1
|
Stoliarov D, Koviarov A, Galiakhmetova D, Rafailov E. Compact femtosecond fiber laser tunable from 800 to 850 nm with pulse energy exceeding 5 nJ. Sci Rep 2025; 15:18867. [PMID: 40442375 PMCID: PMC12122858 DOI: 10.1038/s41598-025-04063-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Accepted: 05/23/2025] [Indexed: 06/02/2025] Open
Abstract
This paper introduces a compact, tunable femtosecond laser based on an Erbium-doped fiber, utilizing the Self-Soliton Frequency Shifted technique and PPLN crystal as a Second Harmonic Generation module. Achieving an unparalleled frequency conversion efficiency up to 55% for the 800 - 850 nm wavelength range, this compact laser emits sub-100 fs pulses. The laser operates simultaneously within the first and third biological windows, delivering pulse energies of 10.4 nJ and 5.1 nJ, respectively. This performance, previously unattained in similar systems, is achieved while maintaining Second Harmonic Generation power stability below 2% RMS. The presented compact laser, developed for bladder cancer detection through multiphoton microscopy, will significantly improve the system's compactness, precision, and cancer detection efficiency.
Collapse
Affiliation(s)
- Dmitrii Stoliarov
- Aston Institute of Photonic Technologies, Aston University, B4 7ET, Birmingham, UK.
| | - Aleksandr Koviarov
- Aston Institute of Photonic Technologies, Aston University, B4 7ET, Birmingham, UK
| | - Diana Galiakhmetova
- Aston Institute of Photonic Technologies, Aston University, B4 7ET, Birmingham, UK
| | - Edik Rafailov
- Aston Institute of Photonic Technologies, Aston University, B4 7ET, Birmingham, UK
| |
Collapse
|
2
|
Yan J, Goncalves CFL, Saha PS, Furdui CM, Zhu C. Optical imaging provides flow-cytometry-like single-cell level analysis of HIF-1 α-mediated metabolic changes in radioresistant head and neck squamous carcinoma cells. BIOPHOTONICS DISCOVERY 2025; 2:012702. [PMID: 39917319 PMCID: PMC11801402 DOI: 10.1117/1.bios.2.1.012702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/09/2025]
Abstract
Significance Radioresistance remains a significant problem for head and neck squamous cell carcinoma (HNSCC) patients. To mitigate this, the cellular and molecular pathways used by radioresistant HNSCC that drive recurrence must be studied. Aim We aim to demonstrate optical imaging strategies to provide flow cytometry-like single-cell level analysis of hypoxia-inducible factor 1-alpha (HIF-1α)-mediated metabolic changes in the radioresistant and radiosensitive HNSCC cells but in a more efficient, cost-effective, and non-destructive manner. Through both optical imaging and flow cytometry studies, we will reveal the role of radiation-induced HIF-1α overexpression and the following metabolic changes in the radioresistance development for HNSCC. Approach We optimized the use of two metabolic probes: 2-[N-(7-nitrobenz-2-oxa-1, 3-diazol-4-yl) amino]-2-deoxy-D-glucose (2-NBDG) (to report glucose uptake) and Tetramethylrhodamine ethyl ester (TMRE) (to report mitochondrial membrane potential) with both a standard fluorescence microscope and a flow cytometry device, to report the changes in metabolism between radioresistant (rSCC-61) and radiosensitive (SCC-61) HNSCC cell lines under radiation stresses with or without HIF-1α inhibition. Results We found that the matched HNSCC cell lines had different baseline metabolic phenotypes, and their metabolism responded differently to radiation stress along with significantly enhanced HIF-1α expressions in the rSCC-61 cells. HIF-1α inhibition during the radiation treatment modulates the metabolic changes and radio-sensitizes the rSCC-61 cells. Through these studies, we demonstrated that a standard fluorescence microscope along with proper image processing methods can provide flow cytometry-like single-cell level analysis of HIF-1α-mediated metabolic changes in the radioresistant and radiosensitive HNSCC cells. Conclusions Our reported optical imaging strategies may enable one to study the role of metabolism reprogramming in cancer therapeutic resistance development at the single-cell level in a more efficient, cost-effective, and non-destructive manner. Our understanding of radiation resistance mechanisms using our imaging methods will offer opportunities to design targeted radiotherapy for improved treatment outcomes for HNSCC patients.
Collapse
Affiliation(s)
- Jing Yan
- University of Kentucky, Department of Biomedical Engineering, Lexington, Kentucky, United States
| | | | - Pranto Soumik Saha
- University of Kentucky, Department of Biomedical Engineering, Lexington, Kentucky, United States
| | - Cristina M. Furdui
- Wake Forest University, Department of Internal Medicine, Winston-Salem, North Carolina, United States
| | - Caigang Zhu
- University of Kentucky, Department of Biomedical Engineering, Lexington, Kentucky, United States
| |
Collapse
|
3
|
Podsednik A, Xu HN, Li LZ. Passage dependence of NADH redox status and reactive oxygen species level in vitro in triple-negative breast cancer cell lines with different invasiveness. TRANSLATIONAL BREAST CANCER RESEARCH : A JOURNAL FOCUSING ON TRANSLATIONAL RESEARCH IN BREAST CANCER 2024; 5:27. [PMID: 39534579 PMCID: PMC11557164 DOI: 10.21037/tbcr-24-36] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024]
Abstract
Background The redox status of nicotinamide adenine dinucleotide (NAD; including oxidized form NAD+ and reduced form NADH) plays key roles in both health and disease and has been actively studied to develop cancer biomarkers and therapeutic strategies. With the optical redox imaging (ORI) technique, we have been investigating the relationship of NADH redox status, reactive oxygen species (ROS), and invasiveness in breast cancer cell cultures, and have associated higher invasiveness with more oxidized NADH redox state. However, the cell cultures may have phenotypic drift and metabolic change with increased passage numbers. Methods We investigated the passage-dependence of NADH redox status and ROS levels in two triple-negative breast cancer (TNBC) cell cultures: the more invasive/metastatic MDA-MB-231 and the less invasive/metastatic HCC1806 cell lines. We measured the NADH redox status, redox plasticity, and cytoplasmic and mitochondrial ROS levels under the basal condition and metabolic perturbations of the mitochondrial electron transport chain. We evaluated the dependence of redox and ROS profiles on the cell passage number by comparing the early (<20 passages) with the late (>60 passages) passage cells. Results (I) NADH redox and ROS baselines are stable and independent of cell passage number, but can vary with passage number under metabolic perturbations depending on specific perturbation and cell line; (II) NADH redox status and intracellular ROS levels can change discordantly in cancer cells; (III) under both basal and metabolically perturbed conditions, the more invasive cell line has a more oxidized NADH redox status with a higher basal cytoplasmic ROS level than the less invasive line, regardless of passage number. Conclusions The general correlation between redox, ROS, and invasiveness in studied TNBC cells is not very sensitive to passage number. These results indicate that NADH redox and basal ROS status in TNBC likely reflect the intrinsic progressive nature of TNBC cells.
Collapse
Affiliation(s)
- Allison Podsednik
- Britton Chance Laboratory of Redox Imaging, Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - He N Xu
- Britton Chance Laboratory of Redox Imaging, Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Lin Z Li
- Britton Chance Laboratory of Redox Imaging, Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
4
|
Restall BS, Haven NJM, Martell MT, Cikaluk BD, Wang J, Kedarisetti P, Tejay S, Adam BA, Sutendra G, Li X, Zemp RJ. Metabolic light absorption, scattering, and emission (MetaLASE) microscopy. SCIENCE ADVANCES 2024; 10:eadl5729. [PMID: 39423271 PMCID: PMC11488571 DOI: 10.1126/sciadv.adl5729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Accepted: 09/13/2024] [Indexed: 10/21/2024]
Abstract
Optical imaging of metabolism can provide key information about health and disease progression in cells and tissues; however, current methods have lacked gold-standard information about histological structure. Conversely, histology and virtual histology methods have lacked metabolic contrast. Here, we present metabolic light absorption, scattering, and emission (MetaLASE) microscopy, which rapidly provides a virtual histology and optical metabolic readout simultaneously. Hematoxylin-like nucleic contrast and eosin-like cytoplasmic contrast are obtained using photoacoustic remote sensing and ultraviolet reflectance microscopy, respectively. The same ultraviolet source excites endogenous Nicotinamide adenine dinucleotide (phosphate), flavin adenine dinucleotide, and collagen autofluorescence, providing a map of optical redox ratios to visualize metabolic variations including in areas of invasive carcinoma. Benign chronic inflammation and glands also are seen to exhibit hypermetabolism. MetaLASE microscopy offers promise for future applications in intraoperative margin analysis and in research applications where greater insights into metabolic activity could be correlated with cell and tissue types.
Collapse
Affiliation(s)
- Brendon S. Restall
- Department of Electrical and Computer Engineering, University of Alberta, 116 Street & 85 Avenue, Edmonton, Alberta T6G 2R3, Canada
| | - Nathaniel J. M. Haven
- Department of Electrical and Computer Engineering, University of Alberta, 116 Street & 85 Avenue, Edmonton, Alberta T6G 2R3, Canada
| | - Matthew T. Martell
- Department of Electrical and Computer Engineering, University of Alberta, 116 Street & 85 Avenue, Edmonton, Alberta T6G 2R3, Canada
| | - Brendyn D. Cikaluk
- Department of Electrical and Computer Engineering, University of Alberta, 116 Street & 85 Avenue, Edmonton, Alberta T6G 2R3, Canada
| | - Joy Wang
- Department of Electrical and Computer Engineering, University of Alberta, 116 Street & 85 Avenue, Edmonton, Alberta T6G 2R3, Canada
| | - Pradyumna Kedarisetti
- Department of Electrical and Computer Engineering, University of Alberta, 116 Street & 85 Avenue, Edmonton, Alberta T6G 2R3, Canada
| | - Saymon Tejay
- Department of Medicine, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Benjamin A. Adam
- Department of Laboratory Medicine and Pathology, University of Alberta, 8440-112 Street, Edmonton, Alberta T6G 2B7, Canada
| | - Gopinath Sutendra
- Department of Medicine, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Xingyu Li
- Department of Electrical and Computer Engineering, University of Alberta, 116 Street & 85 Avenue, Edmonton, Alberta T6G 2R3, Canada
| | - Roger J. Zemp
- Department of Electrical and Computer Engineering, University of Alberta, 116 Street & 85 Avenue, Edmonton, Alberta T6G 2R3, Canada
| |
Collapse
|
5
|
Abir AH, Weckwerth L, Wilhelm A, Thomas J, Reichardt CM, Munoz L, Völkl S, Appelt U, Mroz M, Niesner R, Hauser A, Sophie Fischer R, Pracht K, Jäck HM, Schett G, Krönke G, Mielenz D. Metabolic profiling of single cells by exploiting NADH and FAD fluorescence via flow cytometry. Mol Metab 2024; 87:101981. [PMID: 38971403 PMCID: PMC11300934 DOI: 10.1016/j.molmet.2024.101981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 06/14/2024] [Accepted: 06/27/2024] [Indexed: 07/08/2024] Open
Abstract
OBJECTIVE The metabolism of different cells within the same microenvironment can differ and dictate physiological or pathological adaptions. Current single-cell analysis methods of metabolism are not label-free. METHODS The study introduces a label-free, live-cell analysis method assessing endogenous fluorescence of NAD(P)H and FAD in surface-stained cells by flow cytometry. RESULTS OxPhos inhibition, mitochondrial uncoupling, glucose exposure, genetic inactivation of glucose uptake and mitochondrial respiration alter the optical redox ratios of FAD and NAD(P)H as measured by flow cytometry. Those alterations correlate strongly with measurements obtained by extracellular flux analysis. Consequently, metabolically distinct live B-cell populations can be resolved, showing that human memory B-cells from peripheral blood exhibit a higher glycolytic flexibility than naïve B cells. Moreover, the comparison of blood-derived B- and T-lymphocytes from healthy donors and rheumatoid arthritis patients unleashes rheumatoid arthritis-associated metabolic traits in human naïve and memory B-lymphocytes. CONCLUSIONS Taken together, these data show that the optical redox ratio can depict metabolic differences in distinct cell populations by flow cytometry.
Collapse
Affiliation(s)
- Ariful Haque Abir
- Division of Molecular Immunology, Department of Internal Medicine 3, Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, Nikolaus-Fiebiger-Center, Glückstr. 6, 91054 Erlangen, Germany
| | - Leonie Weckwerth
- Division of Molecular Immunology, Department of Internal Medicine 3, Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, Nikolaus-Fiebiger-Center, Glückstr. 6, 91054 Erlangen, Germany
| | - Artur Wilhelm
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany; Deutsches Zentrum für Immuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Jana Thomas
- Division of Molecular Immunology, Department of Internal Medicine 3, Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, Nikolaus-Fiebiger-Center, Glückstr. 6, 91054 Erlangen, Germany
| | - Clara M Reichardt
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany; Deutsches Zentrum für Immuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Luis Munoz
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany; Deutsches Zentrum für Immuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Simon Völkl
- Department of Internal Medicine 5, Hematology and Oncology, Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, Ulmenweg 18, 91054 Erlangen, Germany
| | - Uwe Appelt
- Flow cytometry core unit, Friedrich-Alexander-Universität Erlangen-Nürnberg, Glückstr. 6, 91054 Erlangen, Germany
| | - Markus Mroz
- Flow cytometry core unit, Friedrich-Alexander-Universität Erlangen-Nürnberg, Glückstr. 6, 91054 Erlangen, Germany
| | - Raluca Niesner
- Deutsches Rheumaforschungszentrum Berlin, Biophysikalische Analytik, Charitéplatz 1, 10117 Berlin, Germany; Freie Universität Berlin, Dynamisches und funktionelles in vivo Imaging, Adresse: Oertzenweg 19b, 14163 Berlin, Germany
| | - Anja Hauser
- Medizinische Klinik mit Schwerpunkt Rheumatologie und Klinische Immunologie, Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany; Deutsches Rheumaforschungszentrum Berlin, Immundynamik, Charitéplatz 1, 10117 Berlin, Germany
| | - Rebecca Sophie Fischer
- Division of Molecular Immunology, Department of Internal Medicine 3, Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, Nikolaus-Fiebiger-Center, Glückstr. 6, 91054 Erlangen, Germany
| | - Katharina Pracht
- Division of Molecular Immunology, Department of Internal Medicine 3, Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, Nikolaus-Fiebiger-Center, Glückstr. 6, 91054 Erlangen, Germany
| | - Hans-Martin Jäck
- Division of Molecular Immunology, Department of Internal Medicine 3, Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, Nikolaus-Fiebiger-Center, Glückstr. 6, 91054 Erlangen, Germany
| | - Georg Schett
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany; Deutsches Zentrum für Immuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Gerhard Krönke
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany; Deutsches Zentrum für Immuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany; Medizinische Klinik mit Schwerpunkt Rheumatologie und Klinische Immunologie, Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Dirk Mielenz
- Division of Molecular Immunology, Department of Internal Medicine 3, Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, Nikolaus-Fiebiger-Center, Glückstr. 6, 91054 Erlangen, Germany.
| |
Collapse
|
6
|
Gallego-López GM, Contreras Guzman E, Desa DE, Knoll LJ, Skala MC. Metabolic changes in Toxoplasma gondii-infected host cells measured by autofluorescence imaging. mBio 2024; 15:e0072724. [PMID: 38975793 PMCID: PMC11323734 DOI: 10.1128/mbio.00727-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Accepted: 05/06/2024] [Indexed: 07/09/2024] Open
Abstract
Toxoplasma gondii, the causative agent of toxoplasmosis, is an obligate intracellular parasite that infects warm-blooded vertebrates across the world. In humans, seropositivity rates of T. gondii range from 10% to 90% across communities. Despite its prevalence, few studies address how T. gondii infection changes the metabolism of host cells. In this study, we investigate how T. gondii manipulates the host cell metabolic environment by monitoring the metabolic response over time using noninvasive autofluorescence lifetime imaging of single cells, metabolite analysis, extracellular flux analysis, and reactive oxygen species (ROS) production. Autofluorescence lifetime imaging indicates that infected host cells become more oxidized and have an increased proportion of bound NAD(P)H compared to uninfected controls. Over time, infected cells also show decreases in levels of intracellular glucose and lactate, increases in oxygen consumption, and variability in ROS production. We further examined changes associated with the pre-invasion "kiss and spit" process using autofluorescence lifetime imaging, which also showed a more oxidized host cell with an increased proportion of bound NAD(P)H over 48 hours compared to uninfected controls, suggesting that metabolic changes in host cells are induced by T. gondii kiss and spit even without invasion.IMPORTANCEThis study sheds light on previously unexplored changes in host cell metabolism induced by T. gondii infection using noninvasive, label-free autofluorescence imaging. In this study, we use optical metabolic imaging (OMI) to measure the optical redox ratio (ORR) in conjunction with fluorescence lifetime imaging microscopy (FLIM) to noninvasively monitor single host cell response to T. gondii infection over 48 hours. Collectively, our results affirm the value of using autofluorescence lifetime imaging to noninvasively monitor metabolic changes in host cells over the time course of a microbial infection. Understanding this metabolic relationship between the host cell and the parasite could uncover new treatment and prevention options for T. gondii infections worldwide.
Collapse
Affiliation(s)
- Gina M. Gallego-López
- Morgridge Institute for Research, Madison, Wisconsin, USA
- Department of Medical Microbiology & Immunology, University of Wisconsin–Madison, Madison, Wisconsin, USA
| | | | | | - Laura J. Knoll
- Department of Medical Microbiology & Immunology, University of Wisconsin–Madison, Madison, Wisconsin, USA
| | - Melissa C. Skala
- Morgridge Institute for Research, Madison, Wisconsin, USA
- Department of Biomedical Engineering, University of Wisconsin–Madison, Madison, Wisconsin, USA
| |
Collapse
|
7
|
Zhou H, Li I, Bramlett CS, Wang B, Hao J, Yen DP, Ando Y, Fraser SE, Lu R, Shen K. Label-free metabolic optical biomarkers track stem cell fate transition in real time. SCIENCE ADVANCES 2024; 10:eadi6770. [PMID: 38718114 PMCID: PMC11078180 DOI: 10.1126/sciadv.adi6770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 04/04/2024] [Indexed: 05/12/2024]
Abstract
Tracking stem cell fate transition is crucial for understanding their development and optimizing biomanufacturing. Destructive single-cell methods provide a pseudotemporal landscape of stem cell differentiation but cannot monitor stem cell fate in real time. We established a metabolic optical metric using label-free fluorescence lifetime imaging microscopy (FLIM), feature extraction and machine learning-assisted analysis, for real-time cell fate tracking. From a library of 205 metabolic optical biomarker (MOB) features, we identified 56 associated with hematopoietic stem cell (HSC) differentiation. These features collectively describe HSC fate transition and detect its bifurcate lineage choice. We further derived a MOB score measuring the "metabolic stemness" of single cells and distinguishing their division patterns. This score reveals a distinct role of asymmetric division in rescuing stem cells with compromised metabolic stemness and a unique mechanism of PI3K inhibition in promoting ex vivo HSC maintenance. MOB profiling is a powerful tool for tracking stem cell fate transition and improving their biomanufacturing from a single-cell perspective.
Collapse
Affiliation(s)
- Hao Zhou
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA 90089, USA
| | - Irene Li
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA 90089, USA
| | - Charles S. Bramlett
- Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Bowen Wang
- Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Jia Hao
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA 90089, USA
| | - Daniel P. Yen
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA 90089, USA
| | - Yuta Ando
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA 90089, USA
| | - Scott E. Fraser
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA 90089, USA
- Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles, CA 90033, USA
- Translational Imaging Center, University of Southern California, Los Angeles, CA 90089, USA
- Molecular and Computational Biology, University of Southern California, Los Angeles, CA 90089, USA
| | - Rong Lu
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA 90089, USA
- Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles, CA 90033, USA
- Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA 90033, USA
- Department of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Keyue Shen
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA 90089, USA
- Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA 90033, USA
- USC Stem Cell, University of Southern California, Los Angeles, CA 90033, USA
| |
Collapse
|
8
|
Kostyuk AI, Rapota DD, Morozova KI, Fedotova AA, Jappy D, Semyanov AV, Belousov VV, Brazhe NA, Bilan DS. Modern optical approaches in redox biology: Genetically encoded sensors and Raman spectroscopy. Free Radic Biol Med 2024; 217:68-115. [PMID: 38508405 DOI: 10.1016/j.freeradbiomed.2024.03.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 02/10/2024] [Accepted: 03/13/2024] [Indexed: 03/22/2024]
Abstract
The objective of the current review is to summarize the current state of optical methods in redox biology. It consists of two parts, the first is dedicated to genetically encoded fluorescent indicators and the second to Raman spectroscopy. In the first part, we provide a detailed classification of the currently available redox biosensors based on their target analytes. We thoroughly discuss the main architecture types of these proteins, the underlying engineering strategies for their development, the biochemical properties of existing tools and their advantages and disadvantages from a practical point of view. Particular attention is paid to fluorescence lifetime imaging microscopy as a possible readout technique, since it is less prone to certain artifacts than traditional intensiometric measurements. In the second part, the characteristic Raman peaks of the most important redox intermediates are listed, and examples of how this knowledge can be implemented in biological studies are given. This part covers such fields as estimation of the redox states and concentrations of Fe-S clusters, cytochromes, other heme-containing proteins, oxidative derivatives of thiols, lipids, and nucleotides. Finally, we touch on the issue of multiparameter imaging, in which biosensors are combined with other visualization methods for simultaneous assessment of several cellular parameters.
Collapse
Affiliation(s)
- Alexander I Kostyuk
- M.M. Shemyakin and Yu.A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, 117997, Russia; Pirogov Russian National Research Medical University, 117997, Moscow, Russia
| | - Diana D Rapota
- M.M. Shemyakin and Yu.A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, 117997, Russia
| | - Kseniia I Morozova
- Faculty of Biology, M.V. Lomonosov Moscow State University, Moscow, 119234, Russia
| | - Anna A Fedotova
- M.M. Shemyakin and Yu.A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, 117997, Russia; Faculty of Biology, M.V. Lomonosov Moscow State University, Moscow, 119234, Russia
| | - David Jappy
- Federal Center of Brain Research and Neurotechnologies, Federal Medical Biological Agency, Moscow, 117997, Russia
| | - Alexey V Semyanov
- M.M. Shemyakin and Yu.A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, 117997, Russia; Faculty of Biology, M.V. Lomonosov Moscow State University, Moscow, 119234, Russia; Sechenov First Moscow State Medical University, Moscow, 119435, Russia; College of Medicine, Jiaxing University, Jiaxing, Zhejiang Province, 314001, China
| | - Vsevolod V Belousov
- M.M. Shemyakin and Yu.A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, 117997, Russia; Pirogov Russian National Research Medical University, 117997, Moscow, Russia; Federal Center of Brain Research and Neurotechnologies, Federal Medical Biological Agency, Moscow, 117997, Russia; Life Improvement by Future Technologies (LIFT) Center, Skolkovo, Moscow, 143025, Russia
| | - Nadezda A Brazhe
- M.M. Shemyakin and Yu.A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, 117997, Russia; Faculty of Biology, M.V. Lomonosov Moscow State University, Moscow, 119234, Russia.
| | - Dmitry S Bilan
- M.M. Shemyakin and Yu.A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, 117997, Russia; Pirogov Russian National Research Medical University, 117997, Moscow, Russia.
| |
Collapse
|
9
|
Peng A, Xu HN, Moon L, Zhang P, Li LZ. Quantitative Optical Redox Imaging of Melanoma Xenografts with Different Metastatic Potentials. Cancers (Basel) 2024; 16:1669. [PMID: 38730620 PMCID: PMC11083304 DOI: 10.3390/cancers16091669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 04/18/2024] [Accepted: 04/22/2024] [Indexed: 05/13/2024] Open
Abstract
To develop imaging biomarkers for tumors aggressiveness, our previous optical redox imaging (ORI) studies of the reduced nicotinamide adenine dinucleotide (NADH) and oxidized flavoproteins (Fp, containing flavin adenine dinucleotide, i.e., FAD) in tumor xenografts of human melanoma associated the high optical redox ratio (ORR = Fp/(Fp + NADH)) and its heterogeneity to the high invasive/metastatic potential, without having reported quantitative results for NADH and Fp. Here, we implemented a calibration procedure to facilitate imaging the nominal concentrations of tissue NADH and Fp in the mouse xenografts of two human melanoma lines, an indolent less metastatic A375P and a more metastatic C8161. Images of the redox indices (NADH, Fp, ORR) revealed the existence of more oxidized areas (OAs) and more reduced areas (RAs) within individual tumors. ORR was found to be higher and NADH lower in C8161 compared to that of A375P xenografts, both globally for the whole tumors and locally in OAs. The ORR in the OA can differentiate xenografts with a higher statistical significance than the global averaged ORR. H&E staining of the tumors indicated that the redox differences we identified were more likely due to intrinsically different cell metabolism, rather than variations in cell density.
Collapse
Affiliation(s)
- April Peng
- Britton Chance Laboratory of Redox Imaging, Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (A.P.); (H.N.X.); (L.M.)
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - He N. Xu
- Britton Chance Laboratory of Redox Imaging, Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (A.P.); (H.N.X.); (L.M.)
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA 19104, USA
- Institute of Translational Medicine and Therapeutics, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Lily Moon
- Britton Chance Laboratory of Redox Imaging, Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (A.P.); (H.N.X.); (L.M.)
| | - Paul Zhang
- Department of Pathology, University of Pennsylvania, Philadelphia, PA 19104, USA;
| | - Lin Z. Li
- Britton Chance Laboratory of Redox Imaging, Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (A.P.); (H.N.X.); (L.M.)
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA 19104, USA
- Institute of Translational Medicine and Therapeutics, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
10
|
Johnson HR, Gunder LC, Gillette A, Sleiman H, Rademacher BL, Meske LM, Culberson WS, Micka JA, Favreau P, Yao E, Matkowskyj KA, Skala MC, Carchman EH. Preclinical Models of Anal Cancer Combined-Modality Therapy. J Surg Res 2024; 294:82-92. [PMID: 37864962 DOI: 10.1016/j.jss.2023.09.053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 09/15/2023] [Accepted: 09/19/2023] [Indexed: 10/23/2023]
Abstract
INTRODUCTION There have been no significant changes in anal cancer treatment options in 4 decades. In this study, we highlight two preclinical models designed to assess anal cancer treatments. MATERIALS AND METHODS Transgenic K14E6/E7 mice were treated with 7, 12-dimethylbenz(a)anthracene until anal tumors developed. Mice were treated with localized radiation in addition to chemotherapy (combined-modality therapy [CMT]) and compared to no treatment control (NTC). K14E6/E7 mouse anal spheroids with and without Pik3ca mutations were isolated and treated with vehicle, LY3023414 (LY3) (a drug previously shown to be effective in cancer prevention), CMT, or CMT + LY3. RESULTS In the in vivo model, there was a significant increase in survival in the CMT group compared to the NTC group (P = 0.0392). In the ex vivo model, there was a significant decrease in the mean diameter of CMT and CMT + LY3-treated spheroids compared to vehicle (P ≤ 0.0001). For LY3 alone compared to vehicle, there was a statistically significant decrease in spheroid size in the K14E6/E7 group without mutation (P = 0.0004). CONCLUSIONS We have provided proof of concept for two preclinical anal cancer treatment models that allow for the future testing of novel therapies for anal cancer.
Collapse
Affiliation(s)
- Hillary R Johnson
- Department of Surgery, University of Wisconsin - Madison, Madison, Wisconsin
| | - Laura C Gunder
- Department of Surgery, University of Wisconsin - Madison, Madison, Wisconsin
| | | | - Hana Sleiman
- Department of Surgery, University of Wisconsin - Madison, Madison, Wisconsin
| | - Brooks L Rademacher
- Department of Surgery, University of Wisconsin - Madison, Madison, Wisconsin
| | - Louise M Meske
- Department of Surgery, University of Wisconsin - Madison, Madison, Wisconsin
| | - Wesley S Culberson
- Department of Medical Physics, School of Medicine and Public Health, University of Wisconsin - Madison, Madison, Wisconsin
| | - John A Micka
- Department of Medical Physics, School of Medicine and Public Health, University of Wisconsin - Madison, Madison, Wisconsin
| | - Peter Favreau
- Morgridge Institute for Research, Madison, Wisconsin
| | - Evan Yao
- Department of Surgery, University of Wisconsin - Madison, Madison, Wisconsin
| | - Kristina A Matkowskyj
- Department of Pathology and Laboratory Medicine, University of Wisconsin Madison, Madison, Wisconsin; Department of Biomedical Engineering, University of Wisconsin - Madison, Madison, Wisconsin; William S. Middleton Memorial Veterans, Madison, Wisconsin
| | - Melissa C Skala
- Morgridge Institute for Research, Madison, Wisconsin; Department of Biomedical Engineering, University of Wisconsin - Madison, Madison, Wisconsin
| | - Evie H Carchman
- Department of Surgery, University of Wisconsin - Madison, Madison, Wisconsin; Carbone Cancer Center, School of Medicine and Public Health, University of Wisconsin - Madison, Madison, Wisconsin; William S. Middleton Memorial Veterans, Madison, Wisconsin.
| |
Collapse
|
11
|
Gallego-López GM, Guzman EC, Knoll LJ, Skala M. Metabolic changes to host cells with Toxoplasma gondii infection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.10.552811. [PMID: 37609172 PMCID: PMC10441426 DOI: 10.1101/2023.08.10.552811] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/24/2023]
Abstract
Toxoplasma gondii, the causative agent of toxoplasmosis, is an obligate intracellular parasite that infects warm-blooded vertebrates across the world. In humans, seropositivity rates of T. gondii range from 10% to 90%. Despite its prevalence, few studies address how T. gondii infection changes the metabolism of host cells. Here, we investigate how T. gondii manipulates the host cell metabolic environment by monitoring metabolic response over time using non-invasive autofluorescence lifetime imaging of single cells, seahorse metabolic flux analysis, reactive oxygen species (ROS) production, and metabolomics. Autofluorescence lifetime imaging indicates that infected host cells become more oxidized and have an increased proportion of bound NAD(P)H with infection. These findings are consistent with changes in mitochondrial and glycolytic function, decrease of intracellular glucose, fluctuations in lactate and ROS production in infected cells over time. We also examined changes associated with the pre-invasion "kiss and spit" process using autofluorescence lifetime imaging, which similarly showed a more oxidized host cell with an increased proportion of bound NAD(P)H over 48 hours. Glucose metabolic flux analysis indicated that these changes are driven by NADH and NADP+ in T. gondii infection. In sum, metabolic changes in host cells with T. gondii infection were similar during full infection, and kiss and spit. Autofluorescence lifetime imaging can non-invasively monitor metabolic changes in host cells over a microbial infection time-course.
Collapse
Affiliation(s)
- Gina M. Gallego-López
- Morgridge Institute for Research, Madison, WI, 53706
- Department of Medical Microbiology & Immunology, University of Wisconsin-Madison, Madison, WI, 53706
| | | | - Laura J. Knoll
- Department of Medical Microbiology & Immunology, University of Wisconsin-Madison, Madison, WI, 53706
| | - Melissa Skala
- Morgridge Institute for Research, Madison, WI, 53706
- Department of Biomedical Engineering, University of Wisconsin- Madison, WI 53706, USA
| |
Collapse
|
12
|
Yan J, Lima Goncalves CF, Korfhage MO, Hasan MZ, Fan TWM, Wang X, Zhu C. Portable optical spectroscopic assay for non-destructive measurement of key metabolic parameters on in vitro cancer cells and organotypic fresh tumor slices. BIOMEDICAL OPTICS EXPRESS 2023; 14:4065-4079. [PMID: 37799678 PMCID: PMC10549737 DOI: 10.1364/boe.497127] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 07/04/2023] [Accepted: 07/05/2023] [Indexed: 10/07/2023]
Abstract
To enable non-destructive metabolic characterizations on in vitro cancer cells and organotypic tumor models for therapeutic studies in an easy-to-access way, we report a highly portable optical spectroscopic assay for simultaneous measurement of glucose uptake and mitochondrial function on various cancer models with high sensitivity. Well-established breast cancer cell lines (MCF-7 and MDA-MB-231) were used to validate the optical spectroscopic assay for metabolic characterizations, while fresh tumor samples harvested from both animals and human cancer patients were used to test the feasibility of our optical metabolic assay for non-destructive measurement of key metabolic parameters on organotypic tumor slices. Our optical metabolic assay captured that MCF-7 cells had higher mitochondrial metabolism, but lower glucose uptake compared to the MDA-MB-231 cells, which is consistent with our microscopy imaging and flow cytometry data, as well as the published Seahorse Assay data. Moreover, we demonstrated that our optical assay could non-destructively measure both glucose uptake and mitochondrial metabolism on the same cancer cell samples at one time, which remains challenging by existing metabolic tools. Our pilot tests on thin fresh tumor slices showed that our optical assay captured increased metabolic activities in tumors compared to normal tissues. Our non-destructive optical metabolic assay provides a cost-effective way for future longitudinal therapeutic studies using patient-derived organotypic fresh tumor slices through the lens of tumor energetics, which will significantly advance translational cancer research.
Collapse
Affiliation(s)
- Jing Yan
- Department of Biomedical Engineering,
University of Kentucky, Lexington, KY 40506, USA
| | | | - Madison O. Korfhage
- Department of Biomedical Engineering,
University of Kentucky, Lexington, KY 40506, USA
| | - Md Zahid Hasan
- Department of Biomedical Engineering,
University of Kentucky, Lexington, KY 40506, USA
| | - Teresa W.-M. Fan
- Center for Environmental and Systems Biochemistry, University of Kentucky, Lexington, KY 40536, USA
- Markey Cancer Center, University of Kentucky, Lexington, KY 40536, USA
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY 40536, USA
| | - Xiaoqin Wang
- Department of Radiology, University of Kentucky, Lexington, KY 40536, USA
| | - Caigang Zhu
- Department of Biomedical Engineering,
University of Kentucky, Lexington, KY 40506, USA
| |
Collapse
|
13
|
Doczi J, Karnok N, Bui D, Azarov V, Pallag G, Nazarian S, Czumbel B, Seyfried TN, Chinopoulos C. Viability of HepG2 and MCF-7 cells is not correlated with mitochondrial bioenergetics. Sci Rep 2023; 13:10822. [PMID: 37402778 DOI: 10.1038/s41598-023-37677-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Accepted: 06/26/2023] [Indexed: 07/06/2023] Open
Abstract
Alterations in metabolism are a hallmark of cancer. It is unclear if oxidative phosphorylation (OXPHOS) is necessary for tumour cell survival. In this study, we investigated the effects of severe hypoxia, site-specific inhibition of respiratory chain (RC) components, and uncouplers on necrotic and apoptotic markers in 2D-cultured HepG2 and MCF-7 tumour cells. Comparable respiratory complex activities were observed in both cell lines. However, HepG2 cells exhibited significantly higher oxygen consumption rates (OCR) and respiratory capacity than MCF-7 cells. Significant non-mitochondrial OCR was observed in MCF-7 cells, which was insensitive to acute combined inhibition of complexes I and III. Pre-treatment of either cell line with RC inhibitors for 24-72 h resulted in the complete abolition of respective complex activities and OCRs. This was accompanied by a time-dependent decrease in citrate synthase activity, suggesting mitophagy. High-content automated microscopy recordings revealed that the viability of HepG2 cells was mostly unaffected by any pharmacological treatment or severe hypoxia. In contrast, the viability of MCF-7 cells was strongly affected by inhibition of complex IV (CIV) or complex V (CV), severe hypoxia, and uncoupling. However, it was only moderately affected by inhibition of complexes I, II, and III. Cell death in MCF-7 cells induced by inhibition of complexes II, III, and IV was partially abrogated by aspartate. These findings indicate that OXPHOS activity and viability are not correlated in these cell lines, suggesting that the connection between OXPHOS and cancer cell survival is dependent on the specific cell type and conditions.
Collapse
Affiliation(s)
- Judit Doczi
- Institute of Biochemistry and Molecular Biology, Department of Biochemistry, Semmelweis University, Budapest, 1094, Hungary
| | - Noemi Karnok
- Institute of Biochemistry and Molecular Biology, Department of Biochemistry, Semmelweis University, Budapest, 1094, Hungary
| | - David Bui
- Institute of Biochemistry and Molecular Biology, Department of Biochemistry, Semmelweis University, Budapest, 1094, Hungary
| | - Victoria Azarov
- Institute of Biochemistry and Molecular Biology, Department of Biochemistry, Semmelweis University, Budapest, 1094, Hungary
| | - Gergely Pallag
- Institute of Biochemistry and Molecular Biology, Department of Biochemistry, Semmelweis University, Budapest, 1094, Hungary
| | - Sara Nazarian
- Institute of Biochemistry and Molecular Biology, Department of Biochemistry, Semmelweis University, Budapest, 1094, Hungary
| | - Bence Czumbel
- Institute of Biochemistry and Molecular Biology, Department of Biochemistry, Semmelweis University, Budapest, 1094, Hungary
| | | | - Christos Chinopoulos
- Institute of Biochemistry and Molecular Biology, Department of Biochemistry, Semmelweis University, Budapest, 1094, Hungary.
| |
Collapse
|
14
|
Yu W, Chen Y, Putluri N, Osman A, Coarfa C, Putluri V, Kamal AHM, Asmussen JK, Katsonis P, Myers JN, Lai SY, Lu W, Stephan CC, Powell RT, Johnson FM, Skinner HD, Kazi J, Ahmed KM, Hu L, Threet A, Meyer MD, Bankson JA, Wang T, Davis J, Parker KR, Harris MA, Baek ML, Echeverria GV, Qi X, Wang J, Frederick AI, Walsh AJ, Lichtarge O, Frederick MJ, Sandulache VC. Evolution of cisplatin resistance through coordinated metabolic reprogramming of the cellular reductive state. Br J Cancer 2023; 128:2013-2024. [PMID: 37012319 PMCID: PMC10205814 DOI: 10.1038/s41416-023-02253-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 03/16/2023] [Accepted: 03/21/2023] [Indexed: 04/05/2023] Open
Abstract
BACKGROUND Cisplatin (CDDP) is a mainstay treatment for advanced head and neck squamous cell carcinomas (HNSCC) despite a high frequency of innate and acquired resistance. We hypothesised that tumours acquire CDDP resistance through an enhanced reductive state dependent on metabolic rewiring. METHODS To validate this model and understand how an adaptive metabolic programme might be imprinted, we performed an integrated analysis of CDDP-resistant HNSCC clones from multiple genomic backgrounds by whole-exome sequencing, RNA-seq, mass spectrometry, steady state and flux metabolomics. RESULTS Inactivating KEAP1 mutations or reductions in KEAP1 RNA correlated with Nrf2 activation in CDDP-resistant cells, which functionally contributed to resistance. Proteomics identified elevation of downstream Nrf2 targets and the enrichment of enzymes involved in generation of biomass and reducing equivalents, metabolism of glucose, glutathione, NAD(P), and oxoacids. This was accompanied by biochemical and metabolic evidence of an enhanced reductive state dependent on coordinated glucose and glutamine catabolism, associated with reduced energy production and proliferation, despite normal mitochondrial structure and function. CONCLUSIONS Our analysis identified coordinated metabolic changes associated with CDDP resistance that may provide new therapeutic avenues through targeting of these convergent pathways.
Collapse
Affiliation(s)
- Wangie Yu
- Bobby R. Alford Department of Otolaryngology Head and Neck Surgery, Baylor College of Medicine, Houston, TX, USA
| | - Yunyun Chen
- Department of Head and Neck Surgery, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Nagireddy Putluri
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Abdullah Osman
- Department of Head and Neck Surgery, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Cristian Coarfa
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
- Center for Precision Environmental Health, Baylor College of Medicine, Houston, TX, USA
- Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Vasanta Putluri
- Advanced Technology core, Dan Duncan Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Abu H M Kamal
- Advanced Technology core, Dan Duncan Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Jennifer Kay Asmussen
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Panagiotis Katsonis
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Jeffrey N Myers
- Department of Head and Neck Surgery, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Stephen Y Lai
- Department of Head and Neck Surgery, University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Molecular and Cellular Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Wuhao Lu
- Bobby R. Alford Department of Otolaryngology Head and Neck Surgery, Baylor College of Medicine, Houston, TX, USA
| | - Clifford C Stephan
- Institute of Biosciences and Technology, Texas A&M Health Science Center, Houston, TX, USA
- Department of Translational Medical Sciences, School of Medicine, Texas A&M University, Houston, TX, USA
| | - Reid T Powell
- Institute of Biosciences and Technology, Texas A&M Health Science Center, Houston, TX, USA
- Department of Translational Medical Sciences, School of Medicine, Texas A&M University, Houston, TX, USA
| | - Faye M Johnson
- Department of Thoracic Head and Neck Medical Oncology, Division of Cancer Medicine, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Heath D Skinner
- Department of Radiation Oncology, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
| | - Jawad Kazi
- Bobby R. Alford Department of Otolaryngology Head and Neck Surgery, Baylor College of Medicine, Houston, TX, USA
| | - Kazi Mokim Ahmed
- Bobby R. Alford Department of Otolaryngology Head and Neck Surgery, Baylor College of Medicine, Houston, TX, USA
| | - Linghao Hu
- Biomedical Engineering, Texas A&M University, College Station, TX, USA
| | - Addison Threet
- Biomedical Engineering, Texas A&M University, College Station, TX, USA
| | - Matthew D Meyer
- Shared Equipment Authority, Rice University, Houston, TX, USA
| | - James A Bankson
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Tony Wang
- Bobby R. Alford Department of Otolaryngology Head and Neck Surgery, Baylor College of Medicine, Houston, TX, USA
| | - Jack Davis
- Bobby R. Alford Department of Otolaryngology Head and Neck Surgery, Baylor College of Medicine, Houston, TX, USA
| | - Kirby R Parker
- Bobby R. Alford Department of Otolaryngology Head and Neck Surgery, Baylor College of Medicine, Houston, TX, USA
| | - Madison A Harris
- Bobby R. Alford Department of Otolaryngology Head and Neck Surgery, Baylor College of Medicine, Houston, TX, USA
| | - Mokryun L Baek
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
- Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, USA
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Gloria V Echeverria
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
- Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, USA
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Xiaoli Qi
- Department of Pharmacology and Chemical Biology, Baylor College of Medicine, Houston, TX, USA
| | - Jin Wang
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
- Department of Pharmacology and Chemical Biology, Baylor College of Medicine, Houston, TX, USA
| | - Andy I Frederick
- School of Electrical and Computer Engineering Undergraduate Department, Cornell University, NY, USA
| | - Alex J Walsh
- Biomedical Engineering, Texas A&M University, College Station, TX, USA
| | - Olivier Lichtarge
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
- Department of Pharmacology and Chemical Biology, Baylor College of Medicine, Houston, TX, USA
- Department of Biochemistry & Molecular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX, USA
- Program in Quantitative and Computational Biosciences, Baylor College of Medicine, One Baylor Plaza, Houston, TX, USA
- Program in Integrative Molecular and Biomedical Sciences, Baylor College of Medicine, One Baylor Plaza, Houston, TX, USA
- Computational and Integrative Biomedical Research Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX, USA
| | - Mitchell J Frederick
- Bobby R. Alford Department of Otolaryngology Head and Neck Surgery, Baylor College of Medicine, Houston, TX, USA.
| | - Vlad C Sandulache
- Bobby R. Alford Department of Otolaryngology Head and Neck Surgery, Baylor College of Medicine, Houston, TX, USA.
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA.
- Center for Translational Research on Inflammatory Diseases, Michael E. DeBakey Veterans Affairs Medical Center, Houston, TX, USA.
| |
Collapse
|
15
|
Shah VS, Hou J, Vinarsky V, Xu J, Surve MV, Lin CP, Rajagopal J. Autofluorescence imaging permits label-free cell type assignment and reveals the dynamic formation of airway secretory cell associated antigen passages (SAPs). eLife 2023; 12:e84375. [PMID: 36994985 PMCID: PMC10154029 DOI: 10.7554/elife.84375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Accepted: 03/29/2023] [Indexed: 03/31/2023] Open
Abstract
The specific functional properties of a tissue are distributed amongst its component cell types. The various cells act coherently, as an ensemble, in order to execute a physiologic response. Modern approaches for identifying and dissecting novel physiologic mechanisms would benefit from an ability to identify specific cell types in live tissues that could then be imaged in real time. Current techniques require the use of fluorescent genetic reporters that are not only cumbersome, but which only allow the study of three or four cell types at a time. We report a non-invasive imaging modality that capitalizes on the endogenous autofluorescence signatures of the metabolic cofactors NAD(P)H and FAD. By marrying morphological characteristics with autofluorescence signatures, all seven of the airway epithelial cell types can be distinguished simultaneously in mouse tracheal explants in real time. Furthermore, we find that this methodology for direct cell type-specific identification avoids pitfalls associated with the use of ostensibly cell type-specific markers that are, in fact, altered by clinically relevant physiologic stimuli. Finally, we utilize this methodology to interrogate real-time physiology and identify dynamic secretory cell associated antigen passages (SAPs) that form in response to cholinergic stimulus. The identical process has been well documented in the intestine where the dynamic formation of SAPs and goblet cell associated antigen passages (GAPs) enable luminal antigen sampling. Airway secretory cells with SAPs are frequently juxtaposed to antigen presenting cells, suggesting that airway SAPs, like their intestinal counterparts, not only sample antigen but convey their cargo for immune cell processing.
Collapse
Affiliation(s)
- Viral S Shah
- Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, Massachusetts General HospitalBostonUnited States
- Center for Regenerative Medicine, Massachusetts General HospitalBostonUnited States
| | - Jue Hou
- Advanced Microscopy Program, Center for Systems Biology and Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical SchoolBostonUnited States
| | - Vladimir Vinarsky
- Center for Regenerative Medicine, Massachusetts General HospitalBostonUnited States
| | - Jiajie Xu
- Center for Regenerative Medicine, Massachusetts General HospitalBostonUnited States
| | - Manalee V Surve
- Center for Regenerative Medicine, Massachusetts General HospitalBostonUnited States
| | - Charles P Lin
- Advanced Microscopy Program, Center for Systems Biology and Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical SchoolBostonUnited States
| | - Jayaraj Rajagopal
- Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, Massachusetts General HospitalBostonUnited States
- Center for Regenerative Medicine, Massachusetts General HospitalBostonUnited States
- Harvard Stem Cell InstituteCambridgeUnited States
- Klarman Cell Observatory, Broad InstituteCambridgeUnited States
| |
Collapse
|
16
|
Karrobi K, Tank A, Fuzail MA, Kalidoss M, Tilbury K, Zaman M, Ferruzzi J, Roblyer D. Fluorescence Lifetime Imaging Microscopy (FLIM) reveals spatial-metabolic changes in 3D breast cancer spheroids. Sci Rep 2023; 13:3624. [PMID: 36869092 PMCID: PMC9984376 DOI: 10.1038/s41598-023-30403-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Accepted: 02/22/2023] [Indexed: 03/05/2023] Open
Abstract
Cancer cells are mechanically sensitive to physical properties of the microenvironment, which can affect downstream signaling to promote malignancy, in part through the modulation of metabolic pathways. Fluorescence Lifetime Imaging Microscopy (FLIM) can be used to measure the fluorescence lifetime of endogenous fluorophores, such as the metabolic co-factors NAD(P)H and FAD, in live samples. We used multiphoton FLIM to investigate the changes in cellular metabolism of 3D breast spheroids derived from MCF-10A and MD-MB-231 cell lines embedded in collagen with varying densities (1 vs. 4 mg/ml) over time (Day 0 vs. Day 3). MCF-10A spheroids demonstrated spatial gradients, with the cells closest to the spheroid edge exhibiting FLIM changes consistent with a shift towards oxidative phosphorylation (OXPHOS) while the spheroid core had changes consistent with a shift towards glycolysis. The MDA-MB-231 spheroids had a large shift consistent with increased OXPHOS with a more pronounced change at the higher collagen concentration. The MDA-MB-231 spheroids invaded into the collagen gel over time and cells that traveled the farthest had the largest changes consistent with a shift towards OXPHOS. Overall, these results suggest that the cells in contact with the extracellular matrix (ECM) and those that migrated the farthest had changes consistent with a metabolic shift towards OXPHOS. More generally, these results demonstrate the ability of multiphoton FLIM to characterize how spheroids metabolism and spatial metabolic gradients are modified by physical properties of the 3D ECM.
Collapse
Affiliation(s)
- Kavon Karrobi
- Department of Biomedical Engineering, Boston University, Boston, MA, 02215, USA
| | - Anup Tank
- Department of Biomedical Engineering, Boston University, Boston, MA, 02215, USA
| | | | - Madhumathi Kalidoss
- Department of Biomedical Engineering, Boston University, Boston, MA, 02215, USA
| | - Karissa Tilbury
- Department of Chemical and Biomedical Engineering, University of Maine, Orono, ME, 04469, USA
| | - Muhammad Zaman
- Department of Biomedical Engineering, Boston University, Boston, MA, 02215, USA
| | - Jacopo Ferruzzi
- Department of Biomedical Engineering, Boston University, Boston, MA, 02215, USA
- Department of Biomedical Engineering, The University of Texas at Dallas, Richardson, TX, 75080, USA
| | - Darren Roblyer
- Department of Biomedical Engineering, Boston University, Boston, MA, 02215, USA.
| |
Collapse
|
17
|
Filon MJ, Gillette AA, Yang B, Khemees TA, Skala MC, Jarrard DF. Prostate cancer cells demonstrate unique metabolism and substrate adaptability acutely after androgen deprivation therapy. Prostate 2022; 82:1547-1557. [PMID: 35980831 PMCID: PMC9804183 DOI: 10.1002/pros.24428] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 07/04/2022] [Indexed: 01/07/2023]
Abstract
BACKGROUND Androgen deprivation therapy (ADT) has been the standard of care for advanced hormone-sensitive prostate cancer (PC), yet tumors invariably develop resistance resulting in castrate-resistant PC. The acute response of cancer cells to ADT includes apoptosis and cell death, but a large fraction remains arrested but viable. In this study, we focused on intensively characterizing the early metabolic changes that result after ADT to define potential metabolic targets for treatment. METHODS A combination of mass spectrometry, optical metabolic imaging which noninvasively measures drug responses in cells, oxygen consumption rate, and protein expression analysis was used to characterize and block metabolic pathways over several days in multiple PC cell lines with variable hormone response status including ADT sensitive lines LNCaP and VCaP, and resistant C4-2 and DU145. RESULTS Mass spectrometry analysis of LNCaP pre- and postexposure to ADT revealed an abundance of glycolytic intermediates after ADT. In LNCaP and VCaP, a reduction in the optical redox ratio [NAD(P)H/FAD], extracellular acidification rate, and a downregulation of key regulatory enzymes for fatty acid and glutamine utilization was acutely observed after ADT. Screening several metabolic inhibitors revealed that blocking fatty acid oxidation and synthesis reversed this stress response in the optical redox ratio seen with ADT alone in LNCaP and VCaP. In contrast, both cell lines demonstrated increased sensitivity to the glycolytic inhibitor 2-Deoxy- d-glucose(2-DG) and maintained sensitivity to electron transport chain inhibitor Malonate after ADT exposure. ADT followed by 2-DG results in synergistic cell death, a result not seen with simultaneous administration. CONCLUSIONS Hormone-sensitive PC cells displayed altered metabolic profiles early after ADT including an overall depression in energy metabolism, induction of a quiescent/senescent phenotype, and sensitivity to selected metabolic inhibitors. Glycolytic blocking agents (e.g., 2-DG) as a sequential treatment after ADT may be promising.
Collapse
Affiliation(s)
- Mikolaj J. Filon
- Department of Urology, School of Medicine and Public HealthUniversity of WisconsinMadisonWisconsinUSA
| | - Amani A. Gillette
- Department of Biomedical EngineeringUniversity of WisconsinMadisonWisconsinUSA
- Morgridge Institute for ResearchMadisonWisconsinUSA
| | - Bing Yang
- Department of Urology, School of Medicine and Public HealthUniversity of WisconsinMadisonWisconsinUSA
| | - Tariq A. Khemees
- Department of Urology, School of Medicine and Public HealthUniversity of WisconsinMadisonWisconsinUSA
| | - Melissa C. Skala
- Department of Biomedical EngineeringUniversity of WisconsinMadisonWisconsinUSA
- Morgridge Institute for ResearchMadisonWisconsinUSA
- Carbone Comprehensive Cancer CenterUniversity of WisconsinMadisonWisconsinUSA
| | - David F. Jarrard
- Department of Urology, School of Medicine and Public HealthUniversity of WisconsinMadisonWisconsinUSA
- Carbone Comprehensive Cancer CenterUniversity of WisconsinMadisonWisconsinUSA
- Molecular and Environmental Toxicology ProgramUniversity of WisconsinMadisonWisconsinUSA
| |
Collapse
|
18
|
Walsh SK, Soni R, Arendt LM, Skala MC, Henak CR. Maturation- and degeneration-dependent articular cartilage metabolism via optical redox ratio imaging. J Orthop Res 2022; 40:1735-1743. [PMID: 34792214 DOI: 10.1002/jor.25214] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 09/24/2021] [Accepted: 11/09/2021] [Indexed: 02/04/2023]
Abstract
From the two metabolic processes in healthy cartilage, glycolysis has been associated with proliferation and oxidative phosphorylation (oxphos) with matrix synthesis. Recently, metabolic dysregulation was significantly correlated with cartilage degradation and osteoarthritis progression. While these findings suggest maturation predisposes cartilage to metabolic instability with consequences for tissue maintenance, these links have not been shown. Therefore, this study sought to address three hypotheses (a) chondrocytes exhibit differential metabolic activity between immaturity (0-4 months), adolescence (5-18 months), and maturity (>18 months); (b) perturbation of metabolic activity has consequences on expression of genes pertinent to cartilage tissue maintenance; and (c) severity of cartilage damage is positively correlated with glycolysis and oxphos activity as well as optical redox ratio in postadolescent cartilage. Porcine femoral cartilage samples from pigs (3 days to 6 years) underwent optical redox ratio imaging, which measures autofluorescence of NAD(P)H and FAD. Gene expression analysis and histological scoring was conducted for comparison against imaging metrics. NAD(P)H and FAD autofluorescence both demonstrated increasing intensity with age, while optical redox ratio was lowest in adolescent samples compared to immature or mature samples. Inhibition of glycolysis suppressed expression of Col2, Col1, ADAMTS4, and ADAMTS5, while oxphos inhibition had no effect. FAD fluorescence and optical redox ratio were positively correlated with histological degeneration. This study demonstrates maturation- and degeneration-dependent metabolic activity in cartilage and explores the consequences of this differential activity on gene expression. This study aids our basic understanding of cartilage biology and highlights opportunity for potential diagnostic applications.
Collapse
Affiliation(s)
- Shannon K Walsh
- Comparative Biomedical Sciences Program, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Rikin Soni
- Department of Mechanical Engineering, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Lisa M Arendt
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Melissa C Skala
- Morgridge Institute for Research, Madison, Wisconsin, USA.,Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Corinne R Henak
- Department of Mechanical Engineering, University of Wisconsin-Madison, Madison, Wisconsin, USA.,Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, Wisconsin, USA.,Department of Orthopedics and Rehabilitation, University of Wisconsin-Madison, Madison, Wisconsin, USA
| |
Collapse
|
19
|
Jacob A, Xu HN, Stout AL, Li LZ. Subcellular analysis of nuclear and cytoplasmic redox indices differentiates breast cancer cell subtypes better than nuclear-to-cytoplasmic area ratio. JOURNAL OF BIOMEDICAL OPTICS 2022; 27:JBO-210375GR. [PMID: 35945669 PMCID: PMC9360498 DOI: 10.1117/1.jbo.27.8.086001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 07/19/2022] [Indexed: 06/15/2023]
Abstract
SIGNIFICANCE Stratification of malignancy is valuable for cancer treatment. Both optical redox imaging (ORI) indices and nuclear-to-cytoplasmic volume/area ratio (N:C ratio) have been investigated to differentiate between cancers with varying aggressiveness, but these two methods have not been directly compared. The redox status in the cell nucleus has not been studied by ORI, and it remains unknown whether nuclear ORI indices add new biological information. AIM We sought to compare the capacity of whole-cell and subcellular ORI indices and N:C ratio to differentiate between breast cancer subtypes with varying aggressiveness and between mitotic and nonmitotic cells. APPROACH ORI indices for whole cell, cytoplasm, and nucleus as well as the N:C area ratio were generated for two triple-negative (more aggressive) and two receptor-positive (less aggressive) breast cancer cell lines by fluorescence microscopy. RESULTS We found positive correlations between nuclear and cytoplasmic ORI indices within individual cells. On average, a nuclear redox status was found to be more oxidized than cytoplasm in triple-negative cells but not in receptor-positive cells. Whole-cell and subcellular ORI indices distinguished between the receptor statuses better than the N:C ratio. However, N:C ratio was a better differentiator between nonmitotic and mitotic triple-negative cells. CONCLUSIONS Subcellular ORI analysis differentiates breast cancer subtypes with varying aggressiveness better than N:C area ratio.
Collapse
Affiliation(s)
- Annemarie Jacob
- University of Pennsylvania, Perelman School of Medicine, Department of Radiology, Britton Chance Laboratory of Redox Imaging, Philadelphia, Pennsylvania, United States
| | - He N. Xu
- University of Pennsylvania, Perelman School of Medicine, Department of Radiology, Britton Chance Laboratory of Redox Imaging, Philadelphia, Pennsylvania, United States
- University of Pennsylvania, Perelman School of Medicine, Institute of Translational Medicine and Therapeutics, Philadelphia, Pennsylvania, United States
| | - Andrea L. Stout
- University of Pennsylvania, Perelman School of Medicine, Department of Cell and Developmental Biology, Philadelphia, Pennsylvania, United States
| | - Lin Z. Li
- University of Pennsylvania, Perelman School of Medicine, Department of Radiology, Britton Chance Laboratory of Redox Imaging, Philadelphia, Pennsylvania, United States
- University of Pennsylvania, Perelman School of Medicine, Institute of Translational Medicine and Therapeutics, Philadelphia, Pennsylvania, United States
| |
Collapse
|
20
|
Gillette AA, DeStefanis RA, Pritzl SL, Deming DA, Skala MC. Inhibition of B-cell lymphoma 2 family proteins alters optical redox ratio, mitochondrial polarization, and cell energetics independent of cell state. JOURNAL OF BIOMEDICAL OPTICS 2022; 27:JBO-210354GR. [PMID: 35643815 PMCID: PMC9142839 DOI: 10.1117/1.jbo.27.5.056505] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 05/09/2022] [Indexed: 05/27/2023]
Abstract
SIGNIFICANCE The optical redox ratio (ORR) [autofluorescence intensity of the reduced form of nicotinamide adenine dinucleotide (phosphate) (NAD(P)H)/flavin adenine dinucleotide (FAD)] provides a label-free method to quantify cellular metabolism. However, it is unclear whether changes in the ORR with B-cell lymphoma 2 (Bcl-2) family protein inhibition are due to metabolic stress alone or compromised cell viability. AIM Determine whether ABT-263 (navitoclax, Bcl-2 family inhibitor) changes the ORR due to changes in mitochondrial function that are independent of changes in cell viability. APPROACH SW48 colon cancer cells were used to investigate changes in ORR, mitochondrial membrane potential, oxygen consumption rates, and cell state (cell growth, viability, proliferation, apoptosis, autophagy, and senescence) with ABT-263, TAK-228 [sapanisertib, mammalian target of rapamycin complex 1/2 (mTORC 1/2) inhibitor], and their combination at 24 h. RESULTS Changes in the ORR with Bcl-2 inhibition are driven by increases in both NAD(P)H and FAD autofluorescence, corresponding with increased basal metabolic rate and increased mitochondrial polarization. ABT-263 treatment does not change cell viability or induce autophagy but does induce a senescent phenotype. The metabolic changes seen with ABT-263 treatment are mitigated by combination with mTORC1/2 inhibition. CONCLUSIONS The ORR is sensitive to increases in mitochondrial polarization, energetic state, and cell senescence, which can change independently from cell viability.
Collapse
Affiliation(s)
- Amani A. Gillette
- University of Wisconsin, Department of Biomedical Engineering, Madison, Wisconsin, United States
| | - Rebecca A. DeStefanis
- University of Wisconsin, McArdle Laboratory for Cancer Research, Department of Oncology, Madison, Wisconsin, United States
| | - Stephanie L. Pritzl
- University of Wisconsin, Division of Hematology, Oncology and Palliative Care, Department of Medicine, Madison, Wisconsin, United States
| | - Dustin A. Deming
- University of Wisconsin, McArdle Laboratory for Cancer Research, Department of Oncology, Madison, Wisconsin, United States
- University of Wisconsin, Division of Hematology, Oncology and Palliative Care, Department of Medicine, Madison, Wisconsin, United States
- University of Wisconsin Carbone Cancer Center, Madison, Wisconsin, United States
| | - Melissa C. Skala
- University of Wisconsin, Department of Biomedical Engineering, Madison, Wisconsin, United States
- Morgridge Institute for Research, Madison, Wisconsin, United States
| |
Collapse
|
21
|
Zhang WQ, Sorvina A, Morrison JL, Darby JRT, Brooks DA, Plush SE, Afshar Vahid S. Development of an optical fiber-based redox monitoring system for tissue metabolism. JOURNAL OF BIOPHOTONICS 2022; 15:e202100304. [PMID: 35038239 DOI: 10.1002/jbio.202100304] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 01/11/2022] [Accepted: 01/12/2022] [Indexed: 06/14/2023]
Abstract
An optical redox ratio can potentially be used to report on the dynamics of cell and tissue metabolism and define altered metabolic conditions for different pathologies. While there are methods to measure the optical redox ratio, they are not particularly suited to real-time in situ or in vivo analysis. Here, we have developed a fiber-optic system to measure redox ratios in cells and tissues and two mathematical models to enable real-time, in vivo redox measurements. The optical redox ratios in tissue explants are correlated directly with endogenous NADH/FAD fluorescence emissions. We apply the mathematical models to the two-photon microscopy data and show consistent results. We also used our fiber-optic system to measure redox in different tissues and show consistent results between the two models, hence demonstrating proof-of-principle. This innovative redox monitoring system will have practical applications for defining different metabolic disease states.
Collapse
Affiliation(s)
- Wen Qi Zhang
- Laser Physics and Photonic Devices Laboratories, School of Engineering, University of South Australia, Adelaide, South Australia, Australia
| | - Alexandra Sorvina
- Clinical and Health Sciences, Sansom Institute for Health Research, University of South Australia, Adelaide, South Australia, Australia
| | - Janna L Morrison
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, UniSA: Clinical and Health Sciences, University of South Australia, Adelaide, South Australia, Australia
| | - Jack R T Darby
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, UniSA: Clinical and Health Sciences, University of South Australia, Adelaide, South Australia, Australia
| | - Doug A Brooks
- Clinical and Health Sciences, Sansom Institute for Health Research, University of South Australia, Adelaide, South Australia, Australia
| | - Sally E Plush
- Clinical and Health Sciences, Sansom Institute for Health Research, University of South Australia, Adelaide, South Australia, Australia
| | - Shahraam Afshar Vahid
- Laser Physics and Photonic Devices Laboratories, School of Engineering, University of South Australia, Adelaide, South Australia, Australia
| |
Collapse
|
22
|
Shirmanova MV, Gavrina AI, Kovaleva TF, Dudenkova VV, Zelenova EE, Shcheslavskiy VI, Mozherov AM, Snopova LB, Lukyanov KA, Zagaynova EV. Insight into redox regulation of apoptosis in cancer cells with multiparametric live-cell microscopy. Sci Rep 2022; 12:4476. [PMID: 35296739 PMCID: PMC8927414 DOI: 10.1038/s41598-022-08509-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 02/25/2022] [Indexed: 02/07/2023] Open
Abstract
Cellular redox status and the level of reactive oxygen species (ROS) are important regulators of apoptotic potential, playing a crucial role in the growth of cancer cell and their resistance to apoptosis. However, the relationships between the redox status and ROS production during apoptosis remain poorly explored. In this study, we present an investigation on the correlations between the production of ROS, the redox ratio FAD/NAD(P)H, the proportions of the reduced nicotinamide cofactors NADH and NADPH, and caspase-3 activity in cancer cells at the level of individual cells. Two-photon excitation fluorescence lifetime imaging microscopy (FLIM) was applied to monitor simultaneously apoptosis using the genetically encoded sensor of caspase-3, mKate2-DEVD-iRFP, and the autofluorescence of redox cofactors in colorectal cancer cells upon stimulation of apoptosis with staurosporine, cisplatin or hydrogen peroxide. We found that, irrespective of the apoptotic stimulus used, ROS accumulation correlated well with both the elevated pool of mitochondrial, enzyme-bound NADH and caspase-3 activation. Meanwhile, a shift in the contribution of bound NADH could develop independently of the apoptosis, and this was observed in the case of cisplatin. An increase in the proportion of bound NADPH was detected only in staurosporine-treated cells, this likely being associated with a high level of ROS production and their resulting detoxification. The results of the study favor the discovery of new therapeutic strategies based on manipulation of the cellular redox balance, which could help improve the anti-tumor activity of drugs and overcome apoptotic resistance.
Collapse
Affiliation(s)
- Marina V Shirmanova
- Privolzhsky Research Medical University, Minin and Pozharsky Sq. 10/1, 603005, Nizhny Novgorod, Russia.
| | - Alena I Gavrina
- Privolzhsky Research Medical University, Minin and Pozharsky Sq. 10/1, 603005, Nizhny Novgorod, Russia
| | - Tatiana F Kovaleva
- Privolzhsky Research Medical University, Minin and Pozharsky Sq. 10/1, 603005, Nizhny Novgorod, Russia
| | - Varvara V Dudenkova
- Privolzhsky Research Medical University, Minin and Pozharsky Sq. 10/1, 603005, Nizhny Novgorod, Russia
| | - Ekaterina E Zelenova
- National Medical Research Radiological Centre of the Ministry of Health of the Russian Federation, 2nd Botkinsky proezd, 3, Moscow, Russia, 125284
| | - Vladislav I Shcheslavskiy
- Privolzhsky Research Medical University, Minin and Pozharsky Sq. 10/1, 603005, Nizhny Novgorod, Russia.,Becker&Hickl GmbH, Nunsdorfer Ring 7-9, 12277, Berlin, Germany
| | - Artem M Mozherov
- Privolzhsky Research Medical University, Minin and Pozharsky Sq. 10/1, 603005, Nizhny Novgorod, Russia
| | - Ludmila B Snopova
- Privolzhsky Research Medical University, Minin and Pozharsky Sq. 10/1, 603005, Nizhny Novgorod, Russia
| | - Konstantin A Lukyanov
- Skolkovo Institute of Science and Technology, Bolshoy Boulevard 30, bld. 1, Moscow, Russia, 121205
| | - Elena V Zagaynova
- Privolzhsky Research Medical University, Minin and Pozharsky Sq. 10/1, 603005, Nizhny Novgorod, Russia.,Lobachevsky State University of Nizhny Novgorod, Gagarin Avenue 23, Nizhny Novgorod, Russia, 603950
| |
Collapse
|
23
|
Duraj T, Carrión-Navarro J, Seyfried TN, García-Romero N, Ayuso-Sacido A. Metabolic therapy and bioenergetic analysis: The missing piece of the puzzle. Mol Metab 2021; 54:101389. [PMID: 34749013 PMCID: PMC8637646 DOI: 10.1016/j.molmet.2021.101389] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 10/29/2021] [Accepted: 11/01/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Aberrant metabolism is recognized as a hallmark of cancer, a pillar necessary for cellular proliferation. Regarding bioenergetics (ATP generation), most cancers display a preference not only toward aerobic glycolysis ("Warburg effect") and glutaminolysis (mitochondrial substrate level-phosphorylation) but also toward other metabolites such as lactate, pyruvate, and fat-derived sources. These secondary metabolites can assist in proliferation but cannot fully cover ATP demands. SCOPE OF REVIEW The concept of a static metabolic profile is challenged by instances of heterogeneity and flexibility to meet fuel/anaplerotic demands. Although metabolic therapies are a promising tool to improve therapeutic outcomes, either via pharmacological targets or press-pulse interventions, metabolic plasticity is rarely considered. Lack of bioenergetic analysis in vitro and patient-derived models is hindering translational potential. Here, we review the bioenergetics of cancer and propose a simple analysis of major metabolic pathways, encompassing both affordable and advanced techniques. A comprehensive compendium of Seahorse XF bioenergetic measurements is presented for the first time. MAJOR CONCLUSIONS Standardization of principal readouts might help researchers to collect a complete metabolic picture of cancer using the most appropriate methods depending on the sample of interest.
Collapse
Affiliation(s)
- Tomás Duraj
- Faculty of Medicine, Institute for Applied Molecular Medicine (IMMA), CEU San Pablo University, 28668, Madrid, Spain.
| | - Josefa Carrión-Navarro
- Faculty of Experimental Sciences, Universidad Francisco de Vitoria, 28223, Madrid, Spain; Brain Tumor Laboratory, Fundación Vithas, Grupo Hospitales Vithas, 28043, Madrid, Spain.
| | - Thomas N Seyfried
- Biology Department, Boston College, 140 Commonwealth Ave, Chestnut Hill, MA, 02467, USA.
| | - Noemí García-Romero
- Faculty of Experimental Sciences, Universidad Francisco de Vitoria, 28223, Madrid, Spain; Brain Tumor Laboratory, Fundación Vithas, Grupo Hospitales Vithas, 28043, Madrid, Spain.
| | - Angel Ayuso-Sacido
- Faculty of Experimental Sciences, Universidad Francisco de Vitoria, 28223, Madrid, Spain; Brain Tumor Laboratory, Fundación Vithas, Grupo Hospitales Vithas, 28043, Madrid, Spain; Faculty of Medicine, Universidad Francisco de Vitoria, 28223, Madrid, Spain.
| |
Collapse
|
24
|
Ratiometric two-photon fluorescence probes for sensing, imaging and biomedicine applications at living cell and small animal levels. Coord Chem Rev 2021. [DOI: 10.1016/j.ccr.2021.214114] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
25
|
Ansari F, Yoval-Sánchez B, Niatsetskaya Z, Sosunov S, Stepanova A, Garcia C, Owusu-Ansah E, Ten V, Wittig I, Galkin A. Quantification of NADH:ubiquinone oxidoreductase (complex I) content in biological samples. J Biol Chem 2021; 297:101204. [PMID: 34543622 PMCID: PMC8503622 DOI: 10.1016/j.jbc.2021.101204] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 08/26/2021] [Accepted: 08/31/2021] [Indexed: 12/14/2022] Open
Abstract
Impairments in mitochondrial energy metabolism have been implicated in human genetic diseases associated with mitochondrial and nuclear DNA mutations, neurodegenerative and cardiovascular disorders, diabetes, and aging. Alteration in mitochondrial complex I structure and activity has been shown to play a key role in Parkinson's disease and ischemia/reperfusion tissue injury, but significant difficulty remains in assessing the content of this enzyme complex in a given sample. The present study introduces a new method utilizing native polyacrylamide gel electrophoresis in combination with flavin fluorescence scanning to measure the absolute content of complex I, as well as α-ketoglutarate dehydrogenase complex, in any preparation. We show that complex I content is 19 ± 1 pmol/mg of protein in the brain mitochondria, whereas varies up to 10-fold in different mouse tissues. Together with the measurements of NADH-dependent specific activity, our method also allows accurate determination of complex I catalytic turnover, which was calculated as 104 min-1 for NADH:ubiquinone reductase in mouse brain mitochondrial preparations. α-ketoglutarate dehydrogenase complex content was determined to be 65 ± 5 and 123 ± 9 pmol/mg protein for mouse brain and bovine heart mitochondria, respectively. Our approach can also be extended to cultured cells, and we demonstrated that about 90 × 103 complex I molecules are present in a single human embryonic kidney 293 cell. The ability to determine complex I content should provide a valuable tool to investigate the enzyme status in samples after in vivo treatment in mutant organisms, cells in culture, or human biopsies.
Collapse
Affiliation(s)
- Fariha Ansari
- Division of Neonatology, Department of Pediatrics, Columbia University Medical Center, New York, New York, USA
| | - Belem Yoval-Sánchez
- Division of Neonatology, Department of Pediatrics, Columbia University Medical Center, New York, New York, USA
| | - Zoya Niatsetskaya
- Division of Neonatology, Department of Pediatrics, Columbia University Medical Center, New York, New York, USA
| | - Sergey Sosunov
- Division of Neonatology, Department of Pediatrics, Columbia University Medical Center, New York, New York, USA
| | - Anna Stepanova
- Division of Neonatology, Department of Pediatrics, Columbia University Medical Center, New York, New York, USA
| | - Christian Garcia
- Department of Physiology & Cellular Biophysics, Columbia University, New York, New York, USA
| | - Edward Owusu-Ansah
- Department of Physiology & Cellular Biophysics, Columbia University, New York, New York, USA
| | - Vadim Ten
- Division of Neonatology, Department of Pediatrics, Columbia University Medical Center, New York, New York, USA
| | - Ilka Wittig
- Functional Proteomics, Institute of Cardiovascular Physiology, Goethe University, Frankfurt am Main, Germany; German Center for Cardiovascular Research (DZHK), Partner site RheinMain, Frankfurt am Main, Germany
| | - Alexander Galkin
- Division of Neonatology, Department of Pediatrics, Columbia University Medical Center, New York, New York, USA; Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, New York, USA.
| |
Collapse
|
26
|
Label-free two-photon imaging of mitochondrial activity in murine macrophages stimulated with bacterial and viral ligands. Sci Rep 2021; 11:14081. [PMID: 34234166 PMCID: PMC8263786 DOI: 10.1038/s41598-021-93043-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2021] [Accepted: 06/16/2021] [Indexed: 11/12/2022] Open
Abstract
Mitochondria are the metabolic hub of the cell, playing a central role in regulating immune responses. Dysfunction of mitochondrial reprogramming can occur during bacterial and viral infections compromising hosts’ immune signaling. Comparative evaluation of these alterations in response to bacterial and viral ligands can provide insights into a cell’s ability to mount pathogen-specific responses. In this study, we used two-photon excitation fluorescence (TPEF) imaging to quantify reduced nicotinamide adenine dinucleotide phosphate (NAD(P)H) and flavin adenine dinucleotide (FAD) levels in the cell and to calculate the optical redox ratio (ORR), an indicator of mitochondrial dysfunction. Analyses were performed on RAW264.7 cells and murine bone marrow derived macrophages (BMM) stimulated with bacterial (LPS) and viral (Poly(I:C)) ligands. Responses were cell type dependent, with primary cells having significantly higher levels of FAD and higher oxygen consumption rates suggesting BMM may be more dependent on mitochondrial metabolism. Our findings also suggest that FAD-TPEF intensity may be a better predictor of mitochondrial activity and localization since it demonstrates unique mitochondrial clustering patterns in LPS vs. Poly(I:C) stimulated macrophages. Collectively, we demonstrate that TPEF imaging is a powerful label-free approach for quantifying changes in mitochondrial function and organization in macrophages following bacterial and viral stimuli.
Collapse
|
27
|
Martirosian V, Deshpande K, Zhou H, Shen K, Smith K, Northcott P, Lin M, Stepanosyan V, Das D, Remsik J, Isakov D, Boire A, De Feyter H, Hurth K, Li S, Wiemels J, Nakamura B, Shao L, Danilov C, Chen T, Neman J. Medulloblastoma uses GABA transaminase to survive in the cerebrospinal fluid microenvironment and promote leptomeningeal dissemination. Cell Rep 2021; 35:109302. [PMID: 34192534 PMCID: PMC8848833 DOI: 10.1016/j.celrep.2021.109302] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 10/02/2020] [Accepted: 06/03/2021] [Indexed: 12/12/2022] Open
Abstract
Medulloblastoma (MB) is a malignant pediatric brain tumor arising in the cerebellum. Although abnormal GABAergic receptor activation has been described in MB, studies have not yet elucidated the contribution of receptor-independent GABA metabolism to MB pathogenesis. We find primary MB tumors globally display decreased expression of GABA transaminase (ABAT), the protein responsible for GABA metabolism, compared with normal cerebellum. However, less aggressive WNT and SHH subtypes express higher ABAT levels compared with metastatic G3 and G4 tumors. We show that elevated ABAT expression results in increased GABA catabolism, decreased tumor cell proliferation, and induction of metabolic and histone characteristics mirroring GABAergic neurons. Our studies suggest ABAT expression fluctuates depending on metabolite changes in the tumor microenvironment, with nutrient-poor conditions upregulating ABAT expression. We find metastatic MB cells require ABAT to maintain viability in the metabolite-scarce cerebrospinal fluid by using GABA as an energy source substitute, thereby facilitating leptomeningeal metastasis formation.
Collapse
Affiliation(s)
- Vahan Martirosian
- Department of Neurological Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA; USC Brain Tumor Center, University of Southern California, Los Angeles, CA 90089, USA
| | - Krutika Deshpande
- Department of Neurological Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA; USC Brain Tumor Center, University of Southern California, Los Angeles, CA 90089, USA
| | - Hao Zhou
- Department of Biomedical Engineering, Viterbi School of Engineering, University of Southern California, Los Angeles, CA 90089, USA
| | - Keyue Shen
- Department of Biomedical Engineering, Viterbi School of Engineering, University of Southern California, Los Angeles, CA 90089, USA; Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA 90089, USA
| | - Kyle Smith
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Paul Northcott
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Michelle Lin
- Department of Neurological Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
| | - Vazgen Stepanosyan
- Department of Neurological Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
| | - Diganta Das
- Department of Neurological Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
| | - Jan Remsik
- Human Oncology and Pathogenesis Program, Department of Neuro-Oncology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Danielle Isakov
- Human Oncology and Pathogenesis Program, Department of Neuro-Oncology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Adrienne Boire
- Human Oncology and Pathogenesis Program, Department of Neuro-Oncology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Henk De Feyter
- Magnetic Resonance Research Center, Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Kyle Hurth
- Department of Pathology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA; USC Brain Tumor Center, University of Southern California, Los Angeles, CA 90089, USA
| | - Shaobo Li
- Center for Genetic Epidemiology, Department of Preventative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
| | - Joseph Wiemels
- Center for Genetic Epidemiology, Department of Preventative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA; Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA 90089, USA
| | - Brooke Nakamura
- Division of Gastrointestinal and Liver Diseases, Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
| | - Ling Shao
- Division of Gastrointestinal and Liver Diseases, Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA; Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA 90089, USA
| | - Camelia Danilov
- Department of Neurological Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
| | - Thomas Chen
- Department of Neurological Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA; USC Brain Tumor Center, University of Southern California, Los Angeles, CA 90089, USA
| | - Josh Neman
- Department of Neurological Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA; Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA 90089, USA; USC Brain Tumor Center, University of Southern California, Los Angeles, CA 90089, USA.
| |
Collapse
|
28
|
Gillette AA, Babiarz CP, VanDommelen AR, Pasch CA, Clipson L, Matkowskyj KA, Deming DA, Skala MC. Autofluorescence Imaging of Treatment Response in Neuroendocrine Tumor Organoids. Cancers (Basel) 2021; 13:cancers13081873. [PMID: 33919802 PMCID: PMC8070804 DOI: 10.3390/cancers13081873] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 03/30/2021] [Accepted: 04/08/2021] [Indexed: 12/30/2022] Open
Abstract
Gastroenteropancreatic neuroendocrine tumors (GEP-NET) account for roughly 60% of all neuroendocrine tumors. Low/intermediate grade human GEP-NETs have relatively low proliferation rates that animal models and cell lines fail to recapitulate. Short-term patient-derived cancer organoids (PDCOs) are a 3D model system that holds great promise for recapitulating well-differentiated human GEP-NETs. However, traditional measurements of drug response (i.e., growth, proliferation) are not effective in GEP-NET PDCOs due to the small volume of tissue and low proliferation rates that are characteristic of the disease. Here, we test a label-free, non-destructive optical metabolic imaging (OMI) method to measure drug response in live GEP-NET PDCOs. OMI captures the fluorescence lifetime and intensity of endogenous metabolic cofactors NAD(P)H and FAD. OMI has previously provided accurate predictions of drug response on a single cell level in other cancer types, but this is the first study to apply OMI to GEP-NETs. OMI tested the response to novel drug combination on GEP-NET PDCOs, specifically ABT263 (navitoclax), a Bcl-2 family inhibitor, and everolimus, a standard GEP-NET treatment that inhibits mTOR. Treatment response to ABT263, everolimus, and the combination were tested in GEP-NET PDCO lines derived from seven patients, using two-photon OMI. OMI measured a response to the combination treatment in 5 PDCO lines, at 72 h post-treatment. In one of the non-responsive PDCO lines, heterogeneous response was identified with two distinct subpopulations of cell metabolism. Overall, this work shows that OMI provides single-cell metabolic measurements of drug response in PDCOs to guide drug development for GEP-NET patients.
Collapse
Affiliation(s)
- Amani A. Gillette
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI 53706, USA;
| | - Christopher P. Babiarz
- Department of Medicine, Division of Hematology, Oncology and Palliative Care, School of Medicine and Public Health, University of Wisconsin, Madison, WI 53705, USA;
| | | | - Cheri A. Pasch
- University of Wisconsin Carbone Cancer Center, Madison, WI 53705, USA; (C.A.P.); (K.A.M.)
| | - Linda Clipson
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin, Madison, WI 53705, USA;
| | - Kristina A. Matkowskyj
- University of Wisconsin Carbone Cancer Center, Madison, WI 53705, USA; (C.A.P.); (K.A.M.)
- Department of Pathology and Laboratory Medicine, University of Wisconsin, Madison, WI 53705, USA
| | - Dustin A. Deming
- Department of Medicine, Division of Hematology, Oncology and Palliative Care, School of Medicine and Public Health, University of Wisconsin, Madison, WI 53705, USA;
- University of Wisconsin Carbone Cancer Center, Madison, WI 53705, USA; (C.A.P.); (K.A.M.)
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin, Madison, WI 53705, USA;
- Correspondence: (D.A.D.); (M.C.S.)
| | - Melissa C. Skala
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI 53706, USA;
- Morgridge Institute for Research, Madison, WI 53715, USA;
- University of Wisconsin Carbone Cancer Center, Madison, WI 53705, USA; (C.A.P.); (K.A.M.)
- Correspondence: (D.A.D.); (M.C.S.)
| |
Collapse
|
29
|
Zhang X, Mariano CF, Ando Y, Shen K. Bioengineering tools for probing intracellular events in T lymphocytes. WIREs Mech Dis 2020; 13:e1510. [PMID: 33073545 DOI: 10.1002/wsbm.1510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 09/14/2020] [Accepted: 09/16/2020] [Indexed: 11/11/2022]
Abstract
T lymphocytes are the central coordinator and executor of many immune functions. The activation and function of T lymphocytes are mediated through the engagement of cell surface receptors and regulated by a myriad of intracellular signaling network. Bioengineering tools, including imaging modalities and fluorescent probes, have been developed and employed to elucidate the cellular events throughout the functional lifespan of T cells. A better understanding of these events can broaden our knowledge in the immune systems biology, as well as accelerate the development of effective diagnostics and immunotherapies. Here we review the commonly used and recently developed techniques and probes for monitoring T lymphocyte intracellular events, following the order of intracellular events in T cells from activation, signaling, metabolism to apoptosis. The techniques introduced here can be broadly applied to other immune cells and cell systems. This article is categorized under: Immune System Diseases > Molecular and Cellular Physiology Immune System Diseases > Biomedical Engineering Infectious Diseases > Biomedical Engineering.
Collapse
Affiliation(s)
- Xinyuan Zhang
- Department of Biomedical Engineering, University of Southern California, Los Angeles, California, USA
| | - Chelsea F Mariano
- Department of Biomedical Engineering, University of Southern California, Los Angeles, California, USA
| | - Yuta Ando
- Department of Biomedical Engineering, University of Southern California, Los Angeles, California, USA
| | - Keyue Shen
- Department of Biomedical Engineering, University of Southern California, Los Angeles, California, USA.,Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, California, USA.,USC Stem Cell, University of Southern California, Los Angeles, California, USA
| |
Collapse
|
30
|
Xu HN, Lin Z, Gandhi CK, Amatya S, Wang Y, Li LZ, Floros J. Sex and SP-A2 Dependent NAD(H) Redox Alterations in Mouse Alveolar Macrophages in Response to Ozone Exposure: Potential Implications for COVID-19. Antioxidants (Basel) 2020; 9:antiox9100915. [PMID: 32992843 PMCID: PMC7601279 DOI: 10.3390/antiox9100915] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 09/08/2020] [Accepted: 09/19/2020] [Indexed: 12/20/2022] Open
Abstract
Co-enzyme nicotinamide adenine dinucleotide (NAD(H)) redox plays a key role in macrophage function. Surfactant protein (SP-) A modulates the functions of alveolar macrophages (AM) and ozone (O3) exposure in the presence or absence of SP-A and reduces mouse survival in a sex-dependent manner. It is unclear whether and how NAD(H) redox status plays a role in the innate immune response in a sex-dependent manner. We investigated the NAD(H) redox status of AM from SP-A2 and SP-A knockout (KO) mice in response to O3 or filtered air (control) exposure using optical redox imaging technique. We found: (i) In SP-A2 mice, the redox alteration of AM in response to O3 showed sex-dependence with AM from males being significantly more oxidized and having a higher level of mitochondrial reactive oxygen species than females; (ii) AM from KO mice were more oxidized after O3 exposure and showed no sex differences; (iii) AM from female KO mice were more oxidized than female SP-A2 mice; and (iv) Two distinct subpopulations characterized by size and redox status were observed in a mouse AM sample. In conclusions, the NAD(H) redox balance in AM responds to O3 in a sex-dependent manner and the innate immune molecule, SP-A2, contributes to this observed sex-specific redox response.
Collapse
Affiliation(s)
- He N. Xu
- Britton Chance Laboratory of Redox Imaging, Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (H.N.X.); (Z.L.)
| | - Zhenwu Lin
- Britton Chance Laboratory of Redox Imaging, Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (H.N.X.); (Z.L.)
| | - Chintan K. Gandhi
- Department of Pediatrics, Center for Host Defense, Inflammation, and Lung Disease, College of Medicine, The Pennsylvania State University, Hershey, PA 17033, USA; (C.K.G.); (S.A.); (Y.W.)
| | - Shaili Amatya
- Department of Pediatrics, Center for Host Defense, Inflammation, and Lung Disease, College of Medicine, The Pennsylvania State University, Hershey, PA 17033, USA; (C.K.G.); (S.A.); (Y.W.)
| | - Yunhua Wang
- Department of Pediatrics, Center for Host Defense, Inflammation, and Lung Disease, College of Medicine, The Pennsylvania State University, Hershey, PA 17033, USA; (C.K.G.); (S.A.); (Y.W.)
| | - Lin Z. Li
- Britton Chance Laboratory of Redox Imaging, Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (H.N.X.); (Z.L.)
- Correspondence: (L.Z.L.); (J.F.)
| | - Joanna Floros
- Departments of Pediatric and Obstetrics and Gynecology, College of Medicine, The Pennsylvania State University, Hershey, PA 17033, USA
- Correspondence: (L.Z.L.); (J.F.)
| |
Collapse
|
31
|
Walsh AJ, Mueller KP, Tweed K, Jones I, Walsh CM, Piscopo NJ, Niemi NM, Pagliarini DJ, Saha K, Skala MC. Classification of T-cell activation via autofluorescence lifetime imaging. Nat Biomed Eng 2020; 5:77-88. [PMID: 32719514 PMCID: PMC7854821 DOI: 10.1038/s41551-020-0592-z] [Citation(s) in RCA: 103] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Accepted: 06/24/2020] [Indexed: 01/20/2023]
Abstract
The function of a T cell depends on its subtype and activation state. Here, we show that the imaging of autofluorescence-lifetime signals from quiescent and activated T cells can be used to classify the cells. T cells isolated from human peripheral blood and activated in culture via a tetrameric antibody against the surface ligands CD2, CD3 and CD28 showed specific activation-state-dependent patterns of autofluorescence lifetime. Logistic-regression models and random-forest models classified T cells according to activation state with 97–99% accuracy, and according to activation state (quiescent or activated) and subtype (CD3+ CD8+ or CD3+ CD4+) with 97% accuracy. Autofluorescence-lifetime imaging could be used to non-destructively determine T-cell function.
Collapse
Affiliation(s)
- Alex J Walsh
- Morgridge Institute for Research, Madison, WI, USA. .,Department of Biomedical Engineering, Texas A&M University, College Station, TX, USA.
| | - Katherine P Mueller
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI, USA.,Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, USA
| | - Kelsey Tweed
- Morgridge Institute for Research, Madison, WI, USA.,Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, USA
| | - Isabel Jones
- Morgridge Institute for Research, Madison, WI, USA
| | - Christine M Walsh
- Morgridge Institute for Research, Madison, WI, USA.,Department of Sociology, State University of New York, Albany, NY, USA
| | - Nicole J Piscopo
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI, USA.,Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, USA
| | - Natalie M Niemi
- Morgridge Institute for Research, Madison, WI, USA.,Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, USA
| | - David J Pagliarini
- Morgridge Institute for Research, Madison, WI, USA.,Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, USA.,Departments of Cell Biology and Physiology, Biochemistry and Molecular Biophysics, and Genetics, Washington University School of Medicine, St Louis, MO, USA
| | - Krishanu Saha
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI, USA.,Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, USA
| | - Melissa C Skala
- Morgridge Institute for Research, Madison, WI, USA. .,Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, USA.
| |
Collapse
|
32
|
Barkauskas DS, Medley G, Liang X, Mohammed YH, Thorling CA, Wang H, Roberts MS. Using in vivo multiphoton fluorescence lifetime imaging to unravel disease-specific changes in the liver redox state. Methods Appl Fluoresc 2020; 8:034003. [PMID: 32422610 DOI: 10.1088/2050-6120/ab93de] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Multiphoton fluorescence lifetime microscopy has revolutionized studies of pathophysiological and xenobiotic dynamics, enabling the spatial and temporal quantification of these processes in intact organs in vivo. We have previously used multiphoton fluorescence lifetime microscopy to characterise the morphology and amplitude weighted mean fluorescence lifetime of the endogenous fluorescent metabolic cofactor nicotinamide adenine dinucleotide (phosphate) (NAD(P)H) of mouse livers in vivo following induction of various disease states. Here, we extend the characterisation of liver disease models by using nonlinear regression to estimate the unbound, bound fluorescence lifetimes for NAD(P)H, flavin adenine dinucleotide (FAD), along with metabolic ratios and examine the impact of using multiple segmentation methods. We found that NAD(P)H amplitude ratio, and fluorescence lifetime redox ratio can be used as discriminators of diseased liver from normal liver. The redox ratio provided a sensitive measure of the changes in hepatic fibrosis and biliary fibrosis. Hepatocellular carcinoma was associated with an increase in spatial heterogeneity and redox ratio coupled with a decrease in mean fluorescence lifetime. We conclude that multiphoton fluorescence lifetime microscopy parameters and metabolic ratios provided insights into the in vivo redox state of diseased compared to normal liver that were not apparent from a global, mean fluorescence lifetime measurement alone.
Collapse
Affiliation(s)
- Deborah S Barkauskas
- Therapeutics Research Group, University of Queensland Diamantina Institute, The University of Queensland, Woolloongabba, QLD 4102, Australia
| | | | | | | | | | | | | |
Collapse
|
33
|
Penjweini R, Roarke B, Alspaugh G, Gevorgyan A, Andreoni A, Pasut A, Sackett DL, Knutson JR. Single cell-based fluorescence lifetime imaging of intracellular oxygenation and metabolism. Redox Biol 2020; 34:101549. [PMID: 32403080 PMCID: PMC7217996 DOI: 10.1016/j.redox.2020.101549] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 04/15/2020] [Accepted: 04/20/2020] [Indexed: 12/02/2022] Open
Abstract
Oxidation-reduction chemistry is fundamental to the metabolism of all living organisms, and hence quantifying the principal redox players is important for a comprehensive understanding of cell metabolism in normal and pathological states. In mammalian cells, this is accomplished by measuring oxygen partial pressure (pO2) in parallel with free and enzyme-bound reduced nicotinamide adenine dinucleotide (phosphate) [H] (NAD(P)H) and flavin adenine dinucleotide (FAD, a proxy for NAD+). Previous optical methods for these measurements had accompanying problems of cytotoxicity, slow speed, population averaging, and inability to measure all redox parameters simultaneously. Herein we present a Förster resonance energy transfer (FRET)-based oxygen sensor, Myoglobin-mCherry, compatible with fluorescence lifetime imaging (FLIM)-based measurement of nicotinamide coenzyme state. This offers a contemporaneous reading of metabolic activity through real-time, non-invasive, cell-by-cell intracellular pO2 and coenzyme status monitoring in living cells. Additionally, this method reveals intracellular spatial heterogeneity and cell-to-cell variation in oxygenation and coenzyme states.
Collapse
Affiliation(s)
- Rozhin Penjweini
- Laboratory of Advanced Microscopy and Biophotonics, National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health (NIH), Building 10, Room 5D14, Bethesda, MD, 20892-1412, USA
| | - Branden Roarke
- Laboratory of Advanced Microscopy and Biophotonics, National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health (NIH), Building 10, Room 5D14, Bethesda, MD, 20892-1412, USA
| | - Greg Alspaugh
- Laboratory of Advanced Microscopy and Biophotonics, National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health (NIH), Building 10, Room 5D14, Bethesda, MD, 20892-1412, USA
| | - Anahit Gevorgyan
- Laboratory of Advanced Microscopy and Biophotonics, National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health (NIH), Building 10, Room 5D14, Bethesda, MD, 20892-1412, USA
| | - Alessio Andreoni
- Laboratory of Advanced Microscopy and Biophotonics, National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health (NIH), Building 10, Room 5D14, Bethesda, MD, 20892-1412, USA; Laboratory of Optical Neurophysiology, Department of Biochemistry and Molecular Medicine, University of California Davis, Tupper Hall, Davis, CA, 95616, USA
| | - Alessandra Pasut
- Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, VIB, Leuven Cancer Institute, KU Leuven, Leuven, 3000, Belgium
| | - Dan L Sackett
- Cytoskeletal Dynamics Group, Division of Basic and Translational Biophysics, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH), Building 9, Room 1E129, Bethesda, MD, 20892-0924, USA
| | - Jay R Knutson
- Laboratory of Advanced Microscopy and Biophotonics, National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health (NIH), Building 10, Room 5D14, Bethesda, MD, 20892-1412, USA.
| |
Collapse
|
34
|
Meyer BO, Stella MPJ, Holst B, Nielsen BS, Holmstrøm K, Andersen PE, Marti D. Selecting optimal spectral bands for improved detection of autofluorescent biomarkers in multiphoton microscopy. JOURNAL OF BIOMEDICAL OPTICS 2020; 25:1-13. [PMID: 32638570 PMCID: PMC7338838 DOI: 10.1117/1.jbo.25.7.071206] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Accepted: 06/22/2020] [Indexed: 05/08/2023]
Abstract
SIGNIFICANCE In multiphoton microscopy, two-photon excited fluorescence (TPEF) spectra carry valuable information on morphological and functional biological features. For measuring these biomarkers, separation of different parts of the fluorescence spectrum into channels is typically achieved by the use of optical band pass filters. However, spectra from different biomarkers can be unknown or overlapping, creating a crosstalk in between the channels. Previously, establishing these channels relied on prior knowledge or heuristic testing. AIM The presented method aims to provide spectral bands with optimal separation between groups of specimens expressing different biomarkers. APPROACH We have developed a system capable of resolving TPEF with high spectral resolution for the characterization of biomarkers. In addition, an algorithm is created to simulate and optimize optical band pass filters for fluorescence detection channels. To demonstrate the potential improvements in cell and tissue classification using these optimized channels, we recorded spectrally resolved images of cancerous (HT29) and normal epithelial colon cells (FHC), cultivated in 2D layers and in 3D to form spheroids. To provide an example of an application, we relate the results with the widely used redox ratio. RESULTS We show that in the case of two detection channels, our system and algorithm enable the selection of optimized band pass filters without the need of knowing involved fluorophores. An improvement of 31,5% in separating different 2D cell cultures is achieved, compared to using established spectral bands that assume NAD(P)H and FAD as main contributors of autofluorescence. The compromise is a reduced SNR in the images. CONCLUSIONS We show that the presented method has the ability to improve imaging contrast and can be used to tailor a given label-free optical imaging system using optical band pass filters targeting a specific biomarker or application.
Collapse
Affiliation(s)
- Björn-Ole Meyer
- Technical University of Denmark, DTU Health Tech, Roskilde, Denmark
| | | | | | | | | | - Peter E. Andersen
- Technical University of Denmark, DTU Health Tech, Roskilde, Denmark
- Address all correspondence to Peter E. Andersen, E-mail:
| | - Dominik Marti
- Technical University of Denmark, DTU Health Tech, Roskilde, Denmark
| |
Collapse
|
35
|
Hou J, Reid NE, Tromberg BJ, Potma EO. Kinetic Analysis of Lipid Metabolism in Breast Cancer Cells via Nonlinear Optical Microscopy. Biophys J 2020; 119:258-264. [PMID: 32610090 DOI: 10.1016/j.bpj.2020.06.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Revised: 06/01/2020] [Accepted: 06/08/2020] [Indexed: 12/26/2022] Open
Abstract
Investigating the behavior of breast cancer cells via reaction kinetics may help unravel the mechanisms that underlie metabolic changes in tumors. However, obtaining human in vivo kinetic data is challenging because of difficulties associated with measuring these parameters. Nondestructive methods of measuring lipid content in live cells provide a novel approach to quantitatively model lipid synthesis and consumption. In this study, coherent Raman scattering microscopy was used to probe de novo intracellular lipid content. Combining nonlinear optical microscopy and Michaelis-Menten kinetics-based simulations, we isolated fatty acid synthesis/consumption rates and elucidated effects of altered lipid metabolism in T47D breast cancer cells. When treated with 17β-estradiol, the lipid utilization in cancer cells jumped by twofold. Meanwhile, the rate of de novo lipid synthesis in cancer cells treated with 17β-estradiol was increased by 42%. To test the model in extreme metabolic conditions, we treated T47D cells with etomoxir. Our kinetic analysis demonstrated that the rate of key enzymatic reactions dropped by 75%. These results underline the capability to probe lipid alterations in live cells with minimum interruption and to characterize lipid metabolism in breast cancer cells via quantitative kinetic models and parameters.
Collapse
Affiliation(s)
- Jue Hou
- Beckman Laser Institute and Medical Center, University of California, Irvine, Irvine, California
| | - Nellone E Reid
- Department of Chemical and Materials Engineering, New Jersey Institute of Technology, Newark, New Jersey
| | - Bruce J Tromberg
- Beckman Laser Institute and Medical Center, University of California, Irvine, Irvine, California
| | - Eric O Potma
- Beckman Laser Institute and Medical Center, University of California, Irvine, Irvine, California.
| |
Collapse
|
36
|
Sharick JT, Walsh CM, Sprackling CM, Pasch CA, Pham DL, Esbona K, Choudhary A, Garcia-Valera R, Burkard ME, McGregor SM, Matkowskyj KA, Parikh AA, Meszoely IM, Kelley MC, Tsai S, Deming DA, Skala MC. Metabolic Heterogeneity in Patient Tumor-Derived Organoids by Primary Site and Drug Treatment. Front Oncol 2020; 10:553. [PMID: 32500020 PMCID: PMC7242740 DOI: 10.3389/fonc.2020.00553] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Accepted: 03/27/2020] [Indexed: 12/16/2022] Open
Abstract
New tools are needed to match cancer patients with effective treatments. Patient-derived organoids offer a high-throughput platform to personalize treatments and discover novel therapies. Currently, methods to evaluate drug response in organoids are limited because they overlook cellular heterogeneity. In this study, non-invasive optical metabolic imaging (OMI) of cellular heterogeneity was characterized in breast cancer (BC) and pancreatic cancer (PC) patient-derived organoids. Baseline heterogeneity was analyzed for each patient, demonstrating that single-cell techniques, such as OMI, are required to capture the complete picture of heterogeneity present in a sample. Treatment-induced changes in heterogeneity were also analyzed, further demonstrating that these measurements greatly complement current techniques that only gauge average cellular response. Finally, OMI of cellular heterogeneity in organoids was evaluated as a predictor of clinical treatment response for the first time. Organoids were treated with the same drugs as the patient's prescribed regimen, and OMI measurements of heterogeneity were compared to patient outcome. OMI distinguished subpopulations of cells with divergent and dynamic responses to treatment in living organoids without the use of labels or dyes. OMI of organoids agreed with long-term therapeutic response in patients. With these capabilities, OMI could serve as a sensitive high-throughput tool to identify optimal therapies for individual patients, and to develop new effective therapies that address cellular heterogeneity in cancer.
Collapse
Affiliation(s)
- Joe T Sharick
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, United States.,Morgridge Institute for Research, Madison, WI, United States
| | | | | | - Cheri A Pasch
- University of Wisconsin Carbone Cancer Center, Madison, WI, United States
| | - Dan L Pham
- Morgridge Institute for Research, Madison, WI, United States.,Department of Biomedical Engineering, University of Wisconsin, Madison, WI, United States
| | - Karla Esbona
- Department of Pathology and Laboratory Medicine, University of Wisconsin, Madison, WI, United States
| | - Alka Choudhary
- University of Wisconsin Carbone Cancer Center, Madison, WI, United States.,Department of Medicine, University of Wisconsin, Madison, WI, United States
| | - Rebeca Garcia-Valera
- University of Wisconsin Carbone Cancer Center, Madison, WI, United States.,Department of Medicine, University of Wisconsin, Madison, WI, United States.,Tecnológico de Monterrey, Escuela de Ingeniería y Ciencias, Zapopan, Mexico
| | - Mark E Burkard
- University of Wisconsin Carbone Cancer Center, Madison, WI, United States.,Department of Medicine, University of Wisconsin, Madison, WI, United States
| | - Stephanie M McGregor
- University of Wisconsin Carbone Cancer Center, Madison, WI, United States.,Department of Pathology and Laboratory Medicine, University of Wisconsin, Madison, WI, United States
| | - Kristina A Matkowskyj
- University of Wisconsin Carbone Cancer Center, Madison, WI, United States.,Department of Pathology and Laboratory Medicine, University of Wisconsin, Madison, WI, United States.,William S. Middleton Memorial Veterans Hospital, Madison, WI, United States
| | - Alexander A Parikh
- Division of Surgical Oncology, East Carolina University Brody School of Medicine, Greenville, NC, United States
| | - Ingrid M Meszoely
- Department of Surgery, Vanderbilt University, Nashville, TN, United States
| | - Mark C Kelley
- Department of Surgery, Vanderbilt University, Nashville, TN, United States
| | - Susan Tsai
- Department of Surgery, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Dustin A Deming
- University of Wisconsin Carbone Cancer Center, Madison, WI, United States.,Division of Hematology and Oncology, Department of Medicine, University of Wisconsin, Madison, WI, United States.,McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin, Madison, WI, United States
| | - Melissa C Skala
- Morgridge Institute for Research, Madison, WI, United States.,University of Wisconsin Carbone Cancer Center, Madison, WI, United States.,Department of Biomedical Engineering, University of Wisconsin, Madison, WI, United States
| |
Collapse
|
37
|
Darby JRT, Sorvina A, Bader CA, Lock MC, Soo JY, Holman SL, Seed M, Kuchel T, Brooks DA, Plush SE, Morrison JL. Detecting metabolic differences in fetal and adult sheep adipose and skeletal muscle tissues. JOURNAL OF BIOPHOTONICS 2020; 13:e201960085. [PMID: 31793184 DOI: 10.1002/jbio.201960085] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Revised: 11/05/2019] [Accepted: 11/29/2019] [Indexed: 06/10/2023]
Abstract
The primary metabolic pathway required to produce ATP differs as a result of tissue type, developmental stage and substrate availability. We utilized molecular and histological techniques to define the metabolic status in foetal and adult, adipose and skeletal muscle tissues. Redox ratios of these tissues were also determined optically by two-photon microscopy. Adult perirenal adipose tissue had a higher optical redox ratio than fetal perirenal adipose tissue, which aligned with glycolysis being used for ATP production; whereas adult skeletal muscle had a lower optical redox ratio than fetal skeletal muscle, which aligned with oxygen demanding oxidative phosphorylation activity being utilized for ATP production. We have compared traditional molecular and microscopy techniques of metabolic tissue characterization with optical redox ratios to provide a more comprehensive report on the dynamics of tissue metabolism.
Collapse
Affiliation(s)
- Jack R T Darby
- Early Origins of Adult Health Research Group, University of South Australia, Adelaide, South Australia, Australia
| | - Alexandra Sorvina
- Mechanisms in Cell Biology and Disease Research Group, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, South Australia, Australia
| | - Christie A Bader
- Mechanisms in Cell Biology and Disease Research Group, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, South Australia, Australia
| | - Mitchell C Lock
- Early Origins of Adult Health Research Group, University of South Australia, Adelaide, South Australia, Australia
| | - Jia Yin Soo
- Early Origins of Adult Health Research Group, University of South Australia, Adelaide, South Australia, Australia
| | - Stacey L Holman
- Early Origins of Adult Health Research Group, University of South Australia, Adelaide, South Australia, Australia
| | - Mike Seed
- The Hospital for Sick Kids, Toronto, Ontario, Canada
| | - Tim Kuchel
- Preclinical Imaging and Research Laboratories, South Australian Health and Medical Research Institute, Adelaide, Australia
| | - Douglas A Brooks
- Mechanisms in Cell Biology and Disease Research Group, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, South Australia, Australia
| | - Sally E Plush
- Mechanisms in Cell Biology and Disease Research Group, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, South Australia, Australia
- Future Industries Institute, University of South Australia, Adelaide, South Australia, Australia
| | - Janna L Morrison
- Early Origins of Adult Health Research Group, University of South Australia, Adelaide, South Australia, Australia
| |
Collapse
|
38
|
Podsednik A, Jacob A, Li LZ, Xu HN. Relationship between Optical Redox Status and Reactive Oxygen Species in Cancer Cells. REACTIVE OXYGEN SPECIES (APEX, N.C.) 2020; 9:95-108. [PMID: 32066994 PMCID: PMC7025870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Shifted NAD(H) redox status and enhanced reactive oxygen species (ROS) scavenging systems have been observed in cancers. However, how such redox shift is related to the ROS level in cancer cells is less clear. Based on collecting the intrinsic fluorescence of oxidized flavoproteins (Fp containing flavin adenine dinucleotide) and reduced nicotinamide adenine dinucleotide (NADH), optical redox imaging (ORI) provides a quantitative measure of the mitochondrial redox state by the optical redox ratio, Fp/(NADH+Fp), a surrogate marker of the NAD+-coupled redox state NAD+/NADH. Our study aims to explore the relationship between NAD(H) redox status and ROS by imaging NADH, Fp, and ROS levels using cultured breast cancer cell models. By manipulating either ROS levels via application of exogenous H2O2 or redox status via metabolic perturbation compounds, we found that: (1) oxidation of NAD(H) redox status correlates with ROS levels at lower H2O2 concentrations (up to ~700 μM), but not necessarily at higher concentrations; (2) an elevated ROS level diminishes NADH and reduces redox ratio plasticity; (3) either more oxidized or more reduced status can correlate to an increased ROS level; and (4) sometimes, a more oxidized status can correlate to a decreased ROS level depending on cell lines. These observations indicated that cellular NAD(H) redox state and ROS are intricately related but can also change separately. This study can benefit cancer research as both NAD(H) redox status and ROS have been implicated in cancer transformation and progression.
Collapse
Affiliation(s)
- Allison Podsednik
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Annemarie Jacob
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Lin Z Li
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - He N Xu
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
39
|
Affiliation(s)
- Alex J Walsh
- Morgridge Institute for Research, Madison, WI, USA
| | | | - Melissa C Skala
- Morgridge Institute for Research, Madison, WI, USA. .,Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, USA.
| |
Collapse
|
40
|
Zhou H, Nguyen L, Arnesano C, Ando Y, Raval M, Rodgers JT, Fraser S, Lu R, Shen K. Non-invasive Optical Biomarkers Distinguish and Track the Metabolic Status of Single Hematopoietic Stem Cells. iScience 2020; 23:100831. [PMID: 31982780 PMCID: PMC6994633 DOI: 10.1016/j.isci.2020.100831] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 12/11/2019] [Accepted: 01/07/2020] [Indexed: 12/28/2022] Open
Abstract
Metabolism is a key regulator of hematopoietic stem cell (HSC) functions. There is a lack of real-time, non-invasive approaches to evaluate metabolism in single HSCs. Using fluorescence lifetime imaging microscopy, we developed a set of metabolic optical biomarkers (MOBs) from the auto-fluorescent properties of metabolic coenzymes NAD(P)H and FAD. The MOBs revealed the enhanced glycolysis, low oxidative metabolism, and distinct mitochondrial localization of HSCs. Importantly, the fluorescence lifetime of enzyme-bound NAD(P)H (τbound) can non-invasively monitor the glycolytic/lactate dehydrogenase activity in single HSCs. As a proof of concept for metabolism-based cell sorting, we further identified HSCs within the Lineage-cKit+Sca1+ (KLS) hematopoietic stem/progenitor population using MOBs and a machine-learning algorithm. Moreover, we revealed the dynamic changes of MOBs, and the association of longer τbound with enhanced glycolysis under HSC stemness-maintaining conditions during HSC culture. Our work thus provides a new paradigm to identify and track the metabolism of single HSCs non-invasively and in real time. Metabolic optical biomarkers non-invasively distinguish HSCs from early progenitors NAD(P)H τbound reflects lactate dehydrogenase activity in single fresh/cultured HSCs pHi correlates with τbound in hematopoietic populations, with HSCs being the highest Optical biomarkers track metabolic changes and response to drugs in cultured HSCs
Collapse
Affiliation(s)
- Hao Zhou
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA 90089, USA
| | - Lisa Nguyen
- Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Cosimo Arnesano
- Translational Imaging Center, University of Southern California, Los Angeles, CA 90089, USA; Molecular and Computational Biology, University of Southern California, Los Angeles, CA 90089, USA
| | - Yuta Ando
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA 90089, USA
| | - Manmeet Raval
- Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Joseph T Rodgers
- Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Scott Fraser
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA 90089, USA; Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles, CA 90033, USA; Translational Imaging Center, University of Southern California, Los Angeles, CA 90089, USA; Molecular and Computational Biology, University of Southern California, Los Angeles, CA 90089, USA
| | - Rong Lu
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA 90089, USA; Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles, CA 90033, USA; Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA 90033, USA; Department of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Keyue Shen
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA 90089, USA; Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA 90033, USA; USC Stem Cell, University of Southern California, Los Angeles, CA 90033, USA.
| |
Collapse
|
41
|
Li LZ, Masek M, Wang T, Xu HN, Nioka S, Baur JA, Ragan TM. Two-Photon Autofluorescence Imaging of Fixed Tissues: Feasibility and Potential Values for Biomedical Applications. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1232:375-381. [PMID: 31893434 DOI: 10.1007/978-3-030-34461-0_48] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The value of optical redox imaging (ORI) of cells/tissues based on the intrinsic fluorescences of NADH (nicotinamide adenine dinucleotide) and oxidized flavoproteins (containing flavin adenine dinucleotide, i.e., FAD) has been demonstrated for potential biomedical applications including diagnosis, prognosis, and determining treatment response. However, the Chance redox scanner (a 3D cryogenic tissue imager) is limited by spatial resolution (~50 μm), and tissue ORI using fluorescence microscopy (single or multi-photon) is limited by the light penetration depth. Furthermore, viable or snap-frozen tissues are usually required. In this project, we aimed to study whether ORI may be achieved for unstained fixed tissue using a state-of-the-art modern Serial Two-Photon (STP) Tomography scanner that can rapidly acquire multi-plane images at micron resolution. Tissue specimens of mouse muscle, liver, and tumor xenografts were harvested and fixed in 4% paraformaldehyde (PFA) for 24 h. Tissue blocks were scanned by STP Tomography under room temperature to acquire the autofluorescence signals (NADH channel: excitation 750 nm, blue emission filter; FAD channel: excitation 860 nm, green emission filter). We observed remarkable signals with significant intra-tissue heterogeneity in images of NADH, FAD and redox ratio (FAD/(NADH+FAD)), which are worthy of further investigation for extracting biological information.
Collapse
Affiliation(s)
- Lin Z Li
- Department of Radiology & Britton Chance Laboratory of Redox Imaging, Johnson Research Foundation, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA. .,Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA. .,Institute of Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| | | | - Ting Wang
- Department of Radiology & Britton Chance Laboratory of Redox Imaging, Johnson Research Foundation, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - He N Xu
- Department of Radiology & Britton Chance Laboratory of Redox Imaging, Johnson Research Foundation, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Shoko Nioka
- Department of Radiology & Britton Chance Laboratory of Redox Imaging, Johnson Research Foundation, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Joseph A Baur
- Department of Physiology, Institute for Diabetes, Obesity and Metabolism, University of Pennsylvania, Philadelphia, PA, USA
| | | |
Collapse
|
42
|
Real-time optical redox imaging of cartilage metabolic response to mechanical loading. Osteoarthritis Cartilage 2019; 27:1841-1850. [PMID: 31513919 DOI: 10.1016/j.joca.2019.08.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 08/27/2019] [Accepted: 08/29/2019] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Metabolic dysregulation has recently been identified as a key feature of osteoarthritis. Mechanical overloading has been postulated as a primary cause of this metabolic response. Current methods of real-time metabolic activity analysis in cartilage are limited and challenging. However, optical redox imaging leverages the autofluorescence of co-enzymes NAD(P)H and FAD to provide dye-free real-time analysis of metabolic activity. This technique has not yet been applied to cartilage. This study aimed to assess the effects of a compressive load on cartilage using optical redox imaging. METHOD Cartilage samples were excised from porcine femoral condyles. To validate this imaging modality in cartilage, glycolysis was inhibited via 2-deoxy-D-glucose (2DG) and oxidative phosphorylation was inhibited by rotenone. Optical redox images were collected pre- and post-inhibition. To assess the effects of mechanical loading, samples were subjected to a compressive load and imaged for approximately 30 min. Load and strain parameters were determined using high-speed camera images in Matlab. A range of loading magnitudes and rates were applied across samples. RESULTS 2DG and rotenone demonstrated the expected inhibitory effects on fluorescence intensity in the channels corresponding to NAD(P)H and FAD, respectively. Mechanical loading induced an increase in NAD(P)H channel fluorescence which subsided by 30 min post-loading. Magnitude of loading parameters had mixed effects on metabolites. CONCLUSIONS Optical redox imaging provides an opportunity to assess real-time metabolic activity in cartilage. This approach revealed a metabolic response to a single load and can be used to provide insight into the role of metabolism in mechanically-mediated cartilage degradation.
Collapse
|
43
|
Dadgar S, Rajaram N. Optical Imaging Approaches to Investigating Radiation Resistance. Front Oncol 2019; 9:1152. [PMID: 31750246 PMCID: PMC6848224 DOI: 10.3389/fonc.2019.01152] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Accepted: 10/16/2019] [Indexed: 12/14/2022] Open
Abstract
Radiation therapy is frequently the first line of treatment for over 50% of cancer patients. While great advances have been made in improving treatment response rates and reducing damage to normal tissue, radiation resistance remains a persistent clinical problem. While hypoxia or a lack of tumor oxygenation has long been considered a key factor in causing treatment failure, recent evidence points to metabolic reprogramming under well-oxygenated conditions as a potential route to promoting radiation resistance. In this review, we present recent studies from our lab and others that use high-resolution optical imaging as well as clinical translational optical spectroscopy to shine light on the biological basis of radiation resistance. Two-photon microscopy of endogenous cellular metabolism has identified key changes in both mitochondrial structure and function that are specific to radiation-resistant cells and help promote cell survival in response to radiation. Optical spectroscopic approaches, such as diffuse reflectance and Raman spectroscopy have demonstrated functional and molecular differences between radiation-resistant and sensitive tumors in response to radiation. These studies have uncovered key changes in metabolic pathways and present a viable route to clinical translation of optical technologies to determine radiation resistance at a very early stage in the clinic.
Collapse
Affiliation(s)
| | - Narasimhan Rajaram
- Department of Biomedical Engineering, University of Arkansas, Fayetteville, AR, United States
| |
Collapse
|
44
|
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
|
45
|
Local redox conditions in cells imaged via non-fluorescent transient states of NAD(P)H. Sci Rep 2019; 9:15070. [PMID: 31636326 PMCID: PMC6803634 DOI: 10.1038/s41598-019-51526-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2019] [Accepted: 09/30/2019] [Indexed: 01/15/2023] Open
Abstract
The autofluorescent coenzyme nicotinamide adenine dinucleotide (NADH) and its phosphorylated form (NADPH) are major determinants of cellular redox balance. Both their fluorescence intensities and lifetimes are extensively used as label-free readouts in cellular metabolic imaging studies. Here, we introduce fluorescence blinking of NAD(P)H, as an additional, orthogonal readout in such studies. Blinking of fluorophores and their underlying dark state transitions are specifically sensitive to redox conditions and oxygenation, parameters of particular relevance in cellular metabolic studies. We show that such dark state transitions in NAD(P)H can be quantified via the average fluorescence intensity recorded upon modulated one-photon excitation, so-called transient state (TRAST) monitoring. Thereby, transitions in NAD(P)H, previously only accessible from elaborate spectroscopic cuvette measurements, can be imaged at subcellular resolution in live cells. We then demonstrate that these transitions can be imaged with a standard laser-scanning confocal microscope and two-photon excitation, in parallel with regular fluorescence lifetime imaging (FLIM). TRAST imaging of NAD(P)H was found to provide additional, orthogonal information to FLIM and allows altered oxidative environments in cells treated with a mitochondrial un-coupler or cyanide to be clearly distinguished. We propose TRAST imaging as a straightforward and widely applicable modality, extending the range of information obtainable from cellular metabolic imaging of NAD(P)H fluorescence.
Collapse
|
46
|
Hai P, Imai T, Xu S, Zhang R, Aft RL, Zou J, Wang LV. High-throughput, label-free, single-cell photoacoustic microscopy of intratumoral metabolic heterogeneity. Nat Biomed Eng 2019; 3:381-391. [PMID: 30936431 PMCID: PMC6544054 DOI: 10.1038/s41551-019-0376-5] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Accepted: 02/26/2019] [Indexed: 12/11/2022]
Abstract
Intratumoral heterogeneity, which is manifested in almost all of the hallmarks of cancer, including the significantly altered metabolic profiles of cancer cells, represents a challenge to effective cancer therapy. High-throughput measurements of the metabolism of individual cancer cells would allow direct visualization and quantification of intratumoral metabolic heterogeneity, yet the throughputs of current measurement techniques are limited to about 120 cells per hour. Here, we show that single-cell photoacoustic microscopy can reach throughputs of approximately 12,000 cells per hour by trapping single cells with blood in an oxygen-diffusion-limited high-density microwell array and by using photoacoustic imaging to measure the haemoglobin oxygen change (that is, the oxygen consumption rate) in the microwells. We demonstrate the capability of this label-free technique by performing high-throughput single-cell oxygen-consumption-rate measurements of cultured cells and by imaging intratumoral metabolic heterogeneity in specimens from patients with breast cancer. High-throughput single-cell photoacoustic microscopy of oxygen consumption rates should enable the faster characterization of intratumoral metabolic heterogeneity.
Collapse
Affiliation(s)
- Pengfei Hai
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, USA
- Caltech Optical Imaging Laboratory, Andrew and Peggy Cherng Department of Medical Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Toru Imai
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, USA
- Caltech Optical Imaging Laboratory, Andrew and Peggy Cherng Department of Medical Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Song Xu
- Department of Electrical and Computer Engineering, Texas A&M University, College Station, TX, USA
| | - Ruiying Zhang
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, USA
| | - Rebecca L Aft
- Department of Surgery, School of Medicine, Washington University, St. Louis, MO, USA
- John Cochran Veterans Hospital, St. Louis, MO, USA
| | - Jun Zou
- Department of Electrical and Computer Engineering, Texas A&M University, College Station, TX, USA.
| | - Lihong V Wang
- Caltech Optical Imaging Laboratory, Andrew and Peggy Cherng Department of Medical Engineering, California Institute of Technology, Pasadena, CA, USA.
- Caltech Optical Imaging Laboratory, Department of Electrical Engineering, California Institute of Technology, Pasadena, CA, USA.
| |
Collapse
|
47
|
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
|
48
|
Madonna MC, Fox DB, Crouch BT, Lee J, Zhu C, Martinez AF, Alvarez JV, Ramanujam N. Optical Imaging of Glucose Uptake and Mitochondrial Membrane Potential to Characterize Her2 Breast Tumor Metabolic Phenotypes. Mol Cancer Res 2019; 17:1545-1555. [PMID: 30902832 DOI: 10.1158/1541-7786.mcr-18-0618] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Revised: 10/09/2018] [Accepted: 03/19/2019] [Indexed: 11/16/2022]
Abstract
With the large number of women diagnosed and treated for breast cancer each year, the importance of studying recurrence has become evident due to most deaths from breast cancer resulting from tumor recurrence following therapy. To mitigate this, cellular and molecular pathways used by residual disease prior to recurrence must be studied. An altered metabolism has long been considered a hallmark of cancer, and several recent studies have gone further to report metabolic dysfunction and alterations as key to understanding the underlying behavior of dormant and recurrent cancer cells. Our group has used two probes, 2-[N-(7-nitrobenz-2-oxa-1, 3-diaxol-4-yl) amino]-2-deoxyglucose (2-NBDG) and tetramethyl rhodamine ethyl ester (TMRE), to image glucose uptake and mitochondrial membrane potential, respectively, to report changes in metabolism between primary tumors, regression, residual disease, and after regrowth in genetically engineered mouse (GEM)-derived mammospheres. Imaging revealed unique metabolic phenotypes across the stages of tumor development. Although primary mammospheres overexpressing Her2 maintained increased glucose uptake ("Warburg effect"), after Her2 downregulation, during regression and residual disease, mammospheres appeared to switch to oxidative phosphorylation. Interestingly, in mammospheres where Her2 overexpression was turned back on to model recurrence, glucose uptake was lowest, indicating a potential change in substrate preference following the reactivation of Her2, reeliciting growth. Our findings highlight the importance of imaging metabolic adaptions to gain insight into the fundamental behaviors of residual and recurrent disease. IMPLICATIONS: This study demonstrates these functional fluorescent probes' ability to report metabolic adaptations during primary tumor growth, regression, residual disease, and regrowth in Her2 breast tumors.
Collapse
Affiliation(s)
- Megan C Madonna
- Department of Biomedical Engineering, Duke University, Durham, North Carolina.
| | - Douglas B Fox
- Department of Pharmacology & Cancer Biology, School of Medicine, Duke University, Durham, North Carolina
| | - Brian T Crouch
- Department of Biomedical Engineering, Duke University, Durham, North Carolina
| | - Jihong Lee
- Department of Biomedical Engineering, Duke University, Durham, North Carolina
| | - Caigang Zhu
- Department of Biomedical Engineering, Duke University, Durham, North Carolina
| | - Amy F Martinez
- Department of Biomedical Engineering, Duke University, Durham, North Carolina
| | - James V Alvarez
- Department of Pharmacology & Cancer Biology, School of Medicine, Duke University, Durham, North Carolina
| | - Nirmala Ramanujam
- Department of Biomedical Engineering, Duke University, Durham, North Carolina.,Department of Pharmacology & Cancer Biology, School of Medicine, Duke University, Durham, North Carolina
| |
Collapse
|
49
|
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
|
50
|
Dravid U A, Mazumder N. Types of advanced optical microscopy techniques for breast cancer research: a review. Lasers Med Sci 2018; 33:1849-1858. [PMID: 30311083 DOI: 10.1007/s10103-018-2659-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Accepted: 10/01/2018] [Indexed: 10/28/2022]
Abstract
A cancerous cell is characterized by morphological and metabolic changes which are the key features of carcinogenesis. Adenosine triphosphate (ATP) in cancer cells is primarily produced by aerobic glycolysis rather than oxidative phosphorylation. In normal cellular metabolism, nicotinamide adenine dinucleotide (NADH) is considered as a principle electron donor and flavin adenine dinucleotide (FAD) as an electron acceptor. During metabolism in a cancerous cell, a net increase in NADH is found as the pathway switched from oxidative phosphorylation to aerobic glycolysis. Often during initiation and progression of cancer, the developmental regulation of extracellular matrix (ECM) is restricted and becomes disorganized. Tumor cell behavior is regulated by the ECM in the tumor micro environment. Collagen, which forms the scaffold of tumor micro-environment also influences its behavior. Advanced optical microscopy techniques are useful for determining the metabolic characteristics of cancerous, normal cells and tissues. They can be used to identify the collagen microstructure and the function of NADH, FAD, and lipids in living system. In this review article, various optical microscopy techniques applied for breast cancer research are discussed including fluorescence, confocal, second harmonic generation (SHG), coherent anti-Stokes Raman scattering (CARS), and fluorescence lifetime imaging (FLIM).
Collapse
Affiliation(s)
- Aparna Dravid U
- Department of Biophysics, School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Nirmal Mazumder
- Department of Biophysics, School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India.
| |
Collapse
|