151
|
Protein-bound NAD(P)H Lifetime is Sensitive to Multiple Fates of Glucose Carbon. Sci Rep 2018; 8:5456. [PMID: 29615678 PMCID: PMC5883019 DOI: 10.1038/s41598-018-23691-x] [Citation(s) in RCA: 76] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Accepted: 03/19/2018] [Indexed: 12/12/2022] Open
Abstract
While NAD(P)H fluorescence lifetime imaging (FLIM) can detect changes in flux through the TCA cycle and electron transport chain (ETC), it remains unclear whether NAD(P)H FLIM is sensitive to other potential fates of glucose. Glucose carbon can be diverted from mitochondria by the pentose phosphate pathway (via glucose 6-phosphate dehydrogenase, G6PDH), lactate production (via lactate dehydrogenase, LDH), and rejection of carbon from the TCA cycle (via pyruvate dehydrogenase kinase, PDK), all of which can be upregulated in cancer cells. Here, we demonstrate that multiphoton NAD(P)H FLIM can be used to quantify the relative concentrations of recombinant LDH and malate dehydrogenase (MDH) in solution. In multiple epithelial cell lines, NAD(P)H FLIM was also sensitive to inhibition of LDH and PDK, as well as the directionality of LDH in cells forced to use pyruvate versus lactate as fuel sources. Among the parameters measurable by FLIM, only the lifetime of protein-bound NAD(P)H (τ2) was sensitive to these changes, in contrast to the optical redox ratio, mean NAD(P)H lifetime, free NAD(P)H lifetime, or the relative amount of free and protein-bound NAD(P)H. NAD(P)H τ2 offers the ability to non-invasively quantify diversions of carbon away from the TCA cycle/ETC, which may support mechanisms of drug resistance.
Collapse
|
152
|
Martinez AF, McCachren SS, Lee M, Murphy HA, Zhu C, Crouch BT, Martin HL, Erkanli A, Rajaram N, Ashcraft KA, Fontanella AN, Dewhirst MW, Ramanujam N. Metaboloptics: Visualization of the tumor functional landscape via metabolic and vascular imaging. Sci Rep 2018. [PMID: 29520098 PMCID: PMC5843602 DOI: 10.1038/s41598-018-22480-w] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Many cancers adeptly modulate metabolism to thrive in fluctuating oxygen conditions; however, current tools fail to image metabolic and vascular endpoints at spatial resolutions needed to visualize these adaptations in vivo. We demonstrate a high-resolution intravital microscopy technique to quantify glucose uptake, mitochondrial membrane potential (MMP), and SO2 to characterize the in vivo phentoypes of three distinct murine breast cancer lines. Tetramethyl rhodamine, ethyl ester (TMRE) was thoroughly validated to report on MMP in normal and tumor-bearing mice. Imaging MMP or glucose uptake together with vascular endpoints revealed that metastatic 4T1 tumors maintained increased glucose uptake across all SO2 (“Warburg effect”), and also showed increased MMP relative to normal tissue. Non-metastatic 67NR and 4T07 tumor lines both displayed increased MMP, but comparable glucose uptake, relative to normal tissue. The 4T1 peritumoral areas also showed a significant glycolytic shift relative to the tumor regions. During a hypoxic stress test, 4T1 tumors showed significant increases in MMP with corresponding significant drops in SO2, indicative of intensified mitochondrial metabolism. Conversely, 4T07 and 67NR tumors shifted toward glycolysis during hypoxia. Our findings underscore the importance of imaging metabolic endpoints within the context of a living microenvironment to gain insight into a tumor’s adaptive behavior.
Collapse
Affiliation(s)
- Amy F Martinez
- Department of Biomedical Engineering, Duke University, Durham, NC, USA.
| | | | - Marianne Lee
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Helen A Murphy
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Caigang Zhu
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Brian T Crouch
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Hannah L Martin
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Alaattin Erkanli
- Department of Biostatistics and Bioinformatics, Duke University Medical Center, Durham, NC, USA
| | | | | | | | | | - Nirmala Ramanujam
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| |
Collapse
|
153
|
Nobis M, Warren SC, Lucas MC, Murphy KJ, Herrmann D, Timpson P. Molecular mobility and activity in an intravital imaging setting - implications for cancer progression and targeting. J Cell Sci 2018; 131:131/5/jcs206995. [PMID: 29511095 DOI: 10.1242/jcs.206995] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Molecular mobility, localisation and spatiotemporal activity are at the core of cell biological processes and deregulation of these dynamic events can underpin disease development and progression. Recent advances in intravital imaging techniques in mice are providing new avenues to study real-time molecular behaviour in intact tissues within a live organism and to gain exciting insights into the intricate regulation of live cell biology at the microscale level. The monitoring of fluorescently labelled proteins and agents can be combined with autofluorescent properties of the microenvironment to provide a comprehensive snapshot of in vivo cell biology. In this Review, we summarise recent intravital microscopy approaches in mice, in processes ranging from normal development and homeostasis to disease progression and treatment in cancer, where we emphasise the utility of intravital imaging to observe dynamic and transient events in vivo We also highlight the recent integration of advanced subcellular imaging techniques into the intravital imaging pipeline, which can provide in-depth biological information beyond the single-cell level. We conclude with an outlook of ongoing developments in intravital microscopy towards imaging in humans, as well as provide an overview of the challenges the intravital imaging community currently faces and outline potential ways for overcoming these hurdles.
Collapse
Affiliation(s)
- Max Nobis
- The Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Cancer Division, St. Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Sydney, NSW 2010, Australia
| | - Sean C Warren
- The Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Cancer Division, St. Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Sydney, NSW 2010, Australia
| | - Morghan C Lucas
- The Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Cancer Division, St. Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Sydney, NSW 2010, Australia
| | - Kendelle J Murphy
- The Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Cancer Division, St. Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Sydney, NSW 2010, Australia
| | - David Herrmann
- The Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Cancer Division, St. Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Sydney, NSW 2010, Australia
| | - Paul Timpson
- The Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Cancer Division, St. Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Sydney, NSW 2010, Australia
| |
Collapse
|
154
|
Wu S, Huang Y, Tang Q, Li Z, Horng H, Li J, Wu Z, Chen Y, Li H. Quantitative evaluation of redox ratio and collagen characteristics during breast cancer chemotherapy using two-photon intrinsic imaging. BIOMEDICAL OPTICS EXPRESS 2018. [PMID: 29541528 PMCID: PMC5846538 DOI: 10.1364/boe.9.001375] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
Preoperative neoadjuvant treatment in locally advanced breast cancer is recognized as an effective adjuvant therapy, as it improves treatment outcomes. However, the potential complications remain a threat, so there is an urgent clinical need to assess both the tumor response and changes in its microenvironment using non-invasive and precise identification techniques. Here, two-photon microscopy was employed to detect morphological alterations in breast cancer progression and recession throughout chemotherapy. The changes in structure were analyzed based on the autofluorescence and collagen of differing statuses. Parameters, including optical redox ratio, the ratio of second harmonic generation and auto-fluorescence signal, collagen density, and collagen shape orientation, were studied. Results indicate that these parameters are potential indicators for evaluating breast tumors and their microenvironment changes during progression and chemotherapy. Combined analyses of these parameters could provide a quantitative, novel method for monitoring tumor therapy.
Collapse
Affiliation(s)
- Shulian Wu
- College of Photonic and Electronic Engineering, Fujian Normal University, Fujian Provincial Key Laboratory of Photonic Technology, Key Laboratory of Optoelectronic Science and Technology for Medicine, Ministry of Education, Fuzhou, Fujian, 350007, China
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, 20742, USA
- These authors contributed equally to this work
| | - Yudian Huang
- Department of Pathology, Fuzhou First Hospital Affiliated to Fujian Medical University, Fuzhou, Fujian, 350009, China
- These authors contributed equally to this work
| | - Qinggong Tang
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, 20742, USA
| | - Zhifang Li
- College of Photonic and Electronic Engineering, Fujian Normal University, Fujian Provincial Key Laboratory of Photonic Technology, Key Laboratory of Optoelectronic Science and Technology for Medicine, Ministry of Education, Fuzhou, Fujian, 350007, China
| | - Hannah Horng
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, 20742, USA
| | - Jiatian Li
- College of Photonic and Electronic Engineering, Fujian Normal University, Fujian Provincial Key Laboratory of Photonic Technology, Key Laboratory of Optoelectronic Science and Technology for Medicine, Ministry of Education, Fuzhou, Fujian, 350007, China
| | - Zaihua Wu
- Department of Pathology, Fuzhou First Hospital Affiliated to Fujian Medical University, Fuzhou, Fujian, 350009, China
| | - Yu Chen
- College of Photonic and Electronic Engineering, Fujian Normal University, Fujian Provincial Key Laboratory of Photonic Technology, Key Laboratory of Optoelectronic Science and Technology for Medicine, Ministry of Education, Fuzhou, Fujian, 350007, China
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, 20742, USA
| | - Hui Li
- College of Photonic and Electronic Engineering, Fujian Normal University, Fujian Provincial Key Laboratory of Photonic Technology, Key Laboratory of Optoelectronic Science and Technology for Medicine, Ministry of Education, Fuzhou, Fujian, 350007, China
| |
Collapse
|
155
|
Liu X, Flinders C, Mumenthaler SM, Hummon AB. MALDI Mass Spectrometry Imaging for Evaluation of Therapeutics in Colorectal Tumor Organoids. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2018; 29:516-526. [PMID: 29209911 PMCID: PMC5839975 DOI: 10.1007/s13361-017-1851-4] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Revised: 10/16/2017] [Accepted: 11/13/2017] [Indexed: 05/03/2023]
Abstract
Patient-derived colorectal tumor organoids (CTOs) closely recapitulate the complex morphological, phenotypic, and genetic features observed in in vivo tumors. Therefore, evaluation of drug distribution and metabolism in this model system can provide valuable information to predict the clinical outcome of a therapeutic response in individual patients. In this report, we applied matrix-assisted laser desorption/ionization mass spectrometry imaging (MALDI-MSI) to examine the spatial distribution of the drug irinotecan and its metabolites in CTOs from two patients. Irinotecan is a prodrug and is often prescribed as part of therapeutic regimes for patients with advanced colorectal cancer. Irinotecan shows a time-dependent and concentration-dependent permeability and metabolism in the CTOs. More interestingly, the active metabolite SN-38 does not co-localize well with the parent drug irinotecan and the inactive metabolite SN-38G. The phenotypic effect of irinotecan metabolism was also confirmed by a viability study showing significantly reduced proliferation in the drug treated CTOs. MALDI-MSI can be used to investigate various pharmaceutical compounds in CTOs derived from different patients. By analyzing multiple CTOs from a patient, this method could be used to predict patient-specific drug responses and help to improve personalized dosing regimens. Graphical Abstract ᅟ.
Collapse
Affiliation(s)
- Xin Liu
- Department of Chemistry and Biochemistry and the Harper Cancer Research Institute, University of Notre Dame, 140 McCourtney Hall, Notre Dame, IN, 46556, USA
| | - Colin Flinders
- Lawrence J. Ellison Institute for Transformative Medicine, University of Southern California, 2250 Alcazar Street, CSC 240, Los Angeles, CA, 90033, USA
| | - Shannon M Mumenthaler
- Lawrence J. Ellison Institute for Transformative Medicine, University of Southern California, 2250 Alcazar Street, CSC 240, Los Angeles, CA, 90033, USA
| | - Amanda B Hummon
- Department of Chemistry and Biochemistry and the Harper Cancer Research Institute, University of Notre Dame, 140 McCourtney Hall, Notre Dame, IN, 46556, USA.
| |
Collapse
|
156
|
Zhang Q, Jeppesen DK, Higginbotham JN, Demory Beckler M, Poulin EJ, Walsh AJ, Skala MC, McKinley ET, Manning HC, Hight MR, Schulte ML, Watt KR, Ayers GD, Wolf MM, Andrejeva G, Rathmell JC, Franklin JL, Coffey RJ. Mutant KRAS Exosomes Alter the Metabolic State of Recipient Colonic Epithelial Cells. Cell Mol Gastroenterol Hepatol 2018; 5:627-629.e6. [PMID: 29930982 PMCID: PMC6009797 DOI: 10.1016/j.jcmgh.2018.01.013] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Accepted: 01/12/2018] [Indexed: 01/18/2023]
Key Words
- 18F-FSPG, (S)-4-(3-[18F]-fluoropropyl)-L-glutamic acid
- Apc, adenomatous polyposis coli
- CRC, colorectal cancer
- DLD-1, Daniel L. Dexter derived 1
- FAD, flavin adenine dinucleotide
- GLUT-1, glucose transporter 1
- KO, knockout
- KRAS, Kirsten rat sarcoma viral oncogene homolog
- NADH, Nicotinamide adenine dinucleotide reduced
- WT, wild-type
Collapse
Affiliation(s)
- Qin Zhang
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Dennis K. Jeppesen
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | | | - Michelle Demory Beckler
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee,Department of Radiology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Emily J. Poulin
- Department of Cell and Developmental Biology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Alex J. Walsh
- Department of Biomedical Engineering, Vanderbilt University Medical Center, Nashville, Tennessee,Morgridge Institute for Research, University of Wisconsin, Madison, Wisconsin
| | - Melissa C. Skala
- Department of Biomedical Engineering, Vanderbilt University Medical Center, Nashville, Tennessee,Morgridge Institute for Research, University of Wisconsin, Madison, Wisconsin
| | - Eliot T. McKinley
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - H. Charles Manning
- Department of Radiology, Vanderbilt University Medical Center, Nashville, Tennessee,Department of Biomedical Engineering, Vanderbilt University Medical Center, Nashville, Tennessee,Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Matthew R. Hight
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, Tennessee,Department of Physics and Astronomy, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Michael L. Schulte
- Department of Radiology, Vanderbilt University Medical Center, Nashville, Tennessee,Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Kimberly R. Watt
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee,Digestive Disease Research Center, Vanderbilt University Medical Center, Nashville, Tennessee
| | - G. Daniel Ayers
- Biostatistics Center, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Melissa M. Wolf
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee,Vanderbilt Center for Immunobiology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Gabriela Andrejeva
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee,Vanderbilt Center for Immunobiology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Jeffrey C. Rathmell
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee,Vanderbilt Center for Immunobiology, Vanderbilt University Medical Center, Nashville, Tennessee,Department of Cancer Biology, Vanderbilt University, Nashville, Tennessee
| | - Jeffrey L. Franklin
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee,Department of Cell and Developmental Biology, Vanderbilt University Medical Center, Nashville, Tennessee,Digestive Disease Research Center, Vanderbilt University Medical Center, Nashville, Tennessee,Department of Veterans Affairs Medical Center, Nashville, Tennessee
| | - Robert J. Coffey
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee,Department of Cell and Developmental Biology, Vanderbilt University Medical Center, Nashville, Tennessee,Department of Veterans Affairs Medical Center, Nashville, Tennessee,Corresponding author:
| |
Collapse
|
157
|
Kapur A, Beres T, Rathi K, Nayak AP, Czarnecki A, Felder M, Gillette A, Ericksen SS, Sampene E, Skala MC, Barroilhet L, Patankar MS. Oxidative stress via inhibition of the mitochondrial electron transport and Nrf-2-mediated anti-oxidative response regulate the cytotoxic activity of plumbagin. Sci Rep 2018; 8:1073. [PMID: 29348410 PMCID: PMC5773707 DOI: 10.1038/s41598-018-19261-w] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Accepted: 12/19/2017] [Indexed: 11/15/2022] Open
Abstract
Plumbagin, an anti-cancer agent, is toxic to cells of multiple species. We investigated if plumbagin targets conserved biochemical processes. Plumbagin induced DNA damage and apoptosis in cells of diverse mutational background with comparable potency. A 3-5 fold increase in intracellular oxygen radicals occurred in response to plumbagin. Neutralization of the reactive oxygen species by N-acetylcysteine blocked apoptosis, indicating a central role for oxidative stress in plumbagin-mediated cell death. Plumbagin docks in the ubiquinone binding sites (Q0 and Qi) of mitochondrial complexes I-III, the major sites for oxygen radicals. Plumbagin decreased oxygen consumption rate, ATP production and optical redox ratio (NAD(P)H/FAD) indicating interference with electron transport downstream of mitochondrial Complex II. Oxidative stress induced by plumbagin triggered an anti-oxidative response via activation of Nrf2. Plumbagin and the Nrf2 inhibitor, brusatol, synergized to inhibit cell proliferation. These data indicate that while inhibition of electron transport is the conserved mechanism responsible for plumbagin's chemotoxicity, activation of Nrf2 is the resulting anti-oxidative response that allows plumbagin to serve as a chemopreventive agent. This study provides the basis for designing potent and selective plumbagin analogs that can be coupled with suitable Nrf2 inhibitors for chemotherapy or administered as single agents to induce Nrf2-mediated chemoprevention.
Collapse
Affiliation(s)
- Arvinder Kapur
- Department of Obstetrics and Gynecology, University of Wisconsin-Madison, Madison, WI, 53792-6188, USA.
| | - Thomas Beres
- Department of Obstetrics and Gynecology, University of Wisconsin-Madison, Madison, WI, 53792-6188, USA
| | - Kavya Rathi
- Department of Obstetrics and Gynecology, University of Wisconsin-Madison, Madison, WI, 53792-6188, USA
| | - Amruta P Nayak
- Department of Obstetrics and Gynecology, University of Wisconsin-Madison, Madison, WI, 53792-6188, USA
- Indian Institute for Science Education and Research, Pune, India
| | - Austin Czarnecki
- Department of Obstetrics and Gynecology, University of Wisconsin-Madison, Madison, WI, 53792-6188, USA
| | - Mildred Felder
- Department of Obstetrics and Gynecology, University of Wisconsin-Madison, Madison, WI, 53792-6188, USA
| | - Amani Gillette
- Morgridge Institute for Research and the Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, USA
| | - Spencer S Ericksen
- Small Molecule Screening Facility, University of Wisconsin Paul P. Carbone Comprehensive Cancer Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Emmanuel Sampene
- Department of Biostatistics, University of Wisconsin-Madison, Madison, WI, USA
| | - Melissa C Skala
- Morgridge Institute for Research and the Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, USA
| | - Lisa Barroilhet
- Department of Obstetrics and Gynecology, University of Wisconsin-Madison, Madison, WI, 53792-6188, USA
| | - Manish S Patankar
- Department of Obstetrics and Gynecology, University of Wisconsin-Madison, Madison, WI, 53792-6188, USA.
| |
Collapse
|
158
|
Chen Y, Qian J, He Q, Zhao H, Toral-Barza L, Shi C, Zhang X, Wu J, Yu K. mTOR complex-2 stimulates acetyl-CoA and de novo lipogenesis through ATP citrate lyase in HER2/PIK3CA-hyperactive breast cancer. Oncotarget 2018; 7:25224-40. [PMID: 27015560 PMCID: PMC5041899 DOI: 10.18632/oncotarget.8279] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Accepted: 03/05/2016] [Indexed: 12/22/2022] Open
Abstract
The mechanistic target of rapamycin (mTOR) is a major regulator of cell growth and is frequently dysregulated in cancer. While mTOR complex-1 (mTORC1) is a validated cancer target, the role of mTOR complex-2 (mTORC2) remains less defined. Here, we reveal mTORC2 as a critical regulator of breast cancer metabolism. We showed that hyperphosphorylation in ATP citrate lyase (ACL) occurs frequently in human breast tumors and correlates well with HER2+ and/or PIK3CA-mutant (HER2+/PIK3CAmut) status in breast tumor cell lines. In HER2+/PIK3CAmut cells, mTORC2 controls Ser-455 phosphorylation of ACL thereby promoting acetyl-CoA production, de novo lipogenesis and mitochondrial physiology, all of which were inhibited by an mTORC1/mTORC2 kinase inhibitor (mTOR-KI) or cellular depletion of mTORC2 or ACL. mTOR-KI but not rapamycin blocked the IGF-1-induced ACL phosphorylation and glucose to lipid conversion. Depletion of mTORC2 but not mTORC1 specifically inhibited the ACL-dependent acetyl-CoA production. In the HER2+/PIK3CAmut MDA361, MDA453, BT-474 and T47D cells, depletion of mTORC2 or ACL led to growth inhibition and mitochondrial hyperpolarization, which were partially rescued by an alternate source of acetyl-CoA. These same changes were not apparent in mTORC2- or ACL-depleted HER2-/PIK3CAwt MDA231 and HCC1806 cells, highlighting a differential dependence of mTORC2-ACL for survival in these two cell types. Moreover, ACL Ser-455 mutants S455E (phosphomimetic) and S455A (non-phosphorylatable) each increased or decreased, respectively, the acetyl-CoA production, mitochondrial homeostasis and survival in ACL-depleted MDA453 cells. These studies define a new and rapamycin-resistant mechanism of mTORC2-ACL in lipogenesis and acetyl-CoA biology and provide a rationale for targeting of mTORC1 and mTORC2 in HER2+/PIK3CAmut breast cancer.
Collapse
Affiliation(s)
- Yaqing Chen
- Department of Pharmacology, Fudan University School of Pharmacy, Shanghai, China
| | - Jianchang Qian
- Department of Pharmacology, Fudan University School of Pharmacy, Shanghai, China
| | - Qun He
- Department of Pharmacology, Fudan University School of Pharmacy, Shanghai, China
| | - Hui Zhao
- Department of Pharmacology, Fudan University School of Pharmacy, Shanghai, China
| | | | - Celine Shi
- Oncology Research, Pfizer Pharmaceuticals, Pearl River, NY, USA
| | - Xuesai Zhang
- Department of Pharmacology, Fudan University School of Pharmacy, Shanghai, China
| | - Jiang Wu
- Oncology Research, Pfizer Pharmaceuticals, Pearl River, NY, USA
| | - Ker Yu
- Department of Pharmacology, Fudan University School of Pharmacy, Shanghai, China
| |
Collapse
|
159
|
Heaster TM, Walsh AJ, Zhao Y, Hiebert SW, Skala MC. Autofluorescence imaging identifies tumor cell-cycle status on a single-cell level. JOURNAL OF BIOPHOTONICS 2018; 11:10.1002/jbio.201600276. [PMID: 28485124 PMCID: PMC5680147 DOI: 10.1002/jbio.201600276] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Revised: 02/13/2017] [Accepted: 02/15/2017] [Indexed: 05/13/2023]
Abstract
The goal of this study is to validate fluorescence intensity and lifetime imaging of metabolic co-enzymes NAD(P)H and FAD (optical metabolic imaging, or OMI) as a method to quantify cell-cycle status of tumor cells. Heterogeneity in tumor cell-cycle status (e. g. proliferation, quiescence, apoptosis) increases drug resistance and tumor recurrence. Cell-cycle status is closely linked to cellular metabolism. Thus, this study applies cell-level metabolic imaging to distinguish proliferating, quiescent, and apoptotic populations. Two-photon microscopy and time-correlated single photon counting are used to measure optical redox ratio (NAD(P)H fluorescence intensity divided by FAD intensity), NAD(P)H and FAD fluorescence lifetime parameters. Redox ratio, NAD(P)H and FAD lifetime parameters alone exhibit significant differences (p<0.05) between population means. To improve separation between populations, linear combination models derived from partial least squares - discriminant analysis (PLS-DA) are used to exploit all measurements together. Leave-one-out cross validation of the model yielded high classification accuracies (92.4 and 90.1 % for two and three populations, respectively). OMI and PLS-DA also identifies each sub-population within heterogeneous samples. These results establish single-cell analysis with OMI and PLS-DA as a label-free method to distinguish cell-cycle status within intact samples. This approach could be used to incorporate cell-level tumor heterogeneity in cancer drug development.
Collapse
Affiliation(s)
- Tiffany M. Heaster
- Department of Biomedical Engineering, University of Wisconsin,
Madison, Wisconsin, 53715, USA
| | - Alex J. Walsh
- National Research Council, JBSA Fort Sam Houston, Texas, 78234,
USA
- 711 Human Performance Wing, Human Effectiveness
Directorate, Bioeffects Division Optical Radiation Branch, Air Force Research Lab,
JBSA Fort Sam Houston, Texas, 78234, USA
| | - Yue Zhao
- Department of Biochemistry, Vanderbilt University School of
Medicine, Nashville, Tennessee, 37232, USA
| | - Scott W. Hiebert
- Department of Biochemistry, Vanderbilt University School of
Medicine, Nashville, Tennessee, 37232, USA
- Vanderbilt-Ingram Cancer Center, Nashville, Tennessee, 37232,
USA
| | - Melissa C. Skala
- Department of Biomedical Engineering, University of Wisconsin,
Madison, Wisconsin, 53715, USA
- Morgridge Institute for Research, Madison, Wisconsin, 53715,
USA
| |
Collapse
|
160
|
Sanchez T, Seidler EA, Gardner DK, Needleman D, Sakkas D. Will noninvasive methods surpass invasive for assessing gametes and embryos? Fertil Steril 2017; 108:730-737. [DOI: 10.1016/j.fertnstert.2017.10.004] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Revised: 09/20/2017] [Accepted: 10/02/2017] [Indexed: 11/27/2022]
|
161
|
Ogrodzinski MP, Bernard JJ, Lunt SY. Deciphering metabolic rewiring in breast cancer subtypes. Transl Res 2017; 189:105-122. [PMID: 28774752 DOI: 10.1016/j.trsl.2017.07.004] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Revised: 06/02/2017] [Accepted: 07/11/2017] [Indexed: 02/07/2023]
Abstract
Metabolic reprogramming, an emerging hallmark of cancer, is observed in breast cancer. Breast cancer cells rewire their cellular metabolism to meet the demands of survival, proliferation, and invasion. However, breast cancer is a heterogeneous disease, and metabolic rewiring is not uniform. Each subtype of breast cancer displays distinct metabolic alterations. Here, we focus on unique metabolic reprogramming associated with subtypes of breast cancer, as well as common features. Therapeutic opportunities based on subtype-specific metabolic alterations are also discussed. Through this discussion, we aim to provide insight into subtype-specific metabolic rewiring and vulnerabilities that have the potential to better guide therapy and improve outcomes for patients.
Collapse
Affiliation(s)
- Martin P Ogrodzinski
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, Mich; Department of Physiology, Michigan State University, East Lansing, Mich
| | - Jamie J Bernard
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Mich
| | - Sophia Y Lunt
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, Mich; Department of Chemical Engineering and Materials Science, Michigan State University, East Lansing, Mich.
| |
Collapse
|
162
|
Zhu C, Martinez AF, Martin HL, Li M, Crouch BT, Carlson DA, Haystead TAJ, Ramanujam N. Near-simultaneous intravital microscopy of glucose uptake and mitochondrial membrane potential, key endpoints that reflect major metabolic axes in cancer. Sci Rep 2017; 7:13772. [PMID: 29062013 PMCID: PMC5653871 DOI: 10.1038/s41598-017-14226-x] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Accepted: 10/06/2017] [Indexed: 12/19/2022] Open
Abstract
While the demand for metabolic imaging has increased in recent years, simultaneous in vivo measurement of multiple metabolic endpoints remains challenging. Here we report on a novel technique that provides in vivo high-resolution simultaneous imaging of glucose uptake and mitochondrial metabolism within a dynamic tissue microenvironment. Two indicators were leveraged; 2-[N-(7-nitrobenz-2-oxa-1, 3-diazol-4-yl) amino]-2-deoxy-D-glucose (2-NBDG) reports on glucose uptake and Tetramethylrhodamine ethyl ester (TMRE) reports on mitochondrial membrane potential. Although we demonstrated that there was neither optical nor chemical crosstalk between 2-NBDG and TMRE, TMRE uptake was significantly inhibited by simultaneous injection with 2-NBDG in vivo. A staggered delivery scheme of the two agents (TMRE injection was followed by 2-NBDG injection after a 10-minute delay) permitted near-simultaneous in vivo microscopy of 2-NBDG and TMRE at the same tissue site by mitigating the interference of 2-NBDG with normal glucose usage. The staggered delivery strategy was evaluated under both normoxic and hypoxic conditions in normal tissues as well as in a murine breast cancer model. The results were consistent with those expected for independent imaging of 2-NBDG and TMRE. This optical imaging technique allows for monitoring of key metabolic endpoints with the unique benefit of repeated, non-destructive imaging within an intact microenvironment.
Collapse
Affiliation(s)
- Caigang Zhu
- Department of Biomedical Engineering, Duke University, Durham, NC, 27708, USA
| | - Amy F Martinez
- Department of Biomedical Engineering, Duke University, Durham, NC, 27708, USA
| | - Hannah L Martin
- Department of Biomedical Engineering, Duke University, Durham, NC, 27708, USA
| | - Martin Li
- Department of Biomedical Engineering, Duke University, Durham, NC, 27708, USA
| | - Brian T Crouch
- Department of Biomedical Engineering, Duke University, Durham, NC, 27708, USA
| | - David A Carlson
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC, 27710, USA
| | - Timothy A J Haystead
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC, 27710, USA
| | - Nimmi Ramanujam
- Department of Biomedical Engineering, Duke University, Durham, NC, 27708, USA.
| |
Collapse
|
163
|
Schaefer PM, Hilpert D, Niederschweiberer M, Neuhauser L, Kalinina S, Calzia E, Rueck A, von Einem B, von Arnim CAF. Mitochondrial matrix pH as a decisive factor in neurometabolic imaging. NEUROPHOTONICS 2017; 4:045004. [PMID: 29181426 PMCID: PMC5685807 DOI: 10.1117/1.nph.4.4.045004] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Accepted: 10/16/2017] [Indexed: 06/07/2023]
Abstract
Alterations of cellular bioenergetics are a common feature in most neurodegenerative disorders. However, there is a selective vulnerability of different brain regions, cell types, and even mitochondrial populations to these metabolic disturbances. Thus, the aim of our study was to establish and validate an in vivo metabolic imaging technique to screen for mitochondrial function on the subcellular level. Based on nicotinamide adenine dinucleotide (phosphate) fluorescence lifetime imaging microscopy [NAD(P)H FLIM], we performed a quantitative correlation to high-resolution respirometry. Thereby, we revealed mitochondrial matrix pH as a decisive factor in imaging NAD(P)H redox state. By combining both parameters, we illustrate a quantitative, high-resolution assessment of mitochondrial function in metabolically modified cells as well as in an amyloid precursor protein-overexpressing model of Alzheimer's disease. Our metabolic imaging technique provides the basis for dissecting mitochondrial deficits not only in a range of neurodegenerative diseases, shedding light onto bioenergetic failures of cells remaining in their metabolic microenvironment.
Collapse
Affiliation(s)
| | - Diana Hilpert
- Ulm University, Department of Neurology, Ulm, Germany
| | | | | | - Sviatlana Kalinina
- Ulm University, Core Facility Confocal and Multiphoton Microscopy, Ulm, Germany
| | - Enrico Calzia
- University Medical School, Institute of Anesthesiological Pathophysiology and Process Engineering, Ulm, Germany
| | - Angelika Rueck
- Ulm University, Core Facility Confocal and Multiphoton Microscopy, Ulm, Germany
| | | | | |
Collapse
|
164
|
Peters S, Griebsch M, Klemm M, Haueisen J, Hammer M. Hydrogen peroxide modulates energy metabolism and oxidative stress in cultures of permanent human Müller cells MIO-M1. JOURNAL OF BIOPHOTONICS 2017; 10:1180-1188. [PMID: 27896951 DOI: 10.1002/jbio.201600201] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Revised: 10/25/2016] [Accepted: 11/14/2016] [Indexed: 05/14/2023]
Abstract
In this study the influence of hydrogen peroxide (H2 O2 ) on the redox state, NADH protein binding, and mitochondrial membrane potential in Müller cells is investigated. Cultures of permanent human Müller cells MIO-M1 were exposed to H2 O2 in 75 µM and 150 µM concentration for two hours. Fluorescence emission spectra and lifetimes were measured by two-photon microscopy (excitation wavelength: 740 nm) at the mitochondria which were identified in the microscopic images by their fluorescence properties (spectra and intensity). Two hours of H2 O2 exposure did not impair viability of MIO-M1 cells in culture. Whereas the ratio of flavine- to NADH fluorescence intensity did not change under either H2 O2 concentration, the mean lifetime was significantly different between controls, not exposed to H2 O2 , and the 150 µM H2 O2 exposure (972 ± 63 ps vs. 1152 ± 64 ps, p = 0.014). One hour after cessation of the H2 O2 exposure, the value retuned to that of the control (983 ± 36 ps). A hyperpolarization of the mitochondrial membrane under 150 µM H2 O2 was found. These findings suggest a shift form free to protein-bound NADH in mitochondria as well as a hyperpolarization of their inner membrane which could be related to an impairment of Müller cell function despite their preserved viability. Exposure of human Müller cells to hydrogen peroxide for two hours results in a reversible change of protein binding of mitochondrial NADH upon unchanged redox ratio. The mitochondrial membrane potential is increased during exposure.
Collapse
Affiliation(s)
- Sven Peters
- University Hospital Jena, Department of Ophthalmology, 07743, Jena, Germany
| | - Max Griebsch
- University Hospital Jena, Department of Ophthalmology, 07743, Jena, Germany
| | - Matthias Klemm
- Technical University Ilmenau, Int. for Biomedical Engineering and Informatics, Gustav-Kirchhoff-Str. 2, 98693, Ilmenau, Germany
| | - Jens Haueisen
- Technical University Ilmenau, Int. for Biomedical Engineering and Informatics, Gustav-Kirchhoff-Str. 2, 98693, Ilmenau, Germany
| | - Martin Hammer
- University Hospital Jena, Department of Ophthalmology, 07743, Jena, Germany
- University of Jena, Center for Biomedical Optics and Photonics, 07740, Jena, Germany
| |
Collapse
|
165
|
Shah AT, Cannon TM, Higginbotham JN, Coffey RJ, Skala MC. Autofluorescence flow sorting of breast cancer cell metabolism. JOURNAL OF BIOPHOTONICS 2017; 10:1026-1033. [PMID: 27730745 PMCID: PMC5547001 DOI: 10.1002/jbio.201600128] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Revised: 07/21/2016] [Accepted: 07/24/2016] [Indexed: 05/05/2023]
Abstract
Clinical cancer treatment aims to target all cell subpopulations within a tumor. Autofluorescence microscopy of the metabolic cofactors NAD(P)H and FAD has shown sensitivity to anti-cancer treatment response. Alternatively, flow cytometry is attractive for high throughput analysis and flow sorting. This study measures cellular autofluorescence in three flow cytometry channels and applies cellular autofluorescence to sort a heterogeneous mixture of breast cancer cells into subpopulations enriched for each phenotype. Sorted cells were grown in culture and sorting was validated by morphology, autofluorescence microscopy, and receptor expression. Ultimately, this method could be applied to improve drug development and personalized treatment planning.
Collapse
Affiliation(s)
- Amy T. Shah
- Department of Biomedical Engineering, Vanderbilt University, Station B, Box 1631, Nashville, TN 37235, USA
| | - Taylor M. Cannon
- Department of Biomedical Engineering, Vanderbilt University, Station B, Box 1631, Nashville, TN 37235, USA
| | - James N. Higginbotham
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Robert J. Coffey
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Melissa C. Skala
- Morgridge Institute for Research, University of Wisconsin, Madison, WI 53715, USA
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI 53715, USA
- Corresponding author: , Phone: 608-316-4108
| |
Collapse
|
166
|
Stringari C, Abdeladim L, Malkinson G, Mahou P, Solinas X, Lamarre I, Brizion S, Galey JB, Supatto W, Legouis R, Pena AM, Beaurepaire E. Multicolor two-photon imaging of endogenous fluorophores in living tissues by wavelength mixing. Sci Rep 2017; 7:3792. [PMID: 28630487 PMCID: PMC5476668 DOI: 10.1038/s41598-017-03359-8] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Accepted: 04/27/2017] [Indexed: 12/26/2022] Open
Abstract
Two-photon imaging of endogenous fluorescence can provide physiological and metabolic information from intact tissues. However, simultaneous imaging of multiple intrinsic fluorophores, such as nicotinamide adenine dinucleotide(phosphate) (NAD(P)H), flavin adenine dinucleotide (FAD) and retinoids in living systems is generally hampered by sequential multi-wavelength excitation resulting in motion artifacts. Here, we report on efficient and simultaneous multicolor two-photon excitation of endogenous fluorophores with absorption spectra spanning the 750-1040 nm range, using wavelength mixing. By using two synchronized pulse trains at 760 and 1041 nm, an additional equivalent two-photon excitation wavelength at 879 nm is generated, and achieves simultaneous excitation of blue, green and red intrinsic fluorophores. This method permits an efficient simultaneous imaging of the metabolic coenzymes NADH and FAD to be implemented with perfect image co-registration, overcoming the difficulties associated with differences in absorption spectra and disparity in concentration. We demonstrate ratiometric redox imaging free of motion artifacts and simultaneous two-photon fluorescence lifetime imaging (FLIM) of NADH and FAD in living tissues. The lifetime gradients of NADH and FAD associated with different cellular metabolic and differentiation states in reconstructed human skin and in the germline of live C. Elegans are thus simultaneously measured. Finally, we present multicolor imaging of endogenous fluorophores and second harmonic generation (SHG) signals during the early stages of Zebrafish embryo development, evidencing fluorescence spectral changes associated with development.
Collapse
Affiliation(s)
- Chiara Stringari
- Laboratory for Optics and Biosciences, Ecole polytechnique, CNRS, INSERM, Université Paris-Saclay, 91128, Palaiseau cedex, France.
| | - Lamiae Abdeladim
- Laboratory for Optics and Biosciences, Ecole polytechnique, CNRS, INSERM, Université Paris-Saclay, 91128, Palaiseau cedex, France
| | - Guy Malkinson
- Laboratory for Optics and Biosciences, Ecole polytechnique, CNRS, INSERM, Université Paris-Saclay, 91128, Palaiseau cedex, France
| | - Pierre Mahou
- Laboratory for Optics and Biosciences, Ecole polytechnique, CNRS, INSERM, Université Paris-Saclay, 91128, Palaiseau cedex, France
| | - Xavier Solinas
- Laboratory for Optics and Biosciences, Ecole polytechnique, CNRS, INSERM, Université Paris-Saclay, 91128, Palaiseau cedex, France
| | - Isabelle Lamarre
- Laboratory for Optics and Biosciences, Ecole polytechnique, CNRS, INSERM, Université Paris-Saclay, 91128, Palaiseau cedex, France
| | | | | | - Willy Supatto
- Laboratory for Optics and Biosciences, Ecole polytechnique, CNRS, INSERM, Université Paris-Saclay, 91128, Palaiseau cedex, France
| | - Renaud Legouis
- Institute for Integrative Biology of the Cell (I2BC), CEA, CNRS, Univ. Paris-Sud, Université Paris-Saclay, 91198, Gif-sur-Yvette, France
| | - Ana-Maria Pena
- L'Oréal Research and Innovation, 93600, Aulnay sous Bois, France
| | - Emmanuel Beaurepaire
- Laboratory for Optics and Biosciences, Ecole polytechnique, CNRS, INSERM, Université Paris-Saclay, 91128, Palaiseau cedex, France.
| |
Collapse
|
167
|
Bhattacharjee A, Datta R, Gratton E, Hochbaum AI. Metabolic fingerprinting of bacteria by fluorescence lifetime imaging microscopy. Sci Rep 2017; 7:3743. [PMID: 28623341 PMCID: PMC5473825 DOI: 10.1038/s41598-017-04032-w] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Accepted: 05/22/2017] [Indexed: 12/21/2022] Open
Abstract
Bacterial populations exhibit a range of metabolic states influenced by their environment, intra- and interspecies interactions. The identification of bacterial metabolic states and transitions between them in their native environment promises to elucidate community behavior and stochastic processes, such as antibiotic resistance acquisition. In this work, we employ two-photon fluorescence lifetime imaging microscopy (FLIM) to create a metabolic fingerprint of individual bacteria and populations. FLIM of autofluorescent reduced nicotinamide adenine dinucleotide (phosphate), NAD(P)H, has been previously exploited for label-free metabolic imaging of mammalian cells. However, NAD(P)H FLIM has not been established as a metabolic proxy in bacteria. Applying the phasor approach, we create FLIM-phasor maps of Escherichia coli, Salmonella enterica serovar Typhimurium, Pseudomonas aeruginosa, Bacillus subtilis, and Staphylococcus epidermidis at the single cell and population levels. The bacterial phasor is sensitive to environmental conditions such as antibiotic exposure and growth phase, suggesting that observed shifts in the phasor are representative of metabolic changes within the cells. The FLIM-phasor approach represents a powerful, non-invasive imaging technique to study bacterial metabolism in situ and could provide unique insights into bacterial community behavior, pathology and antibiotic resistance with sub-cellular resolution.
Collapse
Affiliation(s)
- Arunima Bhattacharjee
- Department of Chemical Engineering and Materials Science, University of California, Irvine, Irvine, CA, 92697, USA
| | - Rupsa Datta
- Laboratory for Fluorescence Dynamics, Department of Biomedical Engineering, University of California, Irvine, Irvine, CA, 92697, USA
| | - Enrico Gratton
- Laboratory for Fluorescence Dynamics, Department of Biomedical Engineering, University of California, Irvine, Irvine, CA, 92697, USA
| | - Allon I Hochbaum
- Department of Chemical Engineering and Materials Science, University of California, Irvine, Irvine, CA, 92697, USA.
- Department of Chemistry, University of California, Irvine, Irvine, CA, 92697, USA.
| |
Collapse
|
168
|
Walsh AJ, Cook RS, Skala MC. Functional Optical Imaging of Primary Human Tumor Organoids: Development of a Personalized Drug Screen. J Nucl Med 2017; 58:1367-1372. [PMID: 28588148 DOI: 10.2967/jnumed.117.192534] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Accepted: 05/26/2017] [Indexed: 12/24/2022] Open
Abstract
Primary tumor organoids are a robust model of individual human cancers and present a unique platform for patient-specific drug testing. Optical imaging is uniquely suited to assess organoid function and behavior because of its subcellular resolution, penetration depth through the entire organoid, and functional endpoints. Specifically, optical metabolic imaging (OMI) is highly sensitive to drug response in organoids, and OMI in tumor organoids correlates with primary tumor drug response. Therefore, an OMI organoid drug screen could enable accurate testing of drug response for individualized cancer treatment. The objective of this perspective is to introduce OMI and tumor organoids to a general audience in order to foster the adoption of these techniques in diverse clinical and laboratory settings.
Collapse
Affiliation(s)
- Alex J Walsh
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee
| | - Rebecca S Cook
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee.,Department of Cancer Biology, Vanderbilt University, Nashville, Tennessee
| | - Melissa C Skala
- Department of Biomedical Engineering, University of Wisconsin, Madison, Wisconsin; and .,Morgridge Institute for Research, Madison, Wisconsin
| |
Collapse
|
169
|
Domingue SR, Bartels RA, Chicco AJ, Wilson JW. Transient absorption imaging of hemes with 2-color, independently tunable visible-wavelength ultrafast source. BIOMEDICAL OPTICS EXPRESS 2017; 8:2807-2821. [PMID: 28663908 PMCID: PMC5480431 DOI: 10.1364/boe.8.002807] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Revised: 04/21/2017] [Accepted: 04/22/2017] [Indexed: 06/07/2023]
Abstract
Pump probe microscopy is a time-resolved multiphoton imaging technique capable of generating contrast between non-fluorescent pigments based on differences in excited-state lifetimes. Here we describe a fiber-based ultrafast system designed for imaging heme proteins with an independently-tunable pulse pair in the visible-wavelength regime. Starting with a 1060 nm fiber amplifier (1.3 W at 63 MHz, 140 fs pulses), visible pulses were produced in the vicinity of 488 nm and 532 nm by doubling the output of a short photonic crystal fiber with a pair of periodically-poled lithium niobate crystals, providing 5-20 mW power in each beam. This was sufficient for acquiring transient absorption images from unstained cryosectioned tissue.
Collapse
Affiliation(s)
- Scott R. Domingue
- Department of Electrical & Computer Engineering, Colorado State University, USA
- Current affiliation: KMLabs, Boulder, CO,
USA
| | - Randy A. Bartels
- Department of Electrical & Computer Engineering, Colorado State University, USA
- School of Biomedical Engineering, Colorado State University, USA
| | - Adam J. Chicco
- Department of Biomedical Sciences, Colorado State University, USA
- School of Biomedical Engineering, Colorado State University, USA
| | - Jesse W. Wilson
- Department of Electrical & Computer Engineering, Colorado State University, USA
- School of Biomedical Engineering, Colorado State University, USA
| |
Collapse
|
170
|
Yu F, Zhuo S, Qu Y, Choudhury D, Wang Z, Iliescu C, Yu H. On chip two-photon metabolic imaging for drug toxicity testing. BIOMICROFLUIDICS 2017; 11:034108. [PMID: 28529673 PMCID: PMC5426952 DOI: 10.1063/1.4983615] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/27/2016] [Accepted: 05/03/2017] [Indexed: 05/03/2023]
Abstract
We have developed a microfluidic system suitable to be incorporated with a metabolic imaging method to monitor the drug response of cells cultured on a chip. The cells were perfusion-cultured to mimic the blood flow in vivo. Label-free optical measurements and imaging of nicotinamide adenine dinucleotide and flavin adenine dinucleotide fluorescence intensity and morphological changes were evaluated non-invasively. Drug responses calculated using redox ratio imaging were compared with the drug toxicity testing results obtained with a traditional well-plate system. We found that our method can accurately monitor the cell viability and drug response and that the IC50 value obtained from imaging analysis was sensitive and comparable with a commonly used cell viability assay: MTS (3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfo-phenyl)-2H-tetrazolium) assay. Our method could serve as a fast, non-invasive, and reliable way for drug screening and toxicity testing as well as enabling real-time monitoring of in vitro cultured cells.
Collapse
Affiliation(s)
| | | | | | - Deepak Choudhury
- Singapore Institute of Manufacturing Technology, ASTAR, 71 Nanyang Dr, Singapore, Singapore, 638075
| | - Zhiping Wang
- Singapore Institute of Manufacturing Technology, ASTAR, 71 Nanyang Dr, Singapore, Singapore, 638075
| | | | | |
Collapse
|
171
|
Conway JRW, Warren SC, Timpson P. Context-dependent intravital imaging of therapeutic response using intramolecular FRET biosensors. Methods 2017; 128:78-94. [PMID: 28435000 DOI: 10.1016/j.ymeth.2017.04.014] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Revised: 03/13/2017] [Accepted: 04/08/2017] [Indexed: 12/18/2022] Open
Abstract
Intravital microscopy represents a more physiologically relevant method for assessing therapeutic response. However, the movement into an in vivo setting brings with it several additional considerations, the primary being the context in which drug activity is assessed. Microenvironmental factors, such as hypoxia, pH, fibrosis, immune infiltration and stromal interactions have all been shown to have pronounced effects on drug activity in a more complex setting, which is often lost in simpler two- or three-dimensional assays. Here we present a practical guide for the application of intravital microscopy, looking at the available fluorescent reporters and their respective expression systems and analysis considerations. Moving in vivo, we also discuss the microscopy set up and methods available for overlaying microenvironmental context to the experimental readouts. This enables a smooth transition into applying higher fidelity intravital imaging to improve the drug discovery process.
Collapse
Affiliation(s)
- James R W Conway
- Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Cancer Division, Sydney, NSW 2010, Australia; St Vincent's Clinical School, Faculty of Medicine, University of NSW, Sydney, NSW 2010, Australia
| | - Sean C Warren
- Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Cancer Division, Sydney, NSW 2010, Australia; St Vincent's Clinical School, Faculty of Medicine, University of NSW, Sydney, NSW 2010, Australia
| | - Paul Timpson
- Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Cancer Division, Sydney, NSW 2010, Australia; St Vincent's Clinical School, Faculty of Medicine, University of NSW, Sydney, NSW 2010, Australia.
| |
Collapse
|
172
|
Cannon TM, Shah AT, Skala MC. Autofluorescence imaging captures heterogeneous drug response differences between 2D and 3D breast cancer cultures. BIOMEDICAL OPTICS EXPRESS 2017; 8:1911-1925. [PMID: 28663873 PMCID: PMC5480588 DOI: 10.1364/boe.8.001911] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Revised: 02/02/2017] [Accepted: 02/12/2017] [Indexed: 05/13/2023]
Abstract
Two-photon microscopy of cellular autofluorescence intensity and lifetime (optical metabolic imaging, or OMI) is a promising tool for preclinical drug development. OMI, which exploits the endogenous fluorescence from the metabolic coenzymes NAD(P)H and FAD, is sensitive to changes in cell metabolism produced by drug treatment. Previous studies have shown that drug response, genetic expression, cell-cell communication, and cell signaling in 3D culture match those of the original in vivo tumor, but not those of 2D culture. The goal of this study is to use OMI to quantify dynamic cell-level metabolic differences in drug response in 2D cell lines vs. 3D organoids generated from xenograft tumors of the same cell origin. BT474 cells and Herceptin-resistant BT474 (HR6) cells were tested. Cells were treated with vehicle control, Herceptin, XL147 (PI3K inhibitor), and the combination. The OMI index was used to quantify response, and is a linear combination of the redox ratio (intensity of NAD(P)H divided by FAD), mean NADH lifetime, and mean FAD lifetime. The results confirm that the OMI index resolves significant differences (p<0.05) in drug response for 2D vs. 3D cultures, specifically for BT474 cells 24 hours after Herceptin treatment, for HR6 cells 24 and 72 hours after combination treatment, and for HR6 cells 72 hours after XL147 treatment. Cell-level analysis of the OMI index also reveals differences in the number of cell sub-populations in 2D vs. 3D culture at 24, 48, and 72 hours post-treatment in control and treated groups. Finally, significant increases (p<0.05) in the mean lifetime of NADH and FAD were measured in 2D vs. 3D for both cell lines at 72 hours post-treatment in control and all treatment groups. These whole-population differences in the mean NADH and FAD lifetimes are supported by differences in the number of cell sub-populations in 2D vs. 3D. Overall, these studies confirm that OMI is sensitive to differences in drug response in 2D vs. 3D, and provides further information on dynamic changes in the relative abundance of metabolic cell sub-populations that contribute to this difference.
Collapse
Affiliation(s)
- T. M. Cannon
- Department of Biomedical Engineering, Vanderbilt University, Station B, Box 1631, Nashville, TN 37235, USA
| | - A. T. Shah
- Department of Biomedical Engineering, Vanderbilt University, Station B, Box 1631, Nashville, TN 37235, USA
| | - M. C. Skala
- Morgridge Institute for Research, University of Wisconsin—Madison, 330 North Orchard Street, Madison, WI 53715, USA
- Department of Biomedical Engineering, University of Wisconsin—Madison, 1415 Engineering Drive, Madison, WI 53706, USA
| |
Collapse
|
173
|
Pastore MN, Studier H, Bonder CS, Roberts MS. Non-invasive metabolic imaging of melanoma progression. Exp Dermatol 2017; 26:607-614. [PMID: 27992081 DOI: 10.1111/exd.13274] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/14/2016] [Indexed: 12/17/2022]
Abstract
Skin cancer is associated with abnormal cellular metabolism which if identified early introduces the possibility of intervention to prevent its progress to a deadly metastatic stage. This study combines multiphoton microscopy with fluorescence lifetime imaging (FLIM) using a syngeneic melanoma mouse model, to detect changes in metabolic state of single epidermal cells as a metabolic marker to monitor the progress of tumor growth. This method utilizes imaging of the ratio of the amounts of the free and protein-bound forms of the intracellular autofluorescent metabolic co-enzyme nicotinamide adenine dinucleotide (NADH). Here, we investigate the impact of the primary tumor lesion on the epidermal layers at three different growth stages of melanoma lesion compared to normal skin as a control. We showed a significant increase in the free-to-bound NADH ratio with the growth of the solid melanoma tumor, while concurrently the short and the long lifetime components of NADH remained constant. These results demonstrate the ability of FLIM for rapid, non-invasive and sensitive assessment of melanoma progression revealing its potential as a diagnostic tool for melanoma detection and as an aid for melanoma staging.
Collapse
Affiliation(s)
- Michael N Pastore
- School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, SA, Australia
| | - Hauke Studier
- School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, SA, Australia
| | - Claudine S Bonder
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, SA, Australia
| | - Michael S Roberts
- School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, SA, Australia.,Therapeutics Research Centre, School of Medicine, Translational Research Institute, University of Queensland, Brisbane, Qld, Australia
| |
Collapse
|
174
|
Bower AJ, Chidester B, Li J, Zhao Y, Marjanovic M, Chaney EJ, Do MN, Boppart SA. A quantitative framework for the analysis of multimodal optical microscopy images. Quant Imaging Med Surg 2017; 7:24-37. [PMID: 28275557 DOI: 10.21037/qims.2017.02.07] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
BACKGROUND Multimodal optical microscopy, a set of imaging techniques based on unique, yet complementary contrast mechanisms and spatially and temporally co-registered data acquisition, has emerged as a powerful biomedical tool. However, the analysis of the dense, high-dimensional datasets acquired by these instruments remains mostly qualitative and restricted to analysis of each modality individually. METHODS Using a custom-built multimodal nonlinear optical microscope, high dimensional datasets were acquired for automated classification of functional cell states as well as identification of histopathological features in tissues slices. Supervised classification of cell death modes was performed through support vector machines (SVM) and semi-supervised classification of tissue slices was performed through the use of the expectation maximization (EM) algorithm. RESULTS Applications of these techniques to the automated classification of cell death modes as well as to the identification of tissue components in fixed ex vivo tissue slices are presented. The analysis techniques developed provide a direct link between multimodal image contrast and biological structure and function, resulting in highly accurate classification in both settings. CONCLUSIONS Quantification of multimodal optical microscopy images through statistical modeling of the high dimensional data acquired gives a strong correlation between biological structure and function and image contrast. These methods are sensitive to the identification of diagnostic, cellular-level features important in a variety of clinical settings.
Collapse
Affiliation(s)
- Andrew J Bower
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL, USA; Department of Electrical and Computer Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Benjamin Chidester
- Department of Electrical and Computer Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Joanne Li
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL, USA; Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Youbo Zhao
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Marina Marjanovic
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL, USA; Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Eric J Chaney
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Minh N Do
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL, USA; Department of Electrical and Computer Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, USA; Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Stephen A Boppart
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL, USA; Department of Electrical and Computer Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, USA; Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL, USA; Department of Internal Medicine, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| |
Collapse
|
175
|
Abstract
Head and neck cancer patients suffer from toxicities, morbidities, and mortalities, and these ailments could be minimized through improved therapies. Drug discovery is a long, expensive, and complex process, so optimized assays can improve the success rate of drug candidates. This study applies optical imaging of cell metabolism to three-dimensional in vitro cultures of head and neck cancer grown from primary tumor tissue (organoids). This technique is advantageous because it measures cell metabolism using intrinsic fluorescence from NAD(P)H and FAD on a single cell level for a three-dimensional in vitro model. Head and neck cancer organoids are characterized alone and after treatment with standard therapies, including an antibody therapy, a chemotherapy, and combination therapy. Additionally, organoid cellular heterogeneity is analyzed quantitatively and qualitatively. Gold standard measures of treatment response, including cell proliferation, cell death, and in vivo tumor volume, validate therapeutic efficacy for each treatment group in a parallel study. Results indicate that optical metabolic imaging is sensitive to therapeutic response in organoids after 1 day of treatment (p<0.05) and resolves cell subpopulations with distinct metabolic phenotypes. Ultimately, this platform could provide a sensitive high-throughput assay to streamline the drug discovery process for head and neck cancer.
Collapse
|
176
|
Sergeeva TF, Shirmanova MV, Zlobovskaya OA, Gavrina AI, Dudenkova VV, Lukina MM, Lukyanov KA, Zagaynova EV. Relationship between intracellular pH, metabolic co-factors and caspase-3 activation in cancer cells during apoptosis. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2017; 1864:604-611. [PMID: 28063999 DOI: 10.1016/j.bbamcr.2016.12.022] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Revised: 12/23/2016] [Accepted: 12/30/2016] [Indexed: 12/21/2022]
Abstract
A complex cascade of molecular events occurs in apoptotic cells but cell-to-cell variability significantly complicates determination of the order and interconnections between different processes. For better understanding of the mechanisms of programmed cell death, dynamic simultaneous registration of several parameters is required. In this paper we used multiparameter fluorescence microscopy to analyze energy metabolism, intracellular pH and caspase-3 activation in living cancer cells in vitro during staurosporine-induced apoptosis. We performed metabolic imaging of two co-factors, NAD(P)H and FAD, and used the genetically encoded pH-indicator SypHer1 and the FRET-based sensor for caspase-3 activity, mKate2-DEVD-iRFP, to visualize these parameters by confocal fluorescence microscopy and two-photon fluorescence lifetime imaging microscopy. The correlation between energy metabolism, intracellular pH and caspase-3 activation and their dynamic changes were studied in CT26 cancer cells during apoptosis. Induction of apoptosis was accompanied by a switch to oxidative phosphorylation, cytosol acidification and caspase-3 activation. We showed that alterations in cytosolic pH and the activation of oxidative phosphorylation are relatively early events associated with the induction of apoptosis.
Collapse
Affiliation(s)
- Tatiana F Sergeeva
- Nizhny Novgorod State Medical Academy, 10/1 Minin and Pozharsky Sq., 603005 Nizhny Novgorod, Russia.
| | - Marina V Shirmanova
- Nizhny Novgorod State Medical Academy, 10/1 Minin and Pozharsky Sq., 603005 Nizhny Novgorod, Russia.
| | - Olga A Zlobovskaya
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry RAS, 16/10 Miklukho-Maklaya St., 117997 Moscow, Russia.
| | - Alena I Gavrina
- Nizhny Novgorod State Medical Academy, 10/1 Minin and Pozharsky Sq., 603005 Nizhny Novgorod, Russia; Nizhny Novgorod State University, Gagarin Ave., 23, 603950 Nizhny Novgorod, Russia.
| | - Varvara V Dudenkova
- Nizhny Novgorod State Medical Academy, 10/1 Minin and Pozharsky Sq., 603005 Nizhny Novgorod, Russia; Nizhny Novgorod State University, Gagarin Ave., 23, 603950 Nizhny Novgorod, Russia.
| | - Maria M Lukina
- Nizhny Novgorod State Medical Academy, 10/1 Minin and Pozharsky Sq., 603005 Nizhny Novgorod, Russia; Nizhny Novgorod State University, Gagarin Ave., 23, 603950 Nizhny Novgorod, Russia.
| | - Konstantin A Lukyanov
- Nizhny Novgorod State Medical Academy, 10/1 Minin and Pozharsky Sq., 603005 Nizhny Novgorod, Russia; Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry RAS, 16/10 Miklukho-Maklaya St., 117997 Moscow, Russia.
| | - Elena V Zagaynova
- Nizhny Novgorod State Medical Academy, 10/1 Minin and Pozharsky Sq., 603005 Nizhny Novgorod, Russia.
| |
Collapse
|
177
|
Bower AJ, Marjanovic M, Zhao Y, Li J, Chaney EJ, Boppart SA. Label-free in vivo cellular-level detection and imaging of apoptosis. JOURNAL OF BIOPHOTONICS 2017; 10:143-150. [PMID: 27089867 PMCID: PMC5071126 DOI: 10.1002/jbio.201600003] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Revised: 03/07/2016] [Accepted: 03/23/2016] [Indexed: 05/18/2023]
Abstract
Cell death plays a critical role in health and homeostasis as well as in the pathogenesis and treatment of a broad spectrum of diseases and can be broadly divided into two main categories: apoptosis, or programmed cell death, and necrosis, or acute cell death. While these processes have been characterized extensively in vitro, label-free detection of apoptosis and necrosis at the cellular level in vivo has yet to be shown. In this study, for the first time, fluorescence lifetime imaging microscopy (FLIM) of intracellular reduced nicotinamide adenine dinucleotide (NADH) was utilized to assess the metabolic response of in vivo mouse epidermal keratinocytes following induction of apoptosis and necrosis. Results show significantly elevated levels of both the mean lifetime of NADH and the intracellular ratio of protein bound-to-free NADH in the apoptotic compared to the necrotic tissue. In addition, the longitudinal profiles of these two cell death processes show remarkable differences. By identifying and extracting these temporal metabolic signatures, apoptosis in single cells can be studied in native tissue environments within the living organism.
Collapse
Affiliation(s)
- Andrew J Bower
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL, USA
- Department of Electrical and Computer Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Marina Marjanovic
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL, USA
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Youbo Zhao
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Joanne Li
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL, USA
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Eric J Chaney
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Stephen A Boppart
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL, USA
- Department of Electrical and Computer Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, USA
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL, USA
- Department of Internal Medicine, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| |
Collapse
|
178
|
Perspectives of Reprogramming Breast Cancer Metabolism. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1026:217-232. [PMID: 29282686 DOI: 10.1007/978-981-10-6020-5_10] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Reprogramming of cellular metabolism is one of the hallmarks of breast cancer. Breast cancer cells remodel metabolic network to maintain their transformed state and survive in a harsh tumor microenvironment. Dysregulated metabolism further interacts with cellular signaling and epigenetics to promote breast cancer development. Meanwhile, breast cancer stem cells exhibit unique metabolic features, which are critical for therapeutic resistance and tumor recurrence. Besides, aberrant metabolism of breast cancer cells reshapes tumor microenvironment, such as promoting cancer vascularization and sabotaging tumor immunity, to accelerate tumor progression. These special metabolic traits not only open vulnerabilities of breast cancer by targeting essential metabolic pathways but also provide promising diagnostic and prognostic biomarkers to facilitate clinical investigations. Studies in the last few decades have significantly advanced our understanding of mechanisms underlying the reprogramming of breast cancer metabolism and metabolic regulation of breast cancer biology. Targeting tumor metabolism serves as a potentially effective therapeutic approach to suppress breast cancer.
Collapse
|
179
|
Awasthi K, Moriya D, Nakabayashi T, Li L, Ohta N. Sensitive detection of intracellular environment of normal and cancer cells by autofluorescence lifetime imaging. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY B-BIOLOGY 2016; 165:256-265. [DOI: 10.1016/j.jphotobiol.2016.10.023] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Revised: 10/18/2016] [Accepted: 10/19/2016] [Indexed: 02/04/2023]
|
180
|
Alhallak K, Rebello LG, Muldoon TJ, Quinn KP, Rajaram N. Optical redox ratio identifies metastatic potential-dependent changes in breast cancer cell metabolism. BIOMEDICAL OPTICS EXPRESS 2016; 7:4364-4374. [PMID: 27895979 PMCID: PMC5119579 DOI: 10.1364/boe.7.004364] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Revised: 09/16/2016] [Accepted: 09/29/2016] [Indexed: 05/20/2023]
Abstract
The development of prognostic indicators of breast cancer metastatic risk could reduce the number of patients receiving chemotherapy for tumors with low metastatic potential. Recent evidence points to a critical role for cell metabolism in driving breast cancer metastasis. Endogenous fluorescence intensity of nicotinamide adenine dinucleotide (NADH) and flavin adenine dinucleotide (FAD) can provide a label-free method for assessing cell metabolism. We report the optical redox ratio of FAD/(FAD + NADH) of four isogenic triple-negative breast cancer cell lines with varying metastatic potential. Under normoxic conditions, the redox ratio increases with increasing metastatic potential (168FARN>4T07>4T1), indicating a shift to more oxidative metabolism in cells capable of metastasis. Reoxygenation following acute hypoxia increased the redox ratio by 43 ± 9% and 33 ± 4% in the 4T1 and 4T07 cells, respectively; in contrast, the redox ratio decreased 14 ± 7% in the non-metastatic 67NR cell line. These results demonstrate that the optical redox ratio is sensitive to the metabolic adaptability of breast cancer cells with high metastatic potential and could potentially be used to measure dynamic functional changes that are indicative of invasive or metastatic potential.
Collapse
|
181
|
Spatial dynamics of SIRT1 and the subnuclear distribution of NADH species. Proc Natl Acad Sci U S A 2016; 113:12715-12720. [PMID: 27791113 DOI: 10.1073/pnas.1609227113] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Sirtuin 1 (SIRT1) is an NAD+-dependent deacetylase that functions as metabolic sensor of cellular energy and modulates biochemical pathways in the adaptation to changes in the environment. SIRT1 substrates include histones and proteins related to enhancement of mitochondrial function as well as antioxidant protection. Fluctuations in intracellular NAD+ levels regulate SIRT1 activity, but how SIRT1 enzymatic activity impacts on NAD+ levels and its intracellular distribution remains unclear. Here, we show that SIRT1 determines the nuclear organization of protein-bound NADH. Using multiphoton microscopy in live cells, we show that free and bound NADH are compartmentalized inside of the nucleus, and its subnuclear distribution depends on SIRT1. Importantly, SIRT6, a chromatin-bound deacetylase of the same class, does not influence NADH nuclear localization. In addition, using fluorescence fluctuation spectroscopy in single living cells, we reveal that NAD+ metabolism in the nucleus is linked to subnuclear dynamics of active SIRT1. These results reveal a connection between NAD+ metabolism, NADH distribution, and SIRT1 activity in the nucleus of live cells and pave the way to decipher links between nuclear organization and metabolism.
Collapse
|
182
|
Shah AT, Diggins KE, Walsh AJ, Irish JM, Skala MC. In Vivo Autofluorescence Imaging of Tumor Heterogeneity in Response to Treatment. Neoplasia 2016; 17:862-870. [PMID: 26696368 PMCID: PMC4688562 DOI: 10.1016/j.neo.2015.11.006] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Revised: 11/11/2015] [Accepted: 11/16/2015] [Indexed: 12/20/2022] Open
Abstract
Subpopulations of cells that escape anti-cancer treatment can cause relapse in cancer patients. Therefore, measurements of cellular-level tumor heterogeneity could enable improved anti-cancer treatment regimens. Cancer exhibits altered cellular metabolism, which affects the autofluorescence of metabolic cofactors NAD(P)H and FAD. The optical redox ratio (fluorescence intensity of NAD(P)H divided by FAD) reflects global cellular metabolism. The fluorescence lifetime (amount of time a fluorophore is in the excited state) is sensitive to microenvironment, particularly protein-binding. High-resolution imaging of the optical redox ratio and fluorescence lifetimes of NAD(P)H and FAD (optical metabolic imaging) enables single-cell analyses. In this study, mice with FaDu tumors were treated with the antibody therapy cetuximab or the chemotherapy cisplatin and imaged in vivo two days after treatment. Results indicate that fluorescence lifetimes of NAD(P)H and FAD are sensitive to early response (two days post-treatment, P < .05), compared with decreases in tumor size (nine days post-treatment, P < .05). Frequency histogram analysis of individual optical metabolic imaging parameters identifies subpopulations of cells, and a new heterogeneity index enables quantitative comparisons of cellular heterogeneity across treatment groups for individual variables. Additionally, a dimensionality reduction technique (viSNE) enables holistic visualization of multivariate optical measures of cellular heterogeneity. These analyses indicate increased heterogeneity in the cetuximab and cisplatin treatment groups compared with the control group. Overall, the combination of optical metabolic imaging and cellular-level analyses provide novel, quantitative insights into tumor heterogeneity.
Collapse
Affiliation(s)
- Amy T Shah
- Department of Biomedical Engineering, Vanderbilt University, Station B, Box 1631, Nashville, TN, 37235, USA
| | - Kirsten E Diggins
- Department of Cancer Biology, Vanderbilt University, Nashville, TN 37232, USA
| | - Alex J Walsh
- Department of Biomedical Engineering, Vanderbilt University, Station B, Box 1631, Nashville, TN, 37235, USA
| | - Jonathan M Irish
- Department of Cancer Biology, Vanderbilt University, Nashville, TN 37232, USA; Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Melissa C Skala
- Department of Biomedical Engineering, Vanderbilt University, Station B, Box 1631, Nashville, TN, 37235, USA; Department of Cancer Biology, Vanderbilt University, Nashville, TN 37232, USA.
| |
Collapse
|
183
|
Williams CH, Hong CC. Zebrafish small molecule screens: Taking the phenotypic plunge. Comput Struct Biotechnol J 2016; 14:350-356. [PMID: 27721960 PMCID: PMC5050293 DOI: 10.1016/j.csbj.2016.09.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Revised: 09/12/2016] [Accepted: 09/13/2016] [Indexed: 12/27/2022] Open
Abstract
Target based chemical screens are a mainstay of modern drug discovery, but the effectiveness of this reductionist approach is being questioned in light of declines in pharmaceutical R & D efficiency. In recent years, phenotypic screens have gained increasing acceptance as a complementary/alternative approach to early drug discovery. We discuss the various model organisms used in phenotypic screens, with particular focus on zebrafish, which has emerged as a leading model of in vivo phenotypic screens. Additionally, we anticipate therapeutic opportunities, particularly in orphan disease space, in the context of rapid advances in human Mendelian genetics, electronic health record (EHR)-enabled genome–phenome associations, and genome editing.
Collapse
Affiliation(s)
- Charles H Williams
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Charles C Hong
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA; Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN 37232, USA; Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA; Vanderbilt Institute of Chemical Biology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA; Research Medicine, Veterans Affairs Tennessee Valley Healthcare System, Nashville, TN 37212, USA
| |
Collapse
|
184
|
Alonzo CA, Karaliota S, Pouli D, Liu Z, Karalis KP, Georgakoudi I. Two-photon excited fluorescence of intrinsic fluorophores enables label-free assessment of adipose tissue function. Sci Rep 2016; 6:31012. [PMID: 27491409 PMCID: PMC4974509 DOI: 10.1038/srep31012] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Accepted: 07/11/2016] [Indexed: 01/24/2023] Open
Abstract
Current methods for evaluating adipose tissue function are destructive or have low spatial resolution. These limit our ability to assess dynamic changes and heterogeneous responses that occur in healthy or diseased subjects, or during treatment. Here, we demonstrate that intrinsic two-photon excited fluorescence enables functional imaging of adipocyte metabolism with subcellular resolution. Steady-state and time-resolved fluorescence from intracellular metabolic co-factors and lipid droplets can distinguish the functional states of excised white, brown, and cold-induced beige fat. Similar optical changes are identified when white and brown fat are assessed in vivo. Therefore, these studies establish the potential of non-invasive, high resolution, endogenous contrast, two-photon imaging to identify distinct adipose tissue types, monitor their functional state, and characterize heterogeneity of induced responses.
Collapse
Affiliation(s)
- Carlo Amadeo Alonzo
- Department of Biomedical Engineering, Tufts University 4 Colby Street, Medford, MA 02155, USA
| | | | - Dimitra Pouli
- Department of Biomedical Engineering, Tufts University 4 Colby Street, Medford, MA 02155, USA
| | - Zhiyi Liu
- Department of Biomedical Engineering, Tufts University 4 Colby Street, Medford, MA 02155, USA
| | - Katia P Karalis
- Biomedical Research Foundation, Academy of Athens, Athens, Greece.,Endocrine Division, Children's Hospital, Boston, MA 02115, USA
| | - Irene Georgakoudi
- Department of Biomedical Engineering, Tufts University 4 Colby Street, Medford, MA 02155, USA
| |
Collapse
|
185
|
Kalinina S, Breymayer J, Schäfer P, Calzia E, Shcheslavskiy V, Becker W, Rück A. Correlative NAD(P)H-FLIM and oxygen sensing-PLIM for metabolic mapping. JOURNAL OF BIOPHOTONICS 2016; 9:800-811. [PMID: 26990032 DOI: 10.1002/jbio.201500297] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Revised: 02/02/2016] [Accepted: 02/21/2016] [Indexed: 06/05/2023]
Abstract
Cellular responses to oxygen tension have been studied extensively. Oxygen tension can be determined by considering the phosphorescence lifetime of a phosphorescence sensor. The simultaneous usage of FLIM of coenzymes as NAD(P)H and FAD(+) and PLIM of oxygen sensors could provide information about correlation of metabolic pathways and oxygen tension. We investigated correlative NAD(P)H-FLIM and oxygen sensing-PLIM for simultaneously analyzing cell metabolism and oxygen tension. Cell metabolism and pO2 were observed under different hypoxic conditions in squamous carcinoma cell cultures and in complex ex vivo systems. Increased hypoxia induced an increase of the phosphorescence lifetime of Ru(BPY)3 and in most cases a decrease in the lifetime of NAD(P)H which is in agreement to the expected decrease of the protein-bound NAD(P)H during hypoxia. Oxygen was modulated directly in the mitochondrial membrane. Blocking of complex III and accumulation of oxygen could be observed by both the decrease of the phosphorescence lifetime of Ru(BPY)3 and a reduction of the lifetime of NAD(P)H which was a clear indication of acute changes in the redox state of the cells. For the first time simultaneous FLIM/PLIM has been shown to be able to visualize intracellular oxygen tension together with a change from oxidative to glycolytic phenotype.
Collapse
Affiliation(s)
- Sviatlana Kalinina
- Ulm University, Core Facility Confocal and Multiphoton Microscopy, N24, Albert Einstein Allee 11, 89081, Ulm, Germany.
| | - Jasmin Breymayer
- Ulm University, Core Facility Confocal and Multiphoton Microscopy, N24, Albert Einstein Allee 11, 89081, Ulm, Germany
| | - Patrick Schäfer
- Zentrum biomedizinische Forschung (ZBF), Ulm University, Institute of Neurology, Helmholtzstr. 8/1, 89081, Ulm, Germany
| | - Enrico Calzia
- Institut für Anästhesiologische Pathophysiologie und Verfahrensentwicklung, Universitätsklinikum Ulm, Helmholtzstr. 8/1, 89081, Ulm, Germany
| | | | - Wolfgang Becker
- Becker & Hickl GmbH, Nahmitzer Damm 30, 12277, Berlin, Germany
| | - Angelika Rück
- Ulm University, Core Facility Confocal and Multiphoton Microscopy, N24, Albert Einstein Allee 11, 89081, Ulm, Germany.
| |
Collapse
|
186
|
Deblois G, Smith HW, Tam IS, Gravel SP, Caron M, Savage P, Labbé DP, Bégin LR, Tremblay ML, Park M, Bourque G, St-Pierre J, Muller WJ, Giguère V. ERRα mediates metabolic adaptations driving lapatinib resistance in breast cancer. Nat Commun 2016; 7:12156. [PMID: 27402251 PMCID: PMC4945959 DOI: 10.1038/ncomms12156] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2015] [Accepted: 06/06/2016] [Indexed: 12/27/2022] Open
Abstract
Despite the initial benefits of treating HER2-amplified breast cancer patients with the tyrosine kinase inhibitor lapatinib, resistance inevitably develops. Here we report that lapatinib induces the degradation of the nuclear receptor ERRα, a master regulator of cellular metabolism, and that the expression of ERRα is restored in lapatinib-resistant breast cancer cells through reactivation of mTOR signalling. Re-expression of ERRα in resistant cells triggers metabolic adaptations favouring mitochondrial energy metabolism through increased glutamine metabolism, as well as ROS detoxification required for cell survival under therapeutic stress conditions. An ERRα inverse agonist counteracts these metabolic adaptations and overcomes lapatinib resistance in a HER2-induced mammary tumour mouse model. This work reveals a molecular mechanism by which ERRα-induced metabolic reprogramming promotes survival of lapatinib-resistant cancer cells and demonstrates the potential of ERRα inhibition as an effective adjuvant therapy in poor outcome HER2-positive breast cancer.
Collapse
Affiliation(s)
- Geneviève Deblois
- Goodman Cancer Research Centre, McGill University, Montréal, Québec, Canada H3A 1A3
- Department of Biochemistry, McGill University, Montréal, Québec, Canada H3G 1Y6
| | - Harvey W. Smith
- Goodman Cancer Research Centre, McGill University, Montréal, Québec, Canada H3A 1A3
| | - Ingrid S. Tam
- Goodman Cancer Research Centre, McGill University, Montréal, Québec, Canada H3A 1A3
| | - Simon-Pierre Gravel
- Goodman Cancer Research Centre, McGill University, Montréal, Québec, Canada H3A 1A3
| | - Maxime Caron
- Department of Human Genetics, McGill University, Montréal, Québec, Canada H3G 1Y6
| | - Paul Savage
- Goodman Cancer Research Centre, McGill University, Montréal, Québec, Canada H3A 1A3
- Department of Medicine, McGill University, Montréal, Québec, Canada H3A 1A3
| | - David P. Labbé
- Goodman Cancer Research Centre, McGill University, Montréal, Québec, Canada H3A 1A3
- Department of Medicine, McGill University, Montréal, Québec, Canada H3A 1A3
| | - Louis R. Bégin
- Service d'anatomopathologie, Hôpital du Sacré-Cœur de Montréal, 5400 Boulevard Gouin Ouest, Montréal, Québec, Canada H4J 1C5
| | - Michel L. Tremblay
- Goodman Cancer Research Centre, McGill University, Montréal, Québec, Canada H3A 1A3
- Department of Biochemistry, McGill University, Montréal, Québec, Canada H3G 1Y6
- Department of Medicine, McGill University, Montréal, Québec, Canada H3A 1A3
- Department of Oncology, McGill University, Montréal, Québec, Canada H2W 1S6
| | - Morag Park
- Goodman Cancer Research Centre, McGill University, Montréal, Québec, Canada H3A 1A3
- Department of Biochemistry, McGill University, Montréal, Québec, Canada H3G 1Y6
- Department of Medicine, McGill University, Montréal, Québec, Canada H3A 1A3
- Department of Oncology, McGill University, Montréal, Québec, Canada H2W 1S6
| | - Guillaume Bourque
- Department of Human Genetics, McGill University, Montréal, Québec, Canada H3G 1Y6
| | - Julie St-Pierre
- Goodman Cancer Research Centre, McGill University, Montréal, Québec, Canada H3A 1A3
- Department of Biochemistry, McGill University, Montréal, Québec, Canada H3G 1Y6
| | - William J. Muller
- Goodman Cancer Research Centre, McGill University, Montréal, Québec, Canada H3A 1A3
- Department of Biochemistry, McGill University, Montréal, Québec, Canada H3G 1Y6
- Department of Medicine, McGill University, Montréal, Québec, Canada H3A 1A3
| | - Vincent Giguère
- Goodman Cancer Research Centre, McGill University, Montréal, Québec, Canada H3A 1A3
- Department of Biochemistry, McGill University, Montréal, Québec, Canada H3G 1Y6
- Department of Medicine, McGill University, Montréal, Québec, Canada H3A 1A3
- Department of Oncology, McGill University, Montréal, Québec, Canada H2W 1S6
| |
Collapse
|
187
|
Lapierre-Landry M, Tucker-Schwartz JM, Skala MC. Depth-resolved analytical model and correction algorithm for photothermal optical coherence tomography. BIOMEDICAL OPTICS EXPRESS 2016; 7:2607-22. [PMID: 27446693 PMCID: PMC4948617 DOI: 10.1364/boe.7.002607] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Revised: 06/02/2016] [Accepted: 06/11/2016] [Indexed: 05/03/2023]
Abstract
Photothermal OCT (PT-OCT) is an emerging molecular imaging technique that occupies a spatial imaging regime between microscopy and whole body imaging. PT-OCT would benefit from a theoretical model to optimize imaging parameters and test image processing algorithms. We propose the first analytical PT-OCT model to replicate an experimental A-scan in homogeneous and layered samples. We also propose the PT-CLEAN algorithm to reduce phase-accumulation and shadowing, two artifacts found in PT-OCT images, and demonstrate it on phantoms and in vivo mouse tumors.
Collapse
|
188
|
Optical Imaging of Drug-Induced Metabolism Changes in Murine and Human Pancreatic Cancer Organoids Reveals Heterogeneous Drug Response. Pancreas 2016; 45:863-9. [PMID: 26495796 PMCID: PMC4874911 DOI: 10.1097/mpa.0000000000000543] [Citation(s) in RCA: 95] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
OBJECTIVES Three-dimensional organoids derived from primary pancreatic ductal adenocarcinomas are an attractive platform for testing potential anticancer drugs on patient-specific tissue. Optical metabolic imaging (OMI) is a novel tool used to assess drug-induced changes in cellular metabolism, and its quantitative end point, the OMI index, is evaluated as a biomarker of drug response in pancreatic cancer organoids. METHODS Optical metabolic imaging is used to assess both malignant cell and fibroblast drug response within primary murine and human pancreatic cancer organoids. RESULTS Anticancer drugs induce significant reductions in the OMI index of murine and human pancreatic cancer organoids. Subpopulation analysis of OMI data revealed heterogeneous drug response and elucidated responding and nonresponding cell populations for a 7-day time course. Optical metabolic imaging index significantly correlates with immunofluorescence detection of cell proliferation and cell death. CONCLUSIONS Optical metabolic imaging of primary pancreatic ductal adenocarcinoma organoids is highly sensitive to drug-induced metabolic changes, provides a nondestructive method for monitoring dynamic drug response, and presents a novel platform for patient-specific drug testing and drug development.
Collapse
|
189
|
Hou J, Wright HJ, Chan N, Tran R, Razorenova OV, Potma EO, Tromberg BJ. Correlating two-photon excited fluorescence imaging of breast cancer cellular redox state with seahorse flux analysis of normalized cellular oxygen consumption. JOURNAL OF BIOMEDICAL OPTICS 2016; 21:60503. [PMID: 27300321 PMCID: PMC4906146 DOI: 10.1117/1.jbo.21.6.060503] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Accepted: 05/23/2016] [Indexed: 05/22/2023]
Abstract
Two-photon excited fluorescence (TPEF) imaging of the cellular cofactors nicotinamide adenine dinucleotide and oxidized flavin adenine dinucleotide is widely used to measure cellular metabolism, both in normal and pathological cells and tissues. When dual-wavelength excitation is used, ratiometric TPEF imaging of the intrinsic cofactor fluorescence provides a metabolic index of cells—the “optical redox ratio” (ORR). With increased interest in understanding and controlling cellular metabolism in cancer, there is a need to evaluate the performance of ORR in malignant cells. We compare TPEF metabolic imaging with seahorse flux analysis of cellular oxygen consumption in two different breast cancer cell lines (MCF-7 and MDA-MB-231). We monitor metabolic index in living cells under both normal culture conditions and, for MCF-7, in response to cell respiration inhibitors and uncouplers. We observe a significant correlation between the TPEF-derived ORR and the flux analyzer measurements (R=0.7901, p<0.001). Our results confirm that the ORR is a valid dynamic index of cell metabolism under a range of oxygen consumption conditions relevant for cancer imaging.
Collapse
Affiliation(s)
- Jue Hou
- University of California, Laser Microbeam and Medical Program, Beckman Laser Institute and Medical Clinic, 1002 Health Sciences Road, Irvine, California 92612, United States
| | - Heather J. Wright
- University of California, Department of Molecular Biology and Biochemistry, 3205 McGaugh Hall, Irvine, California 92697-3900, United States
| | - Nicole Chan
- University of California, Laser Microbeam and Medical Program, Beckman Laser Institute and Medical Clinic, 1002 Health Sciences Road, Irvine, California 92612, United States
| | - Richard Tran
- University of California, Laser Microbeam and Medical Program, Beckman Laser Institute and Medical Clinic, 1002 Health Sciences Road, Irvine, California 92612, United States
| | - Olga V. Razorenova
- University of California, Department of Molecular Biology and Biochemistry, 3205 McGaugh Hall, Irvine, California 92697-3900, United States
| | - Eric O. Potma
- University of California, Laser Microbeam and Medical Program, Beckman Laser Institute and Medical Clinic, 1002 Health Sciences Road, Irvine, California 92612, United States
| | - Bruce J. Tromberg
- University of California, Laser Microbeam and Medical Program, Beckman Laser Institute and Medical Clinic, 1002 Health Sciences Road, Irvine, California 92612, United States
- Address all correspondence to: Bruce J. Tromberg, E-mail:
| |
Collapse
|
190
|
Cavnar SP, Xiao A, Gibbons AE, Rickelmann AD, Neely T, Luker KE, Takayama S, Luker GD. Imaging Sensitivity of Quiescent Cancer Cells to Metabolic Perturbations in Bone Marrow Spheroids. Tomography 2016; 2:146-157. [PMID: 27478871 PMCID: PMC4963031 DOI: 10.18383/j.tom.2016.00157] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Malignant cells from breast cancer and other common cancers such as prostate and melanoma may persist in bone marrow as quiescent, non-dividing cells that remain viable for years or even decades before resuming proliferation to cause recurrent disease. This phenomenon, referred to clinically as tumor dormancy, poses tremendous challenges to curing patients with breast cancer. Quiescent tumor cells resist chemotherapy drugs that predominantly target proliferating cells, limiting success of neo-adjuvant and adjuvant therapies. We recently developed a 3D spheroid model of quiescent breast cancer cells in bone marrow for mechanistic and drug testing studies. We combined this model with optical imaging methods for label-free detection of cells preferentially utilizing glycolysis versus oxidative metabolism to investigate the metabolic state of co-culture spheroids with different bone marrow stromal and breast cancer cells. Through imaging and biochemical assays, we identified different metabolic states of bone marrow stromal cells that control metabolic status and flexibilities of co-cultured breast cancer cells. We tested metabolic stresses and targeted inhibition of specific metabolic pathways to identify approaches to preferentially eliminate quiescent breast cancer cells from bone marrow environments. These studies establish an integrated imaging approach to analyze metabolism in complex tissue environments to identify new metabolically-targeted cancer therapies.
Collapse
Affiliation(s)
- Stephen P. Cavnar
- Department of Biomedical Engineering, University of Michigan College of Engineering, Ann Arbor, Michigan
- Biointerfaces Institute, University of Michigan College of Engineering, Ann Arbor, Michigan
| | - Annie Xiao
- Department of Radiology, Center for Molecular Imaging, University of Michigan Medical School, Ann Arbor, Michigan
| | - Anne E. Gibbons
- Department of Radiology, Center for Molecular Imaging, University of Michigan Medical School, Ann Arbor, Michigan
| | - Andrew D. Rickelmann
- Department of Radiology, Center for Molecular Imaging, University of Michigan Medical School, Ann Arbor, Michigan
| | - Taylor Neely
- Department of Radiology, Center for Molecular Imaging, University of Michigan Medical School, Ann Arbor, Michigan
| | - Kathryn E. Luker
- Department of Radiology, Center for Molecular Imaging, University of Michigan Medical School, Ann Arbor, Michigan
| | - Shuichi Takayama
- Department of Biomedical Engineering, University of Michigan College of Engineering, Ann Arbor, Michigan
- Biointerfaces Institute, University of Michigan College of Engineering, Ann Arbor, Michigan
- Department of Macromolecular Science and Engineering, University of Michigan College of Engineering, Ann Arbor, Michigan; and
| | - Gary D. Luker
- Department of Biomedical Engineering, University of Michigan College of Engineering, Ann Arbor, Michigan
- Department of Radiology, Center for Molecular Imaging, University of Michigan Medical School, Ann Arbor, Michigan
- Department of Macromolecular Science and Engineering, University of Michigan College of Engineering, Ann Arbor, Michigan; and
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan
| |
Collapse
|
191
|
In Vivo Visualization of Stromal Macrophages via label-free FLIM-based metabolite imaging. Sci Rep 2016; 6:25086. [PMID: 27220760 PMCID: PMC4879594 DOI: 10.1038/srep25086] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Accepted: 04/07/2016] [Indexed: 01/02/2023] Open
Abstract
Macrophage infiltration and recruitment in breast tumors has been correlated with poor prognosis in breast cancer patients and has been linked to tumor cell dissemination. Much of our understanding comes from animal models in which macrophages are labeled by expression of an extrinsic fluorophore. However, conventional extrinsic fluorescence labeling approaches are not readily applied to human tissue and clinical use. We report a novel strategy that exploits endogenous fluorescence from the metabolic co-factors NADH and FAD with quantitation from Fluorescence Lifetime Imaging Microscopy (FLIM) as a means to non-invasively identify tumor-associated macrophages in the intact mammary tumor microenvironment. Macrophages were FADHI and demonstrated a glycolytic-like NADH-FLIM signature that was readily separated from the intrinsic fluorescence signature of tumor cells. This non-invasive quantitative technique provides a unique ability to discern specific cell types based upon their metabolic signatures without the use of exogenous fluorescent labels. Not only does this provide high resolution temporal and spatial views of macrophages in live animal breast cancer models, this approach can be extended to other animal disease models where macrophages are implicated and has potential for clinical applications.
Collapse
|
192
|
Druzhkova IN, Shirmanova MV, Lukina MM, Dudenkova VV, Mishina NM, Zagaynova EV. The metabolic interaction of cancer cells and fibroblasts - coupling between NAD(P)H and FAD, intracellular pH and hydrogen peroxide. Cell Cycle 2016; 15:1257-1266. [PMID: 26986068 PMCID: PMC4889290 DOI: 10.1080/15384101.2016.1160974] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Revised: 02/26/2016] [Accepted: 02/26/2016] [Indexed: 12/12/2022] Open
Abstract
Alteration in the cellular energy metabolism is a principal feature of tumors. An important role in modifying cancer cell metabolism belongs to the cancer-associated fibroblasts. However, the regulation of their interaction has been poorly studied to date. In this study we monitored the metabolic status of both cell types by using the optical redox ratio and the fluorescence lifetimes of the metabolic co-factors NAD(P)H and FAD, in addition to the intracellular pH and the hydrogen peroxide levels in the cancer cells, using genetically encoded sensors. In the co-culture of human cervical carcinoma cells HeLa and human fibroblasts we observed a metabolic shift from oxidative phosphorylation toward glycolysis in cancer cells, and from glycolysis toward OXPHOS in fibroblasts, starting from Day 2 of co-culturing. The metabolic switch was accompanied by hydrogen peroxide production and slight acidification of the cytosol in the cancer cells in comparison with that of the corresponding monoculture. Therefore, our HeLa-huFb system demonstrated metabolic behavior similar to Warburg type tumors. To our knowledge, this is the first time that these 3 parameters have been investigated together in a model of tumor-stroma co-evolution. We propose that determination of the start-point of the metabolic alterations and understanding of the mechanisms of their realization can open a new ways for cancer treatment.
Collapse
Affiliation(s)
| | - Marina V. Shirmanova
- Nizhny Novgorod State Medical Academy, Nizhny Novgorod, Russia
- Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, Russia
| | - Maria M. Lukina
- Nizhny Novgorod State Medical Academy, Nizhny Novgorod, Russia
- Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, Russia
| | - Varvara V. Dudenkova
- Nizhny Novgorod State Medical Academy, Nizhny Novgorod, Russia
- Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, Russia
| | - Nataliya M. Mishina
- Nizhny Novgorod State Medical Academy, Nizhny Novgorod, Russia
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry RAS, Moscow, Russia
| | - Elena V. Zagaynova
- Nizhny Novgorod State Medical Academy, Nizhny Novgorod, Russia
- Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, Russia
| |
Collapse
|
193
|
Datta R, Heylman C, George SC, Gratton E. Label-free imaging of metabolism and oxidative stress in human induced pluripotent stem cell-derived cardiomyocytes. BIOMEDICAL OPTICS EXPRESS 2016; 7:1690-701. [PMID: 27231614 PMCID: PMC4871074 DOI: 10.1364/boe.7.001690] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Revised: 03/29/2016] [Accepted: 03/29/2016] [Indexed: 05/20/2023]
Abstract
In this work we demonstrate a label-free optical imaging technique to assess metabolic status and oxidative stress in human induced pluripotent stem cell-derived cardiomyocytes by two-photon fluorescence lifetime imaging of endogenous fluorophores. Our results show the sensitivity of this method to detect shifts in metabolism and oxidative stress in the cardiomyocytes upon pathological stimuli of hypoxia and cardiotoxic drugs. This non-invasive imaging technique could prove beneficial for drug development and screening, especially for in vitro cardiac models created from stem cell-derived cardiomyocytes and to study the pathogenesis of cardiac diseases and therapy.
Collapse
Affiliation(s)
- Rupsa Datta
- Laboratory for Fluorescence Dynamic, University of California, Biomedical Engineering, Irvine, California 92617, USA
| | - Christopher Heylman
- Laboratory for Fluorescence Dynamic, University of California, Biomedical Engineering, Irvine, California 92617, USA
| | - Steven C. George
- Washington University in St. Louis, Biomedical Engineering, St. Louis, Missouri, 63130, USA
| | - Enrico Gratton
- Laboratory for Fluorescence Dynamic, University of California, Biomedical Engineering, Irvine, California 92617, USA
| |
Collapse
|
194
|
Campos D, Peeters W, Nickel K, Burkel B, Bussink J, Kimple RJ, van der Kogel A, Eliceiri KW, Kissick MW. Radiation Promptly Alters Cancer Live Cell Metabolic Fluxes: An In Vitro Demonstration. Radiat Res 2016; 185:496-504. [PMID: 27128739 DOI: 10.1667/rr14093.1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Quantitative data is presented that shows significant changes in cellular metabolism in a head and neck cancer cell line 30 min after irradiation. A head and neck cancer cell line (UM-SCC-22B) and a comparable normal cell line, normal oral keratinocyte (NOK) were each separately exposed to 10 Gy and treated with a control drug for disrupting metabolism (potassium cyanide; KCN). The metabolic changes were measured live by fluorescence lifetime imaging of the intrinsically fluorescent intermediate metabolite nicotinamide adenosine dinucleotide (NADH) fluorescence; this method is sensitive to the ratio of bound to free NADH. The results indicated a prompt shift in metabolic signature in the cancer cell line, but not in the normal cell line. Control KCN treatment demonstrated expected metabolic fluxes due to mitochondrial disruption. The detected radiation shift in the cancer cells was blunted in the presence of both a radical scavenger and a HIF-1α inhibitor. The HIF-1α abundance as detected by immunohistochemical staining also increased substantially for these cancer cells, but not for the normal cells. This type of live-cell metabolic monitoring could be helpful for future real-time studies and in designing adaptive radiotherapy approaches.
Collapse
Affiliation(s)
- David Campos
- a Laboratory for Optical and Computational Instrumentation, Vice Chancellor Office for Graduate Research and Education and Departments of.,b Medical Physics and
| | - Wenny Peeters
- d Department of Radiation Oncology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Kwangok Nickel
- c Human Oncology, University of Wisconsin-Madison, Madison, Wisconsin; and
| | - Brian Burkel
- a Laboratory for Optical and Computational Instrumentation, Vice Chancellor Office for Graduate Research and Education and Departments of
| | - Johan Bussink
- d Department of Radiation Oncology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Randall J Kimple
- c Human Oncology, University of Wisconsin-Madison, Madison, Wisconsin; and
| | | | - Kevin W Eliceiri
- a Laboratory for Optical and Computational Instrumentation, Vice Chancellor Office for Graduate Research and Education and Departments of.,b Medical Physics and
| | - Michael W Kissick
- a Laboratory for Optical and Computational Instrumentation, Vice Chancellor Office for Graduate Research and Education and Departments of.,b Medical Physics and
| |
Collapse
|
195
|
Walsh AJ, Sharick JT, Skala MC, Beier HT. Temporal binning of time-correlated single photon counting data improves exponential decay fits and imaging speed. BIOMEDICAL OPTICS EXPRESS 2016; 7:1385-99. [PMID: 27446663 PMCID: PMC4929649 DOI: 10.1364/boe.7.001385] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Revised: 03/15/2016] [Accepted: 03/15/2016] [Indexed: 05/06/2023]
Abstract
Time-correlated single photon counting (TCSPC) enables acquisition of fluorescence lifetime decays with high temporal resolution within the fluorescence decay. However, many thousands of photons per pixel are required for accurate lifetime decay curve representation, instrument response deconvolution, and lifetime estimation, particularly for two-component lifetimes. TCSPC imaging speed is inherently limited due to the single photon per laser pulse nature and low fluorescence event efficiencies (<10%) required to reduce bias towards short lifetimes. Here, simulated fluorescence lifetime decays are analyzed by SPCImage and SLIM Curve software to determine the limiting lifetime parameters and photon requirements of fluorescence lifetime decays that can be accurately fit. Data analysis techniques to improve fitting accuracy for low photon count data were evaluated. Temporal binning of the decays from 256 time bins to 42 time bins significantly (p<0.0001) improved fit accuracy in SPCImage and enabled accurate fits with low photon counts (as low as 700 photons/decay), a 6-fold reduction in required photons and therefore improvement in imaging speed. Additionally, reducing the number of free parameters in the fitting algorithm by fixing the lifetimes to known values significantly reduced the lifetime component error from 27.3% to 3.2% in SPCImage (p<0.0001) and from 50.6% to 4.2% in SLIM Curve (p<0.0001). Analysis of nicotinamide adenine dinucleotide-lactate dehydrogenase (NADH-LDH) solutions confirmed temporal binning of TCSPC data and a reduced number of free parameters improves exponential decay fit accuracy in SPCImage. Altogether, temporal binning (in SPCImage) and reduced free parameters are data analysis techniques that enable accurate lifetime estimation from low photon count data and enable TCSPC imaging speeds up to 6x and 300x faster, respectively, than traditional TCSPC analysis.
Collapse
Affiliation(s)
- Alex J. Walsh
- National Research Council, JBSA Fort Sam Houston, Texas, 78234, USA
- 711th Human Performance Wing, Human Effectiveness Directorate, Bioeffects Division, Optical Radiation Bioeffects Branch, Air Force Research Lab, JBSA Fort Sam Houston, Texas, 78234, USA
| | - Joe T. Sharick
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee, 37235, USA
| | - Melissa C. Skala
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee, 37235, USA
| | - Hope T. Beier
- 711th Human Performance Wing, Human Effectiveness Directorate, Bioeffects Division, Optical Radiation Bioeffects Branch, Air Force Research Lab, JBSA Fort Sam Houston, Texas, 78234, USA
| |
Collapse
|
196
|
Gutierrez-Navarro O, Campos-Delgado DU, Arce-Santana ER, Jo JA. Quadratic blind linear unmixing: A graphical user interface for tissue characterization. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2016; 124:148-160. [PMID: 26589467 PMCID: PMC4818012 DOI: 10.1016/j.cmpb.2015.10.016] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Revised: 09/20/2015] [Accepted: 10/20/2015] [Indexed: 06/05/2023]
Abstract
Spectral unmixing is the process of breaking down data from a sample into its basic components and their abundances. Previous work has been focused on blind unmixing of multi-spectral fluorescence lifetime imaging microscopy (m-FLIM) datasets under a linear mixture model and quadratic approximations. This method provides a fast linear decomposition and can work without a limitation in the maximum number of components or end-members. Hence this work presents an interactive software which implements our blind end-member and abundance extraction (BEAE) and quadratic blind linear unmixing (QBLU) algorithms in Matlab. The options and capabilities of our proposed software are described in detail. When the number of components is known, our software can estimate the constitutive end-members and their abundances. When no prior knowledge is available, the software can provide a completely blind solution to estimate the number of components, the end-members and their abundances. The characterization of three case studies validates the performance of the new software: ex-vivo human coronary arteries, human breast cancer cell samples, and in-vivo hamster oral mucosa. The software is freely available in a hosted webpage by one of the developing institutions, and allows the user a quick, easy-to-use and efficient tool for multi/hyper-spectral data decomposition.
Collapse
Affiliation(s)
| | - D U Campos-Delgado
- Facultad de Ciencias, Universidad Autonoma de San Luis Potosi, SLP, Mexico.
| | - E R Arce-Santana
- Facultad de Ciencias, Universidad Autonoma de San Luis Potosi, SLP, Mexico
| | - Javier A Jo
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, USA
| |
Collapse
|
197
|
Drug response in organoids generated from frozen primary tumor tissues. Sci Rep 2016; 6:18889. [PMID: 26738962 PMCID: PMC4703961 DOI: 10.1038/srep18889] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Accepted: 11/27/2015] [Indexed: 11/08/2022] Open
Abstract
Primary tumor organoids grown in three-dimensional culture provide an excellent platform for studying tumor progression, invasion, and drug response. However, organoid generation protocols require fresh tumor tissue, which limits organoid research and clinical use. This study investigates cellular morphology, viability, and drug response of organoids derived from frozen tissues. The results demonstrate that viable organoids can be grown from flash-frozen and thawed tissue and from bulk tissues slowly frozen in DMSO supplemented media. While the freezing process affects the basal metabolic rate of the cells, the optical metabolic imaging index correlates between organoids derived from fresh and frozen tissue and can be used to detect drug response of organoids grown from frozen tissues. The slow, DMSO frozen tissue yielded organoids with more accurate drug response than the flash frozen tissues, and thus bulk tissue should be preserved for subsequent organoid generation by slow freezing in DMSO supplemented media.
Collapse
|
198
|
Tain R, Xu HN, Zhou XJ, Li LZ, Cai K. Magnetization Transfer MRI Contrast May Correlate with Tissue Redox State in Prostate Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 923:401-406. [PMID: 27526169 PMCID: PMC5411113 DOI: 10.1007/978-3-319-38810-6_52] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Developing imaging biomarkers for non-invasive measurement of the tissue redox state is a key research area. Recently, we presented the first non-invasive MR imaging method that demonstrated the correlation between the endogenous chemical exchange saturation transfer (CEST) contrast and the tissue redox state. It is well known that the broadband magnetization transfer (MT) can occur via chemical exchange (CEST) and/or dipole-dipole interactions. The present study investigated if the broadband MT also correlated with the tissue redox state. The preliminary result for the prostate tumor xenografts indeed showed a significant correlation between the broadband MT contrast and the NADH redox ratio quantified with the optical redox scanning. In vivo MT contrast, once calibrated, may potentially serve as an imaging biomarker for tissue redox state.
Collapse
Affiliation(s)
- Rongwen Tain
- Department of Radiology, Center for MR Research, University of Illinois at Chicago, 2242 w. Harrison st., Chicago, IL, 60612, USA
| | - He N Xu
- Britton Chance Laboratory of Redox Imaging, Johnson Research Foundation; Department of Radiology, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Xiaohong J Zhou
- Department of Radiology, Center for MR Research, University of Illinois at Chicago, 2242 w. Harrison st., Chicago, IL, 60612, USA
| | - Lin Z Li
- Molecular Imaging Laboratory, Department of Radiology, Britton Chance Laboratory of Redox Imaging, Johnson Research Foundation, Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Kejia Cai
- Department of Radiology, Center for MR Research, University of Illinois at Chicago, 2242 w. Harrison st., Chicago, IL, 60612, USA.
| |
Collapse
|
199
|
Nagathihalli NS, Castellanos JA, Shi C, Beesetty Y, Reyzer ML, Caprioli R, Chen X, Walsh AJ, Skala MC, Moses HL, Merchant NB. Signal Transducer and Activator of Transcription 3, Mediated Remodeling of the Tumor Microenvironment Results in Enhanced Tumor Drug Delivery in a Mouse Model of Pancreatic Cancer. Gastroenterology 2015; 149:1932-1943.e9. [PMID: 26255562 PMCID: PMC4863449 DOI: 10.1053/j.gastro.2015.07.058] [Citation(s) in RCA: 145] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Revised: 07/01/2015] [Accepted: 07/30/2015] [Indexed: 02/08/2023]
Abstract
BACKGROUND & AIMS A hallmark of pancreatic ductal adenocarcinoma (PDAC) is the presence of a dense desmoplastic reaction (stroma) that impedes drug delivery to the tumor. Attempts to deplete the tumor stroma have resulted in formation of more aggressive tumors. We have identified signal transducer and activator of transcription (STAT) 3 as a biomarker of resistance to cytotoxic and molecularly targeted therapy in PDAC. The purpose of this study is to investigate the effects of targeting STAT3 on the PDAC stroma and on therapeutic resistance. METHODS Activated STAT3 protein expression was determined in human pancreatic tissues and tumor cell lines. In vivo effects of AZD1480, a JAK/STAT3 inhibitor, gemcitabine or the combination were determined in Ptf1a(cre/+);LSL-Kras(G12D/+);Tgfbr2(flox/flox) (PKT) mice and in orthotopic tumor xenografts. Drug delivery was analyzed by matrix-assisted laser desorption/ionization imaging mass spectrometry. Collagen second harmonic generation imaging quantified tumor collagen alignment and density. RESULTS STAT3 activation correlates with decreased survival and advanced tumor stage in patients with PDAC. STAT3 inhibition combined with gemcitabine significantly inhibits tumor growth in both an orthotopic and the PKT mouse model of PDAC. This combined therapy attenuates in vivo expression of SPARC, increases microvessel density, and enhances drug delivery to the tumor without depletion of stromal collagen or hyaluronan. Instead, the PDAC tumors demonstrate vascular normalization, remodeling of the tumor stroma, and down-regulation of cytidine deaminase. CONCLUSIONS Targeted inhibition of STAT3 combined with gemcitabine enhances in vivo drug delivery and therapeutic response in PDAC. These effects occur through tumor stromal remodeling and down-regulation of cytidine deaminase without depletion of tumor stromal content.
Collapse
Affiliation(s)
- Nagaraj S. Nagathihalli
- Division of Surgical Oncology, Department of Surgery, University of Miami Miller School of Medicine, Sylvester Comprehensive Cancer Center, Miami, Florida
| | - Jason A. Castellanos
- Department of Surgery, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Chanjuan Shi
- Department of Pathology, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Yugandhar Beesetty
- Department of Surgery, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Michelle L. Reyzer
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Richard Caprioli
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Xi Chen
- Department of Biostatistics, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Alex J. Walsh
- Department of Biomedical Engineering, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Melissa C. Skala
- Department of Biomedical Engineering, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Harold L. Moses
- Department of Cancer Biology, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Nipun B. Merchant
- Division of Surgical Oncology, Department of Surgery, University of Miami Miller School of Medicine, Sylvester Comprehensive Cancer Center, Miami, Florida
| |
Collapse
|
200
|
Campos-Delgado DU, Navarro OG, Arce-Santana ER, Walsh AJ, Skala MC, Jo JA. Deconvolution of fluorescence lifetime imaging microscopy by a library of exponentials. OPTICS EXPRESS 2015; 23:23748-67. [PMID: 26368470 PMCID: PMC4646519 DOI: 10.1364/oe.23.023748] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Revised: 08/08/2015] [Accepted: 08/22/2015] [Indexed: 05/18/2023]
Abstract
Fluorescence lifetime microscopy imaging (FLIM) is an optic technique that allows a quantitative characterization of the fluorescent components of a sample. However, for an accurate interpretation of FLIM, an initial processing step is required to deconvolve the instrument response of the system from the measured fluorescence decays. In this paper, we present a novel strategy for the deconvolution of FLIM data based on a library of exponentials. Our approach searches for the scaling coefficients of the library by non-negative least squares approximations plus Thikonov/l2 or l1 regularization terms. The parameters of the library are given by the lower and upper bounds in the characteristic lifetimes of the exponential functions and the size of the library, where we observe that this last variable is not a limiting factor in the resulting fitting accuracy. We compare our proposal to nonlinear least squares and global non-linear least squares estimations with a multi-exponential model, and also to constrained Laguerre-base expansions, where we visualize an advantage of our proposal based on Thikonov/l2 regularization in terms of estimation accuracy, computational time, and tuning strategy. Our validation strategy considers synthetic datasets subject to both shot and Gaussian noise and samples with different lifetime maps, and experimental FLIM data of ex-vivo atherosclerotic plaques and human breast cancer cells.
Collapse
Affiliation(s)
| | | | - E. R. Arce-Santana
- Facultad de Ciencias, Universidad Autonoma de San Luis Potosi, SLP, Mexico
| | - Alex J. Walsh
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee,
USA
| | - Melissa C. Skala
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee,
USA
| | - Javier A. Jo
- Department of Biomedical Engineering, Texas A& M University, College Station, Texas,
USA
| |
Collapse
|