- Start Up Unit, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Marilena V Iorio
- Start Up Unit, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy.
| | | |
Collapse
|
147
|
Zhu C, Martin HL, Crouch BT, Martinez AF, Li M, Palmer GM, Dewhirst MW, Ramanujam N. Near-simultaneous quantification of glucose uptake, mitochondrial membrane potential, and vascular parameters in murine flank tumors using quantitative diffuse reflectance and fluorescence spectroscopy. BIOMEDICAL OPTICS EXPRESS 2018; 9:3399-3412. [PMID: 29984105 PMCID: PMC6033552 DOI: 10.1364/boe.9.003399] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Revised: 06/14/2018] [Accepted: 06/18/2018] [Indexed: 05/24/2023]
Abstract
The shifting metabolic landscape of aggressive tumors, with fluctuating oxygenation conditions and temporal changes in glycolysis and mitochondrial metabolism, is a critical phenomenon to study in order to understand negative treatment outcomes. Recently, we have demonstrated near-simultaneous optical imaging of mitochondrial membrane potential (MMP) and glucose uptake in non-tumor window chambers, using the fluorescent probes tetramethylrhodamine ethyl ester (TMRE) and 2-N-(7-nitrobenz-2-oxa-1,3-diazol-4-yl)amino)-2-deoxyglucose (2-NBDG). Here, we demonstrate a complementary technique to perform near-simultaneous in vivo optical spectroscopy of tissue vascular parameters, glucose uptake, and MMP in a solid tumor model that is most often used for therapeutic studies. Our study demonstrates the potential of optical spectroscopy as an effective tool to quantify the vascular and metabolic characteristics of a tumor, which is an important step towards understanding the mechanisms underlying cancer progression, metastasis, and resistance to therapies.
Collapse
Affiliation(s)
- Caigang Zhu
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
| | - Hannah L. Martin
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
| | - Brian T. Crouch
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
| | - Amy F. Martinez
- Currently with Office of Research, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Martin Li
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
| | - Gregory M. Palmer
- Department of Radiation Oncology, Duke University, Durham, NC 27710, USA
| | - Mark W. Dewhirst
- Department of Radiation Oncology, Duke University, Durham, NC 27710, USA
| | - Nimmi Ramanujam
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
| |
Collapse
|
148
|
A Radiosensitizing Inhibitor of HIF-1 alters the Optical Redox State of Human Lung Cancer Cells In Vitro. Sci Rep 2018; 8:8815. [PMID: 29891977 PMCID: PMC5995847 DOI: 10.1038/s41598-018-27262-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Accepted: 05/31/2018] [Indexed: 12/21/2022] Open
Abstract
Treatment failure caused by a radiation-resistant cell phenotype remains an impediment to the success of radiation therapy in cancer. We recently showed that a radiation-resistant isogenic line of human A549 lung cancer cells had significantly elevated expression of hypoxia-inducible factor (HIF-1α), and increased glucose catabolism compared with the parental, radiation-sensitive cell line. The objective of this study was to investigate the longitudinal metabolic changes in radiation-resistant and sensitive A549 lung cancer cells after treatment with a combination of radiation therapy and YC-1, a potent HIF-1 inhibitor. Using label-free two-photon excited fluorescence microscopy, we determined changes in the optical redox ratio of FAD/(NADH and FAD) over a period of 24 hours following treatment with YC-1, radiation, and both radiation and YC-1. To complement the optical redox ratio, we also evaluated changes in mitochondrial organization, glucose uptake, reactive oxygen species (ROS), and reduced glutathione. We observed significant differences in the optical redox ratio of radiation-resistant and sensitive A549 cells in response to radiation or YC-1 treatment alone; however, combined treatment eliminated these differences. Our results demonstrate that the optical redox ratio can elucidate radiosensitization of previously radiation-resistant A549 cancer cells, and provide a method for evaluating treatment response in patient-derived tumor biopsies.
Collapse
|
149
|
Hou J, Williams J, Botvinick EL, Potma EO, Tromberg BJ. Visualization of Breast Cancer Metabolism Using Multimodal Nonlinear Optical Microscopy of Cellular Lipids and Redox State. Cancer Res 2018; 78:2503-2512. [PMID: 29535219 PMCID: PMC5955854 DOI: 10.1158/0008-5472.can-17-2618] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Revised: 01/12/2018] [Accepted: 03/09/2018] [Indexed: 12/22/2022]
Abstract
Label-free nonlinear optical microscopy (NLOM) based on two-photon excited fluorescence (TPEF) from cofactors nicotinamide adenine dinucleotide (NADH) and flavin adenine dinucleotide (FAD+) is widely used for high-resolution cellular redox imaging. In this work, we combined three label-free NLOM imaging methods to quantitatively characterize breast cancer cells and their relative invasive potential: (i) TPEF optical redox ratio (ORR = FAD+/NADH + FAD+), (ii) coherent Raman scattering of cellular lipids, and (iii) second harmonic generation of extracellular matrix (ECM) collagen. 3D spheroid models of primary mammary epithelial (PME) cells and breast cancer cell lines (T47D and MDA-MB-231) were characterized based on their unique ORR and lipid volume fraction signatures. Treatment with 17β-estradiol (E2) increased glycolysis in both PME and T47D ER+ breast cancer acini. However, PME cells displayed increased lipid content with no effect on ECM, while T47D cells had decreased lipid storage (P < 0.001) and significant reorganization of collagen. By measuring deuterated lipids synthesized from exogenously administered deuterium-labeled glucose, treatment of T47D cells with E2 increased both lipid synthesis and consumption rates. These results confirm that glucose is a significant source for the cellular synthesis of lipid in glycolytic breast cancer cells, and that the combination of cellular redox and lipid fraction imaging endpoints is a powerful approach with new and complementary information content.Significance: These findings provide unique insight into metabolic processes, revealing correlations between cancer metastasis and cellular redox state, lipid metabolism, and extracellular matrix. Cancer Res; 78(10); 2503-12. ©2018 AACR.
Collapse
Affiliation(s)
- Jue Hou
- Laser Microbeam and Medical Program (LAMMP), Beckman Laser Institute and Medical Clinic, University of California Irvine, Irvine, California
| | - Joshua Williams
- Laser Microbeam and Medical Program (LAMMP), Beckman Laser Institute and Medical Clinic, University of California Irvine, Irvine, California
| | - Elliot L Botvinick
- Laser Microbeam and Medical Program (LAMMP), Beckman Laser Institute and Medical Clinic, University of California Irvine, Irvine, California
- Bio-Engineering of Advanced Mechanical Systems (BEAMS) Laboratory, University of California Irvine, Irvine, California
| | - Eric O Potma
- Laser Microbeam and Medical Program (LAMMP), Beckman Laser Institute and Medical Clinic, University of California Irvine, Irvine, California
- Department of Chemistry, University of California Irvine, Irvine, California
| | - Bruce J Tromberg
- Laser Microbeam and Medical Program (LAMMP), Beckman Laser Institute and Medical Clinic, University of California Irvine, Irvine, California.
| |
Collapse
|
150
|
Giskeødegård GF, Madssen TS, Euceda LR, Tessem MB, Moestue SA, Bathen TF. NMR-based metabolomics of biofluids in cancer. NMR IN BIOMEDICINE 2018; 32:e3927. [PMID: 29672973 DOI: 10.1002/nbm.3927] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Revised: 02/13/2018] [Accepted: 03/07/2018] [Indexed: 06/08/2023]
Abstract
This review describes the current status of NMR-based metabolomics of biofluids with respect to cancer risk assessment, detection, disease characterization, prognosis, and treatment monitoring. While the metabolism of cancer cells is altered compared with that of non-proliferating cells, the metabolome of blood and urine reflects the entire organism. We conclude that many studies show impressive associations between biofluid metabolomics and cancer progression, but translation to clinical practice is currently hindered by lack of validation, difficulties in biological interpretation, and non-standardized analytical procedures.
Collapse
Affiliation(s)
- Guro F Giskeødegård
- Department of Circulation and Medical Imaging, Norwegian University of Science and Technology-NTNU, Trondheim, Norway
| | - Torfinn S Madssen
- Department of Circulation and Medical Imaging, Norwegian University of Science and Technology-NTNU, Trondheim, Norway
| | - Leslie R Euceda
- Department of Circulation and Medical Imaging, Norwegian University of Science and Technology-NTNU, Trondheim, Norway
| | - May-Britt Tessem
- Department of Circulation and Medical Imaging, Norwegian University of Science and Technology-NTNU, Trondheim, Norway
| | - Siver A Moestue
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology-NTNU, Trondheim, Norway
- Department of Health Science, Nord University, Bodø, Norway
| | - Tone F Bathen
- Department of Circulation and Medical Imaging, Norwegian University of Science and Technology-NTNU, Trondheim, Norway
| |
Collapse
|