1
|
Hemalatha A, Li Z, Gonzalez DG, Matte-Martone C, Tai K, Lathrop E, Gil D, Ganesan S, Gonzalez LE, Skala M, Perry RJ, Greco V. Metabolic rewiring in skin epidermis drives tolerance to oncogenic mutations. Nat Cell Biol 2025; 27:218-231. [PMID: 39762578 PMCID: PMC11821535 DOI: 10.1038/s41556-024-01574-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 11/01/2024] [Indexed: 02/06/2025]
Abstract
Skin epithelial stem cells correct aberrancies induced by oncogenic mutations. Oncogenes invoke different strategies of epithelial tolerance; while wild-type cells outcompete β-catenin-gain-of-function (βcatGOF) cells, HrasG12V cells outcompete wild-type cells. Here we ask how metabolic states change as wild-type stem cells interface with mutant cells and drive different cell-competition outcomes. By tracking the endogenous redox ratio (NAD(P)H/FAD) with single-cell resolution in the same mouse over time, we discover that βcatGOF and HrasG12V mutations, when interfaced with wild-type epidermal stem cells, lead to a rapid drop in redox ratios, indicating more oxidized cellular redox. However, the resultant redox differential persists through time in βcatGOF, whereas it is flattened rapidly in the HrasG12Vmodel. Using 13C liquid chromatography-tandem mass spectrometry, we find that the βcatGOF and HrasG12V mutant epidermis increase the fractional contribution of glucose through the oxidative tricarboxylic acid cycle. Treatment with metformin, a modifier of cytosolic redox, inhibits downstream mutant phenotypes and reverses cell-competition outcomes of both mutant models.
Collapse
Affiliation(s)
| | - Zongyu Li
- Departments of Cellular & Molecular Physiology and Internal Medicine (Endocrinology), Yale School of Medicine, New Haven, CT, USA
| | - David G Gonzalez
- Department of Genetics, Yale School of Medicine, New Haven, CT, USA
| | | | - Karen Tai
- Department of Genetics, Yale School of Medicine, New Haven, CT, USA
| | | | - Daniel Gil
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI, USA
- Morgridge Institute for Research, Madison, WI, USA
| | - Smirthy Ganesan
- Department of Genetics, Yale School of Medicine, New Haven, CT, USA
| | | | - Melissa Skala
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI, USA
- Morgridge Institute for Research, Madison, WI, USA
| | - Rachel J Perry
- Departments of Cellular & Molecular Physiology and Internal Medicine (Endocrinology), Yale School of Medicine, New Haven, CT, USA.
| | - Valentina Greco
- Department of Genetics, Yale School of Medicine, New Haven, CT, USA.
- Departments of Cell Biology and Dermatology, Yale Stem Cell Center, Yale Cancer Center, Yale School of Medicine, New Haven, CT, USA.
- Howard Hughes Medical Institute (HHMI), Chevy Chase, MD, USA.
| |
Collapse
|
2
|
Woessner AE, Witt NJ, Jones JD, Sander EA, Quinn KP. Quantification of age-related changes in the structure and mechanical function of skin with multiscale imaging. GeroScience 2024; 46:4869-4882. [PMID: 38761286 PMCID: PMC11336155 DOI: 10.1007/s11357-024-01199-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 05/08/2024] [Indexed: 05/20/2024] Open
Abstract
The mechanical properties of skin change during aging but the relationships between structure and mechanical function remain poorly understood. Previous work has shown that young skin exhibits a substantial decrease in tissue volume, a large macro-scale Poisson's ratio, and an increase in micro-scale collagen fiber alignment during mechanical stretch. In this study, label-free multiphoton microscopy was used to quantify how the microstructure and fiber kinematics of aged mouse skin affect its mechanical function. In an unloaded state, aged skin was found to have less collagen alignment and more non-enzymatic collagen fiber crosslinks. Skin samples were then loaded in uniaxial tension and aged skin exhibited a lower mechanical stiffness compared to young skin. Aged tissue also demonstrated less volume reduction and a lower macro-scale Poisson's ratio at 10% uniaxial strain, but not at 20% strain. The magnitude of 3D fiber realignment in the direction of loading was not different between age groups, and the amount of realignment in young and aged skin was less than expected based on theoretical fiber kinematics affine to the local deformation. These findings provide key insights on how the collagen fiber microstructure changes with age, and how those changes affect the mechanical function of skin, findings which may help guide wound healing or anti-aging treatments.
Collapse
Affiliation(s)
- Alan E Woessner
- Department of Biomedical Engineering, University of Arkansas, 123 John A. White Jr. Engineering Hall, Fayetteville, AR, 72701, USA
- Arkansas Integrative Metabolic Research Center, University of Arkansas, Fayetteville, AR, USA
| | - Nathan J Witt
- Roy J. Carver Department of Biomedical Engineering, University of Iowa, Iowa City, IA, USA
| | - Jake D Jones
- Department of Biomedical Engineering, University of Arkansas, 123 John A. White Jr. Engineering Hall, Fayetteville, AR, 72701, USA
| | - Edward A Sander
- Roy J. Carver Department of Biomedical Engineering, University of Iowa, Iowa City, IA, USA
| | - Kyle P Quinn
- Department of Biomedical Engineering, University of Arkansas, 123 John A. White Jr. Engineering Hall, Fayetteville, AR, 72701, USA.
- Arkansas Integrative Metabolic Research Center, University of Arkansas, Fayetteville, AR, USA.
| |
Collapse
|
3
|
Tandon I, Woessner AE, Ferreira LA, Shamblin C, Vaca-Diez G, Walls A, Kuczwara P, Applequist A, Nascimento DF, Tandon S, Kim JW, Rausch M, Timek T, Padala M, Kinter MT, Province D, Byrum SD, Quinn KP, Balachandran K. A three-dimensional valve-on-chip microphysiological system implicates cell cycle progression, cholesterol metabolism and protein homeostasis in early calcific aortic valve disease progression. Acta Biomater 2024; 186:167-184. [PMID: 39084496 DOI: 10.1016/j.actbio.2024.07.036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 07/18/2024] [Accepted: 07/23/2024] [Indexed: 08/02/2024]
Abstract
BACKGROUND Calcific aortic valve disease (CAVD) is one of the most common forms of valvulopathy, with a 50 % elevated risk of a fatal cardiovascular event, and greater than 15,000 annual deaths in North America alone. The treatment standard is valve replacement as early diagnostic, mitigation, and drug strategies remain underdeveloped. The development of early diagnostic and therapeutic strategies requires the fabrication of effective in vitro valve mimetic models to elucidate early CAVD mechanisms. METHODS In this study, we developed a multilayered physiologically relevant 3D valve-on-chip (VOC) system that incorporated aortic valve mimetic extracellular matrix (ECM), porcine aortic valve interstitial cell (VIC) and endothelial cell (VEC) co-culture and dynamic mechanical stimuli. Collagen and glycosaminoglycan (GAG) based hydrogels were assembled in a bilayer to mimic healthy or diseased compositions of the native fibrosa and spongiosa. Multiphoton imaging and proteomic analysis of healthy and diseased VOCs were performed. RESULTS Collagen-based bilayered hydrogel maintained the phenotype of the VICs. Proteins related to cellular processes like cell cycle progression, cholesterol biosynthesis, and protein homeostasis were found to be significantly altered and correlated with changes in cell metabolism in diseased VOCs. This study suggested that diseased VOCs may represent an early, adaptive disease initiation stage, which was corroborated by human aortic valve proteomic assessment. CONCLUSIONS In this study, we developed a collagen-based bilayered hydrogel to mimic healthy or diseased compositions of the native fibrosa and spongiosa layers. When the gels were assembled in a VOC with VECs and VICs, the diseased VOCs revealed key insights about the CAVD initiation process. STATEMENT OF SIGNIFICANCE Calcific aortic valve disease (CAVD) elevates the risk of death due to cardiovascular pathophysiology by 50 %, however, prevention and mitigation strategies are lacking, clinically. Developing tools to assess early disease would significantly aid in the prevention of disease and in the development of therapeutics. Previously, studies have utilized collagen and glycosaminoglycan-based hydrogels for valve cell co-cultures, valve cell co-cultures in dynamic environments, and inorganic polymer-based multilayered hydrogels; however, these approaches have not been combined to make a physiologically relevant model for CAVD studies. We fabricated a bi-layered hydrogel that closely mimics the aortic valve and used it for valve cell co-culture in a dynamic platform to gain mechanistic insights into the CAVD initiation process using proteomic and multiphoton imaging assessment.
Collapse
Affiliation(s)
- Ishita Tandon
- Department of Biomedical Engineering, University of Arkansas, Fayetteville, AR, USA
| | - Alan E Woessner
- Arkansas Integrative Metabolic Research Center, University of Arkansas, Fayetteville, AR, USA
| | - Laίs A Ferreira
- Department of Biomedical Engineering, University of Arkansas, Fayetteville, AR, USA
| | | | - Gustavo Vaca-Diez
- Department of Biomedical Engineering, University of Arkansas, Fayetteville, AR, USA
| | - Amanda Walls
- Department of Biomedical Engineering, University of Arkansas, Fayetteville, AR, USA
| | - Patrick Kuczwara
- Department of Biological and Agricultural Engineering, Materials Science & Engineering, University of Arkansas, Fayetteville, AR, USA
| | - Alexis Applequist
- Department of Biomedical Engineering, University of Arkansas, Fayetteville, AR, USA
| | - Denise F Nascimento
- Department of Biomedical Engineering, University of Arkansas, Fayetteville, AR, USA
| | - Swastika Tandon
- Department of Biomedical Engineering, University of Arkansas, Fayetteville, AR, USA
| | - Jin-Woo Kim
- Department of Biological and Agricultural Engineering, Materials Science & Engineering, University of Arkansas, Fayetteville, AR, USA
| | - Manuel Rausch
- Departments of Aerospace Engineering and Engineering Mechanics and Biomedical Engineering, Institute for Computational Engineering and Science, University of Texas at Austin, Austin, TX, USA
| | - Tomasz Timek
- Meijer Heart and Vascular Institute at Spectrum Health, Grand Rapids, MI, USA
| | - Muralidhar Padala
- Division of Cardiothoracic Surgery, Joseph P. Whitehead Department of Surgery, Emory University, Atlanta, GA, USA
| | - Michael T Kinter
- Aging & Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Dennis Province
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Stephanie D Byrum
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Kyle P Quinn
- Department of Biomedical Engineering, University of Arkansas, Fayetteville, AR, USA; Arkansas Integrative Metabolic Research Center, University of Arkansas, Fayetteville, AR, USA
| | - Kartik Balachandran
- Department of Biomedical Engineering, University of Arkansas, Fayetteville, AR, USA.
| |
Collapse
|
4
|
Song N, Gu B. in vivo and in situ robust quantitative optical biopsy of hepatocellular carcinoma and metastasis based on two-photon autofluorescence imaging. OPTICS LETTERS 2024; 49:4054-4057. [PMID: 39008774 DOI: 10.1364/ol.529284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 06/17/2024] [Indexed: 07/17/2024]
Abstract
Two-photon autofluorescence (TPAF) imaging is able to offer precise cellular metabolic information with high spatiotemporal resolution, making it a promising biopsy tool. The technique is greatly hampered by the complexity of either the optical system or data processing. Here, the excitation wavelength was optimized to simultaneously excite both flavin adenine dinucleotide and nicotinamide adenine dinucleotide and eliminate the unexpected TPAF. The optical redox ratio (ORR) images were robustly achieved without additional calibration under the optimized single-wavelength excitation. The in vitro, ex vivo, and in vivo biopsy by the TPAF method were systematically studied and compared using hepato-cellular carcinoma and metastasis as examples. It was demonstrated that the proposed TPAF method simplified the optical system, improved the robustness of ORR, and enabled early-stage cancer diagnosis, showing distinguished advantages as compared with previous methods.
Collapse
|
5
|
Ivers JD, Puvvada N, Quick CM, Rajaram N. Investigating the relationship between hypoxia, hypoxia-inducible factor 1, and the optical redox ratio in response to radiation therapy. BIOPHOTONICS DISCOVERY 2024; 1:015003. [PMID: 40109884 PMCID: PMC11922545 DOI: 10.1117/1.bios.1.1.015003] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/22/2025]
Abstract
Significance Radiation resistance is a major contributor to cancer treatment failure and is likely driven by multiple pathways. Multivariate visualization that preserves the spatial co-localization of factors could aid in understanding mechanisms of resistance and identifying biomarkers of response. Aim We aim to investigate the spatial and temporal relationship between hypoxia, hypoxia-inducible factor 1 (HIF-1α), and metabolism in response to radiation therapy in two cell lines of known radiation resistance and sensitivity. Approach Two-photon excited fluorescence and fluorescence lifetime imaging microscopy were used to quantify the optical redox ratio (ORR) and NAD(P)H fluorescent lifetime and bound fraction in frozen tumor sections and co-registered with immunohistochemical stain-based imaging of hypoxic fraction and HIF-1α. Results Histogram analysis of hypoxia, HIF-1α, and ORR revealed an increase in the ORR in regions of low hypoxia and high HIF-1α, indicating that the stabilization of HIF-1α is likely due to an increase in reactive oxygen species following radiation therapy. In addition, the bound NAD(P)H fraction was higher in regions with a low ORR in resistant tumors following radiation, suggesting an increase in fatty acid synthesis. Conclusions A multivariate histogram approach can reveal hidden trends not observed in bulk analysis of tumor images and may be useful in understanding biomarkers and mechanisms of radiation resistance.
Collapse
Affiliation(s)
- Jesse D Ivers
- University of Arkansas, Department of Biomedical Engineering, Fayetteville, Arkansas, United States
| | - Nagavenkatasai Puvvada
- University of Arkansas, Department of Biomedical Engineering, Fayetteville, Arkansas, United States
| | - Charles M Quick
- University of Arkansas for Medical Sciences, Department of Pathology, Little Rock, Arkansas, United States
| | - Narasimhan Rajaram
- University of Arkansas, Department of Biomedical Engineering, Fayetteville, Arkansas, United States
| |
Collapse
|
6
|
Bess SN, Igoe MJ, Muldoon TJ. Live-Cell Imaging Quantifies Changes in Function and Metabolic NADH Autofluorescence During Macrophage-Mediated Phagocytosis of Tumor Cells. Immunol Invest 2024; 53:210-223. [PMID: 37999933 PMCID: PMC10959688 DOI: 10.1080/08820139.2023.2284369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2023]
Abstract
BACKGROUND The immune system has evolved to detect foreign antigens and deliver coordinated responses, while minimizing "friendly fire." Until recently, studies investigating the behavior of immune cells were limited to static in vitro measurements. Although static measurements allow for real-time imaging, results are often difficult to translate to an in vivo setting. Multiphoton microscopy is an emerging method to capture spatial information on subcellular events and characterize the local microenvironment. Previous studies have shown that multiphoton microscopy can monitor changes in single-cell macrophage heterogeneity during differentiation. Therefore, there is a need to use multiphoton microscopy to monitor molecular interactions during immunological activities like phagocytosis. Here we investigate the correlation between phagocytic function and changes in endogenous optical reporters during phagocytosis. METHODS In vitro autofluorescence imaging of nicotinamide adenine dinucleotide (NADH) and flavin adenine dinucleotide (FAD) was used to detect metabolic changes in macrophages during phagocytosis. More specifically, optical redox ratio, mean NADH fluorescence lifetime and ratio of free to protein-bound NADH were used to quantify changes in metabolism. RESULTS Results show that IFN-γ (M1) macrophages showed decreased optical redox ratios and mean NADH lifetime while phagocytosing immunogenic cancer cells compared to metastatic cells. To validate phagocytic function, a fluorescence microscopy-based protocol using a pH-sensitive fluorescent probe was used. Results indicate that M0 and M1 macrophages show similar trends in phagocytic potential. CONCLUSION Overall, this work demonstrates that in vitro multiphoton imaging can be used to longitudinally track changes in phagocytosis and endogenous metabolic cofactors.
Collapse
Affiliation(s)
- Shelby N Bess
- Department of Biomedical Engineering, University of Arkansas, Fayetteville, Arkansas, USA
| | - Matthew J Igoe
- Department of Biomedical Engineering, University of Arkansas, Fayetteville, Arkansas, USA
| | - Timothy J Muldoon
- Department of Biomedical Engineering, University of Arkansas, Fayetteville, Arkansas, USA
| |
Collapse
|
7
|
Bess SN, Igoe MJ, Denison AC, Muldoon TJ. Autofluorescence imaging of endogenous metabolic cofactors in response to cytokine stimulation of classically activated macrophages. Cancer Metab 2023; 11:22. [PMID: 37957679 PMCID: PMC10644562 DOI: 10.1186/s40170-023-00325-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 11/05/2023] [Indexed: 11/15/2023] Open
Abstract
BACKGROUND Macrophages are one of the most prevalent subsets of immune cells within the tumor microenvironment and perform a range of functions depending on the cytokines and chemokines released by surrounding cells and tissues. Recent research has revealed that macrophages can exhibit a spectrum of phenotypes, making them highly plastic due to their ability to alter their physiology in response to environmental cues. Recent advances in examining heterogeneous macrophage populations include optical metabolic imaging, such as fluorescence lifetime imaging (FLIM), and multiphoton microscopy. However, the method of detection for these systems is reliant upon the coenzymes NAD(P)H and FAD, which can be affected by factors other than cytoplasmic metabolic changes. In this study, we seek to validate these optical measures of metabolism by comparing optical results to more standard methods of evaluating cellular metabolism, such as extracellular flux assays and the presence of metabolic intermediates. METHODS Here, we used autofluorescence imaging of endogenous metabolic co-factors via multiphoton microscopy and FLIM in conjunction with oxygen consumption rate and extracellular acidification rate through Seahorse extracellular flux assays to detect changes in cellular metabolism in quiescent and classically activated macrophages in response to cytokine stimulation. RESULTS Based on our Seahorse XFP flux analysis, M0 and M1 macrophages exhibit comparable trends in oxygen consumption rate (OCR) and extracellular acidification rate (ECAR). Autofluorescence imaging of M0 and M1 macrophages was not only able to show acute changes in the optical redox ratio from pre-differentiation (0 hours) to 72 hours post-cytokine differentiation (M0: 0.320 to 0.258 and M1: 0.316 to 0.386), mean NADH lifetime (M0: 1.272 ns to 1.379 ns and M1: 1.265 ns to 1.206 ns), and A1/A2 ratio (M0: 3.452 to ~ 4 and M1: 3.537 to 4.529) but could also detect heterogeneity within each macrophage population. CONCLUSIONS Overall, the findings of this study suggest that autofluorescence metabolic imaging could be a reliable technique for longitudinal tracking of immune cell metabolism during activation post-cytokine stimulation.
Collapse
Affiliation(s)
- Shelby N Bess
- Department of Biomedical Engineering, University of Arkansas, Fayetteville, AR, USA
| | - Matthew J Igoe
- Department of Biomedical Engineering, University of Arkansas, Fayetteville, AR, USA
| | - Abby C Denison
- Department of Biomedical Engineering, University of Arkansas, Fayetteville, AR, USA
| | - Timothy J Muldoon
- Department of Biomedical Engineering, University of Arkansas, Fayetteville, AR, USA.
| |
Collapse
|
8
|
Colboc H, Moguelet P, Bazin D, Letavernier E, Sun C, Chessel A, Carvalho P, Lok C, Dillies AS, Chaby G, Maillard H, Kottler D, Goujon E, Jurus C, Panaye M, Tang E, Courville P, Boury A, Monfort JB, Chasset F, Senet P, Schanne-Klein MC. Elastic fiber alterations and calcifications in calcific uremic arteriolopathy. Sci Rep 2023; 13:15519. [PMID: 37726292 PMCID: PMC10509184 DOI: 10.1038/s41598-023-42492-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 09/11/2023] [Indexed: 09/21/2023] Open
Abstract
Calcific uremic arteriolopathy (CUA) is a severely morbid disease, affecting mostly dialyzed end-stage renal disease (ESRD) patients, associated with calcium deposits in the skin. Calcifications have been identified in ESRD patients without CUA, indicating that their presence is not specific to the disease. The objective of this retrospective multicenter study was to compare elastic fiber structure and skin calcifications in ESRD patients with CUA to those without CUA using innovative structural techniques. Fourteen ESRD patients with CUA were compared to 12 ESRD patients without CUA. Analyses of elastic fiber structure and skin calcifications using multiphoton microscopy followed by machine-learning analysis and field-emission scanning electron microscopy coupled with energy dispersive X-ray were performed. Elastic fibers specifically appeared fragmented in CUA. Quantitative analyses of multiphoton images showed that they were significantly straighter in ESRD patients with CUA than without CUA. Interstitial and vascular calcifications were observed in both groups of ESRD patients, but vascular calcifications specifically appeared massive and circumferential in CUA. Unlike interstitial calcifications, massive circumferential vascular calcifications and elastic fibers straightening appeared specific to CUA. The origins of such specific elastic fiber's alteration are still to be explored and may involve relationships with ischemic vascular or inflammatory processes.
Collapse
Affiliation(s)
- Hester Colboc
- Sorbonne Université, Hôpital Rothschild, Service Plaies et Cicatrisation, UMRS_1155, 5, Rue Santerre, 75012, Paris, France.
| | - Philippe Moguelet
- Sorbonne Université, Hôpital Tenon, Anatomie et Cytologie Pathologiques, Paris, France
| | - Dominique Bazin
- Université Paris-Saclay, CNRS, Institut de Chimie Physique, 91405, Orsay, France
| | - Emmanuel Letavernier
- Sorbonne Université, Hôpital Tenon, Service des Explorations Fonctionnelles Multidisciplinaires, UMRS_1155, Paris, France
| | - Chenyu Sun
- Laboratoire d'Optique et Biosciences, CNRS, Inserm, Ecole Polytechnique, Institut Polytechnique de Paris, Palaiseau, France
| | - Anatole Chessel
- Laboratoire d'Optique et Biosciences, CNRS, Inserm, Ecole Polytechnique, Institut Polytechnique de Paris, Palaiseau, France
| | - Priscille Carvalho
- Centre Hospitalier Universitaire de Rouen, Service de Dermatologie, Rouen, France
| | - Catherine Lok
- Centre Hospitalier Universitaire d'Amiens, Service de Dermatologie, Amiens, France
| | | | - Guillaume Chaby
- Centre Hospitalier Universitaire d'Amiens, Service de Dermatologie, Amiens, France
| | - Hervé Maillard
- Centre Hospitalier du Mans, Service de Dermatologie, Le Mans, France
| | - Diane Kottler
- Centre Hospitalier Universitaire de Caen, Service de Dermatologie, Caen, France
| | - Elisa Goujon
- Centre Hospitalier de Chalon-sur-Saône, Service de Dermatologie, Chalon, France
| | - Christine Jurus
- Clinique du Tonkin, Service de Médecine Vasculaire, Villeurbanne, France
| | - Marine Panaye
- Clinique du Tonkin, Service de Médecine Vasculaire, Villeurbanne, France
| | - Ellie Tang
- Sorbonne Université, Hôpital Tenon, Service des Explorations Fonctionnelles Multidisciplinaires, UMRS_1155, Paris, France
| | - Philippe Courville
- Centre Hospitalier Universitaire de Rouen, Anatomie et Cytologie Pathologiques, Rouen, France
| | - Antoine Boury
- Université Paris-Saclay, CNRS, Institut de Chimie Physique, 91405, Orsay, France
| | - Jean-Benoit Monfort
- Sorbonne Université, Faculté de Médecine, Service de Dermatologie et Allergologie, Hôpital Tenon, Paris, France
| | - François Chasset
- Sorbonne Université, Faculté de Médecine, Service de Dermatologie 3t Allergologie, Hôpital Tenon, INSERM U1135, CIMI, Paris, France
| | - Patricia Senet
- Sorbonne Université, Faculté de Médecine, Service de Dermatologie et Allergologie, Hôpital Tenon, Paris, France
| | - Marie-Claire Schanne-Klein
- Laboratoire d'Optique et Biosciences, CNRS, Inserm, Ecole Polytechnique, Institut Polytechnique de Paris, Palaiseau, France
| |
Collapse
|
9
|
Abstract
Over the last half century, the autofluorescence of the metabolic cofactors NADH (reduced nicotinamide adenine dinucleotide) and FAD (flavin adenine dinucleotide) has been quantified in a variety of cell types and disease states. With the spread of nonlinear optical microscopy techniques in biomedical research, NADH and FAD imaging has offered an attractive solution to noninvasively monitor cell and tissue status and elucidate dynamic changes in cell or tissue metabolism. Various tools and methods to measure the temporal, spectral, and spatial properties of NADH and FAD autofluorescence have been developed. Specifically, an optical redox ratio of cofactor fluorescence intensities and NADH fluorescence lifetime parameters have been used in numerous applications, but significant work remains to mature this technology for understanding dynamic changes in metabolism. This article describes the current understanding of our optical sensitivity to different metabolic pathways and highlights current challenges in the field. Recent progress in addressing these challenges and acquiring more quantitative information in faster and more metabolically relevant formats is also discussed.
Collapse
Affiliation(s)
- Irene Georgakoudi
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts, USA;
- Genetics, Molecular and Cellular Biology Program, Graduate School of Biomedical Sciences, Tufts University, Boston, Massachusetts, USA
| | - Kyle P Quinn
- Department of Biomedical Engineering and the Arkansas Integrative Metabolic Research Center, University of Arkansas, Fayetteville, Arkansas, USA
| |
Collapse
|
10
|
Ghosh B, Agarwal K. Viewing life without labels under optical microscopes. Commun Biol 2023; 6:559. [PMID: 37231084 PMCID: PMC10212946 DOI: 10.1038/s42003-023-04934-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 05/12/2023] [Indexed: 05/27/2023] Open
Abstract
Optical microscopes today have pushed the limits of speed, quality, and observable space in biological specimens revolutionizing how we view life today. Further, specific labeling of samples for imaging has provided insight into how life functions. This enabled label-based microscopy to percolate and integrate into mainstream life science research. However, the use of labelfree microscopy has been mostly limited, resulting in testing for bio-application but not bio-integration. To enable bio-integration, such microscopes need to be evaluated for their timeliness to answer biological questions uniquely and establish a long-term growth prospect. The article presents key label-free optical microscopes and discusses their integrative potential in life science research for the unperturbed analysis of biological samples.
Collapse
|
11
|
Lim SY, Jang JI, Yoon H, Kim HM. Spectroscopic Study of Time-Varying Optical Redox Ratio in NADH/FAD Solution. J Phys Chem B 2022; 126:9840-9849. [PMID: 36399328 DOI: 10.1021/acs.jpcb.2c05292] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Autofluorescence imaging has been widely applied as advanced noninvasive diagnostics for in vivo and ex vivo tissues. The optical redox ratio (ORR), which is defined as the fluorescence intensity ratio between reduced nicotine adenine dinucleotide (NADH) and oxidized flavin adenine dinucleotide (FAD), has been used as a diagnostic parameter strongly, because NADH and FAD play an important role in energetic and respiratory metabolism as coenzymes. The ORR method has provided successful assessment in cancer diagnosis including breast, cervical, and oral cancer; few studies have been reported about optical and chemical interference between two molecules resulting in a change in ORR values. In this study, we investigated the variations in ORR values of NADH/FAD mixtures dissolved in tris(hydroxymethyl)aminomethane, phosphate buffer, and deionized water environments. In vitro solutions were prepared in various concentration ratios and the experimental and theoretical ORR values were obtained from fluorescence and absorption spectra in time series. Based on the spectroscopic analysis, we concluded that the inner filter effect causes an instant decrease in FAD fluorescence just after dissolution and that the oxidation-reduction coupled with oxygenation reaction results in time-varying decreases in NADH fluorescence and FAD emission.
Collapse
Affiliation(s)
- Soo Yeong Lim
- Department of Chemistry, Kookmin University, 77, Jeongneung-ro, Seongbuk-gu, Seoul02707, Republic of Korea
| | - Jin Il Jang
- Department of Chemistry, Kookmin University, 77, Jeongneung-ro, Seongbuk-gu, Seoul02707, Republic of Korea
| | - Hongman Yoon
- Division of Convergence Technology, National Cancer Center, 323 Ilsan-ro, Ilsandong-gu, Goyang-si, Gyeonggi-do10408, Republic of Korea
| | - Hyung Min Kim
- Department of Chemistry, Kookmin University, 77, Jeongneung-ro, Seongbuk-gu, Seoul02707, Republic of Korea
| |
Collapse
|
12
|
Dhekane R, Mhade S, Kaushik KS. Adding a new dimension: Multi-level structure and organization of mixed-species Pseudomonas aeruginosa and Staphylococcus aureus biofilms in a 4-D wound microenvironment. Biofilm 2022; 4:100087. [PMID: 36324526 PMCID: PMC9618786 DOI: 10.1016/j.bioflm.2022.100087] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Revised: 09/20/2022] [Accepted: 10/11/2022] [Indexed: 11/06/2022] Open
Abstract
Biofilms in wounds typically consist of aggregates of bacteria, most often Pseudomonas aeruginosa and Staphylococcus aureus, in close association with each other and the host microenvironment. Given this, the interplay across host and microbial elements, including the biochemical and nutrient profile of the microenvironment, likely influences the structure and organization of wound biofilms. While clinical studies, in vivo and ex vivo model systems have provided insights into the distribution of P. aeruginosa and S. aureus in wounds, they are limited in their ability to provide a detailed characterization of biofilm structure and organization across the host-microbial interface. On the other hand, biomimetic in vitro systems, such as host cell surfaces and simulant media conditions, albeit reductionist, have been shown to support the co-existence of P. aeruginosa and S. aureus biofilms, with species-dependent localization patterns and interspecies interactions. Therefore, composite in vitro models that bring together key features of the wound microenvironment could provide unprecedented insights into the structure and organization of mixed-species biofilms. We have built a four-dimensional (4-D) wound microenvironment consisting of a 3-D host cell scaffold of co-cultured human epidermal keratinocytes and dermal fibroblasts, and an in vitro wound milieu (IVWM); the IVWM provides the fourth dimension that represents the biochemical and nutrient profile of the wound infection state. We leveraged this 4-D wound microenvironment, in comparison with biofilms in IVWM alone and standard laboratory media, to probe the structure of mixed-species P. aeruginosa and S. aureus biofilms across multiple levels of organization such as aggregate dimensions and biomass thickness, species co-localization and spatial organization within the biomass, overall biomass composition and interspecies interactions. In doing so, the 4-D wound microenvironment platform provides multi-level insights into the structure of mixed-species biofilms, which we incorporate into the current understanding of P. aeruginosa and S. aureus organization in the wound bed.
Collapse
Affiliation(s)
- Radhika Dhekane
- Department of Biotechnology, Savitribai Phule Pune University, Pune, India
| | - Shreeya Mhade
- Department of Bioinformatics, Guru Nanak Khalsa College of Arts, Science and Commerce (Autonomous), Mumbai, India
| | | |
Collapse
|
13
|
Hristu R, Eftimie LG, Stanciu SG, Glogojeanu RR, Gheorghita P, Stanciu GA. Assessment of Extramammary Paget Disease by Two-Photon Microscopy. Front Med (Lausanne) 2022; 9:839786. [PMID: 35280872 PMCID: PMC8913931 DOI: 10.3389/fmed.2022.839786] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 01/24/2022] [Indexed: 12/29/2022] Open
Abstract
Two-photon microscopy techniques are non-linear optical imaging methods which are gaining momentum in the investigation of fixed tissue sections, fresh tissue or even for in vivo experiments. Two-photon excited fluorescence and second harmonic generation are two non-linear optical contrast mechanisms which can be simultaneously used for offering complementary information on the tissue architecture. While the former can originate from endogenous autofluorescence sources (e.g., NADH, FAD, elastin, keratin, lipofuscins, or melanin), or exogenous eosin, the latter is generated in fibrillar structures within living organisms (e.g., collagen and myosin). Here we test the ability of both these contrast mechanisms to highlight features of the extramammary Paget disease on fixed tissue sections prepared for standard histological examination using immunohistochemical markers and hematoxylin and eosin staining. We also demonstrate the label-free abilities of both imaging techniques to highlight histological features on unstained fixed tissue sections. The study demonstrated that two-photon microscopy can detect specific cellular features of the extramammary Paget disease in good correlation with histopathological results.
Collapse
Affiliation(s)
- Radu Hristu
- Center for Microscopy-Microanalysis and Information Processing, University Politehnica of Bucharest, Bucharest, Romania
| | - Lucian G. Eftimie
- Center for Microscopy-Microanalysis and Information Processing, University Politehnica of Bucharest, Bucharest, Romania
- Pathology Department, Central University Emergency Military Hospital, Bucharest, Romania
| | - Stefan G. Stanciu
- Center for Microscopy-Microanalysis and Information Processing, University Politehnica of Bucharest, Bucharest, Romania
| | - Remus R. Glogojeanu
- Department of Special Motricity and Medical Recovery, The National University of Physical Education and Sports, Bucharest, Romania
| | - Pavel Gheorghita
- Center for Microscopy-Microanalysis and Information Processing, University Politehnica of Bucharest, Bucharest, Romania
- Faculty of Energetics, University Politehnica of Bucharest, Bucharest, Romania
| | - George A. Stanciu
- Center for Microscopy-Microanalysis and Information Processing, University Politehnica of Bucharest, Bucharest, Romania
| |
Collapse
|
14
|
Datta R, Sivanand S, Lau AN, Florek LV, Barbeau AM, Wyckoff J, Skala MC, Vander Heiden MG. Interactions with stromal cells promote a more oxidized cancer cell redox state in pancreatic tumors. SCIENCE ADVANCES 2022; 8:eabg6383. [PMID: 35061540 PMCID: PMC8782446 DOI: 10.1126/sciadv.abg6383] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Accepted: 11/29/2021] [Indexed: 05/27/2023]
Abstract
Access to electron acceptors supports oxidized biomass synthesis and can be limiting for cancer cell proliferation, but how cancer cells overcome this limitation in tumors is incompletely understood. Nontransformed cells in tumors can help cancer cells overcome metabolic limitations, particularly in pancreatic cancer, where pancreatic stellate cells (PSCs) promote cancer cell proliferation and tumor growth. However, whether PSCs affect the redox state of cancer cells is not known. By taking advantage of the endogenous fluorescence properties of reduced nicotinamide adenine dinucleotide and oxidized flavin adenine dinucleotide cofactors we use optical imaging to assess the redox state of pancreatic cancer cells and PSCs and find that direct interactions between PSCs and cancer cells promote a more oxidized state in cancer cells. This suggests that metabolic interaction between cancer cells and PSCs is a mechanism to overcome the redox limitations of cell proliferation in pancreatic cancer.
Collapse
Affiliation(s)
- Rupsa Datta
- Morgridge Institute for Research, Madison, WI 53715, USA
| | - Sharanya Sivanand
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Allison N. Lau
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | | | - Anna M. Barbeau
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Jeffrey Wyckoff
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Melissa C. Skala
- Morgridge Institute for Research, Madison, WI 53715, USA
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI 53715, USA
| | - Matthew G. Vander Heiden
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
| |
Collapse
|
15
|
Jones JD, Rodriguez MR, Quinn KP. Automated Extraction of Skin Wound Healing Biomarkers From In Vivo Label-Free Multiphoton Microscopy Using Convolutional Neural Networks. Lasers Surg Med 2021; 53:1086-1095. [PMID: 33442889 PMCID: PMC8275674 DOI: 10.1002/lsm.23375] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 12/06/2020] [Accepted: 12/23/2020] [Indexed: 12/16/2022]
Abstract
BACKGROUND AND OBJECTIVES Histological analysis is a gold standard technique for studying impaired skin wound healing. Label-free multiphoton microscopy (MPM) can provide natural image contrast similar to histological sections and quantitative metabolic information using NADH and FAD autofluorescence. However, MPM analysis requires time-intensive manual segmentation of specific wound tissue regions limiting the practicality and usage of the technology for monitoring wounds. The goal of this study was to train a series of convolutional neural networks (CNNs) to segment MPM images of skin wounds to automate image processing and quantification of wound geometry and metabolism. STUDY DESIGN/MATERIALS AND METHODS Two CNNs with a 4-layer U-Net architecture were trained to segment unstained skin wound tissue sections and in vivo z-stacks of the wound edge. The wound section CNN used 380 distinct MPM images while the in vivo CNN used 5,848 with both image sets being randomly distributed to training, validation, and test sets following a 70%, 20%, and 10% split. The accuracy of each network was evaluated on the test set of images, and the effectiveness of automated measurement of wound geometry and optical redox ratio were compared with hand traced outputs of six unstained wound sections and 69 wound edge z-stacks from eight mice. RESULTS The MPM wound section CNN had an overall accuracy of 92.83%. Measurements of epidermal/dermal thickness, wound depth, wound width, and % re-epithelialization were within 10% error when evaluated on six full wound sections from days 3, 5, and 10 post-wounding that were not included in the training set. The in vivo wound z-stack CNN had an overall accuracy of 89.66% and was able to isolate the wound edge epithelium in z-stacks from eight mice across post-wound time points to quantify the optical redox ratio within 5% of what was recorded by manual segmentations. CONCLUSION The CNNs trained and presented in this study can accurately segment MPM imaged wound sections and in vivo z-stacks to enable automated and rapid calculation of wound geometry and metabolism. Although MPM is a noninvasive imaging modality well suited to imaging living wound tissue, its use has been limited by time-intensive user segmentation. The use of CNNs for automated image segmentation demonstrate that it is possible for MPM to deliver near real-time quantitative readouts of tissue structure and function. Lasers Surg. Med. © 2021 Wiley Periodicals LLC.
Collapse
Affiliation(s)
- Jake D. Jones
- Department of Biomedical Engineering, University of Arkansas, Fayetteville, AR, USA
| | - Marcos R. Rodriguez
- Department of Biomedical Engineering, University of Arkansas, Fayetteville, AR, USA
| | - Kyle P. Quinn
- Department of Biomedical Engineering, University of Arkansas, Fayetteville, AR, USA
| |
Collapse
|
16
|
Lee M, Kannan S, Muniraj G, Rosa V, Lu WF, Fuh JYH, Sriram G, Cao T. Two-Photon Fluorescence Microscopy and Applications in Angiogenesis and Related Molecular Events. TISSUE ENGINEERING PART B-REVIEWS 2021; 28:926-937. [PMID: 34541887 DOI: 10.1089/ten.teb.2021.0140] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The role of angiogenesis in health and disease have gained considerable momentum in recent years. Visualizing angiogenic patterns and associated events of surrounding vascular beds in response to therapeutic and laboratory-grade biomolecules have become a commonplace in regenerative medicine and the biosciences. To aid imaging investigations in angiogenesis, the two-photon excitation fluorescence microscopy (2PEF), or multiphoton fluorescence microscopy is increasingly utilized in scientific investigations. The 2PEF microscope confers several distinct imaging advantages over other fluorescence excitation microscopy techniques - for the observation of in-depth, three-dimensional vascularity in a variety of tissue formats, including fixed tissue specimens and in vivo vasculature in live specimens. Understanding morphological and subcellular changes that occur in cells and tissues during angiogenesis will provide insights to behavioral responses in diseased states, advance the engineering of physiologically-relevant tissue models and provide biochemical clues for the design of therapeutic strategies. We review the applicability and limitations of the 2PEF microscope on the biophysical and molecular-level signatures of angiogenesis in various tissue models. Imaging techniques and strategies for best practices in 2PEF microscopy will be reviewed.
Collapse
Affiliation(s)
- Marcus Lee
- Faculty of Dentistry, National University of Singapore, Singapore, Singapore
| | - Sathya Kannan
- Faculty of Dentistry, National University of Singapore, Singapore, Singapore
| | - Giridharan Muniraj
- Faculty of Dentistry, National University of Singapore, Singapore, Singapore
| | - Vinicius Rosa
- Faculty of Dentistry, National University of Singapore, Singapore, Singapore
| | - Wen Feng Lu
- Department of Mechanical Engineering, National University of Singapore, Singapore, Singapore
| | - Jerry Y H Fuh
- Department of Mechanical Engineering, National University of Singapore, Singapore, Singapore
| | - Gopu Sriram
- Faculty of Dentistry, National University of Singapore, Singapore, Singapore
| | - Tong Cao
- Faculty of Dentistry, National University of Singapore, Singapore, Singapore
| |
Collapse
|
17
|
Seeger M, Dehner C, Jüstel D, Ntziachristos V. Label-free concurrent 5-modal microscopy (Co5M) resolves unknown spatio-temporal processes in wound healing. Commun Biol 2021; 4:1040. [PMID: 34489513 PMCID: PMC8421396 DOI: 10.1038/s42003-021-02573-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Accepted: 08/18/2021] [Indexed: 02/07/2023] Open
Abstract
The non-invasive investigation of multiple biological processes remains a methodological challenge as it requires capturing different contrast mechanisms, usually not available with any single modality. Intravital microscopy has played a key role in dynamically studying biological morphology and function, but it is generally limited to resolving a small number of contrasts, typically generated by the use of transgenic labels, disturbing the biological system. We introduce concurrent 5-modal microscopy (Co5M), illustrating a new concept for label-free in vivo observations by simultaneously capturing optoacoustic, two-photon excitation fluorescence, second and third harmonic generation, and brightfield contrast. We apply Co5M to non-invasively visualize multiple wound healing biomarkers and quantitatively monitor a number of processes and features, including longitudinal changes in wound shape, microvascular and collagen density, vessel size and fractality, and the plasticity of sebaceous glands. Analysis of these parameters offers unique insights into the interplay of wound closure, vasodilation, angiogenesis, skin contracture, and epithelial reformation in space and time, inaccessible by other methods. Co5M challenges the conventional concept of biological observation by yielding multiple simultaneous parameters of pathophysiological processes in a label-free mode.
Collapse
Affiliation(s)
- Markus Seeger
- Chair of Biological Imaging, Central Institute for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, Munich, Germany
- Institute of Biological and Medical Imaging, Helmholtz Zentrum München, Neuherberg, Germany
| | - Christoph Dehner
- Chair of Biological Imaging, Central Institute for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, Munich, Germany
- Institute of Biological and Medical Imaging, Helmholtz Zentrum München, Neuherberg, Germany
| | - Dominik Jüstel
- Chair of Biological Imaging, Central Institute for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, Munich, Germany
- Institute of Biological and Medical Imaging, Helmholtz Zentrum München, Neuherberg, Germany
- Institute of Computational Biology, Helmholtz Zentrum München, Neuherberg, Germany
| | - Vasilis Ntziachristos
- Chair of Biological Imaging, Central Institute for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, Munich, Germany.
- Institute of Biological and Medical Imaging, Helmholtz Zentrum München, Neuherberg, Germany.
| |
Collapse
|
18
|
Kim S, Le VH, Kim B, Kim CJ, Im SH, Kim KH. Longitudinal Label-Free Two-Photon Microscopy of Cellular Healing Processes in Non-Ablative Fractional Laser Wounds. Lasers Surg Med 2021; 53:1413-1426. [PMID: 34139024 DOI: 10.1002/lsm.23445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 05/26/2021] [Accepted: 05/31/2021] [Indexed: 11/07/2022]
Abstract
BACKGROUND AND OBJECTIVES Wound healing is an important biomedical problem with various associated complications. Although cutaneous wound healing has been studied in vivo extensively using various optical imaging methods, early-stage cellular healing processes were difficult to study due to scab formation. The objective of this study is to demonstrate that minimal laser wounds and optical microscopy can access the detailed cellular healing processes of cutaneous wounds from the early stage. STUDY DESIGN/MATERIALS AND METHODS A non-ablative fractional laser (NAFL) and label-free two-photon microscopy (TPM) were used to induce minimal cutaneous wounds and to image the wounds in three-dimension. Sixteen hairless mice and a single human volunteer were used. NAFL wounds were induced in the hindlimb skin of the mice and in the forearm skin of the human subject. The NAFL wounds were longitudinally imaged during the healing period, starting from an hour post wound induction in the earliest and until 21 days. Cells in the wound and surrounding normal skin were visualized based on two-photon excited auto-fluorescence (TPAF), and cellular changes were tracked by analyzing longitudinal TPM images both qualitatively and quantitatively. Damage and recovery in the skin dermis were tracked by using the second harmonic generation (SHG) signal of collagen. Immunofluorescence and hematoxylin and eosin histology analysis were conducted to validate the TPM results of the murine skin. RESULTS Cellular healing processes in NAFL wounds and surroundings could be observed by longitudinal TPM. In the case of murine skin, various healing phases including inflammation, re-epithelization, granulation tissue formation, and late remodeling phase including collagen regeneration were observed in the same wounds owing to minimal or no scab formation. The re-epithelization process was analyzed quantitatively by measuring cell density and thickness of the epithelium in the wound surroundings. In the case of the human skin, the access inside the wound was blocked for a few days post wound induction due to scabs but the cellular changes in the wound surroundings were observed from the early stage. Cellular healing processes in the NAFL wound of the human skin were similar to those in murine skin. CONCLUSIONS The minimal NAFL wound model and label-free TPM demonstrated the cell level assessment of wound healing processes with applicability to human subjects. © 2021 Wiley Periodicals LLC.
Collapse
Affiliation(s)
- Seonghan Kim
- Department of Mechanical Engineering, Pohang University of Science and Technology, 77 Cheongam-Ro, Nam-gu, Pohang, Gyeongbuk, 37673, Republic of Korea
| | - Viet-Hoan Le
- Department of Life Sciences & Division of Integrative Biosciences and Biotechnology, Pohang University of Science and Technology, Pohang, 37673, Republic of Korea
| | - Bumju Kim
- Department of Life Sciences & Division of Integrative Biosciences and Biotechnology, Pohang University of Science and Technology, Pohang, 37673, Republic of Korea
| | - Chan Johng Kim
- Department of Life Sciences & Division of Integrative Biosciences and Biotechnology, Pohang University of Science and Technology, Pohang, 37673, Republic of Korea
| | - Sin-Hyeog Im
- Department of Life Sciences & Division of Integrative Biosciences and Biotechnology, Pohang University of Science and Technology, Pohang, 37673, Republic of Korea.,ImmunoBiome Inc. Bio Open Innovation Center, 47 Jigok-ro, Nam-gu, Pohang, Gyeongbukdo, 37673, Republic of Korea
| | - Ki Hean Kim
- Department of Mechanical Engineering, Pohang University of Science and Technology, 77 Cheongam-Ro, Nam-gu, Pohang, Gyeongbuk, 37673, Republic of Korea
| |
Collapse
|
19
|
Tandon I, Quinn KP, Balachandran K. Label-Free Multiphoton Microscopy for the Detection and Monitoring of Calcific Aortic Valve Disease. Front Cardiovasc Med 2021; 8:688513. [PMID: 34179147 PMCID: PMC8226007 DOI: 10.3389/fcvm.2021.688513] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 05/17/2021] [Indexed: 12/12/2022] Open
Abstract
Calcific aortic valve disease (CAVD) is the most common valvular heart disease. CAVD results in a considerable socio-economic burden, especially considering the aging population in Europe and North America. The only treatment standard is surgical valve replacement as early diagnostic, mitigation, and drug strategies remain underdeveloped. Novel diagnostic techniques and biomarkers for early detection and monitoring of CAVD progression are thus a pressing need. Additionally, non-destructive tools are required for longitudinal in vitro and in vivo assessment of CAVD initiation and progression that can be translated into clinical practice in the future. Multiphoton microscopy (MPM) facilitates label-free and non-destructive imaging to obtain quantitative, optical biomarkers that have been shown to correlate with key events during CAVD progression. MPM can also be used to obtain spatiotemporal readouts of metabolic changes that occur in the cells. While cellular metabolism has been extensively explored for various cardiovascular disorders like atherosclerosis, hypertension, and heart failure, and has shown potential in elucidating key pathophysiological processes in heart valve diseases, it has yet to gain traction in the study of CAVD. Furthermore, MPM also provides structural, functional, and metabolic readouts that have the potential to correlate with key pathophysiological events in CAVD progression. This review outlines the applicability of MPM and its derived quantitative metrics for the detection and monitoring of early CAVD progression. The review will further focus on the MPM-detectable metabolic biomarkers that correlate with key biological events during valve pathogenesis and their potential role in assessing CAVD pathophysiology.
Collapse
Affiliation(s)
- Ishita Tandon
- Department of Biomedical Engineering, University of Arkansas, Fayetteville, AR, United States
| | - Kyle P Quinn
- Department of Biomedical Engineering, University of Arkansas, Fayetteville, AR, United States
| | - Kartik Balachandran
- Department of Biomedical Engineering, University of Arkansas, Fayetteville, AR, United States
| |
Collapse
|
20
|
Haensel D, Jin S, Sun P, Cinco R, Dragan M, Nguyen Q, Cang Z, Gong Y, Vu R, MacLean AL, Kessenbrock K, Gratton E, Nie Q, Dai X. Defining Epidermal Basal Cell States during Skin Homeostasis and Wound Healing Using Single-Cell Transcriptomics. Cell Rep 2021; 30:3932-3947.e6. [PMID: 32187560 PMCID: PMC7218802 DOI: 10.1016/j.celrep.2020.02.091] [Citation(s) in RCA: 147] [Impact Index Per Article: 36.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2019] [Revised: 01/07/2020] [Accepted: 02/25/2020] [Indexed: 01/17/2023] Open
Abstract
Our knowledge of transcriptional heterogeneities in epithelial stem and progenitor cell compartments is limited. Epidermal basal cells sustain cutaneous tissue maintenance and drive wound healing. Previous studies have probed basal cell heterogeneity in stem and progenitor potential, but a comprehensive dissection of basal cell dynamics during differentiation is lacking. Using single-cell RNA sequencing coupled with RNAScope and fluorescence lifetime imaging, we identify three non-proliferative and one proliferative basal cell state in homeostatic skin that differ in metabolic preference and become spatially partitioned during wound re-epithelialization. Pseudotemporal trajectory and RNA velocity analyses predict a quasi-linear differentiation hierarchy where basal cells progress from Col17a1Hi/Trp63Hi state to early-response state, proliferate at the juncture of these two states, or become growth arrested before differentiating into spinous cells. Wound healing induces plasticity manifested by dynamic basal-spinous interconversions at multiple basal transcriptional states. Our study provides a systematic view of epidermal cellular dynamics, supporting a revised “hierarchical-lineage” model of homeostasis. Haensel et al. performed a comprehensive dissection of the cellular makeup of skin during homeostasis and wound healing and the molecular heterogeneity and cellular dynamics within its stem-cell-containing epidermal basal layer. Their work provides insights and stimulates further investigation into the mechanism of skin maintenance and repair.
Collapse
Affiliation(s)
- Daniel Haensel
- Department of Biological Chemistry, School of Medicine, University of California, Irvine, CA 92697, USA
- The NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, CA 92627, USA
- These authors contributed equally
| | - Suoqin Jin
- Department of Mathematics, University of California, Irvine, CA 92697, USA
- These authors contributed equally
| | - Peng Sun
- Department of Biological Chemistry, School of Medicine, University of California, Irvine, CA 92697, USA
| | - Rachel Cinco
- Laboratory for Fluorescence Dynamics, Department of Biomedical Engineering, University of California, Irvine, CA 92697, USA
| | - Morgan Dragan
- Department of Biological Chemistry, School of Medicine, University of California, Irvine, CA 92697, USA
- The NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, CA 92627, USA
| | - Quy Nguyen
- Department of Biological Chemistry, School of Medicine, University of California, Irvine, CA 92697, USA
| | - Zixuan Cang
- The NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, CA 92627, USA
- Department of Mathematics, University of California, Irvine, CA 92697, USA
| | - Yanwen Gong
- Department of Biological Chemistry, School of Medicine, University of California, Irvine, CA 92697, USA
- Center for Complex Biological Systems, University of California, Irvine, CA 92697, USA
| | - Remy Vu
- Department of Biological Chemistry, School of Medicine, University of California, Irvine, CA 92697, USA
- The NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, CA 92627, USA
| | - Adam L. MacLean
- Department of Mathematics, University of California, Irvine, CA 92697, USA
| | - Kai Kessenbrock
- Department of Biological Chemistry, School of Medicine, University of California, Irvine, CA 92697, USA
| | - Enrico Gratton
- Laboratory for Fluorescence Dynamics, Department of Biomedical Engineering, University of California, Irvine, CA 92697, USA
| | - Qing Nie
- The NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, CA 92627, USA
- Department of Mathematics, University of California, Irvine, CA 92697, USA
- Department of Developmental and Cell Biology, University of California, Irvine, CA 92697, USA
- Correspondence: (Q.N.), (X.D.)
| | - Xing Dai
- Department of Biological Chemistry, School of Medicine, University of California, Irvine, CA 92697, USA
- The NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, CA 92627, USA
- Lead Contact
- Correspondence: (Q.N.), (X.D.)
| |
Collapse
|
21
|
Gil DA, Deming D, Skala MC. Patient-derived cancer organoid tracking with wide-field one-photon redox imaging to assess treatment response. JOURNAL OF BIOMEDICAL OPTICS 2021; 26:JBO-200400R. [PMID: 33754540 PMCID: PMC7983069 DOI: 10.1117/1.jbo.26.3.036005] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Accepted: 02/24/2021] [Indexed: 05/04/2023]
Abstract
SIGNIFICANCE Accessible tools are needed for rapid, non-destructive imaging of patient-derived cancer organoid (PCO) treatment response to accelerate drug discovery and streamline treatment planning for individual patients. AIM To segment and track individual PCOs with wide-field one-photon redox imaging to extract morphological and metabolic variables of treatment response. APPROACH Redox imaging of the endogenous fluorophores, nicotinamide dinucleotide (NADH), nicotinamide dinucleotide phosphate (NADPH), and flavin adenine dinucleotide (FAD), was used to monitor the metabolic state and morphology of PCOs. Redox imaging was performed on a wide-field one-photon epifluorescence microscope to evaluate drug response in two colorectal PCO lines. An automated image analysis framework was developed to track PCOs across multiple time points over 48 h. Variables quantified for each PCO captured metabolic and morphological response to drug treatment, including the optical redox ratio (ORR) and organoid area. RESULTS The ORR (NAD(P)H/(FAD + NAD(P)H)) was independent of PCO morphology pretreatment. Drugs that induced cell death decreased the ORR and growth rate compared to control. Multivariate analysis of redox and morphology variables identified distinct PCO subpopulations. Single-organoid tracking improved sensitivity to drug treatment compared to pooled organoid analysis. CONCLUSIONS Wide-field one-photon redox imaging can monitor metabolic and morphological changes on a single organoid-level, providing an accessible, non-destructive tool to screen drugs in patient-matched samples.
Collapse
Affiliation(s)
- Daniel A. Gil
- University of Wisconsin, Department of Biomedical Engineering, Madison, Wisconsin, United States
- Morgridge Institute for Research, Madison, Wisconsin, United States
| | - Dustin Deming
- University of Wisconsin Carbone Cancer Center, Madison, Wisconsin, United States
- University of Wisconsin, Division of Hematology and Oncology, Department of Medicine, Madison, Wisconsin, United States
- University of Wisconsin, McArdle Laboratory for Cancer Research, Madison, Wisconsin, United States
- William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin, United States
| | - Melissa C. Skala
- University of Wisconsin, Department of Biomedical Engineering, Madison, Wisconsin, United States
- Morgridge Institute for Research, Madison, Wisconsin, United States
- University of Wisconsin Carbone Cancer Center, Madison, Wisconsin, United States
- Address all correspondence to Melissa C. Skala,
| |
Collapse
|
22
|
Ayuso JM, Rehman S, Virumbrales-Munoz M, McMinn PH, Geiger P, Fitzgerald C, Heaster T, Skala MC, Beebe DJ. Microfluidic tumor-on-a-chip model to evaluate the role of tumor environmental stress on NK cell exhaustion. SCIENCE ADVANCES 2021; 7:7/8/eabc2331. [PMID: 33597234 PMCID: PMC7888951 DOI: 10.1126/sciadv.abc2331] [Citation(s) in RCA: 81] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Accepted: 12/28/2020] [Indexed: 05/09/2023]
Abstract
Solid tumors generate a suppressive environment that imposes an overwhelming burden on the immune system. Nutrient depletion, waste product accumulation, hypoxia, and pH acidification severely compromise the capacity of effector immune cells such as T and natural killer (NK) cells to destroy cancer cells. However, the specific molecular mechanisms driving immune suppression, as well as the capacity of immune cells to adapt to the suppressive environment, are not completely understood. Thus, here, we used an in vitro microfluidic tumor-on-a-chip platform to evaluate how NK cells respond to the tumor-induced suppressive environment. The results demonstrated that the suppressive environment created by the tumor gradually eroded NK cell cytotoxic capacity, leading to compromised NK cell surveillance and tumor tolerance. Further, NK cell exhaustion persisted for an extended period of time after removing NK cells from the microfluidic platform. Last, the addition of checkpoint inhibitors and immunomodulatory agents alleviated NK cell exhaustion.
Collapse
Affiliation(s)
- Jose M Ayuso
- Department of Pathology & Laboratory Medicine, University of Wisconsin, Madison, WI, USA.
| | - Shujah Rehman
- Morgridge Institute for Research, 330 N Orchard Street, Madison, WI, USA
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI, USA
- The University of Wisconsin Carbone Cancer Center, University of Wisconsin, Madison, WI, USA
| | | | - Patrick H McMinn
- Department of Pathology & Laboratory Medicine, University of Wisconsin, Madison, WI, USA
| | - Peter Geiger
- Department of Pathology & Laboratory Medicine, University of Wisconsin, Madison, WI, USA
| | - Cate Fitzgerald
- Department of Pathology & Laboratory Medicine, University of Wisconsin, Madison, WI, USA
| | - Tiffany Heaster
- Morgridge Institute for Research, 330 N Orchard Street, Madison, WI, USA
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI, USA
| | - Melissa C Skala
- Morgridge Institute for Research, 330 N Orchard Street, Madison, WI, USA.
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI, USA
- The University of Wisconsin Carbone Cancer Center, University of Wisconsin, Madison, WI, USA
| | - David J Beebe
- Department of Pathology & Laboratory Medicine, University of Wisconsin, Madison, WI, USA.
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI, USA
- The University of Wisconsin Carbone Cancer Center, University of Wisconsin, Madison, WI, USA
| |
Collapse
|
23
|
Tandon I, Johns S, Woessner A, Perez J, Cross D, Ozkizilcik A, Muldoon TJ, Vallurupalli S, Padala M, Quinn KP, Balachandran K. Label-free optical biomarkers detect early calcific aortic valve disease in a wild-type mouse model. BMC Cardiovasc Disord 2020; 20:521. [PMID: 33308143 PMCID: PMC7731510 DOI: 10.1186/s12872-020-01776-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Accepted: 11/08/2020] [Indexed: 12/31/2022] Open
Abstract
Background Calcific aortic valve disease (CAVD) pathophysiology is a complex, multistage process, usually diagnosed at advanced stages after significant anatomical and hemodynamic changes in the valve. Early detection of disease progression is thus pivotal in the development of prevention and mitigation strategies. In this study, we developed a diet-based, non-genetically modified mouse model for early CAVD progression, and explored the utility of two-photon excited fluorescence (TPEF) microscopy for early detection of CAVD progression. TPEF imaging provides label-free, non-invasive, quantitative metrics with the potential to correlate with multiple stages of CAVD pathophysiology including calcium deposition, collagen remodeling and osteogenic differentiation. Methods Twenty-week old C57BL/6J mice were fed either a control or pro-calcific diet for 16 weeks and monitored via echocardiography, histology, immunohistochemistry, and quantitative polarized light imaging. Additionally, TPEF imaging was used to quantify tissue autofluorescence (A) at 755 nm, 810 nm and 860 nm excitation, to calculate TPEF 755–860 ratio (A860/525/(A755/460 + A860/525)) and TPEF Collagen-Calcium ratio (A810/525/(A810/460 + A810/525)) in the murine valves. In a separate experiment, animals were fed the above diets till 28 weeks to assess for later-stage calcification. Results Pro-calcific mice showed evidence of lipid deposition at 4 weeks and calcification at 16 weeks at the valve commissures. The valves of pro-calcific mice also showed positive expression for markers of osteogenic differentiation, myofibroblast activation, proliferation, inflammatory cytokines and collagen remodeling. Pro-calcific mice exhibited lower TPEF autofluorescence ratios, at locations coincident with calcification, that correlated with increased collagen disorganization and positive expression of osteogenic markers. Additionally, locations with lower TPEF autofluorescence ratios at 4 and 16 weeks exhibited increased calcification at later 28-week timepoints. Conclusions This study suggests the potential of TPEF autofluorescence metrics to serve as a label-free tool for early detection and monitoring of CAVD pathophysiology.
Collapse
Affiliation(s)
- Ishita Tandon
- Department of Biomedical Engineering, University of Arkansas, 122 John A. White Jr. Engineering Hall, Fayetteville, AR, 72701, USA
| | - Shelby Johns
- Department of Biomedical Engineering, University of Arkansas, 122 John A. White Jr. Engineering Hall, Fayetteville, AR, 72701, USA
| | - Alan Woessner
- Department of Biomedical Engineering, University of Arkansas, 122 John A. White Jr. Engineering Hall, Fayetteville, AR, 72701, USA
| | - Jessica Perez
- Department of Biomedical Engineering, University of Arkansas, 122 John A. White Jr. Engineering Hall, Fayetteville, AR, 72701, USA
| | - Delaney Cross
- Department of Biomedical Engineering, University of Arkansas, 122 John A. White Jr. Engineering Hall, Fayetteville, AR, 72701, USA
| | - Asya Ozkizilcik
- Department of Biomedical Engineering, University of Arkansas, 122 John A. White Jr. Engineering Hall, Fayetteville, AR, 72701, USA
| | - Timothy J Muldoon
- Department of Biomedical Engineering, University of Arkansas, 122 John A. White Jr. Engineering Hall, Fayetteville, AR, 72701, USA
| | - Srikanth Vallurupalli
- Division of Cardiology, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA
| | - Muralidhar Padala
- Division of Cardiothoracic Surgery, Joseph P. Whitehead Department of Surgery, Emory University, Atlanta, GA, 30322, USA
| | - Kyle P Quinn
- Department of Biomedical Engineering, University of Arkansas, 122 John A. White Jr. Engineering Hall, Fayetteville, AR, 72701, USA
| | - Kartik Balachandran
- Department of Biomedical Engineering, University of Arkansas, 122 John A. White Jr. Engineering Hall, Fayetteville, AR, 72701, USA.
| |
Collapse
|
24
|
Wang Z, Wang Y, Bradbury N, Gonzales Bravo C, Schnabl B, Di Nardo A. Skin wound closure delay in metabolic syndrome correlates with SCF deficiency in keratinocytes. Sci Rep 2020; 10:21732. [PMID: 33303806 PMCID: PMC7728784 DOI: 10.1038/s41598-020-78244-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Accepted: 11/23/2020] [Indexed: 12/15/2022] Open
Abstract
Poor wound closure due to diabetes, aging, stress, obesity, alcoholism, and chronic disease affects millions of people worldwide. Reasons wounds will not close are still unclear, and current therapies are limited. Although stem cell factor (SCF), a cytokine, is known to be important for wound repair, the cellular and molecular mechanisms of SCF in wound closure remain poorly understood. Here, we found that SCF expression in the epidermis is decreased in mouse models of delayed wound closure intended to mimic old age, obesity, and alcoholism. By using SCF conditionally knocked out mice, we demonstrated that keratinocytes' autocrine production of SCF activates a transient c-kit receptor in keratinocytes. Transient activation of the c-kit receptor induces the expression of growth factors and chemokines to promote wound re-epithelialization by increasing migration of skin cells (keratinocytes and fibroblasts) and immune cells (neutrophils) to the wound bed 24-48 h post-wounding. Our results demonstrate that keratinocyte-produced SCF is essential to wound closure due to the increased recruitment of a unique combination of skin cells and immune cells in the early phase after wounding. This discovery is imperative for developing clinical strategies that might improve the body's natural repair mechanisms for treating patients with wound-closure pathologies.
Collapse
Affiliation(s)
- Zhenping Wang
- Department of Dermatology, School of Medicine, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Yanhan Wang
- Division of Gastroenterology, School of Medicine, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Nicholas Bradbury
- Department of Dermatology, School of Medicine, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Carolina Gonzales Bravo
- Department of Dermatology, School of Medicine, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Bernd Schnabl
- Division of Gastroenterology, School of Medicine, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Anna Di Nardo
- Department of Dermatology, School of Medicine, University of California, San Diego, La Jolla, CA, 92093, USA.
| |
Collapse
|
25
|
Jones JD, Quinn KP. Automated Quantitative Analysis of Wound Histology Using Deep-Learning Neural Networks. J Invest Dermatol 2020; 141:1367-1370. [PMID: 33121938 DOI: 10.1016/j.jid.2020.10.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 09/27/2020] [Accepted: 10/07/2020] [Indexed: 01/20/2023]
Affiliation(s)
- Jake D Jones
- Department of Biomedical Engineering, University of Arkansas, Fayetteville, Arkansas, USA
| | - Kyle P Quinn
- Department of Biomedical Engineering, University of Arkansas, Fayetteville, Arkansas, USA.
| |
Collapse
|
26
|
Kim CS, Ding X, Allmeroth K, Biggs LC, Kolenc OI, L'Hoest N, Chacón-Martínez CA, Edlich-Muth C, Giavalisco P, Quinn KP, Denzel MS, Eming SA, Wickström SA. Glutamine Metabolism Controls Stem Cell Fate Reversibility and Long-Term Maintenance in the Hair Follicle. Cell Metab 2020; 32:629-642.e8. [PMID: 32905798 DOI: 10.1016/j.cmet.2020.08.011] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 06/30/2020] [Accepted: 08/13/2020] [Indexed: 12/12/2022]
Abstract
Stem cells reside in specialized niches that are critical for their function. Upon activation, hair follicle stem cells (HFSCs) exit their niche to generate the outer root sheath (ORS), but a subset of ORS progeny returns to the niche to resume an SC state. Mechanisms of this fate reversibility are unclear. We show that the ability of ORS cells to return to the SC state requires suppression of a metabolic switch from glycolysis to oxidative phosphorylation and glutamine metabolism that occurs during early HFSC lineage progression. HFSC fate reversibility and glutamine metabolism are regulated by the mammalian target of rapamycin complex 2 (mTORC2)-Akt signaling axis within the niche. Deletion of mTORC2 results in a failure to re-establish the HFSC niche, defective hair follicle regeneration, and compromised long-term maintenance of HFSCs. These findings highlight the importance of spatiotemporal control of SC metabolic states in organ homeostasis.
Collapse
Affiliation(s)
- Christine S Kim
- Max Planck Institute for Biology of Ageing, Cologne, Germany; Cluster of Excellence Cellular Stress Responses in Ageing-associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Xiaolei Ding
- Department of Dermatology, University of Cologne, Cologne, Germany
| | - Kira Allmeroth
- Max Planck Institute for Biology of Ageing, Cologne, Germany; Cluster of Excellence Cellular Stress Responses in Ageing-associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Leah C Biggs
- Helsinki Institute of Life Science, Biomedicum Helsinki, University of Helsinki, Helsinki, Finland; Wihuri Research Institute, Biomedicum Helsinki, University of Helsinki, Helsinki, Finland; Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Olivia I Kolenc
- Department of Biomedical Engineering, University of Arkansas, Fayetteville, AR, USA
| | - Nina L'Hoest
- Max Planck Institute for Biology of Ageing, Cologne, Germany; Cluster of Excellence Cellular Stress Responses in Ageing-associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Carlos Andrés Chacón-Martínez
- Max Planck Institute for Biology of Ageing, Cologne, Germany; Cluster of Excellence Cellular Stress Responses in Ageing-associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | | | | | - Kyle P Quinn
- Department of Biomedical Engineering, University of Arkansas, Fayetteville, AR, USA
| | - Martin S Denzel
- Max Planck Institute for Biology of Ageing, Cologne, Germany; Cluster of Excellence Cellular Stress Responses in Ageing-associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Sabine A Eming
- Cluster of Excellence Cellular Stress Responses in Ageing-associated Diseases (CECAD), University of Cologne, Cologne, Germany; Department of Dermatology, University of Cologne, Cologne, Germany; Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany; Institute of Zoology, Developmental Biology Unit, University of Cologne, Cologne, Germany.
| | - Sara A Wickström
- Max Planck Institute for Biology of Ageing, Cologne, Germany; Cluster of Excellence Cellular Stress Responses in Ageing-associated Diseases (CECAD), University of Cologne, Cologne, Germany; Helsinki Institute of Life Science, Biomedicum Helsinki, University of Helsinki, Helsinki, Finland; Wihuri Research Institute, Biomedicum Helsinki, University of Helsinki, Helsinki, Finland; Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland.
| |
Collapse
|
27
|
Attia ABE, Bi R, Dev K, Du Y, Olivo M. Clinical noninvasive imaging and spectroscopic tools for dermatological applications: Review of recent progress. TRANSLATIONAL BIOPHOTONICS 2020. [DOI: 10.1002/tbio.202000010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Affiliation(s)
- Amalina Binte Ebrahim Attia
- Lab of Bio‐Optical Imaging, Singapore Bioimaging Consortium (SBIC) Agency for Science Technology and Research (A*STAR) Singapore Singapore
| | - Renzhe Bi
- Lab of Bio‐Optical Imaging, Singapore Bioimaging Consortium (SBIC) Agency for Science Technology and Research (A*STAR) Singapore Singapore
| | - Kapil Dev
- Lab of Bio‐Optical Imaging, Singapore Bioimaging Consortium (SBIC) Agency for Science Technology and Research (A*STAR) Singapore Singapore
| | | | - Malini Olivo
- Lab of Bio‐Optical Imaging, Singapore Bioimaging Consortium (SBIC) Agency for Science Technology and Research (A*STAR) Singapore Singapore
| |
Collapse
|
28
|
Barkauskas DS, Medley G, Liang X, Mohammed YH, Thorling CA, Wang H, Roberts MS. Using in vivo multiphoton fluorescence lifetime imaging to unravel disease-specific changes in the liver redox state. Methods Appl Fluoresc 2020; 8:034003. [PMID: 32422610 DOI: 10.1088/2050-6120/ab93de] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Multiphoton fluorescence lifetime microscopy has revolutionized studies of pathophysiological and xenobiotic dynamics, enabling the spatial and temporal quantification of these processes in intact organs in vivo. We have previously used multiphoton fluorescence lifetime microscopy to characterise the morphology and amplitude weighted mean fluorescence lifetime of the endogenous fluorescent metabolic cofactor nicotinamide adenine dinucleotide (phosphate) (NAD(P)H) of mouse livers in vivo following induction of various disease states. Here, we extend the characterisation of liver disease models by using nonlinear regression to estimate the unbound, bound fluorescence lifetimes for NAD(P)H, flavin adenine dinucleotide (FAD), along with metabolic ratios and examine the impact of using multiple segmentation methods. We found that NAD(P)H amplitude ratio, and fluorescence lifetime redox ratio can be used as discriminators of diseased liver from normal liver. The redox ratio provided a sensitive measure of the changes in hepatic fibrosis and biliary fibrosis. Hepatocellular carcinoma was associated with an increase in spatial heterogeneity and redox ratio coupled with a decrease in mean fluorescence lifetime. We conclude that multiphoton fluorescence lifetime microscopy parameters and metabolic ratios provided insights into the in vivo redox state of diseased compared to normal liver that were not apparent from a global, mean fluorescence lifetime measurement alone.
Collapse
Affiliation(s)
- Deborah S Barkauskas
- Therapeutics Research Group, University of Queensland Diamantina Institute, The University of Queensland, Woolloongabba, QLD 4102, Australia
| | | | | | | | | | | | | |
Collapse
|
29
|
Tandon I, Kolenc OI, Cross D, Vargas I, Johns S, Quinn KP, Balachandran K. Label-free metabolic biomarkers for assessing valve interstitial cell calcific progression. Sci Rep 2020; 10:10317. [PMID: 32587322 PMCID: PMC7316720 DOI: 10.1038/s41598-020-66960-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Accepted: 05/29/2020] [Indexed: 12/13/2022] Open
Abstract
Calcific aortic valve disease (CAVD) is the most common form of valve disease where the only available treatment strategy is surgical valve replacement. Technologies for the early detection of CAVD would benefit the development of prevention, mitigation and alternate therapeutic strategies. Two-photon excited fluorescence (TPEF) microscopy is a label-free, non-destructive imaging technique that has been shown to correlate with multiple markers for cellular differentiation and phenotypic changes in cancer and wound healing. Here we show how specific TPEF markers, namely, the optical redox ratio and mitochondrial fractal dimension, correlate with structural, functional and phenotypic changes occurring in the aortic valve interstitial cells (VICs) during osteogenic differentiation. The optical redox ratio, and fractal dimension of mitochondria were assessed and correlated with gene expression and nuclear morphology of VICs. The optical redox ratio decreased for VICs during early osteogenic differentiation and correlated with biological markers for CAVD progression. Fractal dimension correlated with structural and osteogenic markers as well as measures of nuclear morphology. Our study suggests that TPEF imaging markers, specifically the optical redox ratio and mitochondrial fractal dimension, can be potentially used as a tool for assessing early CAVD progression in vitro.
Collapse
Affiliation(s)
- Ishita Tandon
- Department of Biomedical Engineering, University of Arkansas, Fayetteville, AR, 72701, USA
| | - Olivia I Kolenc
- Department of Biomedical Engineering, University of Arkansas, Fayetteville, AR, 72701, USA
| | - Delaney Cross
- Department of Biomedical Engineering, University of Arkansas, Fayetteville, AR, 72701, USA
| | - Isaac Vargas
- Department of Biomedical Engineering, University of Arkansas, Fayetteville, AR, 72701, USA
| | - Shelby Johns
- Department of Biomedical Engineering, University of Arkansas, Fayetteville, AR, 72701, USA
| | - Kyle P Quinn
- Department of Biomedical Engineering, University of Arkansas, Fayetteville, AR, 72701, USA.
| | - Kartik Balachandran
- Department of Biomedical Engineering, University of Arkansas, Fayetteville, AR, 72701, USA.
| |
Collapse
|
30
|
Datta R, Heaster TM, Sharick JT, Gillette AA, Skala MC. Fluorescence lifetime imaging microscopy: fundamentals and advances in instrumentation, analysis, and applications. JOURNAL OF BIOMEDICAL OPTICS 2020; 25:1-43. [PMID: 32406215 PMCID: PMC7219965 DOI: 10.1117/1.jbo.25.7.071203] [Citation(s) in RCA: 399] [Impact Index Per Article: 79.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Accepted: 04/24/2020] [Indexed: 05/18/2023]
Abstract
SIGNIFICANCE Fluorescence lifetime imaging microscopy (FLIM) is a powerful technique to distinguish the unique molecular environment of fluorophores. FLIM measures the time a fluorophore remains in an excited state before emitting a photon, and detects molecular variations of fluorophores that are not apparent with spectral techniques alone. FLIM is sensitive to multiple biomedical processes including disease progression and drug efficacy. AIM We provide an overview of FLIM principles, instrumentation, and analysis while highlighting the latest developments and biological applications. APPROACH This review covers FLIM principles and theory, including advantages over intensity-based fluorescence measurements. Fundamentals of FLIM instrumentation in time- and frequency-domains are summarized, along with recent developments. Image segmentation and analysis strategies that quantify spatial and molecular features of cellular heterogeneity are reviewed. Finally, representative applications are provided including high-resolution FLIM of cell- and organelle-level molecular changes, use of exogenous and endogenous fluorophores, and imaging protein-protein interactions with Förster resonance energy transfer (FRET). Advantages and limitations of FLIM are also discussed. CONCLUSIONS FLIM is advantageous for probing molecular environments of fluorophores to inform on fluorophore behavior that cannot be elucidated with intensity measurements alone. Development of FLIM technologies, analysis, and applications will further advance biological research and clinical assessments.
Collapse
Affiliation(s)
- Rupsa Datta
- Morgridge Institute for Research, Madison, Wisconsin, United States
| | - Tiffany M. Heaster
- Morgridge Institute for Research, Madison, Wisconsin, United States
- University of Wisconsin, Department of Biomedical Engineering, Madison, Wisconsin, United States
| | - Joe T. Sharick
- Morgridge Institute for Research, Madison, Wisconsin, United States
| | - Amani A. Gillette
- Morgridge Institute for Research, Madison, Wisconsin, United States
- University of Wisconsin, Department of Biomedical Engineering, Madison, Wisconsin, United States
| | - Melissa C. Skala
- Morgridge Institute for Research, Madison, Wisconsin, United States
- University of Wisconsin, Department of Biomedical Engineering, Madison, Wisconsin, United States
| |
Collapse
|
31
|
Synthetic Retinoid Seletinoid G Improves Skin Barrier Function through Wound Healing and Collagen Realignment in Human Skin Equivalents. Int J Mol Sci 2020; 21:ijms21093198. [PMID: 32366052 PMCID: PMC7247558 DOI: 10.3390/ijms21093198] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 04/21/2020] [Accepted: 04/29/2020] [Indexed: 12/17/2022] Open
Abstract
The outer epidermal skin is a primary barrier that protects the body from extrinsic factors, such as ultraviolet (UV) radiation, chemicals and pollutants. The complete epithelialization of a wound by keratinocytes is essential for restoring the barrier function of the skin. However, age-related alterations predispose the elderly to impaired wound healing. Therefore, wound-healing efficacy could be also considered as a potent function of an anti-aging reagent. Here, we examine the epidermal wound-healing efficacy of the fourth-generation retinoid, seletinoid G, using HaCaT keratinocytes and skin tissues. We found that seletinoid G promoted the proliferation and migration of keratinocytes in scratch assays and time-lapse imaging. It also increased the gene expression levels of several keratinocyte proliferation-regulating factors. In human skin equivalents, seletinoid G accelerated epidermal wound closure, as assessed using optical coherence tomography (OCT) imaging. Moreover, second harmonic generation (SHG) imaging revealed that seletinoid G recovered the reduced dermal collagen deposition seen in ultraviolet B (UVB)-irradiated human skin equivalents. Taken together, these results indicate that seletinoid G protects the skin barrier by accelerating wound healing in the epidermis and by repairing collagen deficiency in the dermis. Thus, seletinoid G could be a potent anti-aging agent for protecting the skin barrier.
Collapse
|
32
|
Borghi F, Silva C, da Silva PC, Ferrucci DL, Morais CL, Conceição-Vertamatti AG, Carvalho HF, Fonseca MDC, Vieira AS, Grassi-Kassisse DM. The influence of hypertensive environment on adipose tissue remodeling measured by fluorescence lifetime imaging in spontaneously hypertensive rats. Mol Cell Endocrinol 2020; 506:110758. [PMID: 32057944 DOI: 10.1016/j.mce.2020.110758] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 02/10/2020] [Accepted: 02/10/2020] [Indexed: 12/20/2022]
Abstract
There is a lack of information correlating low adiposity with hypertension experienced by Spontaneous Hypertensive Rats (SHR) or overweight and normotension in Wistar-Kyoto (WKY). We aimed to investigate this lipodystrophy phenomenon by measuring fluorescence lifetime (FLIM), optical redox ratio (ORR), serum levels of hypothalamic-pituitary-adrenal (HPA) and/or hypothalamic-pituitary-thyroid (HPT) hormones axes between Wistar, WKY and SHR before and after establishment of hypertension. Under high blood pressure, we evaluated serum adipokines. Brown adipose tissue was characterized as lower ORR and shorter FLIM compared to white adipose tissue. HPT axis showed a crucial role in the SHR adipose tissue configuration by attenuating whitening. The increased adiposity in WKY may act as a preventive agent for hypertension, since SHR, with low adiposity, establishes the disease. The hypertensive environment can highlight key adipokines that may result in new therapeutic approaches to the treatment of adiposity dysfunctions and hypertension.
Collapse
Affiliation(s)
- Filipy Borghi
- LABEEST, Laboratory of Stress Study, Department of Structural and Functional Biology, Institute of Biology, University of Campinas, UNICAMP, 13083-862, Campinas, SP, Brazil
| | - Carolina Silva
- LABEEST, Laboratory of Stress Study, Department of Structural and Functional Biology, Institute of Biology, University of Campinas, UNICAMP, 13083-862, Campinas, SP, Brazil
| | - Priscila Cristina da Silva
- LABEEST, Laboratory of Stress Study, Department of Structural and Functional Biology, Institute of Biology, University of Campinas, UNICAMP, 13083-862, Campinas, SP, Brazil
| | - Danilo Lopes Ferrucci
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas, UNICAMP, 13083-862, Campinas, SP, Brazil
| | - Camila Lidiane Morais
- LABEEST, Laboratory of Stress Study, Department of Structural and Functional Biology, Institute of Biology, University of Campinas, UNICAMP, 13083-862, Campinas, SP, Brazil
| | - Ana Gabriela Conceição-Vertamatti
- LABEEST, Laboratory of Stress Study, Department of Structural and Functional Biology, Institute of Biology, University of Campinas, UNICAMP, 13083-862, Campinas, SP, Brazil
| | - Hernandes Faustino Carvalho
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas, UNICAMP, 13083-862, Campinas, SP, Brazil
| | - Matheus de Castro Fonseca
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), 13083-970, Campinas, Sao Paulo, Brazil
| | - André Schwambach Vieira
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas, UNICAMP, 13083-862, Campinas, SP, Brazil
| | - Dora Maria Grassi-Kassisse
- LABEEST, Laboratory of Stress Study, Department of Structural and Functional Biology, Institute of Biology, University of Campinas, UNICAMP, 13083-862, Campinas, SP, Brazil.
| |
Collapse
|
33
|
Morone D, Autilia FD, Schorn T, Erreni M, Doni A. Evaluation of cell metabolic adaptation in wound and tumour by Fluorescence Lifetime Imaging Microscopy. Sci Rep 2020; 10:6289. [PMID: 32286404 PMCID: PMC7156395 DOI: 10.1038/s41598-020-63203-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Accepted: 03/20/2020] [Indexed: 12/12/2022] Open
Abstract
Acidic pH occurs in acute wounds progressing to healing as consequence of a cell metabolic adaptation in response to injury-induced tissue hypoperfusion. In tumours, high metabolic rate leads to acidosis affecting cancer progression. Acidic pH affects activities of remodelling cells in vitro. The pH measurement predicts healing in pathological wounds and success of surgical treatment of burns and chronic ulcers. However, current methods are limited to skin surface or based on detection of fluorescence intensity of specific sensitive probes that suffer of microenvironment factors. Herein, we ascertained relevance in vivo of cell metabolic adaptation in skin repair by interfering with anaerobic glycolysis. Moreover, a custom-designed skin imaging chamber, 2-Photon microscopy (2PM), fluorescence lifetime imaging (FLIM) and data mapping analyses were used to correlate maps of glycolytic activity in vivo as measurement of NADH intrinsic lifetime with areas of hypoxia and acidification in models of skin injury and cancer. The method was challenged by measuring the NADH profile by interfering with anaerobic glycolysis and oxidative phosphorylation in the mitochondrial respiratory chain. Therefore, intravital NADH FLIM represents a tool for investigating cell metabolic adaptation occurring in wounds, as well as the relationship between cell metabolism and cancer.
Collapse
Affiliation(s)
- Diego Morone
- Unit of Advanced Optical Microscopy, IRCCS, Humanitas Clinical and Research Center, Rozzano, Milan, Italy.,Faculty of Biomedical Sciences, Institute for Research in Biomedicine, Università della Svizzera italiana (USI), Bellinzona, Switzerland
| | - Francesca D' Autilia
- Unit of Advanced Optical Microscopy, IRCCS, Humanitas Clinical and Research Center, Rozzano, Milan, Italy
| | - Tilo Schorn
- Unit of Advanced Optical Microscopy, IRCCS, Humanitas Clinical and Research Center, Rozzano, Milan, Italy
| | - Marco Erreni
- Unit of Advanced Optical Microscopy, IRCCS, Humanitas Clinical and Research Center, Rozzano, Milan, Italy
| | - Andrea Doni
- Unit of Advanced Optical Microscopy, IRCCS, Humanitas Clinical and Research Center, Rozzano, Milan, Italy.
| |
Collapse
|
34
|
Rico-Jimenez J, Lee JH, Alex A, Musaad S, Chaney E, Barkalifa R, Spillman DR, Olson E, Adams D, Marjanovic M, Arp Z, Boppart SA. Non-invasive monitoring of pharmacodynamics during the skin wound healing process using multimodal optical microscopy. BMJ Open Diabetes Res Care 2020; 8:8/1/e000974. [PMID: 32327442 PMCID: PMC7202789 DOI: 10.1136/bmjdrc-2019-000974] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 01/13/2020] [Accepted: 02/22/2020] [Indexed: 12/12/2022] Open
Abstract
OBJECTIVE Impaired diabetic wound healing is one of the serious complications associated with diabetes. In patients with diabetes, this impairment is characterized by several physiological abnormalities such as metabolic changes, reduced collagen production, and diminished angiogenesis. We designed and developed a multimodal optical imaging system that can longitudinally monitor formation of new blood vessels, metabolic changes, and collagen deposition in a non-invasive, label-free manner. RESEARCH DESIGN AND METHODS The closure of a skin wound in (db/db) mice, which presents delayed wound healing pathologically similar to conditions in human type 2 diabetes mellitus, was non-invasively followed using the custom-built multimodal microscope. In this microscope, optical coherence tomography angiography was used for studying neovascularization, fluorescence lifetime imaging microscopy for nicotinamide adenine dinucleotide (phosphate) (NAD(P)H) assessment, fluorescence intensity changes of NAD(P)H and flavin adenine dinucleotide (FAD) cofactors for evaluating metabolic changes, and second harmonic generation microscopy for analyzing collagen deposition and organization. The animals were separated into four groups: control, placebo, low concentration (LC), and high concentration (HC) treatment. Images of the wound and surrounding areas were acquired at different time points during a 28-day period. RESULTS Various physiological changes measured using the optical imaging modalities at different phases of wound healing were compared. A statistically significant improvement in the functional relationship between angiogenesis, metabolism, and structural integrity was observed in the HC group. CONCLUSIONS This study demonstrated the capability of multimodal optical imaging to non-invasively monitor various physiological aspects of the wound healing process, and thus become a promising tool in the development of better diagnostic, treatment, and monitoring strategies for diabetic wound care.
Collapse
Affiliation(s)
- Jose Rico-Jimenez
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - Jang Hyuk Lee
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - Aneesh Alex
- GlaxoSmithKline, Philadelphia, Pennsylvania, USA
| | - Salma Musaad
- Interdisciplinary Health Sciences Institute, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - Eric Chaney
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - Ronit Barkalifa
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - Darold R Spillman
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - Eric Olson
- GlaxoSmithKline, Philadelphia, Pennsylvania, USA
| | - David Adams
- GlaxoSmithKline, Philadelphia, Pennsylvania, USA
| | - Marina Marjanovic
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - Zane Arp
- GlaxoSmithKline, Philadelphia, Pennsylvania, USA
| | - Stephen A Boppart
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| |
Collapse
|
35
|
Blair MJ, Jones JD, Woessner AE, Quinn KP. Skin Structure-Function Relationships and the Wound Healing Response to Intrinsic Aging. Adv Wound Care (New Rochelle) 2020; 9:127-143. [PMID: 31993254 DOI: 10.1089/wound.2019.1021] [Citation(s) in RCA: 85] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Accepted: 08/09/2019] [Indexed: 02/06/2023] Open
Abstract
Significance: Chronic wounds, such as diabetic foot ulcers, venous stasis ulcers, and pressure ulcers affect millions of Americans each year, and disproportionately afflict our increasingly older population. Older individuals are predisposed to wound infection, repeated trauma, and the development of chronic wounds. However, a complete understanding of how the attributes of aging skin affect the wound healing process has remained elusive. Recent Advances: A variety of studies have demonstrated that the dermal matrix becomes thinner, increasingly crosslinked, and fragmented with advanced age. These structural changes, as well as an increase in cell senescence, result in altered collagen fiber remodeling and increased stiffness. Studies combining mechanical testing with advanced imaging techniques are providing new insights into the relationships between these age-related changes. Emerging research into the mechanobiology of aging and the wound healing process indicate that the altered mechanical environment of aged skin may have a significant effect on age-related delays in healing. Critical Issues: The interpretation and synthesis of clinical studies is confounded by the effects of common comorbidities that also contribute to the development of chronic wounds. A lack of quantitative biomarkers of wound healing and age-related changes makes understanding structure-function relationships during the wound healing process challenging. Future Directions: Additional work is needed to establish quantitative and mechanistic relationships among age-related changes in the skin microstructure, mechanical function, and the cellular responses to wound healing.
Collapse
Affiliation(s)
- Michael J. Blair
- Department of Biomedical Engineering, University of Arkansas, Fayetteville, Arkansas
| | - Jake D. Jones
- Department of Biomedical Engineering, University of Arkansas, Fayetteville, Arkansas
| | - Alan E. Woessner
- Department of Biomedical Engineering, University of Arkansas, Fayetteville, Arkansas
| | - Kyle P. Quinn
- Department of Biomedical Engineering, University of Arkansas, Fayetteville, Arkansas
| |
Collapse
|
36
|
Li J, Wilson MN, Bower AJ, Marjanovic M, Chaney EJ, Barkalifa R, Boppart SA. Video-rate multimodal multiphoton imaging and three-dimensional characterization of cellular dynamics in wounded skin. JOURNAL OF INNOVATIVE OPTICAL HEALTH SCIENCES 2020; 13:2050007. [PMID: 33584862 PMCID: PMC7880242 DOI: 10.1142/s1793545820500078] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
To date, numerous studies have been performed to elucidate the complex cellular dynamics in skin diseases, but few have attempted to characterize these cellular events under conditions similar to the native environment. To address this challenge, a three-dimensional (3D) multimodal analysis platform was developed for characterizing in vivo cellular dynamics in skin, which was then utilized to process in vivo wound healing data to demonstrate its applicability. Special attention is focused on in vivo biological parameters that are difficult to study with ex vivo analysis, including 3D cell tracking and techniques to connect biological information obtained from different imaging modalities. These results here open new possibilities for evaluating 3D cellular dynamics in vivo, and can potentially provide new tools for characterizing the skin microenvironment and pathologies in the future.
Collapse
Affiliation(s)
- Joanne Li
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL, U.S.A
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL, U.S.A
| | - Madison N. Wilson
- Department of Electrical and Computer Engineering, University of Illinois at Urbana-Champaign Urbana, IL, U.S.A
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL, U.S.A
| | - Andrew J. Bower
- Department of Electrical and Computer Engineering, University of Illinois at Urbana-Champaign Urbana, IL, U.S.A
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL, U.S.A
| | - Marina Marjanovic
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL, U.S.A
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL, U.S.A
| | - Eric J. Chaney
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL, U.S.A
| | - Ronit Barkalifa
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL, U.S.A
| | - Stephen A. Boppart
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL, U.S.A
- Department of Electrical and Computer Engineering, University of Illinois at Urbana-Champaign Urbana, IL, U.S.A
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL, U.S.A
- Carle Illinois College of Medicine, University of Illinois at Urbana-Champaign, Urbana, IL, U.S.A
| |
Collapse
|
37
|
Jones JD, Ramser HE, Woessner AE, Veves A, Quinn KP. Quantifying Age-Related Changes in Skin Wound Metabolism Using In Vivo Multiphoton Microscopy. Adv Wound Care (New Rochelle) 2020; 9:90-102. [PMID: 31993251 PMCID: PMC6985773 DOI: 10.1089/wound.2019.1030] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Accepted: 07/28/2019] [Indexed: 12/27/2022] Open
Abstract
Objective: The elderly are at high risk for developing chronic skin wounds, but the effects of intrinsic aging on skin healing are difficult to isolate due to common comorbidities like diabetes. Our objective is to use multiphoton microscopy (MPM) to find endogenous, noninvasive biomarkers to differentiate changes in skin wound healing metabolism between young and aged mice in vivo. Approach: We utilized MPM to monitor skin metabolism at the edge of full-thickness, excisional wounds in 24- and 4-month-old mice of both sexes for 10 days. MPM can assess quantitative biomarkers of cellular metabolism in vivo by utilizing autofluorescence from the cofactors nicotinamide adenine dinucleotide (NADH) and flavin adenine dinucleotide (FAD). Results: An optical redox ratio of FAD/(NADH+FAD) autofluorescence and NADH fluorescence lifetime imaging revealed dynamic changes in keratinocyte function during healing. Aged female mice demonstrated an attenuation of keratinocyte proliferation during wound healing detectable optically through a higher redox ratio and longer NADH fluorescence lifetime. By measuring the correlation between NADH lifetime and the optical redox ratio at each day, we also demonstrate sensitivity to the proliferative phase of wound healing. Innovation: Label-free MPM was used to longitudinally monitor individual wounds in vivo, which revealed age-dependent differences in wound metabolism. Conclusion: These results indicate in vivo MPM can provide quantitative biomarkers of age-related delays in healing, which can be used in the future to provide patient-specific wound care.
Collapse
Affiliation(s)
- Jake D. Jones
- Department of Biomedical Engineering, University of Arkansas, Fayetteville, Arkansas
| | - Hallie E. Ramser
- Department of Biomedical Engineering, University of Arkansas, Fayetteville, Arkansas
| | - Alan E. Woessner
- Department of Biomedical Engineering, University of Arkansas, Fayetteville, Arkansas
| | - Aristidis Veves
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Kyle P. Quinn
- Department of Biomedical Engineering, University of Arkansas, Fayetteville, Arkansas
| |
Collapse
|
38
|
Mohammed YH, Barkauskas DS, Holmes A, Grice J, Roberts MS. Noninvasive in vivo human multiphoton microscopy: a key method in proving nanoparticulate zinc oxide sunscreen safety. JOURNAL OF BIOMEDICAL OPTICS 2020; 25:1-19. [PMID: 31939224 PMCID: PMC7008509 DOI: 10.1117/1.jbo.25.1.014509] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Accepted: 12/03/2019] [Indexed: 05/27/2023]
Abstract
We describe the contribution of our in vivo multiphoton microscopy (MPM) studies over the last ten years with DermaInspect;® (JenLab, Germany), a CE-certified medical tomograph based on detection of fluorescent biomolecules, to the assessment of possible penetration of nanoparticulate zinc oxide in sunscreen through human skin. At the time we started our work, there was a strong movement for the precautionary principle to be applied to the use of nanoparticles in consumer products due to a lack of knowledge. The combined application of different MPM modalities, including spectral imaging, fluorescence lifetime imaging, second harmonic fluorescence generation, and phosphorescence microscopy, has provided overwhelming evidence that nanoparticle zinc oxide particles do not penetrate human skin when applied to various skin types with a range of methods of topical sunscreen application. MPM has also been used to study the viable epidermal morphology and redox state in supporting the safe use of topical zinc oxide nanoparticles. The impact of this work is emphasized by the recent proposed rule by the United States FDA on Sunscreen Drug Products for Over-the-Counter Human Use, which listed only zinc oxide and titanium dioxide of the currently marketed products to be generally recognized as safe and effective.
Collapse
Affiliation(s)
- Yousuf H. Mohammed
- University of Queensland, University of Queensland Diamantina Institute, Therapeutics Research Group, Woolloongabba, Queensland, Australia
| | - Deborah S. Barkauskas
- University of Queensland, University of Queensland Diamantina Institute, Therapeutics Research Group, Woolloongabba, Queensland, Australia
| | - Amy Holmes
- University of South Australia, Basil Hetzel Institute for Translational Medical Research, The Queen Elizabeth Hospital, School of Pharmacy and Medical Sciences, Adelaide, Australia
| | - Jeffrey Grice
- University of Queensland, University of Queensland Diamantina Institute, Therapeutics Research Group, Woolloongabba, Queensland, Australia
| | - Michael S. Roberts
- University of Queensland, University of Queensland Diamantina Institute, Therapeutics Research Group, Woolloongabba, Queensland, Australia
- University of South Australia, Basil Hetzel Institute for Translational Medical Research, The Queen Elizabeth Hospital, School of Pharmacy and Medical Sciences, Adelaide, Australia
| |
Collapse
|
39
|
Yuan Y, Yan Z, Miao J, Cai R, Zhang M, Wang Y, Wang L, Dang W, Wang D, Xiang D, Wang Y, Zhang P, Cui Y, Bian X, Ma Q. Autofluorescence of NADH is a new biomarker for sorting and characterizing cancer stem cells in human glioma. Stem Cell Res Ther 2019; 10:330. [PMID: 31747975 PMCID: PMC6865050 DOI: 10.1186/s13287-019-1467-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 10/14/2019] [Accepted: 10/22/2019] [Indexed: 02/07/2023] Open
Abstract
Background The existing cell surface markers used for sorting glioma stem cells (GSCs) have obvious limitations, such as vulnerability to the enzymatic digestion and time-consuming labeling procedure. Reduced nicotinamide adenine dinucleotide (NADH) as a cellular metabolite with property of autofluorescence has the potential to be used as a new biomarker for sorting GSCs. Methods A method for sorting GSCs was established according to the properties of the autofluorescence of NADH. Then, the NADHhigh and NADHlow subpopulations were sorted. The stem-like properties of the subpopulations were evaluated by qRT-PCR, western blot analyses, limiting dilution assay, cell viability assay, bioluminescence imaging, and immunofluorescence analysis in vitro and in vivo. The relationship between CD133+/CD15+ cells and NADHhigh subpopulation was also assessed. Results NADHhigh cells expressed higher stem-related genes, formed more tumor spheres, and harbored stronger pluripotency in vitro and higher tumorigenicity in vivo, compared to NADHlow subpopulation. NADHhigh glioma cells had the similar stemness with CD133+ or CD15+ GSCs, but the three subpopulations less overlaid each other. Also, NADHhigh glioma cells were more invasive and more resistant to chemotherapeutic drug temozolomide (TMZ) than NADHlow cells. In addition, the autofluorescence of NADH might be an appropriate marker to sort cancer stem cells (CSCs) in other cancer types, such as breast and colon cancer. Conclusion Our findings demonstrate that intracellular autofluorescence of NADH is a non-labeling, sensitive maker for isolating GSCs, even for other CSCs.
Collapse
Affiliation(s)
- Ye Yuan
- Institute of Pathology and Southwest Cancer Center, Key Laboratory of the Ministry of Education, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Zexuan Yan
- Institute of Pathology and Southwest Cancer Center, Key Laboratory of the Ministry of Education, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Jingya Miao
- Institute of Pathology and Southwest Cancer Center, Key Laboratory of the Ministry of Education, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Ruili Cai
- Institute of Pathology and Southwest Cancer Center, Key Laboratory of the Ministry of Education, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Mengsi Zhang
- Institute of Pathology and Southwest Cancer Center, Key Laboratory of the Ministry of Education, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Yanxia Wang
- Institute of Pathology and Southwest Cancer Center, Key Laboratory of the Ministry of Education, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Lihong Wang
- Institute of Pathology and Southwest Cancer Center, Key Laboratory of the Ministry of Education, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Weiqi Dang
- Institute of Pathology and Southwest Cancer Center, Key Laboratory of the Ministry of Education, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Di Wang
- Institute of Pathology and Southwest Cancer Center, Key Laboratory of the Ministry of Education, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Dongfang Xiang
- Institute of Pathology and Southwest Cancer Center, Key Laboratory of the Ministry of Education, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Yan Wang
- Institute of Pathology and Southwest Cancer Center, Key Laboratory of the Ministry of Education, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Peng Zhang
- Institute of Pathology and Southwest Cancer Center, Key Laboratory of the Ministry of Education, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Youhong Cui
- Institute of Pathology and Southwest Cancer Center, Key Laboratory of the Ministry of Education, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China.
| | - Xiuwu Bian
- Institute of Pathology and Southwest Cancer Center, Key Laboratory of the Ministry of Education, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China.
| | - Qinghua Ma
- Institute of Pathology and Southwest Cancer Center, Key Laboratory of the Ministry of Education, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China.
| |
Collapse
|
40
|
Dadgar S, Rajaram N. Optical Imaging Approaches to Investigating Radiation Resistance. Front Oncol 2019; 9:1152. [PMID: 31750246 PMCID: PMC6848224 DOI: 10.3389/fonc.2019.01152] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Accepted: 10/16/2019] [Indexed: 12/14/2022] Open
Abstract
Radiation therapy is frequently the first line of treatment for over 50% of cancer patients. While great advances have been made in improving treatment response rates and reducing damage to normal tissue, radiation resistance remains a persistent clinical problem. While hypoxia or a lack of tumor oxygenation has long been considered a key factor in causing treatment failure, recent evidence points to metabolic reprogramming under well-oxygenated conditions as a potential route to promoting radiation resistance. In this review, we present recent studies from our lab and others that use high-resolution optical imaging as well as clinical translational optical spectroscopy to shine light on the biological basis of radiation resistance. Two-photon microscopy of endogenous cellular metabolism has identified key changes in both mitochondrial structure and function that are specific to radiation-resistant cells and help promote cell survival in response to radiation. Optical spectroscopic approaches, such as diffuse reflectance and Raman spectroscopy have demonstrated functional and molecular differences between radiation-resistant and sensitive tumors in response to radiation. These studies have uncovered key changes in metabolic pathways and present a viable route to clinical translation of optical technologies to determine radiation resistance at a very early stage in the clinic.
Collapse
Affiliation(s)
| | - Narasimhan Rajaram
- Department of Biomedical Engineering, University of Arkansas, Fayetteville, AR, United States
| |
Collapse
|
41
|
Woessner AE, McGee JD, Jones JD, Quinn KP. Characterizing differences in the collagen fiber organization of skin wounds using quantitative polarized light imaging. Wound Repair Regen 2019; 27:711-714. [PMID: 31418977 DOI: 10.1111/wrr.12758] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Accepted: 08/09/2019] [Indexed: 11/29/2022]
Abstract
Collagen fiber organization requires characterization in many biomedical applications, but it is difficult to objectively quantify in standard histology tissue sections. Quantitative polarized light imaging is a low-cost technique that allows for rapid measurement of collagen fiber orientation and thickness. In this study, we utilize a quantitative polarized light imaging system to characterize fiber orientation and thickness from wound sections. Full thickness skin wound sections that were previously stained with hematoxylin and eosin were used to assess collagen fiber content and organization at different points during the wound healing process. Overall, wounds exhibited a measurable increase in collagen fiber thickness and a nonlinear change in fiber reorganization within the wound. Our study demonstrates that quantitative polarized light imaging is an inexpensive alternative or supplement to standard histology protocols, requiring no additional stains or dyes, and yields repeatable quantitative assessments of collagen organization.
Collapse
Affiliation(s)
- Alan E Woessner
- Department of Biomedical Engineering, University of Arkansas, Fayetteville, Arkansas
| | - James D McGee
- Department of Biomedical Engineering, University of Arkansas, Fayetteville, Arkansas
| | - Jake D Jones
- Department of Biomedical Engineering, University of Arkansas, Fayetteville, Arkansas
| | - Kyle P Quinn
- Department of Biomedical Engineering, University of Arkansas, Fayetteville, Arkansas
| |
Collapse
|
42
|
Ranawat H, Pal S, Mazumder N. Recent trends in two-photon auto-fluorescence lifetime imaging (2P-FLIM) and its biomedical applications. Biomed Eng Lett 2019; 9:293-310. [PMID: 31456890 PMCID: PMC6694381 DOI: 10.1007/s13534-019-00119-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 05/30/2019] [Accepted: 06/27/2019] [Indexed: 02/07/2023] Open
Abstract
Two photon fluorescence microscopy and the numerous technical advances to it have served as valuable tools in biomedical research. The fluorophores (exogenous or endogenous) absorb light and emit lower energy photons than the absorption energy and the emission (fluorescence) signal is measured using a fluorescence decay graph. Additionally, high spatial resolution images can be acquired in two photon fluorescence lifetime imaging (2P-FLIM) with improved penetration depth which helps in detection of fluorescence signal in vivo. 2P-FLIM is a non-invasive imaging technique in order to visualize cellular metabolic, by tracking intrinsic fluorophores present in it, such as nicotinamide adenine dinucleotide, flavin adenine dinucleotide and tryptophan etc. 2P-FLIM of these molecules enable the visualization of metabolic alterations, non-invasively. This comprehensive review discusses the numerous applications of 2P-FLIM towards cancer, neuro-degenerative, infectious diseases, and wound healing.
Collapse
Affiliation(s)
- Harsh Ranawat
- Department of Biophysics, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka 576104 India
| | - Sagnik Pal
- Department of Biophysics, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka 576104 India
| | - Nirmal Mazumder
- Department of Biophysics, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka 576104 India
| |
Collapse
|
43
|
Lam NT, Tandon I, Balachandran K. The role of fibroblast growth factor 1 and 2 on the pathological behavior of valve interstitial cells in a three-dimensional mechanically-conditioned model. J Biol Eng 2019; 13:45. [PMID: 31149027 PMCID: PMC6537403 DOI: 10.1186/s13036-019-0168-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Accepted: 04/11/2019] [Indexed: 01/08/2023] Open
Abstract
Background More than five million Americans suffer from heart valve disease annually, a condition that worsens cardiac function and gradually leads to heart failure if appropriate treatment is not performed on time. Currently no medication can cure heart valve disease, leaving surgical intervention as the only viable option for patients at late stages of cardiac valve disease. Tremendous efforts have been undertaken to elucidate how resident cells in the valves respond to pathological stimulation as well as the underlying mechanisms that regulate these responses, to identify potential therapeutic targets for non-surgical treatment of valvular heart disease. Results Cardiac valve interstitial cells (VICs) naturally reside in a complex three-dimensional environment under varying hemodynamics, which is difficult to replicate in vitro. As a result, most cell signaling studies in the field have traditionally been conducted on two-dimensional models or in the absence of hemodynamic forces. Previously, we reported the fabrication of a hydrogel scaffold that could be used to culture valve cells under dynamic mechanical stimulation in a valve-mimetic environment. This model, therefore appeared to be suitable for VIC signaling studies as it provided cells a three-dimensional environment with the ability to incorporate mechanical stretching stimulation. Utilizing this model, we investigated the possible role of fibroblast growth factor 1 and 2 (FGF1 and FGF2) via FGFR1 receptor signaling in regulating valve cell activation under physiological (10% stretch) and pathological (20% stretch) mechanical conditions as well as in mediating cell proliferation and metabolism via the Akt/mTOR pathways. We reported that 1) FGF1 and FGF2 treatment was able to maintain the quiescent phenotype of VICs; 2) Cells increased proliferation as determined by optical redox ratios under elevated cyclic stretch via Akt/mTOR pathways; and 3) FGF1 and 2 signaling via the FGFR1 reduced VIC proliferation and activation under elevated cyclic stretch conditions. Conclusions Overall, these results suggested that targeting FGFR1 receptor signaling may represent a possible therapeutic strategy for preventing heart valve disease progression. Electronic supplementary material The online version of this article (10.1186/s13036-019-0168-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Ngoc Thien Lam
- 1Cell and Molecular Biology Program, University of Arkansas, Fayetteville, AR 72701 USA
| | - Ishita Tandon
- 2Department of Biomedical Engineering, University of Arkansas, 122 John A. White Jr. Engineering Hall, Fayetteville, AR 72701 USA
| | - Kartik Balachandran
- 1Cell and Molecular Biology Program, University of Arkansas, Fayetteville, AR 72701 USA.,2Department of Biomedical Engineering, University of Arkansas, 122 John A. White Jr. Engineering Hall, Fayetteville, AR 72701 USA
| |
Collapse
|