1
|
Alves JL, Reis PM, Quinta-Ferreira RM, Quinta-Ferreira ME, Matias CM. Changes in reactive oxygen species and autofluorescence under hypoxia at the hippocampal CA3 area: role of calcium and zinc influxes. Neurochem Int 2024; 180:105882. [PMID: 39413928 DOI: 10.1016/j.neuint.2024.105882] [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: 05/09/2024] [Revised: 09/12/2024] [Accepted: 10/13/2024] [Indexed: 10/18/2024]
Abstract
Reactive oxygen species (ROS) have an important role in cellular biology, being involved, in a way that depends on their levels, in cell signaling processes or in oxidative stress, probably associated with neurodegenerative and other diseases. Most of the studies about ROS formation were performed in ischemic conditions, and thus, there is limited knowledge about ROS formation in less severe hypoxic conditions. This study investigates neuronal ROS generation and autofluorescence changes in hypoxic conditions, focusing on the involvement of calcium and zinc. Using hippocampal slices from Wistar rats, ROS production was monitored by the permeant fluorescent indicator H2DCFDA under different oxygenation levels. Moderate hypoxia (40% O2) led to a small ROS increase, while severe hypoxia (0% O2) showed a more pronounced rise. KCl-induced depolarization significantly enhanced ROS formation, particularly under severe hypoxia. Inhibition of NMDA receptors reduced ROS generation without affecting autofluorescence, while chelation of zinc ions decreased ROS production and increased flavin adenine dinucleotide (FAD) autofluorescence. These findings suggest that, in hypoxic conditions, ROS formation is mediated by calcium entry through NMDA receptors and also by zinc influxes. Thus, these ions play a crucial role in oxidative stress, which may be related with neurodegenerative diseases associated with ROS dysregulation.
Collapse
Affiliation(s)
- João L Alves
- Department of Life Sciences, University of Coimbra, Portugal; CNC-UC - Center for Neurosciences and Cell Biology, University of Coimbra, Portugal
| | - Patrícia M Reis
- CERES - Chemical Engineering and Renewable Resources for Sustainability, Department of Chemical Engineering, University of Coimbra, Portugal
| | - Rosa M Quinta-Ferreira
- CERES - Chemical Engineering and Renewable Resources for Sustainability, Department of Chemical Engineering, University of Coimbra, Portugal
| | - M Emília Quinta-Ferreira
- CNC-UC - Center for Neurosciences and Cell Biology, University of Coimbra, Portugal; Department of Physics, University of Coimbra, Portugal
| | - Carlos M Matias
- CNC-UC - Center for Neurosciences and Cell Biology, University of Coimbra, Portugal; Department of Physics, UTAD, Vila Real, Portugal.
| |
Collapse
|
2
|
Baigildin V, Shakirova J, Zharskaia N, Ivanova E, Silonov S, Sokolov V, Tunik S. Design and Preparation of Lifetime-Based Dual Fluorescent/Phosphorescent Sensor of pH and Oxygen and its Exploration in Model Physiological Solutions and Cells. Macromol Biosci 2024; 24:e2400225. [PMID: 38987922 DOI: 10.1002/mabi.202400225] [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: 05/07/2024] [Revised: 06/14/2024] [Indexed: 07/12/2024]
Abstract
In the present report, a novel dual pH-O2 sensor based on covalent conjugate of rhodamine 6G and cyclometalated iridium complex with poly(vinylpyrrolidone-block-vinyltetrazole) copolymer is reported. In model physiological solutions the sensor chromophores display independent phosphorescent and fluorescent lifetime responses onto variations in oxygen concentration and pH, respectively. Colocalization studies on Chinese hamster ovary cells demonstrate the preferential localization in endosomes and lysosomes. The fluorescent lifetime imaging microscopy-phosphorescent lifetime imaging microscopy (FLIM-PLIM) experiments show that the phosphorescent O2 sensor provides unambiguous information onto hypoxia versus normoxia cell status as well as semi-quantitative data on the oxygen concentration in cells in between these two states. However, the results of FLIM measurements indicate that dynamic lifetime interval of the sensor (≈0.5 ns between pH values 5.0 and 8.0) is insufficient even for qualitative estimation of pH in living cells because half-width of lifetime distribution in the studied samples is higher than the sensor dynamic interval. Nevertheless, the variations in rhodamine emission intensity are much higher and allow rough discrimination of acidic and neutral cell conditions. Thus, the results of this study indicate that the suggested approach to the design of dual pH-O2 sensors makes possible to prepare the biocompatible and water-soluble conjugate with fast cellular uptake.
Collapse
Affiliation(s)
- Vadim Baigildin
- Institute of Chemistry, Saint-Petersburg State University, Universitetsky Pr. 26, St. Petersburg, 198504, Russia
| | - Julia Shakirova
- Institute of Chemistry, Saint-Petersburg State University, Universitetsky Pr. 26, St. Petersburg, 198504, Russia
| | - Nina Zharskaia
- Institute of Chemistry, Saint-Petersburg State University, Universitetsky Pr. 26, St. Petersburg, 198504, Russia
| | - Elena Ivanova
- Institute of Chemistry, Saint-Petersburg State University, Universitetsky Pr. 26, St. Petersburg, 198504, Russia
| | - Sergey Silonov
- Institute of Chemistry, Saint-Petersburg State University, Universitetsky Pr. 26, St. Petersburg, 198504, Russia
- Laboratory of Structural Dynamics, Stability and Folding of Proteins, Institute of Cytology, Russian Academy of Sciences, 4 Tikhoretsky Ave., St. Petersburg, 194064, Russia
| | - Viktor Sokolov
- Institute of Chemistry, Saint-Petersburg State University, Universitetsky Pr. 26, St. Petersburg, 198504, Russia
| | - Sergey Tunik
- Institute of Chemistry, Saint-Petersburg State University, Universitetsky Pr. 26, St. Petersburg, 198504, Russia
| |
Collapse
|
3
|
Kühl M, Nielsen DA, Borisov SM. In Vivo Lifetime Imaging of the Internal O 2 Dynamics in Corals with near-Infrared-Emitting Sensor Nanoparticles. ACS Sens 2024; 9:4671-4679. [PMID: 39179239 PMCID: PMC11443520 DOI: 10.1021/acssensors.4c01029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/26/2024]
Abstract
Mapping of O2 with luminescent sensors within intact animals is challenging due to attenuation of excitation and emission light caused by tissue absorption and scattering as well as interfering background fluorescence. Here we show the application of luminescent O2 sensor nanoparticles (∼50-70 nm) composed of the O2 indicator platinum(II) tetra(4-fluoro)phenyltetrabenzoporphyrin (PtTPTBPF) immobilized in poly(methyl methacrylate-co-methacrylic acid) (PMMA-MA). We injected the sensor nanoparticles into the gastrovascular system of intact colony fractions of reef-building tropical corals that harbor photosynthetic microalgae in their tissues. The sensor nanoparticles are excited by red LED light (617 nm) and emit in the near-infrared (780 nm), which enhances the transmission of excitation and emission light through biological materials. This enabled us to map the internal O2 concentration via time-domain luminescence lifetime imaging through the outer tissue layers across several coral polyps in flowing seawater. After injection, nanoparticles dispersed within the coral tissue for several hours. While luminescence intensity imaging showed some local aggregation of sensor particles, lifetime imaging showed a more homogeneous O2 distribution across a larger area of the coral colony. Local stimulation of symbiont photosynthesis in corals induced oxygenation of illuminated tissue areas and formation of lateral O2 gradients toward surrounding respiring tissues, which were dissipated rapidly after the onset of darkness. Such measurements are key to improving our understanding of how corals regulate their internal chemical microenvironment and metabolic activity, and how they are affected by environmental stress such as ocean warming, acidification, and deoxygenation. Our experimental approach can also be adapted for in vivo O2 imaging in other natural systems such as biofilms, plant and animal tissues, as well as in organoids and other cell constructs, where imaging internal O2 conditions are relevant and challenging due to high optical density and background fluorescence.
Collapse
Affiliation(s)
- Michael Kühl
- Marine Biological Section, Department of Biology, University of Copenhagen, Strandpromenaden 5, 3000 Helsingør, Denmark
- Climate Change Cluster, University of Technology Sydney, Broadway 2007, Australia
| | | | - Sergey M Borisov
- Institute of Analytical Chemistry and Food Chemistry, Graz University of Technology, Stremayrgasse 9, A-8010 Graz, Austria
| |
Collapse
|
4
|
Bing Y, Józsa TI, Payne SJ. Parameter quantification for oxygen transport in the human brain. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2024; 257:108433. [PMID: 39362064 DOI: 10.1016/j.cmpb.2024.108433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 09/02/2024] [Accepted: 09/18/2024] [Indexed: 10/05/2024]
Abstract
BACKGROUND AND OBJECTIVE Oxygen is carried to the brain by blood flow through generations of vessels across a wide range of length scales. This multi-scale nature of blood flow and oxygen transport poses challenges on investigating the mechanisms underlying both healthy and pathological states through imaging techniques alone. Recently, multi-scale models describing whole brain perfusion and oxygen transport have been developed. Such models rely on effective parameters that represent the microscopic properties. While parameters of the perfusion models have been characterised, those for oxygen transport are still lacking. In this study, we set to quantify the parameters associated with oxygen transport and their uncertainties. METHODS Effective parameter values of a continuum-based porous multi-scale, multi-compartment oxygen transport model are systematically estimated. In particular, geometric parameters that capture the microvascular topologies are obtained through statistically accurate capillary networks. Maximum consumption rates of oxygen are optimised to uniquely define the oxygen distribution over depth. Simulations are then carried out within a one-dimensional tissue column and a three-dimensional patient-specific brain mesh using the finite element method. RESULTS Effective values of the geometric parameters, vessel volume fraction and surface area to volume ratio, are found to be 1.42% and 627 [mm2/mm3], respectively. These values compare well with those acquired from human and monkey vascular samples. Simulation results of the one-dimensional tissue column show qualitative agreement with experimental measurements of tissue oxygen partial pressure in rats. Differences between the oxygenation level in the tissue column and the brain mesh are observed, which highlights the importance of anatomical accuracy. Finally, one-at-a-time sensitivity analysis reveals that the oxygen model is not sensitive to most of its parameters; however, perturbations in oxygen solubilities and plasma to whole blood oxygen concentration ratio have a considerable impact on the tissue oxygenation. CONCLUSIONS The findings of this study demonstrate the validity of using a porous continuum approach to model organ-scale oxygen transport and draw attention to the significance of anatomy and parameters associated with inter-compartment diffusion.
Collapse
Affiliation(s)
- Yun Bing
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford, UK
| | - Tamás I Józsa
- Centre for Computational Engineering Sciences, School of Aerospace, Transport and Manufacturing, Cranfield University, Cranfield, UK.
| | - Stephen J Payne
- Institute of Applied Mechanics, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
5
|
Charest J, Walsh M, Genois É, Sévigny E, Schwarz PO, Gagnon L, Desjardins M. Comparison of compartmental analytical Blood-Oxygen-Level-Dependent functional Magnetic Resonance Imaging models against Monte Carlo simulations performed over cortical micro-angiograms. NMR IN BIOMEDICINE 2024:e5252. [PMID: 39245649 DOI: 10.1002/nbm.5252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 08/06/2024] [Accepted: 08/19/2024] [Indexed: 09/10/2024]
Abstract
Blood oxygen level-dependent functional magnetic resonance imaging (BOLD fMRI) arises from a physiological and physical cascade of events taking place at the level of the cortical microvasculature which constitutes a medium with complex geometry. Several analytical models of the BOLD contrast have been developed, but these have not been compared directly against detailed bottom-up modeling methods. Using a 3D modeling method based on experimentally measured images of mice microvasculature and Monte Carlo simulations, we quantified the accuracy of two analytical models to predict the amplitude of the BOLD response from 1.5 to 7 T, for different echo time (TE) and for both gradient echo and spin echo acquisition protocols. We also showed that accounting for the tridimensional structure of the microvasculature results in more accurate prediction of the BOLD amplitude, even if the values for SO2 were averaged across individual vascular compartments. A secondary finding is that modeling the venous compartment as two individual compartments results in more accurate prediction of the BOLD amplitude compared with standard homogenous venous modeling, arising from the bimodal distribution of venous SO2 across the microvasculature in our data.
Collapse
Affiliation(s)
- Jordan Charest
- Department of Physics, Engineering Physics and Optics, Université Laval, Quebec, Canada
| | - Mathieu Walsh
- Department of Physics, Engineering Physics and Optics, Université Laval, Quebec, Canada
| | - Élie Genois
- Department of Physics, Université de Sherbrooke, Sherbrooke, Canada
| | - Emmanuelle Sévigny
- Department of Radiology and Nuclear Medicine, Université Laval, Quebec, Canada
| | | | - Louis Gagnon
- Department of Physics, Engineering Physics and Optics, Université Laval, Quebec, Canada
- Department of Radiology and Nuclear Medicine, Université Laval, Quebec, Canada
| | - Michèle Desjardins
- Department of Physics, Engineering Physics and Optics, Université Laval, Quebec, Canada
- Oncology Division, Centre de recherche du CHU de Québec-Université Laval, Quebec, Canada
| |
Collapse
|
6
|
Tomar A, Engelmann SA, Woods AL, Dunn AK. Non-degenerate two-photon imaging of deep rodent cortex using indocyanine green in the water absorption window. BIOMEDICAL OPTICS EXPRESS 2024; 15:5053-5066. [PMID: 39296386 PMCID: PMC11407249 DOI: 10.1364/boe.520977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 07/15/2024] [Accepted: 07/20/2024] [Indexed: 09/21/2024]
Abstract
We present a novel approach for deep vascular imaging in rodent cortex at excitation wavelengths susceptible to water absorption using two-photon microscopy with photons of dissimilar wavelengths. We demonstrate that non-degenerate two-photon excitation (ND-2PE) enables imaging in the water absorption window from 1400-1550 nm using two excitation sources with temporally overlapped pulses at 1300 nm and 1600 nm that straddle the absorption window. We explore the brightness spectra of indocyanine green (ICG) and assess its suitability for imaging in the water absorption window. Further, we demonstrate in vivo imaging of the rodent cortex vascular structure up to 1.2 mm using ND-2PE. Lastly, a comparative analysis of ND-2PE at 1435 nm and single-wavelength, two-photon imaging at 1300 nm and 1435 nm is presented. Our work extends the excitation range for fluorescent dyes to include water absorption regimes and underscores the feasibility of deep two-photon imaging at these wavelengths.
Collapse
Affiliation(s)
- Alankrit Tomar
- Department of Eletrical and Computer Engineering, The University of Texas at Austin, 2501 Speedway, Austin, TX 78712, USA
| | - Shaun A Engelmann
- Department of Biomedical Engineering, The University of Texas at Austin, 107 W. Dean Keeton, Austin, TX 78712, USA
| | - Aaron L Woods
- Department of Biomedical Engineering, The University of Texas at Austin, 107 W. Dean Keeton, Austin, TX 78712, USA
| | - Andrew K Dunn
- Department of Eletrical and Computer Engineering, The University of Texas at Austin, 2501 Speedway, Austin, TX 78712, USA
- Department of Biomedical Engineering, The University of Texas at Austin, 107 W. Dean Keeton, Austin, TX 78712, USA
| |
Collapse
|
7
|
Zhou H, Wang J, Zhu Z, Hu L, An E, Lu J, Zhao H. A New Perspective on Stroke Research: Unraveling the Role of Brain Oxygen Dynamics in Stroke Pathophysiology. Aging Dis 2024:AD.2024.0548. [PMID: 39226161 DOI: 10.14336/ad.2024.0548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 08/02/2024] [Indexed: 09/05/2024] Open
Abstract
Stroke, a leading cause of death and disability, often results from ischemic events that cut off the brain blood flow, leading to neuron death. Despite treatment advancements, survivors frequently endure lasting impairments. A key focus is the ischemic penumbra, the area around the stroke that could potentially recover with prompt oxygenation; yet its monitoring is complex. Recent progress in bioluminescence-based oxygen sensing, particularly through the Green enhanced Nano-lantern (GeNL), offers unprecedented views of oxygen fluctuations in vivo. Utilized in awake mice, GeNL has uncovered hypoxic pockets within the cerebral cortex, revealing the brain's oxygen environment as a dynamic landscape influenced by physiological states and behaviors like locomotion and wakefulness. These findings illuminate the complexity of oxygen dynamics and suggest the potential impact of hypoxic pockets on ischemic injury and recovery, challenging existing paradigms and highlighting the importance of microenvironmental oxygen control in stroke resilience. This review examines the implications of these novel findings for stroke research, emphasizing the criticality of understanding pre-existing oxygen dynamics for addressing brain ischemia. The presence of hypoxic pockets in non-stroke conditions indicates a more intricate hypoxic scenario in ischemic brains, suggesting strategies to alleviate hypoxia could lead to more effective treatments and rehabilitation. By bridging gaps in our knowledge, especially concerning microenvironmental changes post-stroke, and leveraging new technologies like GeNL, we can pave the way for therapeutic innovations that significantly enhance outcomes for stroke survivors, promising a future where an understanding of cerebral oxygenation dynamics profoundly informs stroke therapy.
Collapse
Affiliation(s)
- Hongmei Zhou
- Anesthesiology Department, The Second Hospital of Jiaxing, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Jialing Wang
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Joint Innovation Center for Brain Disorders, Capital Medical University, Beijing, China
| | - Zhipeng Zhu
- Anesthesiology Department, The Second Hospital of Jiaxing, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Li Hu
- Anesthesiology Department, The Second Hospital of Jiaxing, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Erdan An
- Anesthesiology Department, The Second Hospital of Jiaxing, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Jian Lu
- Anesthesiology Department, The Second Hospital of Jiaxing, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Heng Zhao
- Anesthesiology Department, The Second Hospital of Jiaxing, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Joint Innovation Center for Brain Disorders, Capital Medical University, Beijing, China
| |
Collapse
|
8
|
Lee LCC, Lo KKW. Shining New Light on Biological Systems: Luminescent Transition Metal Complexes for Bioimaging and Biosensing Applications. Chem Rev 2024; 124:8825-9014. [PMID: 39052606 PMCID: PMC11328004 DOI: 10.1021/acs.chemrev.3c00629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/27/2024]
Abstract
Luminescence imaging is a powerful and versatile technique for investigating cell physiology and pathology in living systems, making significant contributions to life science research and clinical diagnosis. In recent years, luminescent transition metal complexes have gained significant attention for diagnostic and therapeutic applications due to their unique photophysical and photochemical properties. In this Review, we provide a comprehensive overview of the recent development of luminescent transition metal complexes for bioimaging and biosensing applications, with a focus on transition metal centers with a d6, d8, and d10 electronic configuration. We elucidate the structure-property relationships of luminescent transition metal complexes, exploring how their structural characteristics can be manipulated to control their biological behavior such as cellular uptake, localization, biocompatibility, pharmacokinetics, and biodistribution. Furthermore, we introduce the various design strategies that leverage the interesting photophysical properties of luminescent transition metal complexes for a wide variety of biological applications, including autofluorescence-free imaging, multimodal imaging, organelle imaging, biological sensing, microenvironment monitoring, bioorthogonal labeling, bacterial imaging, and cell viability assessment. Finally, we provide insights into the challenges and perspectives of luminescent transition metal complexes for bioimaging and biosensing applications, as well as their use in disease diagnosis and treatment evaluation.
Collapse
Affiliation(s)
- Lawrence Cho-Cheung Lee
- Department of Chemistry, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong, P. R. China
- Laboratory for Synthetic Chemistry and Chemical Biology Limited, Units 1503-1511, 15/F, Building 17W, Hong Kong Science Park, New Territories, Hong Kong, P. R. China
| | - Kenneth Kam-Wing Lo
- Department of Chemistry, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong, P. R. China
- State Key Laboratory of Terahertz and Millimeter Waves, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong, P. R. China
| |
Collapse
|
9
|
Rathbone E, Fu D. Quantitative Optical Imaging of Oxygen in Brain Vasculature. J Phys Chem B 2024; 128:6975-6989. [PMID: 38991095 DOI: 10.1021/acs.jpcb.4c01277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/13/2024]
Abstract
The intimate relationship between neuronal activity and cerebral oxygenation underpins fundamental brain functions like cognition, sensation, and motor control. Optical imaging offers a noninvasive approach to assess brain oxygenation and often serves as an indirect proxy for neuronal activity. However, deciphering neurovascular coupling─the intricate interplay between neuronal activity, blood flow, and oxygen delivery─necessitates independent, high spatial resolution, and high temporal resolution measurements of both microvasculature oxygenation and neuronal activation. This Perspective examines the established optical techniques employed for brain oxygen imaging, specifically functional near-infrared spectroscopy, photoacoustic imaging, optical coherence tomography, and two-photon phosphorescent lifetime microscopy, highlighting their fundamental principles, strengths, and limitations. Several other emerging optical techniques are also introduced. Finally, we discuss key technological challenges and future directions for quantitative optical oxygen imaging, paving the way for a deeper understanding of oxygen metabolism in the brain.
Collapse
Affiliation(s)
- Emily Rathbone
- Department of Chemistry, University of Washington, Seattle, Washington 98195, United States
| | - Dan Fu
- Department of Chemistry, University of Washington, Seattle, Washington 98195, United States
| |
Collapse
|
10
|
Liu C, Cárdenas-Rivera A, Teitelbaum S, Birmingham A, Alfadhel M, Yaseen MA. Neuroinflammation increases oxygen extraction in a mouse model of Alzheimer's disease. Alzheimers Res Ther 2024; 16:78. [PMID: 38600598 PMCID: PMC11005245 DOI: 10.1186/s13195-024-01444-5] [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: 09/29/2023] [Accepted: 03/31/2024] [Indexed: 04/12/2024]
Abstract
BACKGROUND Neuroinflammation, impaired metabolism, and hypoperfusion are fundamental pathological hallmarks of early Alzheimer's disease (AD). Numerous studies have asserted a close association between neuroinflammation and disrupted cerebral energetics. During AD progression and other neurodegenerative disorders, a persistent state of chronic neuroinflammation reportedly exacerbates cytotoxicity and potentiates neuronal death. Here, we assessed the impact of a neuroinflammatory challenge on metabolic demand and microvascular hemodynamics in the somatosensory cortex of an AD mouse model. METHODS We utilized in vivo 2-photon microscopy and the phosphorescent oxygen sensor Oxyphor 2P to measure partial pressure of oxygen (pO2) and capillary red blood cell flux in cortical microvessels of awake mice. Intravascular pO2 and capillary RBC flux measurements were performed in 8-month-old APPswe/PS1dE9 mice and wildtype littermates on days 0, 7, and 14 of a 14-day period of lipopolysaccharide-induced neuroinflammation. RESULTS Before the induced inflammatory challenge, AD mice demonstrated reduced metabolic demand but similar capillary red blood cell flux as their wild type counterparts. Neuroinflammation provoked significant reductions in cerebral intravascular oxygen levels and elevated oxygen extraction in both animal groups, without significantly altering red blood cell flux in capillaries. CONCLUSIONS This study provides evidence that neuroinflammation alters cerebral oxygen demand at the early stages of AD without substantially altering vascular oxygen supply. The results will guide our understanding of neuroinflammation's influence on neuroimaging biomarkers for early AD diagnosis.
Collapse
Affiliation(s)
- Chang Liu
- Department of Bioengineering, Northeastern University, Boston, MA, 02115, USA
| | | | - Shayna Teitelbaum
- Department of Bioengineering, Northeastern University, Boston, MA, 02115, USA
| | - Austin Birmingham
- Department of Bioengineering, Northeastern University, Boston, MA, 02115, USA
| | - Mohammed Alfadhel
- Department of Bioengineering, Northeastern University, Boston, MA, 02115, USA
| | - Mohammad A Yaseen
- Department of Bioengineering, Northeastern University, Boston, MA, 02115, USA.
| |
Collapse
|
11
|
Longden TA, Lederer WJ. Electro-metabolic signaling. J Gen Physiol 2024; 156:e202313451. [PMID: 38197953 PMCID: PMC10783436 DOI: 10.1085/jgp.202313451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 10/27/2023] [Accepted: 12/14/2023] [Indexed: 01/11/2024] Open
Abstract
Precise matching of energy substrate delivery to local metabolic needs is essential for the health and function of all tissues. Here, we outline a mechanistic framework for understanding this critical process, which we refer to as electro-metabolic signaling (EMS). All tissues exhibit changes in metabolism over varying spatiotemporal scales and have widely varying energetic needs and reserves. We propose that across tissues, common signatures of elevated metabolism or increases in energy substrate usage that exceed key local thresholds rapidly engage mechanisms that generate hyperpolarizing electrical signals in capillaries that then relax contractile elements throughout the vasculature to quickly adjust blood flow to meet changing needs. The attendant increase in energy substrate delivery serves to meet local metabolic requirements and thus avoids a mismatch in supply and demand and prevents metabolic stress. We discuss in detail key examples of EMS that our laboratories have discovered in the brain and the heart, and we outline potential further EMS mechanisms operating in tissues such as skeletal muscle, pancreas, and kidney. We suggest that the energy imbalance evoked by EMS uncoupling may be central to cellular dysfunction from which the hallmarks of aging and metabolic diseases emerge and may lead to generalized organ failure states-such as diverse flavors of heart failure and dementia. Understanding and manipulating EMS may be key to preventing or reversing these dysfunctions.
Collapse
Affiliation(s)
- Thomas A. Longden
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, USA
- Laboratory of Neurovascular Interactions, Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - W. Jonathan Lederer
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, USA
- Laboratory of Molecular Cardiology, Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
12
|
Mikkelsen SH, Skøtt MV, Gutierrez E, Postnov DD. Laser speckle imaging of the hippocampus. BIOMEDICAL OPTICS EXPRESS 2024; 15:1268-1277. [PMID: 38404300 PMCID: PMC10890870 DOI: 10.1364/boe.507371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 12/15/2023] [Accepted: 12/20/2023] [Indexed: 02/27/2024]
Abstract
Research on hippocampal blood flow is essential for gaining insight into its involvement in learning and memory and its role in age-related cognitive impairment and dementia. In this study, we applied laser speckle contrast imaging (LSCI) and dynamic light scattering imaging (DLSI) to monitor perfusion in mouse hippocampus via a chronic, optically transparent window. LSCI scans showed hippocampal blood vessels appear more out of focus than similar caliber vessels in the mouse cortex. We hypothesize that it is caused by the inverse vascular topology and increased contribution of multiply-scattered photons detected from the upper layers of the hippocampus. We support the hypothesis with DLSI, showing a 1300% increased contribution of multiple-scattering unordered dynamics regime in large hippocampal vessels.
Collapse
Affiliation(s)
- Signe H. Mikkelsen
- Center of Functionally Integrative Neuroscience, Aarhus University, Aarhus, Denmark
| | - Mia V. Skøtt
- Center of Functionally Integrative Neuroscience, Aarhus University, Aarhus, Denmark
| | | | | |
Collapse
|
13
|
Tomar A, Engelmann SA, Woods AL, Dunn AK. Non-Degenerate Two-Photon Imaging of Deep Rodent Cortex using Indocyanine Green in the water absorption window. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.13.575485. [PMID: 38293101 PMCID: PMC10827096 DOI: 10.1101/2024.01.13.575485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2024]
Abstract
We present a novel approach for deep vascular imaging in rodent cortex at excitation wavelengths susceptible to water absorption using two-photon microscopy with photons of dissimilar wavelengths. We demonstrate that non-degenerate two-photon excitation (ND-2PE) enables imaging in the water absorption window from 1400-1550 nm using two synchronized excitation sources at 1300 nm and 1600 nm that straddle the absorption window. We explore the brightness spectra of indocyanine green (ICG) and assess its suitability for imaging in the water absorption window. Further, we demonstrate in vivo imaging of the rodent cortex vascular structure up to 1.2 mm using ND-2PE. Lastly, a comparative analysis of ND-2PE at 1435 nm and single-wavelength, two-photon imaging at 1300 nm and 1435 nm is presented. Our work extends the excitation range for fluorescent dyes to include water absorption regimes and underscores the feasibility of deep two-photon imaging at these wavelengths.
Collapse
Affiliation(s)
- Alankrit Tomar
- Department of Eletrical and Computer Engineering, The University of Texas at Austin, 2501 Speedway, Austin, TX 78712, USA
| | - Shaun A. Engelmann
- Department of Biomedical Engineering, The University of Texas at Austin, 107 W. Dean Keeton, Austin, TX 78712, USA
| | - Aaron L. Woods
- Department of Biomedical Engineering, The University of Texas at Austin, 107 W. Dean Keeton, Austin, TX 78712, USA
| | - Andrew K. Dunn
- Department of Eletrical and Computer Engineering, The University of Texas at Austin, 2501 Speedway, Austin, TX 78712, USA
- Department of Biomedical Engineering, The University of Texas at Austin, 107 W. Dean Keeton, Austin, TX 78712, USA
| |
Collapse
|
14
|
Qureshi MM, Allam N, Im J, Kwon HS, Chung E, Vitkin IA. Advances in laser speckle imaging: From qualitative to quantitative hemodynamic assessment. JOURNAL OF BIOPHOTONICS 2024; 17:e202300126. [PMID: 37545037 DOI: 10.1002/jbio.202300126] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 07/03/2023] [Accepted: 07/27/2023] [Indexed: 08/08/2023]
Abstract
Laser speckle imaging (LSI) techniques have emerged as a promising method for visualizing functional blood vessels and tissue perfusion by analyzing the speckle patterns generated by coherent light interacting with living biological tissue. These patterns carry important biophysical tissue information including blood flow dynamics. The noninvasive, label-free, and wide-field attributes along with relatively simple instrumental schematics make it an appealing imaging modality in preclinical and clinical applications. The review outlines the fundamentals of speckle physics and the three categories of LSI techniques based on their degree of quantification: qualitative, semi-quantitative and quantitative. Qualitative LSI produces microvascular maps by capturing speckle contrast variations between blood vessels containing moving red blood cells and the surrounding static tissue. Semi-quantitative techniques provide a more accurate analysis of blood flow dynamics by accounting for the effect of static scattering on spatiotemporal parameters. Quantitative LSI such as optical speckle image velocimetry provides quantitative flow velocity measurements, which is inspired by the particle image velocimetry in fluid mechanics. Additionally, discussions regarding the prospects of future innovations in LSI techniques for optimizing the vascular flow quantification with associated clinical outlook are presented.
Collapse
Affiliation(s)
- Muhammad Mohsin Qureshi
- Division of Biophysics and Bioimaging, Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
| | - Nader Allam
- Division of Biophysics and Bioimaging, Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Canada
| | - Jeongmyo Im
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, Republic of Korea
| | - Hyuk-Sang Kwon
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, Republic of Korea
| | - Euiheon Chung
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, Republic of Korea
- AI Graduate School, Gwangju Institute of Science and Technology, Gwangju, Republic of Korea
| | - I Alex Vitkin
- Division of Biophysics and Bioimaging, Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Canada
- Department of Radiation Oncology, University of Toronto, Toronto, Canada
| |
Collapse
|
15
|
Amra LN, Mächler P, Fomin-Thunemann N, Kılıç K, Saisan P, Devor A, Thunemann M. Tissue Oxygen Depth Explorer: an interactive database for microscopic oxygen imaging data. Front Neuroinform 2023; 17:1278787. [PMID: 38088985 PMCID: PMC10711099 DOI: 10.3389/fninf.2023.1278787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Accepted: 11/07/2023] [Indexed: 02/01/2024] Open
Affiliation(s)
- Layth N. Amra
- Department of Biomedical Engineering, Boston University, Boston, MA, United States
| | - Philipp Mächler
- Department of Physics, University of California, San Diego, La Jolla, CA, United States
| | | | - Kıvılcım Kılıç
- Department of Biomedical Engineering, Boston University, Boston, MA, United States
| | - Payam Saisan
- Department of Radiology, University of California, San Diego, La Jolla, CA, United States
| | - Anna Devor
- Department of Biomedical Engineering, Boston University, Boston, MA, United States
- Harvard Medical School, Martinos Center for Biomedical Imaging, MGH, Charlestown, MA, United States
| | - Martin Thunemann
- Department of Biomedical Engineering, Boston University, Boston, MA, United States
| |
Collapse
|
16
|
Banerji R, Grifno GN, Shi L, Smolen D, LeBourdais R, Muhvich J, Eberman C, Hiller BE, Lee J, Regan K, Zheng S, Zhang S, Jiang J, Raslan AA, Breda JC, Pihl R, Traber K, Mazzilli S, Ligresti G, Mizgerd JP, Suki B, Nia HT. Crystal ribcage: a platform for probing real-time lung function at cellular resolution. Nat Methods 2023; 20:1790-1801. [PMID: 37710017 PMCID: PMC10860663 DOI: 10.1038/s41592-023-02004-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Accepted: 08/10/2023] [Indexed: 09/16/2023]
Abstract
Understanding the dynamic pathogenesis and treatment response in pulmonary diseases requires probing the lung at cellular resolution in real time. Despite advances in intravital imaging, optical imaging of the lung during active respiration and circulation has remained challenging. Here, we introduce the crystal ribcage: a transparent ribcage that allows multiscale optical imaging of the functioning lung from whole-organ to single-cell level. It enables the modulation of lung biophysics and immunity through intravascular, intrapulmonary, intraparenchymal and optogenetic interventions, and it preserves the three-dimensional architecture, air-liquid interface, cellular diversity and respiratory-circulatory functions of the lung. Utilizing these capabilities on murine models of pulmonary pathologies we probed remodeling of respiratory-circulatory functions at the single-alveolus and capillary levels during disease progression. The crystal ribcage and its broad applications presented here will facilitate further studies of nearly any pulmonary disease as well as lead to the identification of new targets for treatment strategies.
Collapse
Affiliation(s)
- Rohin Banerji
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Gabrielle N Grifno
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Linzheng Shi
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Dylan Smolen
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Rob LeBourdais
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Johnathan Muhvich
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Cate Eberman
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Bradley E Hiller
- Pulmonary Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Jisu Lee
- Pulmonary Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Kathryn Regan
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Siyi Zheng
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Sue Zhang
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - John Jiang
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Ahmed A Raslan
- Pulmonary Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Zoology, Faculty of Science, Assiut University, Assiut, Egypt
| | - Julia C Breda
- Section of Computational Biomedicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Riley Pihl
- Pulmonary Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Katrina Traber
- Pulmonary Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Sarah Mazzilli
- Section of Computational Biomedicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Giovanni Ligresti
- Pulmonary Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Joseph P Mizgerd
- Pulmonary Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Béla Suki
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Hadi T Nia
- Department of Biomedical Engineering, Boston University, Boston, MA, USA.
- Pulmonary Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA.
| |
Collapse
|
17
|
Zhdanov AV, Sen R, Devoy C, Li L, Tangney M, Papkovsky DB. Analysis of tumour oxygenation in model animals on a phosphorescence lifetime based macro-imager. Sci Rep 2023; 13:18732. [PMID: 37907625 PMCID: PMC10618169 DOI: 10.1038/s41598-023-46224-7] [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: 11/30/2022] [Accepted: 10/30/2023] [Indexed: 11/02/2023] Open
Abstract
Monitoring of tissue O2 is essential for cancer development and treatment, as hypoxic tumour regions develop resistance to radio- and chemotherapy. We describe a minimally invasive technique for the monitoring of tissue oxygenation in developing grafted tumours, which uses the new phosphorescence lifetime based Tpx3Cam imager. CT26 cells stained with a near-infrared emitting nanoparticulate O2 probe NanO2-IR were injected into mice to produce grafted tumours with characteristic phosphorescence. The tumours were allowed to develop for 3, 7, 10 and 17 days, with O2 imaging experiments performed on live and euthanised animals at different time points. Despite a marked trend towards decreased O2 in dead animals, their tumour areas produced phosphorescence lifetime values between 44 and 47 µs, which corresponded to hypoxic tissue with 5-20 μM O2. After the O2 imaging in animals, confocal Phosphorescence Lifetime Imaging Microscopy was conducted to examine the distribution of NanO2-IR probe in the tumours, which were excised, fixed and sliced for the purpose. The probe remained visible as bright and discrete 'islands' embedded in the tumour tissue until day 17 of tumour growth. Overall, this O2 macro-imaging method using NanO2-IR holds promise for long-term studies with grafted tumours in live animal models, providing quantitative 2D mapping of tissue O2.
Collapse
Affiliation(s)
- Alexander V Zhdanov
- School of Biochemistry and Cell Biology, University College Cork, Pharmacy Building, College Road, Cork, Ireland
| | - Rajannya Sen
- School of Biochemistry and Cell Biology, University College Cork, Pharmacy Building, College Road, Cork, Ireland
| | - Ciaran Devoy
- Cancer Research @UCC, University College Cork, Cork, Ireland
| | - Liang Li
- School of Biochemistry and Cell Biology, University College Cork, Pharmacy Building, College Road, Cork, Ireland
| | - Mark Tangney
- Cancer Research @UCC, University College Cork, Cork, Ireland
| | - Dmitri B Papkovsky
- School of Biochemistry and Cell Biology, University College Cork, Pharmacy Building, College Road, Cork, Ireland.
| |
Collapse
|
18
|
Liu C, Cardenas-Rivera A, Teitelbaum S, Birmingham A, Alfadhel M, Yaseen MA. Neuroinflammation increases oxygen extraction in a mouse model of Alzheimer's disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.16.562353. [PMID: 37905082 PMCID: PMC10614808 DOI: 10.1101/2023.10.16.562353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/02/2023]
Abstract
Neuroinflammation, impaired metabolism, and hypoperfusion are fundamental pathological hallmarks of early Alzheimer's disease (AD). Numerous studies have asserted a close association between neuroinflammation and disrupted cerebral energetics. During AD progression and other neurodegenerative disorders, a persistent state of chronic neuroinflammation reportedly exacerbates cytotoxicity and potentiates neuronal death. Here, we assessed the impact of a neuroinflammatory challenge on metabolic demand and microvascular hemodynamics in the somatosensory cortex of an AD mouse model. We utilized in vivo 2-photon microscopy and the phosphorescent oxygen sensor Oxyphor 2P to measure partial pressure of oxygen (pO2) and capillary red blood cell flux in cortical microvessels of awake mice. Intravascular pO2 and capillary RBC flux measurements were performed in 8-month-old APPswe/PS1dE9 mice and wildtype littermates on days 0, 7, and 14 of a 14-day period of lipopolysaccaride-induced neuroinflammation. Before the induced inflammatory challenge, AD mice demonstrated reduced metabolic demand but similar capillary red blood cell flux as their wild type counterparts. Neuroinflammation provoked significant reductions in cerebral intravascular oxygen levels and elevated oxygen extraction in both animal groups, without significantly altering red blood cell flux in capillaries. This study provides evidence that neuroinflammation alters cerebral oxygen demand at the early stages of AD without substantially altering vascular oxygen supply. The results will guide our understanding of neuroinflammation's influence on neuroimaging biomarkers for early AD diagnosis.
Collapse
|
19
|
Delafontaine-Martel P, Zhang C, Lu X, Damseh R, Lesage F, Marchand PJ. Targeted capillary photothrombosis via multiphoton excitation of Rose Bengal. J Cereb Blood Flow Metab 2023; 43:1713-1725. [PMID: 36647768 PMCID: PMC10581236 DOI: 10.1177/0271678x231151560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 12/12/2022] [Accepted: 12/19/2022] [Indexed: 01/18/2023]
Abstract
Microvascular stalling, the process occurring when a capillary temporarily loses perfusion, has gained increasing interest in recent years through its demonstrated presence in various neuropathologies. Studying the impact of such stalls on the surrounding brain tissue is of paramount importance to understand their role in such diseases. Despite efforts trying to study the stalling events, investigations are hampered by their elusiveness and scarcity. In an attempt to alleviate these hurdles, we present here a novel methodology enabling transient occlusions of targeted microvascular segments through multiphoton excitation of Rose Bengal, an established photothrombotic agent. With n = 7 mice C57BL/6 J (5 males and 2 females) and 95 photothrombosis trials, we demonstrate the ability of triggering reversible blockages by illuminating a capillary segment during ∼300 s at 1000 nm, using a standard Ti:Sapphire femtosecond laser. Furthermore, we performed concurrent Optical Coherence Microscopy (OCM) angiography imaging of the microvascular network to highlight the specificity of the targeted occlusion and its duration. Through comparison with a control group, we conclude that blood flow cessation is indeed created by the photothrombotic agent via multiphoton excitation and is temporary, followed by a flow recovery in less than 24 h. Moreover, Immunohistology points toward a stalling mechanism driven by adherence of the neutrophil in the vascular lumen. This observation seems to be promoted by the inflammation locally created via multiphoton activation of Rose Bengal.
Collapse
Affiliation(s)
- Patrick Delafontaine-Martel
- Department of Electrical Engineering, Polytechnique Montreal, Montreal, Canada
- Research Center, Montreal Heart Institute, Montreal, Canada
| | - Cong Zhang
- Department of Electrical Engineering, Polytechnique Montreal, Montreal, Canada
- Research Center, Montreal Heart Institute, Montreal, Canada
| | - Xuecong Lu
- Research Center, Montreal Heart Institute, Montreal, Canada
- DeGroote School of Business – McMaster University, Ontario, Canada
| | - Rafat Damseh
- Research Center, Montreal Heart Institute, Montreal, Canada
- College of Information Technology, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Frédéric Lesage
- Department of Electrical Engineering, Polytechnique Montreal, Montreal, Canada
- Research Center, Montreal Heart Institute, Montreal, Canada
| | - Paul J Marchand
- Department of Electrical Engineering, Polytechnique Montreal, Montreal, Canada
- Research Center, Montreal Heart Institute, Montreal, Canada
- École polytechnique fédérale de Lausanne- EPFL, Lausanne, Switzerland
| |
Collapse
|
20
|
Shin P, Pian Q, Ishikawa H, Hamanaka G, Mandeville ET, Guo S, Fu B, Alfadhel M, Allu SR, Şencan-Eğilmez I, Li B, Ran C, Vinogradov SA, Ayata C, Lo E, Arai K, Devor A, Sakadžić S. Aerobic exercise reverses aging-induced depth-dependent decline in cerebral microcirculation. eLife 2023; 12:e86329. [PMID: 37402178 PMCID: PMC10319437 DOI: 10.7554/elife.86329] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 06/16/2023] [Indexed: 07/06/2023] Open
Abstract
Aging is a major risk factor for cognitive impairment. Aerobic exercise benefits brain function and may promote cognitive health in older adults. However, underlying biological mechanisms across cerebral gray and white matter are poorly understood. Selective vulnerability of the white matter to small vessel disease and a link between white matter health and cognitive function suggests a potential role for responses in deep cerebral microcirculation. Here, we tested whether aerobic exercise modulates cerebral microcirculatory changes induced by aging. To this end, we carried out a comprehensive quantitative examination of changes in cerebral microvascular physiology in cortical gray and subcortical white matter in mice (3-6 vs. 19-21 months old), and asked whether and how exercise may rescue age-induced deficits. In the sedentary group, aging caused a more severe decline in cerebral microvascular perfusion and oxygenation in deep (infragranular) cortical layers and subcortical white matter compared with superficial (supragranular) cortical layers. Five months of voluntary aerobic exercise partly renormalized microvascular perfusion and oxygenation in aged mice in a depth-dependent manner, and brought these spatial distributions closer to those of young adult sedentary mice. These microcirculatory effects were accompanied by an improvement in cognitive function. Our work demonstrates the selective vulnerability of the deep cortex and subcortical white matter to aging-induced decline in microcirculation, as well as the responsiveness of these regions to aerobic exercise.
Collapse
Affiliation(s)
- Paul Shin
- Athinoula A Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical SchoolCharlestownUnited States
| | - Qi Pian
- Athinoula A Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical SchoolCharlestownUnited States
| | - Hidehiro Ishikawa
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital, Harvard Medical SchoolCharlestownUnited States
| | - Gen Hamanaka
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital, Harvard Medical SchoolCharlestownUnited States
| | - Emiri T Mandeville
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital, Harvard Medical SchoolCharlestownUnited States
| | - Shuzhen Guo
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital, Harvard Medical SchoolCharlestownUnited States
| | - Buyin Fu
- Athinoula A Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical SchoolCharlestownUnited States
| | - Mohammed Alfadhel
- Athinoula A Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical SchoolCharlestownUnited States
- Department of Bioengineering, Northeastern UniversityBostonUnited States
| | - Srinivasa Rao Allu
- Department of Biochemistry and Biophysics, University of PennsylvaniaPhiladelphiaUnited States
- Department of Chemistry, University of PennsylvaniaPhiladelphiaUnited States
| | - Ikbal Şencan-Eğilmez
- Athinoula A Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical SchoolCharlestownUnited States
- Biophotonics Research Center, Mallinckrodt Institute of Radiology, Department of Radiology, Washington University School of MedicineSt. LouisUnited States
| | - Baoqiang Li
- Athinoula A Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical SchoolCharlestownUnited States
- Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of SciencesShenzhenChina
| | - Chongzhao Ran
- Athinoula A Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical SchoolCharlestownUnited States
| | - Sergei A Vinogradov
- Department of Biochemistry and Biophysics, University of PennsylvaniaPhiladelphiaUnited States
- Department of Chemistry, University of PennsylvaniaPhiladelphiaUnited States
| | - Cenk Ayata
- Neurovascular Research Laboratory, Department of Radiology, Massachusetts General Hospital, Harvard Medical SchoolCharlestownUnited States
- Stroke Service, Department of Neurology, Massachusetts General Hospital, Harvard Medical SchoolCharlestownUnited States
| | - Eng Lo
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital, Harvard Medical SchoolCharlestownUnited States
| | - Ken Arai
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital, Harvard Medical SchoolCharlestownUnited States
| | - Anna Devor
- Athinoula A Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical SchoolCharlestownUnited States
- Department of Biomedical Engineering, Boston UniversityBostonUnited States
| | - Sava Sakadžić
- Athinoula A Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical SchoolCharlestownUnited States
| |
Collapse
|
21
|
Gareau DS, RochaKim N, Choudhury A, Bamkole M, Snuderl M, Zou J, Yaroslavsky A, Jacques SL, Strickland S, Krueger JG, Ahn HJ. Fiberoptic hemodynamic spectroscopy reveals abnormal cerebrovascular reactivity in a freely moving mouse model of Alzheimer's disease. Front Mol Neurosci 2023; 16:1163447. [PMID: 37465366 PMCID: PMC10350529 DOI: 10.3389/fnmol.2023.1163447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 05/22/2023] [Indexed: 07/20/2023] Open
Abstract
Many Alzheimer's disease (AD) patients suffer from altered cerebral blood flow and damaged cerebral vasculature. Cerebrovascular dysfunction could play an important role in this disease. However, the mechanism underlying a vascular contribution in AD is still unclear. Cerebrovascular reactivity (CVR) is a critical mechanism that maintains cerebral blood flow and brain homeostasis. Most current methods to analyze CVR require anesthesia which is known to hamper the investigation of molecular mechanisms underlying CVR. We therefore combined spectroscopy, spectral analysis software, and an implantable device to measure cerebral blood volume fraction (CBVF) and oxygen saturation (SO2) in unanesthetized, freely-moving mice. Then, we analyzed basal CBVF and SO2, and CVR of 5-month-old C57BL/6 mice during hypercapnia as well as during basic behavior such as grooming, walking and running. Moreover, we analyzed the CVR of freely-moving AD mice and their wildtype (WT) littermates during hypercapnia and could find impaired CVR in AD mice compared to WT littermates. Our results suggest that this optomechanical approach to reproducibly getting light into the brain enabled us to successfully measure CVR in unanesthetized freely-moving mice and to find impaired CVR in a mouse model of AD.
Collapse
Affiliation(s)
- Daniel S. Gareau
- Laboratory of Investigative Dermatology, The Rockefeller University, New York, NY, United States
| | - Nicholas RochaKim
- Department of Pharmacology, Physiology and Neuroscience, Rutgers-New Jersey Medical School, Newark, NJ, United States
| | - Arnab Choudhury
- Department of Pharmacology, Physiology and Neuroscience, Rutgers-New Jersey Medical School, Newark, NJ, United States
| | - Michael Bamkole
- Department of Pharmacology, Physiology and Neuroscience, Rutgers-New Jersey Medical School, Newark, NJ, United States
| | - Matija Snuderl
- Department of Pathology, NYU Langone Health and Grossman School of Medicine, New York, NY, United States
| | - Julia Zou
- Laboratory of Investigative Dermatology, The Rockefeller University, New York, NY, United States
| | - Anna Yaroslavsky
- Department of Physics and Applied Physics, University of Massachusetts, Lowell, MA, United States
| | - Steven L. Jacques
- Department of Bioengineering, University of Washington, Seattle, WA, United States
| | - Sidney Strickland
- Laboratory of Neurobiology and Genetics, The Rockefeller University, New York, NY, United States
| | - James G. Krueger
- Laboratory of Investigative Dermatology, The Rockefeller University, New York, NY, United States
| | - Hyung Jin Ahn
- Department of Pharmacology, Physiology and Neuroscience, Rutgers-New Jersey Medical School, Newark, NJ, United States
- Brain Health Institute, Rutgers University, Piscataway, NJ, United States
| |
Collapse
|
22
|
Pian Q, Alfadhel M, Tang J, Lee GV, Li B, Fu B, Ayata Y, Yaseen MA, Boas DA, Secomb TW, Sakadzic S. Cortical microvascular blood flow velocity mapping by combining dynamic light scattering optical coherence tomography and two-photon microscopy. JOURNAL OF BIOMEDICAL OPTICS 2023; 28:076003. [PMID: 37484973 PMCID: PMC10362155 DOI: 10.1117/1.jbo.28.7.076003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 05/30/2023] [Accepted: 06/05/2023] [Indexed: 07/25/2023]
Abstract
Significance The accurate large-scale mapping of cerebral microvascular blood flow velocity is crucial for a better understanding of cerebral blood flow (CBF) regulation. Although optical imaging techniques enable both high-resolution microvascular angiography and fast absolute CBF velocity measurements in the mouse cortex, they usually require different imaging techniques with independent system configurations to maximize their performances. Consequently, it is still a challenge to accurately combine functional and morphological measurements to co-register CBF speed distribution from hundreds of microvessels with high-resolution microvascular angiograms. Aim We propose a data acquisition and processing framework to co-register a large set of microvascular blood flow velocity measurements from dynamic light scattering optical coherence tomography (DLS-OCT) with the corresponding microvascular angiogram obtained using two-photon microscopy (2PM). Approach We used DLS-OCT to first rapidly acquire a large set of microvascular velocities through a sealed cranial window in mice and then to acquire high-resolution microvascular angiograms using 2PM. The acquired data were processed in three steps: (i) 2PM angiogram coregistration with the DLS-OCT angiogram, (ii) 2PM angiogram segmentation and graphing, and (iii) mapping of the CBF velocities to the graph representation of the 2PM angiogram. Results We implemented the developed framework on the three datasets acquired from the mice cortices to facilitate the coregistration of the large sets of DLS-OCT flow velocity measurements with 2PM angiograms. We retrieved the distributions of red blood cell velocities in arterioles, venules, and capillaries as a function of the branching order from precapillary arterioles and postcapillary venules from more than 1000 microvascular segments. Conclusions The proposed framework may serve as a useful tool for quantitative analysis of large microvascular datasets obtained by OCT and 2PM in studies involving normal brain functioning, progression of various diseases, and numerical modeling of the oxygen advection and diffusion in the realistic microvascular networks.
Collapse
Affiliation(s)
- Qi Pian
- Massachusetts General Hospital, Harvard Medical School, Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Charlestown, Massachusetts, United States
| | - Mohammed Alfadhel
- Massachusetts General Hospital, Harvard Medical School, Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Charlestown, Massachusetts, United States
- Northeastern University, Department of Bioengineering, Boston, Massachusetts, United States
| | - Jianbo Tang
- Southern University of Science and Technology, Department of Biomedical Engineering, Shenzhen, China
| | - Grace V. Lee
- University of Arizona, Program in Applied Mathematics, Tucson, Arizona, United States
| | - Baoqiang Li
- Massachusetts General Hospital, Harvard Medical School, Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Charlestown, Massachusetts, United States
- Chinese Academy of Sciences, Shenzhen Institute of Advanced Technology, Brain Cognition and Brain Disease Institute; Shenzhen Fundamental Research Institutions, Shenzhen–Hong Kong Institute of Brain Science, Shenzhen, Guangdong, China
| | - Buyin Fu
- Massachusetts General Hospital, Harvard Medical School, Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Charlestown, Massachusetts, United States
| | - Yagmur Ayata
- Massachusetts General Hospital, Harvard Medical School, Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Charlestown, Massachusetts, United States
| | - Mohammad Abbas Yaseen
- Massachusetts General Hospital, Harvard Medical School, Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Charlestown, Massachusetts, United States
- Northeastern University, Department of Bioengineering, Boston, Massachusetts, United States
| | - David A. Boas
- Boston University, Department of Biomedical Engineering, Boston, Massachusetts, United States
| | - Timothy W. Secomb
- University of Arizona, Program in Applied Mathematics, Tucson, Arizona, United States
- University of Arizona, Department of Mathematics, Tucson, Arizona, United States
- University of Arizona, Department of Physiology, Tucson, Arizona, United States
| | - Sava Sakadzic
- Massachusetts General Hospital, Harvard Medical School, Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Charlestown, Massachusetts, United States
| |
Collapse
|
23
|
Li B, Leng J, Şencan-Eğilmez I, Takase H, Alfadhel MAH, Fu B, Shahidi M, Lo EH, Arai K, Sakadžić S. Differential reductions in the capillary red-blood-cell flux between retina and brain under chronic global hypoperfusion. NEUROPHOTONICS 2023; 10:035001. [PMID: 37323511 PMCID: PMC10266089 DOI: 10.1117/1.nph.10.3.035001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 04/13/2023] [Accepted: 04/26/2023] [Indexed: 06/17/2023]
Abstract
Significance It has been hypothesized that abnormal microcirculation in the retina might predict the risk of ischemic damages in the brain. Direct comparison between the retinal and the cerebral microcirculation using similar animal preparation and under similar experimental conditions would help test this hypothesis. Aim We investigated capillary red-blood-cell (RBC) flux changes under controlled conditions and bilateral-carotid-artery-stenosis (BCAS)-induced hypoperfusion, and then compared them with our previous measurements performed in the brain. Approach We measured capillary RBC flux in mouse retina with two-photon microscopy using a fluorescence-labeled RBC-passage approach. Key physiological parameters were monitored during experiments to ensure stable physiology. Results We found that under the controlled conditions, capillary RBC flux in the retina was much higher than in the brain (i.e., cerebral cortical gray matter and subcortical white matter), and that BCAS induced a much larger decrease in capillary RBC flux in the retina than in the brain. Conclusions We demonstrated a two-photon microscopy-based technique to efficiently measure capillary RBC flux in the retina. Since cerebral subcortical white matter often exhibits early pathological developments due to global hypoperfusion, our results suggest that retinal microcirculation may be utilized as an early marker of brain diseases involving global hypoperfusion.
Collapse
Affiliation(s)
- Baoqiang Li
- Chinese Academy of Sciences, Shenzhen Institute of Advanced Technology, Brain Cognition and Brain Disease Institute; Shenzhen Fundamental Research Institutions, Shenzhen–Hong Kong Institute of Brain Science, Shenzhen, Guangdong, China
- Harvard Medical School, Massachusetts General Hospital, Athinoula A. Martinos Center for Biomedical Imaging, Charlestown, Massachusetts, United States
| | - Ji Leng
- Chinese Academy of Sciences, Shenzhen Institute of Advanced Technology, Brain Cognition and Brain Disease Institute; Shenzhen Fundamental Research Institutions, Shenzhen–Hong Kong Institute of Brain Science, Shenzhen, Guangdong, China
| | - Ikbal Şencan-Eğilmez
- Harvard Medical School, Massachusetts General Hospital, Athinoula A. Martinos Center for Biomedical Imaging, Charlestown, Massachusetts, United States
| | - Hajime Takase
- Harvard Medical School, Massachusetts General Hospital, Department of Radiology, Charlestown, Massachusetts, United States
- Harvard Medical School, Massachusetts General Hospital, Department of Neurology, Charlestown, Massachusetts, United States
| | - Mohammed Ali H. Alfadhel
- Harvard Medical School, Massachusetts General Hospital, Athinoula A. Martinos Center for Biomedical Imaging, Charlestown, Massachusetts, United States
| | - Buyin Fu
- Harvard Medical School, Massachusetts General Hospital, Athinoula A. Martinos Center for Biomedical Imaging, Charlestown, Massachusetts, United States
| | - Mahnaz Shahidi
- University of Southern California, Department of Ophthalmology, Los Angeles, California, United States
| | - Eng H. Lo
- Harvard Medical School, Massachusetts General Hospital, Department of Radiology, Charlestown, Massachusetts, United States
- Harvard Medical School, Massachusetts General Hospital, Department of Neurology, Charlestown, Massachusetts, United States
| | - Ken Arai
- Harvard Medical School, Massachusetts General Hospital, Department of Radiology, Charlestown, Massachusetts, United States
- Harvard Medical School, Massachusetts General Hospital, Department of Neurology, Charlestown, Massachusetts, United States
| | - Sava Sakadžić
- Harvard Medical School, Massachusetts General Hospital, Athinoula A. Martinos Center for Biomedical Imaging, Charlestown, Massachusetts, United States
| |
Collapse
|
24
|
Samandarsangari M, Kozina DO, Sokolov VV, Komarova AD, Shirmanova MV, Kritchenkov IS, Tunik SP. Biocompatible Phosphorescent O 2 Sensors Based on Ir(III) Complexes for In Vivo Hypoxia Imaging. BIOSENSORS 2023; 13:680. [PMID: 37504079 PMCID: PMC10377268 DOI: 10.3390/bios13070680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 06/12/2023] [Accepted: 06/22/2023] [Indexed: 07/29/2023]
Abstract
In this work, we obtained three new phosphorescent iridium complexes (Ir1-Ir3) of general stoichiometry [Ir(N^C)2(N^N)]Cl decorated with oligo(ethylene glycol) fragments to make them water-soluble and biocompatible, as well as to protect them from aggregation with biomolecules such as albumin. The major photophysical characteristics of these phosphorescent complexes are determined by the nature of two cyclometallating ligands (N^C) based on 2-pyridine-benzothiophene, since quantum chemical calculations revealed that the electronic transitions responsible for the excitation and emission are localized mainly at these fragments. However, the use of various diimine ligands (N^N) proved to affect the quantum yield of phosphorescence and allowed for changing the complexes' sensitivity to oxygen, due to the variations in the steric accessibility of the chromophore center for O2 molecules. It was also found that the N^N ligands made it possible to tune the biocompatibility of the resulting compounds. The wavelengths of the Ir1-Ir3 emission maxima fell in the range of 630-650 nm, the quantum yields reached 17% (Ir1) in a deaerated solution, and sensitivity to molecular oxygen, estimated as the ratio of emission lifetime in deaerated and aerated water solutions, displayed the highest value, 8.2, for Ir1. The obtained complexes featured low toxicity, good water solubility and the absence of a significant effect of biological environment components on the parameters of their emission. Of the studied compounds, Ir1 and Ir2 were chosen for in vitro and in vivo biological experiments to estimate oxygen concentration in cell lines and tumors. These sensors have demonstrated their effectiveness for mapping the distribution of oxygen and for monitoring hypoxia in the biological objects studied.
Collapse
Affiliation(s)
- Mozhgan Samandarsangari
- Institute of Chemistry, St. Petersburg State University, Universitetskaya Embankment 7-9, 199034 St. Petersburg, Russia
| | - Daria O Kozina
- Institute of Chemistry, St. Petersburg State University, Universitetskaya Embankment 7-9, 199034 St. Petersburg, Russia
| | - Victor V Sokolov
- Institute of Chemistry, St. Petersburg State University, Universitetskaya Embankment 7-9, 199034 St. Petersburg, Russia
| | - Anastasia D Komarova
- Institute of Experimental Oncology and Biomedical Technologies, Privolzhskiy Research Medical University, Minin and Pozharsky Sq. 10/1, 603005 Nizhny Novgorod, Russia
- Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, Gagarina Av., 23, 603950 Nizhny Novgorod, Russia
| | - Marina V Shirmanova
- Institute of Experimental Oncology and Biomedical Technologies, Privolzhskiy Research Medical University, Minin and Pozharsky Sq. 10/1, 603005 Nizhny Novgorod, Russia
| | - Ilya S Kritchenkov
- Institute of Chemistry, St. Petersburg State University, Universitetskaya Embankment 7-9, 199034 St. Petersburg, Russia
| | - Sergey P Tunik
- Institute of Chemistry, St. Petersburg State University, Universitetskaya Embankment 7-9, 199034 St. Petersburg, Russia
| |
Collapse
|
25
|
Li B, Yabluchanskiy A, Tarantini S, Allu SR, Şencan-Eğilmez I, Leng J, Alfadhel MAH, Porter JE, Fu B, Ran C, Erdener SE, Boas DA, Vinogradov SA, Sonntag WE, Csiszar A, Ungvari Z, Sakadžić S. Measurements of cerebral microvascular blood flow, oxygenation, and morphology in a mouse model of whole-brain irradiation-induced cognitive impairment by two-photon microscopy and optical coherence tomography: evidence for microvascular injury in the cerebral white matter. GeroScience 2023; 45:1491-1510. [PMID: 36792820 PMCID: PMC10400746 DOI: 10.1007/s11357-023-00735-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 01/17/2023] [Indexed: 02/17/2023] Open
Abstract
Whole-brain irradiation (WBI, also known as whole-brain radiation therapy) is a mainstay treatment modality for patients with multiple brain metastases. It is also used as a prophylactic treatment for microscopic tumors that cannot be detected by magnetic resonance imaging. WBI induces a progressive cognitive decline in ~ 50% of the patients surviving over 6 months, significantly compromising the quality of life. There is increasing preclinical evidence that radiation-induced injury to the cerebral microvasculature and accelerated neurovascular senescence plays a central role in this side effect of WBI. To better understand this side effect, male C57BL/6 mice were first subjected to a clinically relevant protocol of fractionated WBI (5 Gy, two doses per week, for 4 weeks). Nine months post the WBI treatment, we applied two-photon microscopy and Doppler optical coherence tomography to measure capillary red-blood-cell (RBC) flux, capillary morphology, and microvascular oxygen partial pressure (PO2) in the cerebral somatosensory cortex in the awake, head-restrained, WPI-treated mice and their age-matched controls, through a cover-glass-sealed chronic cranial window. Thanks to the extended penetration depth with the fluorophore - Alexa680, measurements of capillary blood flow properties (e.g., RBC flux, speed, and linear density) in the cerebral subcortical white matter were enabled. We found that the WBI-treated mice exhibited a significantly decreased capillary RBC flux in the white matter. WBI also caused a significant reduction in capillary diameter, as well as a large (although insignificant) reduction in segment density at the deeper cortical layers (e.g., 600-700 μm), while the other morphological properties (e.g., segment length and tortuosity) were not obviously affected. In addition, we found that PO2 measured in the arterioles and venules, as well as the calculated oxygen saturation and oxygen extraction fraction, were not obviously affected by WBI. Lastly, WBI was associated with a significant increase in the erythrocyte-associated transients of PO2, while the changes of other cerebral capillary PO2 properties (e.g., capillary mean-PO2, RBC-PO2, and InterRBC-PO2) were not significant. Collectively, our findings support the notion that WBI results in persistent cerebral white matter microvascular impairment, which likely contributes to the WBI-induced brain injury and cognitive decline. Further studies are warranted to assess the WBI-induced changes in brain tissue oxygenation and malfunction of the white matter microvasculature as well.
Collapse
Affiliation(s)
- Baoqiang Li
- Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, Guangdong, China
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, 02129, USA
| | - Andriy Yabluchanskiy
- Vascular Cognitive Impairment and Neurodegeneration Program, Oklahoma Center for Geroscience and Healthy Brain Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
- The Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Stefano Tarantini
- Vascular Cognitive Impairment and Neurodegeneration Program, Oklahoma Center for Geroscience and Healthy Brain Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
- The Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, Budapest, 1083, Hungary
| | - Srinivasa Rao Allu
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Department of Chemistry, School of Arts and Sciences, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Ikbal Şencan-Eğilmez
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, 02129, USA
- Biophotonics Research Center, Department of Radiology, Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Ji Leng
- Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, Guangdong, China
| | - Mohammed Ali H Alfadhel
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, 02129, USA
| | - Jason E Porter
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, 02129, USA
| | - Buyin Fu
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, 02129, USA
| | - Chongzhao Ran
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, 02129, USA
| | - Sefik Evren Erdener
- Department of Biomedical Engineering, Boston University, Boston, MA, 02215, USA
| | - David A Boas
- Department of Biomedical Engineering, Boston University, Boston, MA, 02215, USA
| | - Sergei A Vinogradov
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Department of Chemistry, School of Arts and Sciences, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - William E Sonntag
- Vascular Cognitive Impairment and Neurodegeneration Program, Oklahoma Center for Geroscience and Healthy Brain Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
- The Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Anna Csiszar
- Vascular Cognitive Impairment and Neurodegeneration Program, Oklahoma Center for Geroscience and Healthy Brain Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
- The Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Translational Medicine, Semmelweis University, Budapest, 1083, Hungary
| | - Zoltan Ungvari
- Vascular Cognitive Impairment and Neurodegeneration Program, Oklahoma Center for Geroscience and Healthy Brain Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA.
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA.
- The Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA.
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, Budapest, 1083, Hungary.
| | - Sava Sakadžić
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, 02129, USA.
| |
Collapse
|
26
|
Hosford PS, Jalan R, Hadjihambi A. Reply to: "Does ammonia really disrupt brain oxygen homeostasis?". JHEP Rep 2023; 5:100666. [PMID: 37096141 PMCID: PMC10121447 DOI: 10.1016/j.jhepr.2022.100666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 12/16/2022] [Indexed: 01/01/2023] Open
Affiliation(s)
| | - Rajiv Jalan
- Liver Failure Group, Institute for Liver and Digestive Health, University College London, Royal Free Campus, London, United Kingdom
- European Foundation for the Study of Chronic Liver Failure (EF Clif), Spain
| | - Anna Hadjihambi
- The Roger Williams Institute of Hepatology London, Foundation for Liver Research, London, United Kingdom
- Faculty of Life Sciences and Medicine, King’s College London, United Kingdom
| |
Collapse
|
27
|
Arora S, Nagpal R, Gusain M, Singh B, Pan Y, Yadav D, Ahmed I, Kumar V, Parshad B. Organic-Inorganic Porphyrinoid Frameworks for Biomolecule Sensing. ACS Sens 2023; 8:443-464. [PMID: 36683281 DOI: 10.1021/acssensors.2c02408] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Porphyrinoids and their analogous compounds play an important role in biosensing applications on account of their unique and versatile catalytic, coordination, photophysical, and electrochemical properties. Their remarkable arrays of properties can be finely tuned by synthetically modifying the porphyrinoid ring and varying the various structural parameters such as peripheral functionalization, metal coordination, and covalent or physical conjugation with other organic or inorganic scaffolds such as nanoparticles, metal-organic frameworks, and polymers. Porphyrinoids and their organic-inorganic conjugates are not only used as responsive materials but also utilized for the immobilization and embedding of biomolecules for applications in wearable devices, fast sensing devices, and other functional materials. The present review delineates the impact of different porphyrinoid conjugates on their physicochemical properties and their specificity as biosensors in a range of applications. The newest porphyrinoid types and their synthesis, modification, and functionalization are presented along with their advantages and performance improvements.
Collapse
Affiliation(s)
- Smriti Arora
- Institut für Chemie und Biochemie Organische Chemie, Freie Universität Berlin, Takustr. 3, 14195 Berlin, Germany
| | - Ritika Nagpal
- Department of Chemistry, SRM University, 39, Rajiv Gandhi Education City, Delhi-NCR, Sonipat, Haryana 131029, India
| | - Meenakshi Gusain
- Centre of Micro-Nano System, School of Information Science and Technology, Fudan University, 200433 Shanghai, China
| | | | - Yuanwei Pan
- Department of Diagnostic Radiology, Department of Chemical and Biomolecular Engineering, and Department of Biomedical Engineering, Yong Loo Lin School of Medicine and Faculty of Engineering, National University of Singapore, Singapore 119074, Singapore
| | - Deepak Yadav
- Department of Chemistry, Gurugram University, Gurugram, Haryana 122003, India
| | - Ishtiaq Ahmed
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge CB3 0AS, U.K
| | - Vinod Kumar
- Department of Chemistry, Central University of Haryana, Mahendergarh, Haryana 123031, India
| | - Badri Parshad
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge CB3 0AS, U.K
| |
Collapse
|
28
|
Shin P, Pian Q, Ishikawa H, Hamanaka G, Mandeville ET, Shuzhen G, Buyin F, Alfadhel M, Allu SR, Şencan-Eğilmez I, Li B, Ran C, Vinogradov SA, Ayata C, Lo EH, Arai K, Devor A, Sakadžić S. Aerobic exercise reverses aging-induced depth-dependent decline in cerebral microcirculation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.12.528244. [PMID: 36824939 PMCID: PMC9949059 DOI: 10.1101/2023.02.12.528244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/19/2023]
Abstract
Aging is a major risk factor for cognitive impairment. Aerobic exercise benefits brain function and may promote cognitive health in older adults. However, underlying biological mechanisms across cerebral gray and white matter are poorly understood. Selective vulnerability of the white matter to small vessel disease and a link between white matter health and cognitive function suggests a potential role for responses in deep cerebral microcirculation. Here, we tested whether aerobic exercise modulates cerebral microcirculatory changes induced by aging. To this end, we carried out a comprehensive quantitative examination of changes in cerebral microvascular physiology in cortical gray and subcortical white matter in mice (3-6 vs. 19-21 months old), and asked whether and how exercise may rescue age-induced deficits. In the sedentary group, aging caused a more severe decline in cerebral microvascular perfusion and oxygenation in deep (infragranular) cortical layers and subcortical white matter compared with superficial (supragranular) cortical layers. Five months of voluntary aerobic exercise partly renormalized microvascular perfusion and oxygenation in aged mice in a depth-dependent manner, and brought these spatial distributions closer to those of young adult sedentary mice. These microcirculatory effects were accompanied by an improvement in cognitive function. Our work demonstrates the selective vulnerability of the deep cortex and subcortical white matter to aging-induced decline in microcirculation, as well as the responsiveness of these regions to aerobic exercise.
Collapse
Affiliation(s)
- Paul Shin
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
- Corresponding author:
| | - Qi Pian
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Hidehiro Ishikawa
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Gen Hamanaka
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Emiri T Mandeville
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Guo Shuzhen
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Fu Buyin
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Mohammed Alfadhel
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
- Department of Bioengineering, Northeastern University, Boston, MA, USA
| | - Srinivasa Rao Allu
- Department of Biochemistry and Biophysics, University of Pennsylvania, Philadelphia, PA, USA
- Department of Chemistry, University of Pennsylvania, Philadelphia, PA, USA
| | - Ikbal Şencan-Eğilmez
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
- Biophotonics Research Center, Mallinckrodt Institute of Radiology, Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Baoqiang Li
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
- Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, China
| | - Chongzhao Ran
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Sergei A Vinogradov
- Department of Biochemistry and Biophysics, University of Pennsylvania, Philadelphia, PA, USA
- Department of Chemistry, University of Pennsylvania, Philadelphia, PA, USA
| | - Cenk Ayata
- Neurovascular Research Laboratory, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
- Stroke Service, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Eng H Lo
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Ken Arai
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Anna Devor
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Sava Sakadžić
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| |
Collapse
|
29
|
Ferrer Ortas J, Mahou P, Escot S, Stringari C, David NB, Bally-Cuif L, Dray N, Négrerie M, Supatto W, Beaurepaire E. Label-free imaging of red blood cells and oxygenation with color third-order sum-frequency generation microscopy. LIGHT, SCIENCE & APPLICATIONS 2023; 12:29. [PMID: 36702815 PMCID: PMC9879988 DOI: 10.1038/s41377-022-01064-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 12/09/2022] [Accepted: 12/22/2022] [Indexed: 06/18/2023]
Abstract
Mapping red blood cells (RBCs) flow and oxygenation is of key importance for analyzing brain and tissue physiology. Current microscopy methods are limited either in sensitivity or in spatio-temporal resolution. In this work, we introduce a novel approach based on label-free third-order sum-frequency generation (TSFG) and third-harmonic generation (THG) contrasts. First, we propose a novel experimental scheme for color TSFG microscopy, which provides simultaneous measurements at several wavelengths encompassing the Soret absorption band of hemoglobin. We show that there is a strong three-photon (3P) resonance related to the Soret band of hemoglobin in THG and TSFG signals from zebrafish and human RBCs, and that this resonance is sensitive to RBC oxygenation state. We demonstrate that our color TSFG implementation enables specific detection of flowing RBCs in zebrafish embryos and is sensitive to RBC oxygenation dynamics with single-cell resolution and microsecond pixel times. Moreover, it can be implemented on a 3P microscope and provides label-free RBC-specific contrast at depths exceeding 600 µm in live adult zebrafish brain. Our results establish a new multiphoton contrast extending the palette of deep-tissue microscopy.
Collapse
Affiliation(s)
- Júlia Ferrer Ortas
- Laboratory for Optics and Biosciences, CNRS, INSERM, École polytechnique, IP Paris, 91128, Palaiseau, France
| | - Pierre Mahou
- Laboratory for Optics and Biosciences, CNRS, INSERM, École polytechnique, IP Paris, 91128, Palaiseau, France
| | - Sophie Escot
- Laboratory for Optics and Biosciences, CNRS, INSERM, École polytechnique, IP Paris, 91128, Palaiseau, France
| | - Chiara Stringari
- Laboratory for Optics and Biosciences, CNRS, INSERM, École polytechnique, IP Paris, 91128, Palaiseau, France
| | - Nicolas B David
- Laboratory for Optics and Biosciences, CNRS, INSERM, École polytechnique, IP Paris, 91128, Palaiseau, France
| | - Laure Bally-Cuif
- Zebrafish Neurogenetics Unit, team supported by Ligue Nationale contre le Cancer, Institut Pasteur, CNRS, 75015, Paris, France
| | - Nicolas Dray
- Zebrafish Neurogenetics Unit, team supported by Ligue Nationale contre le Cancer, Institut Pasteur, CNRS, 75015, Paris, France
| | - Michel Négrerie
- Laboratory for Optics and Biosciences, CNRS, INSERM, École polytechnique, IP Paris, 91128, Palaiseau, France
| | - Willy Supatto
- Laboratory for Optics and Biosciences, CNRS, INSERM, École polytechnique, IP Paris, 91128, Palaiseau, France
| | - Emmanuel Beaurepaire
- Laboratory for Optics and Biosciences, CNRS, INSERM, École polytechnique, IP Paris, 91128, Palaiseau, France.
| |
Collapse
|
30
|
Lin J, Cheng Z, Yang G, Cui M. Optical gearbox enabled versatile multiscale high-throughput multiphoton functional imaging. Nat Commun 2022; 13:6564. [PMID: 36323707 PMCID: PMC9630539 DOI: 10.1038/s41467-022-34472-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Accepted: 10/19/2022] [Indexed: 11/05/2022] Open
Abstract
To understand the function and mechanism of biological systems, it is crucial to observe the cellular dynamics at high spatiotemporal resolutions within live animals. The recent advances in genetically encoded function indicators have significantly improved the response rate to a near millisecond time scale. However, the widely employed in vivo imaging systems often lack the temporal solution to capture the fast biological dynamics. To broadly enable the capability of high-speed in vivo deep-tissue imaging, we developed an optical gearbox. As an add-on module, the optical gearbox can convert the common multiphoton imaging systems for versatile multiscale high-throughput imaging applications. In this work, we demonstrate in vivo 2D and 3D function imaging in mammalian brains at frame rates ranging from 50 to 1000 Hz. The optical gearbox's versatility and compatibility with the widely employed imaging components will be highly valuable to a variety of deep tissue imaging applications.
Collapse
Affiliation(s)
- Jianian Lin
- School of Electrical and Computer Engineering, Purdue University, West Lafayette, IN, 47907, USA
- Bindley Bioscience Center, Purdue University, West Lafayette, IN, 47907, USA
| | - Zongyue Cheng
- School of Electrical and Computer Engineering, Purdue University, West Lafayette, IN, 47907, USA
- Bindley Bioscience Center, Purdue University, West Lafayette, IN, 47907, USA
| | - Guang Yang
- Department of Anesthesiology, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Meng Cui
- School of Electrical and Computer Engineering, Purdue University, West Lafayette, IN, 47907, USA.
- Bindley Bioscience Center, Purdue University, West Lafayette, IN, 47907, USA.
- Department of Biology, Purdue University, West Lafayette, IN, 47907, USA.
| |
Collapse
|
31
|
Mächler P, Fomin-Thunemann N, Thunemann M, Sætra MJ, Desjardins M, Kılıç K, Amra LN, Martin EA, Chen IA, Şencan-Eğilmez I, Li B, Saisan P, Jiang JX, Cheng Q, Weldy KL, Boas DA, Buxton RB, Einevoll GT, Dale AM, Sakadžić S, Devor A. Baseline oxygen consumption decreases with cortical depth. PLoS Biol 2022; 20:e3001440. [PMID: 36301995 PMCID: PMC9642908 DOI: 10.1371/journal.pbio.3001440] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Revised: 11/08/2022] [Accepted: 09/30/2022] [Indexed: 11/05/2022] Open
Abstract
The cerebral cortex is organized in cortical layers that differ in their cellular density, composition, and wiring. Cortical laminar architecture is also readily revealed by staining for cytochrome oxidase-the last enzyme in the respiratory electron transport chain located in the inner mitochondrial membrane. It has been hypothesized that a high-density band of cytochrome oxidase in cortical layer IV reflects higher oxygen consumption under baseline (unstimulated) conditions. Here, we tested the above hypothesis using direct measurements of the partial pressure of O2 (pO2) in cortical tissue by means of 2-photon phosphorescence lifetime microscopy (2PLM). We revisited our previously developed method for extraction of the cerebral metabolic rate of O2 (CMRO2) based on 2-photon pO2 measurements around diving arterioles and applied this method to estimate baseline CMRO2 in awake mice across cortical layers. To our surprise, our results revealed a decrease in baseline CMRO2 from layer I to layer IV. This decrease of CMRO2 with cortical depth was paralleled by an increase in tissue oxygenation. Higher baseline oxygenation and cytochrome density in layer IV may serve as an O2 reserve during surges of neuronal activity or certain metabolically active brain states rather than reflecting baseline energy needs. Our study provides to our knowledge the first quantification of microscopically resolved CMRO2 across cortical layers as a step towards better understanding of brain energy metabolism.
Collapse
Affiliation(s)
- Philipp Mächler
- Department of Biomedical Engineering, Boston University, Boston, Massachusetts, United States of America
| | - Natalie Fomin-Thunemann
- Department of Biomedical Engineering, Boston University, Boston, Massachusetts, United States of America
| | - Martin Thunemann
- Department of Biomedical Engineering, Boston University, Boston, Massachusetts, United States of America
| | - Marte Julie Sætra
- Department of Numerical Analysis and Scientific Computing, Simula Research Laboratory, Oslo, Norway
| | - Michèle Desjardins
- Département de Physique, de Génie Physique et d’Optique and Axe Oncologie, Centre de Recherche du CHU de Québec–Université Laval, Université Laval, Québec, Canada
| | - Kıvılcım Kılıç
- Department of Biomedical Engineering, Boston University, Boston, Massachusetts, United States of America
| | - Layth N. Amra
- Department of Biomedical Engineering, Boston University, Boston, Massachusetts, United States of America
| | - Emily A. Martin
- Department of Biomedical Engineering, Boston University, Boston, Massachusetts, United States of America
| | - Ichun Anderson Chen
- Department of Biomedical Engineering, Boston University, Boston, Massachusetts, United States of America
| | - Ikbal Şencan-Eğilmez
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Harvard Medical School, Massachusetts General Hospital, Charlestown, Massachusetts, United States of America
| | - Baoqiang Li
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Harvard Medical School, Massachusetts General Hospital, Charlestown, Massachusetts, United States of America
| | - Payam Saisan
- Department of Neurosciences, University of California San Diego, La Jolla, California, United States of America
| | - John X. Jiang
- Department of Biomedical Engineering, Boston University, Boston, Massachusetts, United States of America
| | - Qun Cheng
- Department of Neurosciences, University of California San Diego, La Jolla, California, United States of America
| | - Kimberly L. Weldy
- Department of Neurosciences, University of California San Diego, La Jolla, California, United States of America
| | - David A. Boas
- Department of Biomedical Engineering, Boston University, Boston, Massachusetts, United States of America
| | - Richard B. Buxton
- Department of Radiology, University of California San Diego, La Jolla, California, United States of America
| | - Gaute T. Einevoll
- Department of Physics, University of Oslo, Oslo, Norway
- Department of Physics, Norwegian University of Life Sciences, Ås, Norway
| | - Anders M. Dale
- Department of Neurosciences, University of California San Diego, La Jolla, California, United States of America
- Department of Radiology, University of California San Diego, La Jolla, California, United States of America
| | - Sava Sakadžić
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Harvard Medical School, Massachusetts General Hospital, Charlestown, Massachusetts, United States of America
- * E-mail: (SS); (AD)
| | - Anna Devor
- Department of Biomedical Engineering, Boston University, Boston, Massachusetts, United States of America
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Harvard Medical School, Massachusetts General Hospital, Charlestown, Massachusetts, United States of America
- * E-mail: (SS); (AD)
| |
Collapse
|
32
|
Guyon J, Fernandez‐Moncada I, Larrieu CM, Bouchez CL, Pagano Zottola AC, Galvis J, Chouleur T, Burban A, Joseph K, Ravi VM, Espedal H, Røsland GV, Daher B, Barre A, Dartigues B, Karkar S, Rudewicz J, Romero‐Garmendia I, Klink B, Grützmann K, Derieppe M, Molinié T, Obad N, Léon C, Seano G, Miletic H, Heiland DH, Marsicano G, Nikolski M, Bjerkvig R, Bikfalvi A, Daubon T. Lactate dehydrogenases promote glioblastoma growth and invasion via a metabolic symbiosis. EMBO Mol Med 2022; 14:e15343. [PMID: 36278433 PMCID: PMC9728051 DOI: 10.15252/emmm.202115343] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 09/26/2022] [Accepted: 09/27/2022] [Indexed: 12/14/2022] Open
Abstract
Lactate is a central metabolite in brain physiology but also contributes to tumor development. Glioblastoma (GB) is the most common and malignant primary brain tumor in adults, recognized by angiogenic and invasive growth, in addition to its altered metabolism. We show herein that lactate fuels GB anaplerosis by replenishing the tricarboxylic acid (TCA) cycle in absence of glucose. Lactate dehydrogenases (LDHA and LDHB), which we found spatially expressed in GB tissues, catalyze the interconversion of pyruvate and lactate. However, ablation of both LDH isoforms, but not only one, led to a reduction in tumor growth and an increase in mouse survival. Comparative transcriptomics and metabolomics revealed metabolic rewiring involving high oxidative phosphorylation (OXPHOS) in the LDHA/B KO group which sensitized tumors to cranial irradiation, thus improving mouse survival. When mice were treated with the antiepileptic drug stiripentol, which targets LDH activity, tumor growth decreased. Our findings unveil the complex metabolic network in which both LDHA and LDHB are integrated and show that the combined inhibition of LDHA and LDHB strongly sensitizes GB to therapy.
Collapse
Affiliation(s)
- Joris Guyon
- University Bordeaux, INSERM U1312, BRICPessacFrance
| | | | | | | | | | - Johanna Galvis
- University Bordeaux, CNRS, IBGC, UMR 5095BordeauxFrance,Bordeaux Bioinformatic Center CBiBUniversity of BordeauxBordeauxFrance
| | | | - Audrey Burban
- University Bordeaux, CNRS, IBGC, UMR 5095BordeauxFrance
| | - Kevin Joseph
- Microenvironment and Immunology Research Laboratory, Medical CenterUniversity of FreiburgFreiburgGermany,Department of Neurosurgery, Medical CenterUniversity of FreiburgFreiburgGermany,Faculty of Medicine, University of FreiburgFreiburgGermany,Translational NeuroOncology Research Group, Medical CenterUniversity of FreiburgFreiburgGermany,Center of Advanced Surgical Tissue Analysis (CAST)University of FreiburgFreiburgGermany
| | - Vidhya M Ravi
- Microenvironment and Immunology Research Laboratory, Medical CenterUniversity of FreiburgFreiburgGermany,Department of Neurosurgery, Medical CenterUniversity of FreiburgFreiburgGermany,Faculty of Medicine, University of FreiburgFreiburgGermany,Translational NeuroOncology Research Group, Medical CenterUniversity of FreiburgFreiburgGermany,Center of Advanced Surgical Tissue Analysis (CAST)University of FreiburgFreiburgGermany,Freiburg Institute for Advanced Studies (FRIAS)University of FreiburgFreiburgGermany
| | - Heidi Espedal
- NorLux Neuro‐Oncology, Department of BiomedicineUniversity of BergenBergenNorway
| | | | | | - Aurélien Barre
- Bordeaux Bioinformatic Center CBiBUniversity of BordeauxBordeauxFrance
| | | | - Slim Karkar
- Bordeaux Bioinformatic Center CBiBUniversity of BordeauxBordeauxFrance
| | - Justine Rudewicz
- Bordeaux Bioinformatic Center CBiBUniversity of BordeauxBordeauxFrance
| | | | - Barbara Klink
- Department of OncologyLuxembourg Institute of HealthLuxembourgLuxembourg,German Cancer Consortium (DKTK)DresdenGermany,Core Unit for Molecular Tumor Diagnostics (CMTD)National Center for Tumor Diseases (NCT)DresdenGermany
| | - Konrad Grützmann
- Core Unit for Molecular Tumor Diagnostics (CMTD)National Center for Tumor Diseases (NCT)DresdenGermany
| | | | | | - Nina Obad
- NorLux Neuro‐Oncology, Department of BiomedicineUniversity of BergenBergenNorway
| | - Céline Léon
- University Bordeaux, INSERM U1312, BRICPessacFrance
| | - Giorgio Seano
- Institut Curie, INSERM U1021, CNRS UMR3347, Tumor Microenvironment LabUniversity Paris‐SaclayOrsayFrance
| | - Hrvoje Miletic
- NorLux Neuro‐Oncology, Department of BiomedicineUniversity of BergenBergenNorway,Department of PathologyHaukeland University HospitalBergenNorway
| | - Dieter Henrik Heiland
- Microenvironment and Immunology Research Laboratory, Medical CenterUniversity of FreiburgFreiburgGermany,Department of Neurosurgery, Medical CenterUniversity of FreiburgFreiburgGermany,Faculty of Medicine, University of FreiburgFreiburgGermany,Translational NeuroOncology Research Group, Medical CenterUniversity of FreiburgFreiburgGermany,German Cancer Consortium (DKTK), partner site FreiburgFreiburgGermany
| | | | - Macha Nikolski
- University Bordeaux, CNRS, IBGC, UMR 5095BordeauxFrance,Bordeaux Bioinformatic Center CBiBUniversity of BordeauxBordeauxFrance
| | - Rolf Bjerkvig
- NorLux Neuro‐Oncology, Department of BiomedicineUniversity of BergenBergenNorway
| | | | - Thomas Daubon
- University Bordeaux, INSERM U1312, BRICPessacFrance,University Bordeaux, CNRS, IBGC, UMR 5095BordeauxFrance
| |
Collapse
|
33
|
Chong SH, Ong YH, El Khatib M, Allu SR, Parthasarathy AB, Greenberg JH, Yodh AG, Vinogradov SA. Real-time tracking of brain oxygen gradients and blood flow during functional activation. NEUROPHOTONICS 2022; 9:045006. [PMID: 36457848 PMCID: PMC9704417 DOI: 10.1117/1.nph.9.4.045006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Accepted: 11/01/2022] [Indexed: 06/11/2023]
Abstract
Significance Cerebral metabolic rate of oxygen ( CMRO 2 ) consumption is a key physiological variable that characterizes brain metabolism in a steady state and during functional activation. Aim We aim to develop a minimally invasive optical technique for real-time measurement of CMRO 2 concurrently with cerebral blood flow (CBF). Approach We used a pair of macromolecular phosphorescent probes with nonoverlapping optical spectra, which were localized in the intra- and extravascular compartments of the brain tissue, thus providing a readout of oxygen gradients between these two compartments. In parallel, we measured CBF using laser speckle contrast imaging. Results The method enables computation and tracking of CMRO 2 during functional activation with high temporal resolution ( ∼ 7 Hz ). In contrast to other approaches, our assessment of CMRO 2 does not require measurements of CBF or hemoglobin oxygen saturation. Conclusions The independent records of intravascular and extravascular partial pressures of oxygen, CBF, and CMRO 2 provide information about the physiological events that accompany neuronal activation, creating opportunities for dynamic quantification of brain metabolism.
Collapse
Affiliation(s)
- Sang Hoon Chong
- University of Pennsylvania, Department of Physics and Astronomy, Philadelphia, Pennsylvania, United States
| | - Yi Hong Ong
- University of Pennsylvania, Department of Physics and Astronomy, Philadelphia, Pennsylvania, United States
- University of Pennsylvania, Department of Radiation Oncology, Philadelphia, Pennsylvania, United States
| | - Mirna El Khatib
- University of Pennsylvania, Perelman School of Medicine, Department of Biochemistry and Biophysics, Philadelphia, Pennsylvania, United States
- University of Pennsylvania, School of Arts and Sciences, Department of Chemistry, Philadelphia, Pennsylvania, United States
| | - Srinivasa Rao Allu
- University of Pennsylvania, Perelman School of Medicine, Department of Biochemistry and Biophysics, Philadelphia, Pennsylvania, United States
- University of Pennsylvania, School of Arts and Sciences, Department of Chemistry, Philadelphia, Pennsylvania, United States
| | - Ashwin B. Parthasarathy
- University of South Florida, Department of Electrical Engineering, Tampa, Florida, United States
| | - Joel H. Greenberg
- University of Pennsylvania, Department of Neurology, Philadelphia, Pennsylvania, United States
| | - Arjun G. Yodh
- University of Pennsylvania, Department of Physics and Astronomy, Philadelphia, Pennsylvania, United States
| | - Sergei A. Vinogradov
- University of Pennsylvania, Perelman School of Medicine, Department of Biochemistry and Biophysics, Philadelphia, Pennsylvania, United States
- University of Pennsylvania, School of Arts and Sciences, Department of Chemistry, Philadelphia, Pennsylvania, United States
| |
Collapse
|
34
|
Qin Q, Liu Y, Yang Z, Aimaijiang M, Ma R, Yang Y, Zhang Y, Zhou Y. Hypoxia-Inducible Factors Signaling in Osteogenesis and Skeletal Repair. Int J Mol Sci 2022; 23:ijms231911201. [PMID: 36232501 PMCID: PMC9569554 DOI: 10.3390/ijms231911201] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 09/10/2022] [Accepted: 09/14/2022] [Indexed: 11/23/2022] Open
Abstract
Sufficient oxygen is required to maintain normal cellular and physiological function, such as a creature’s development, breeding, and homeostasis. Lately, some researchers have reported that both pathological hypoxia and environmental hypoxia might affect bone health. Adaptation to hypoxia is a pivotal cellular event in normal cell development and differentiation and in pathological settings such as ischemia. As central mediators of homeostasis, hypoxia-inducible transcription factors (HIFs) can allow cells to survive in a low-oxygen environment and are essential for the regulation of osteogenesis and skeletal repair. From this perspective, we summarized the role of HIF-1 and HIF-2 in signaling pathways implicated in bone development and skeletal repair and outlined the molecular mechanism of regulation of downstream growth factors and protein molecules such as VEGF, EPO, and so on. All of these present an opportunity for developing therapies for bone regeneration.
Collapse
|
35
|
Kajtez J, Wesseler MF, Birtele M, Khorasgani FR, Rylander Ottosson D, Heiskanen A, Kamperman T, Leijten J, Martínez‐Serrano A, Larsen NB, Angelini TE, Parmar M, Lind JU, Emnéus J. Embedded 3D Printing in Self-Healing Annealable Composites for Precise Patterning of Functionally Mature Human Neural Constructs. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2201392. [PMID: 35712780 PMCID: PMC9443452 DOI: 10.1002/advs.202201392] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 05/14/2022] [Indexed: 05/02/2023]
Abstract
Human in vitro models of neural tissue with tunable microenvironment and defined spatial arrangement are needed to facilitate studies of brain development and disease. Towards this end, embedded printing inside granular gels holds great promise as it allows precise patterning of extremely soft tissue constructs. However, granular printing support formulations are restricted to only a handful of materials. Therefore, there has been a need for novel materials that take advantage of versatile biomimicry of bulk hydrogels while providing high-fidelity support for embedded printing akin to granular gels. To address this need, Authors present a modular platform for bioengineering of neuronal networks via direct embedded 3D printing of human stem cells inside Self-Healing Annealable Particle-Extracellular matrix (SHAPE) composites. SHAPE composites consist of soft microgels immersed in viscous extracellular-matrix solution to enable precise and programmable patterning of human stem cells and consequent generation mature subtype-specific neurons that extend projections into the volume of the annealed support. The developed approach further allows multi-ink deposition, live spatial and temporal monitoring of oxygen levels, as well as creation of vascular-like channels. Due to its modularity and versatility, SHAPE biomanufacturing toolbox has potential to be used in applications beyond functional modeling of mechanically sensitive neural constructs.
Collapse
Affiliation(s)
- Janko Kajtez
- Department of Experimental Medical SciencesWallenberg Neuroscience CenterDivision of Neurobiology and Lund Stem Cell CenterLund UniversityLundS‐221 84Sweden
- Department of Biotechnology and Biomedicine (DTU Bioengineering)Technical University of DenmarkKongens Lyngby2800Denmark
| | - Milan Finn Wesseler
- Department of Health Technology (DTU Health Tech)Technical University of DenmarkKongens Lyngby2800Denmark
| | - Marcella Birtele
- Department of Experimental Medical SciencesWallenberg Neuroscience CenterDivision of Neurobiology and Lund Stem Cell CenterLund UniversityLundS‐221 84Sweden
| | - Farinaz Riyahi Khorasgani
- Department of Biotechnology and Biomedicine (DTU Bioengineering)Technical University of DenmarkKongens Lyngby2800Denmark
| | - Daniella Rylander Ottosson
- Department of Experimental Medical SciencesWallenberg Neuroscience CenterDivision of Neurobiology and Lund Stem Cell CenterLund UniversityLundS‐221 84Sweden
| | - Arto Heiskanen
- Department of Biotechnology and Biomedicine (DTU Bioengineering)Technical University of DenmarkKongens Lyngby2800Denmark
| | - Tom Kamperman
- Department of Developmental BioEngineeringFaculty of Science and TechnologyTechnical Medical CentreUniversity of TwenteEnschede7522The Netherlands
| | - Jeroen Leijten
- Department of Developmental BioEngineeringFaculty of Science and TechnologyTechnical Medical CentreUniversity of TwenteEnschede7522The Netherlands
| | - Alberto Martínez‐Serrano
- Department of Molecular BiologyUniversidad Autónoma de Madridand Division of HomeostasisCenter of Molecular Biology Severo Ochoa (UAM‐CSIC)Madrid28049Spain
| | - Niels B. Larsen
- Department of Health Technology (DTU Health Tech)Technical University of DenmarkKongens Lyngby2800Denmark
| | - Thomas E. Angelini
- Department of Mechanical and Aerospace EngineeringUniversity of FloridaGainsvilleFL32611USA
| | - Malin Parmar
- Department of Experimental Medical SciencesWallenberg Neuroscience CenterDivision of Neurobiology and Lund Stem Cell CenterLund UniversityLundS‐221 84Sweden
| | - Johan U. Lind
- Department of Health Technology (DTU Health Tech)Technical University of DenmarkKongens Lyngby2800Denmark
| | - Jenny Emnéus
- Department of Biotechnology and Biomedicine (DTU Bioengineering)Technical University of DenmarkKongens Lyngby2800Denmark
| |
Collapse
|
36
|
Mikkelsen SH, Wied B, Dashkovskyi V, Lindhardt TB, Hirschler L, Warnking JM, Barbier EL, Postnov D, Hansen B, Gutiérrez-Jiménez E. Head holder and cranial window design for sequential magnetic resonance imaging and optical imaging in awake mice. Front Neurosci 2022; 16:926828. [PMID: 36051645 PMCID: PMC9425635 DOI: 10.3389/fnins.2022.926828] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Accepted: 07/14/2022] [Indexed: 11/28/2022] Open
Abstract
Medical imaging techniques are widely used in preclinical research as diagnostic tools to detect physiological abnormalities and assess the progression of neurovascular disease in animal models. Despite the wealth of imaging options in magnetic resonance imaging (MRI), interpretation of imaging-derived parameters regarding underlying tissue properties is difficult due to technical limitations or lack of parameter specificity. To address the challenge of interpretation, we present an animal preparation protocol to achieve quantitative measures from both MRI and advanced optical techniques, including laser speckle contrast imaging and two-photon microscopy, in murine models. In this manner, non-translatable methods support and improve interpretation of less specific, translatable methods, i.e., MRI. Combining modalities for improved clinical interpretation involves satisfying the requirements of various methods. Furthermore, physiology unperturbed by anesthetics is a prerequisite for the strategy to succeed. Awake animal imaging with restraint provides an alternative to anesthesia and facilitates translatability of cerebral measurements. The method outlines design requirements for the setup and a corresponding reproducible surgical procedure for implanting a 3D printed head holder and cranial window to enable repeated multimodal imaging. We document the development, application, and validation of the method and provide examples confirming the usefulness of the design in acquiring high quality data from multiple modalities for quantification of a wide range of metrics of cerebral physiology in the same animal. The method contributes to preclinical small animal imaging, enabling sequential imaging of previously mutually exclusive techniques.
Collapse
Affiliation(s)
- Signe H. Mikkelsen
- Center of Functionally Integrative Neuroscience, Aarhus University, Aarhus, Denmark
| | - Boris Wied
- Center of Functionally Integrative Neuroscience, Aarhus University, Aarhus, Denmark
| | - Vitalii Dashkovskyi
- Center of Functionally Integrative Neuroscience, Aarhus University, Aarhus, Denmark
| | | | | | - Jan M. Warnking
- Univ. Grenoble Alpes, Inserm, U1216, GIN, Grenoble Institut des Neurosciences, La Tronche, France
| | - Emmanuel L. Barbier
- Univ. Grenoble Alpes, Inserm, U1216, GIN, Grenoble Institut des Neurosciences, La Tronche, France
| | - Dmitry Postnov
- Center of Functionally Integrative Neuroscience, Aarhus University, Aarhus, Denmark
| | - Brian Hansen
- Center of Functionally Integrative Neuroscience, Aarhus University, Aarhus, Denmark
- *Correspondence: Brian Hansen,
| | - Eugenio Gutiérrez-Jiménez
- Center of Functionally Integrative Neuroscience, Aarhus University, Aarhus, Denmark
- Eugenio Gutiérrez-Jiménez,
| |
Collapse
|
37
|
Martin JD, Lanning RM, Chauhan VP, Martin MR, Mousa AS, Kamoun WS, Han HS, Lee H, Stylianopoulos T, Bawendi MG, Duda DG, Brown EB, Padera TP, Fukumura D, Jain RK. Multiphoton Phosphorescence Quenching Microscopy Reveals Kinetics of Tumor Oxygenation during Antiangiogenesis and Angiotensin Signaling Inhibition. Clin Cancer Res 2022; 28:3076-3090. [PMID: 35584239 PMCID: PMC9355624 DOI: 10.1158/1078-0432.ccr-22-0486] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Revised: 04/14/2022] [Accepted: 05/11/2022] [Indexed: 01/07/2023]
Abstract
PURPOSE The abnormal function of tumor blood vessels causes tissue hypoxia, promoting disease progression and treatment resistance. Although tumor microenvironment normalization strategies can alleviate hypoxia globally, how local oxygen levels change is not known because of the inability to longitudinally assess vascular and interstitial oxygen in tumors with sufficient resolution. Understanding the spatial and temporal heterogeneity should help improve the outcome of various normalization strategies. EXPERIMENTAL DESIGN We developed a multiphoton phosphorescence quenching microscopy system using a low-molecular-weight palladium porphyrin probe to measure perfused vessels, oxygen tension, and their spatial correlations in vivo in mouse skin, bone marrow, and four different tumor models. Further, we measured the temporal and spatial changes in oxygen and vessel perfusion in tumors in response to an anti-VEGFR2 antibody (DC101) and an angiotensin-receptor blocker (losartan). RESULTS We found that vessel function was highly dependent on tumor type. Although some tumors had vessels with greater oxygen-carrying ability than those of normal skin, most tumors had inefficient vessels. Further, intervessel heterogeneity in tumors is associated with heterogeneous response to DC101 and losartan. Using both vascular and stromal normalizing agents, we show that spatial heterogeneity in oxygen levels persists, even with reductions in mean extravascular hypoxia. CONCLUSIONS High-resolution spatial and temporal responses of tumor vessels to two agents known to improve vascular perfusion globally reveal spatially heterogeneous changes in vessel structure and function. These dynamic vascular changes should be considered in optimizing the dose and schedule of vascular and stromal normalizing strategies to improve the therapeutic outcome.
Collapse
Affiliation(s)
- John D. Martin
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts.,Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Ryan M. Lanning
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts.,Harvard-Massachusetts Institute of Technology Division of Health Sciences and Technology, Cambridge, Massachusetts
| | - Vikash P. Chauhan
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts.,School of Engineering and Applied Sciences, Harvard University, Cambridge, Massachusetts
| | - Margaret R. Martin
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Ahmed S. Mousa
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Walid S. Kamoun
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Hee-Sun Han
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Hang Lee
- Biostatistics Center, Harvard Medical School, Massachusetts General Hospital, Boston, Massachusetts
| | - Triantafyllos Stylianopoulos
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Moungi G. Bawendi
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Dan G. Duda
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Edward B. Brown
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Timothy P. Padera
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Dai Fukumura
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Rakesh K. Jain
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts.,Corresponding Author: Rakesh K. Jain, Department of Radiation Oncology, 100 Blossom Street, Cox 7, Boston, MA 02114. E-mail:
| |
Collapse
|
38
|
Erlebach E, Ravotto L, Wyss MT, Condrau J, Troxler T, Vinogradov SA, Weber B. Measurement of cerebral oxygen pressure in living mice by two-photon phosphorescence lifetime microscopy. STAR Protoc 2022; 3:101370. [PMID: 35573482 PMCID: PMC9092998 DOI: 10.1016/j.xpro.2022.101370] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
The ability to quantify partial pressure of oxygen (pO2) is of primary importance for studies of metabolic processes in health and disease. Here, we present a protocol for imaging of oxygen distributions in tissue and vasculature of the cerebral cortex of anesthetized and awake mice. We describe in vivo two-photon phosphorescence lifetime microscopy (2PLM) of oxygen using the probe Oxyphor 2P. This minimally invasive protocol outperforms existing approaches in terms of accuracy, resolution, and imaging depth. For complete details on the use and execution of this protocol, please refer to Esipova et al. (2019). Two-photon phosphorescence imaging of Oxyphor 2P allows for oxygen measurement in vivo Oxygen imaging can be performed in anesthetized or awake, behaving mice Intravenous injection enables oxygen imaging in the vasculature Cisterna magna injection enables extra- and intravascular oxygen imaging in the brain
Collapse
Affiliation(s)
- Eva Erlebach
- Institute of Pharmacology and Toxicology, University of Zurich, 8057 Zürich, Switzerland
| | - Luca Ravotto
- Institute of Pharmacology and Toxicology, University of Zurich, 8057 Zürich, Switzerland
| | - Matthias T. Wyss
- Institute of Pharmacology and Toxicology, University of Zurich, 8057 Zürich, Switzerland
| | - Jacqueline Condrau
- Institute of Pharmacology and Toxicology, University of Zurich, 8057 Zürich, Switzerland
| | - Thomas Troxler
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Chemistry, School of Arts and Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Sergei A. Vinogradov
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Chemistry, School of Arts and Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA
- Corresponding author
| | - Bruno Weber
- Institute of Pharmacology and Toxicology, University of Zurich, 8057 Zürich, Switzerland
- Corresponding author
| |
Collapse
|
39
|
Zhu X, Huang Q, DiSpirito A, Vu T, Rong Q, Peng X, Sheng H, Shen X, Zhou Q, Jiang L, Hoffmann U, Yao J. Real-time whole-brain imaging of hemodynamics and oxygenation at micro-vessel resolution with ultrafast wide-field photoacoustic microscopy. LIGHT, SCIENCE & APPLICATIONS 2022; 11:138. [PMID: 35577780 PMCID: PMC9110749 DOI: 10.1038/s41377-022-00836-2] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 04/27/2022] [Accepted: 05/04/2022] [Indexed: 05/10/2023]
Abstract
High-speed high-resolution imaging of the whole-brain hemodynamics is critically important to facilitating neurovascular research. High imaging speed and image quality are crucial to visualizing real-time hemodynamics in complex brain vascular networks, and tracking fast pathophysiological activities at the microvessel level, which will enable advances in current queries in neurovascular and brain metabolism research, including stroke, dementia, and acute brain injury. Further, real-time imaging of oxygen saturation of hemoglobin (sO2) can capture fast-paced oxygen delivery dynamics, which is needed to solve pertinent questions in these fields and beyond. Here, we present a novel ultrafast functional photoacoustic microscopy (UFF-PAM) to image the whole-brain hemodynamics and oxygenation. UFF-PAM takes advantage of several key engineering innovations, including stimulated Raman scattering (SRS) based dual-wavelength laser excitation, water-immersible 12-facet-polygon scanner, high-sensitivity ultrasound transducer, and deep-learning-based image upsampling. A volumetric imaging rate of 2 Hz has been achieved over a field of view (FOV) of 11 × 7.5 × 1.5 mm3 with a high spatial resolution of ~10 μm. Using the UFF-PAM system, we have demonstrated proof-of-concept studies on the mouse brains in response to systemic hypoxia, sodium nitroprusside, and stroke. We observed the mouse brain's fast morphological and functional changes over the entire cortex, including vasoconstriction, vasodilation, and deoxygenation. More interestingly, for the first time, with the whole-brain FOV and micro-vessel resolution, we captured the vasoconstriction and hypoxia simultaneously in the spreading depolarization (SD) wave. We expect the new imaging technology will provide a great potential for fundamental brain research under various pathological and physiological conditions.
Collapse
Affiliation(s)
- Xiaoyi Zhu
- Department of Biomedical Engineering, Duke University, Durham, NC, 27708, USA
| | - Qiang Huang
- Department of Biomedical Engineering, Duke University, Durham, NC, 27708, USA
- Department of Pediatric Surgery, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Anthony DiSpirito
- Department of Biomedical Engineering, Duke University, Durham, NC, 27708, USA
| | - Tri Vu
- Department of Biomedical Engineering, Duke University, Durham, NC, 27708, USA
| | - Qiangzhou Rong
- Department of Biomedical Engineering, Duke University, Durham, NC, 27708, USA
| | - Xiaorui Peng
- Department of Biomedical Engineering, Duke University, Durham, NC, 27708, USA
| | - Huaxin Sheng
- Roski Eye Institute, Department of Ophthalmology, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
| | - Xiling Shen
- Department of Biomedical Engineering, Duke University, Durham, NC, 27708, USA
| | - Qifa Zhou
- Roski Eye Institute, Department of Ophthalmology, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, 90089, USA
| | - Laiming Jiang
- Roski Eye Institute, Department of Ophthalmology, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA.
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, 90089, USA.
| | - Ulrike Hoffmann
- Department of Anesthesiology, Duke University, Durham, NC, 27708, USA.
| | - Junjie Yao
- Department of Biomedical Engineering, Duke University, Durham, NC, 27708, USA.
| |
Collapse
|
40
|
Cui S, Pratx G. 3D computational model of oxygen depletion kinetics in brain vasculature during FLASH RT, and its implications for in vivo oximetry experiments. Med Phys 2022; 49:3914-3925. [PMID: 35393643 DOI: 10.1002/mp.15642] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 02/19/2022] [Accepted: 03/23/2022] [Indexed: 11/07/2022] Open
Abstract
PURPOSE Ultra-high dose rate irradiation, also known as FLASH, has been shown to improve the therapeutic ratio of radiation therapy (RT). The mechanism behind this effect has been partially explained by the radiochemical oxygen depletion (ROD) hypothesis, which attributes the protection of the normal tissue to the induction of transient hypoxia by ROD. To better understand the contribution of oxygen to the FLASH effect, it is necessary to measure oxygen (O2 ) in vivo during FLASH irradiation. This study's goal is to determine the temporal resolution required to accurately measure the rapidly changing oxygen concentration immediately after FLASH irradiation. METHODS We conducted a computational simulation of oxygen dynamics using a real vascular model that was constructed from a public fluorescence microscopy dataset. The dynamic distribution of oxygen tension (po2 ) during and after FLASH RT was modeled by a partial differential equation (PDE) considering oxygen diffusion, metabolism, and ROD. The underestimation of ROD due to oxygen recovery was evaluated assuming either complete or partial depletion, and a range of possible values for parameters such as oxygen diffusion, consumption, vascular po2 and vessel density. RESULT The O2 concentration recovers rapidly after FLASH RT. Assuming a temporal resolution of 0.5 s, the estimated ROD is only 50.7% and 36.7% of its actual value in cases of partial and complete depletion, respectively. Additionally, the underestimation of ROD is highly dependent on the vascular density. To estimate ROD rate with 90% accuracy, temporal resolution on the order of milliseconds is required considering the uncertainty in parameters involved, especially, the diverse vascular density of the tissue. CONCLUSION The rapid recovery of O2 poses a great challenge for in vivo ROD measurements during FLASH RT. Temporal resolution on the order of milliseconds is recommended for ROD measurements in the normal tissue. Further work is warranted to investigate whether the same requirements apply to tumors, given their irregular vasculature. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Sunan Cui
- Department of Radiation Oncology, Stanford University, Palo Alto, California, USA
| | - Guillem Pratx
- Department of Radiation Oncology, Stanford University, Palo Alto, California, USA
| |
Collapse
|
41
|
Rane V. Harnessing Electron Spin Hyperpolarization in Chromophore-Radical Spin Probes for Subcellular Resolution in Electron Paramagnetic Resonance Imaging: Concept and Feasibility. J Phys Chem B 2022; 126:2715-2728. [PMID: 35353514 DOI: 10.1021/acs.jpcb.1c10920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Obtaining a subcellular resolution for biological samples doped with stable radicals at room temperature (RT) is a long-sought goal in electron paramagnetic resonance imaging (EPRI). The spatial resolution in current EPRI methods is constrained either because of low electron spin polarization at RT or the experimental limitations associated with the field gradients and the radical linewidth. Inspired by the recent demonstration of a large electron spin hyperpolarization in chromophore-nitroxyl spin probe molecules, the present work proposes a novel optically hyperpolarized EPR imaging (OH-EPRI) method, which combines the optical method of two-photon confocal microscopy for hyperpolarization generation and the rapid scan (RS) EPR method for signal detection. An important aspect of OH-EPRI is that it is not limited by the abovementioned restrictions of conventional EPRI since the large hyperpolarization in the spin probes overcomes the poor thermal spin polarization at RT, and the use of two-photon optical excitation of the chromophore naturally generates the required spatial resolution, without the need for any magnetic field gradient. Simulations based on time-dependent Bloch equations, which took into account both the RS field modulation and the hyperpolarization generation by optical means, were performed to examine the feasibility of OH-EPRI. The simulation results revealed that a spatial resolution of up to 2 fL can be achieved in OH-EPRI at RT under in vitro conditions. Notably, the majority of the requirements for an OH-EPRI experiment can be fulfilled by the currently available technologies, thereby paving the way for its easy implementation. Thus, the proposed method could potentially bridge the sensitivity gap between the optical and magnetic imaging techniques.
Collapse
Affiliation(s)
- Vinayak Rane
- Radiochemistry Division, Bhabha Atomic Research Centre, Trombay, Mumbai 400085, India
| |
Collapse
|
42
|
Şencan İ, Esipova T, Kılıç K, Li B, Desjardins M, Yaseen MA, Wang H, Porter JE, Kura S, Fu B, Secomb TW, Boas DA, Vinogradov SA, Devor A, Sakadžić S. Optical measurement of microvascular oxygenation and blood flow responses in awake mouse cortex during functional activation. J Cereb Blood Flow Metab 2022; 42:510-525. [PMID: 32515672 PMCID: PMC8985437 DOI: 10.1177/0271678x20928011] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The cerebral cortex has a number of conserved morphological and functional characteristics across brain regions and species. Among them, the laminar differences in microvascular density and mitochondrial cytochrome c oxidase staining suggest potential laminar variability in the baseline O2 metabolism and/or laminar variability in both O2 demand and hemodynamic response. Here, we investigate the laminar profile of stimulus-induced intravascular partial pressure of O2 (pO2) transients to stimulus-induced neuronal activation in fully awake mice using two-photon phosphorescence lifetime microscopy. Our results demonstrate that stimulus-induced changes in intravascular pO2 are conserved across cortical layers I-IV, suggesting a tightly controlled neurovascular response to provide adequate O2 supply across cortical depth. In addition, we observed a larger change in venular O2 saturation (ΔsO2) compared to arterioles, a gradual increase in venular ΔsO2 response towards the cortical surface, and absence of the intravascular "initial dip" previously reported under anesthesia. This study paves the way for quantification of layer-specific cerebral O2 metabolic responses, facilitating investigation of brain energetics in health and disease and informed interpretation of laminar blood oxygen level dependent functional magnetic resonance imaging signals.
Collapse
Affiliation(s)
- İkbal Şencan
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Tatiana Esipova
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.,Department of Chemistry, School of Arts and Sciences, University of Pennsylvania, Philadelphia, PA, USA
| | - Kıvılcım Kılıç
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Baoqiang Li
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Michèle Desjardins
- Department of Physics, Engineering Physics and Optics, Université Laval, QC, Canada
| | - Mohammad A Yaseen
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Hui Wang
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Jason E Porter
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Sreekanth Kura
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Buyin Fu
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Timothy W Secomb
- Department of Physiology, University of Arizona, Tucson, AZ, USA
| | - David A Boas
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Sergei A Vinogradov
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.,Department of Chemistry, School of Arts and Sciences, University of Pennsylvania, Philadelphia, PA, USA
| | - Anna Devor
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA.,Department of Biomedical Engineering, Boston University, Boston, MA, USA.,Department of Neurosciences, University of California San Diego, La Jolla, CA, USA.,Department of Radiology, University of California San Diego, La Jolla, CA, USA
| | - Sava Sakadžić
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| |
Collapse
|
43
|
Khatib ME, Van Slyke AL, Velalopoulou A, Kim MM, Shoniyozov K, Allu SR, Diffenderfer EE, Busch TM, Wiersma RD, Koch CJ, Vinogradov SA. Ultrafast Tracking of Oxygen Dynamics during Proton FLASH. Int J Radiat Oncol Biol Phys 2022; 113:624-634. [DOI: 10.1016/j.ijrobp.2022.03.016] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 02/01/2022] [Accepted: 03/12/2022] [Indexed: 10/18/2022]
|
44
|
Abstract
Metabolism has been studied mainly in cultured cells or at the level of whole tissues or whole organisms in vivo. Consequently, our understanding of metabolic heterogeneity among cells within tissues is limited, particularly when it comes to rare cells with biologically distinct properties, such as stem cells. Stem cell function, tissue regeneration and cancer suppression are all metabolically regulated, although it is not yet clear whether there are metabolic mechanisms unique to stem cells that regulate their activity and function. Recent work has, however, provided evidence that stem cells do have a metabolic signature that is distinct from that of restricted progenitors and that metabolic changes influence tissue homeostasis and regeneration. Stem cell maintenance throughout life in many tissues depends upon minimizing anabolic pathway activation and cell division. Consequently, stem cell activation by tissue injury is associated with changes in mitochondrial function, lysosome activity and lipid metabolism, potentially at the cost of eroding self-renewal potential. Stem cell metabolism is also regulated by the environment: stem cells metabolically interact with other cells in their niches and are able to sense and adapt to dietary changes. The accelerating understanding of stem cell metabolism is revealing new aspects of tissue homeostasis with the potential to promote tissue regeneration and cancer suppression.
Collapse
|
45
|
Conjugated polymer nanoparticles and their nanohybrids as smart photoluminescent and photoresponsive material for biosensing, imaging, and theranostics. Mikrochim Acta 2022; 189:83. [PMID: 35118576 DOI: 10.1007/s00604-021-05153-w] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 12/13/2021] [Indexed: 02/06/2023]
Abstract
The emergence of conjugated polymers (CPs) has provided a pathway to attain smart multifunctional conjugated polymer nanoparticles (CPNs) with enhanced properties and diverse applications. CPNs based on π-extended CPs exhibit high fluorescence brightness, low cytotoxicity, excellent photostability, reactive oxygen species (ROS) generation ability, high photothermal conversion efficiency (PCE), etc. which endorse them as an excellent theranostic tool. Furthermore, the unique light-harvesting and energy transfer properties of CPNs enables their transformation into smart functional nanohybrids with augmented performance. Owing to such numerous features, simple preparation method and an easy separation process, the CPNs and their hybrids have been constantly rising as a frontrunner in the domain of medicine and much work has been done in the respective research area. This review summarizes the recent progress that has been made in the field of CPNs for biological and biomedical applications with special emphasis on biosensing, imaging, and theranostics. Following an introduction into the field, a first large section provides overview of the conventional as well as recently established synthetic methods for various types of CPNs. Then, the CPNs-based fluorometric assays for biomolecules based on different detection strategies have been described. Later on, examples of CPNs-based probes for imaging, both in vitro and in vivo using cancer cells and animal models have been explored. The next section highlighted the vital theranostic applications of CPNs and corresponding nanohybrids, mainly via imaging-guided photodynamic therapy (PDT), photothermal therapy (PTT) and drug delivery. The last section summarizes the current challenges and gives an outlook on the potential future trends on CPNs as advanced healthcare material.
Collapse
|
46
|
Brender JR, Saida Y, Devasahayam N, Krishna MC, Kishimoto S. Hypoxia Imaging As a Guide for Hypoxia-Modulated and Hypoxia-Activated Therapy. Antioxid Redox Signal 2022; 36:144-159. [PMID: 34428981 PMCID: PMC8856011 DOI: 10.1089/ars.2021.0176] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 08/16/2021] [Indexed: 01/03/2023]
Abstract
Significance: Oxygen imaging techniques, which can probe the spatiotemporal heterogeneity of tumor oxygenation, could be of significant clinical utility in radiation treatment planning and in evaluating the effectiveness of hypoxia-activated prodrugs. To fulfill these goals, oxygen imaging techniques should be noninvasive, quantitative, and capable of serial imaging, as well as having sufficient temporal resolution to detect the dynamics of tumor oxygenation to distinguish regions of chronic and acute hypoxia. Recent Advances: No current technique meets all these requirements, although all have strengths in certain areas. The current status of positron emission tomography (PET)-based hypoxia imaging, oxygen-enhanced magnetic resonance imaging (MRI), 19F MRI, and electron paramagnetic resonance (EPR) oximetry are reviewed along with their strengths and weaknesses for planning hypoxia-guided, intensity-modulated radiation therapy and detecting treatment response for hypoxia-targeted prodrugs. Critical Issues: Spatial and temporal resolution emerges as a major concern for these areas along with specificity and quantitative response. Although multiple oxygen imaging techniques have reached the investigative stage, clinical trials to test the therapeutic effectiveness of hypoxia imaging have been limited. Future Directions: Imaging elements of the redox environment besides oxygen by EPR and hyperpolarized MRI may have a significant impact on our understanding of the basic biology of the reactive oxygen species response and may extend treatment possibilities.
Collapse
Affiliation(s)
- Jeffrey R. Brender
- Radiation Biology Branch, Center for Cancer Research, National
Institutes of Health, Bethesda, Maryland, USA
| | - Yu Saida
- Radiation Biology Branch, Center for Cancer Research, National
Institutes of Health, Bethesda, Maryland, USA
| | - Nallathamby Devasahayam
- Radiation Biology Branch, Center for Cancer Research, National
Institutes of Health, Bethesda, Maryland, USA
| | - Murali C. Krishna
- Radiation Biology Branch, Center for Cancer Research, National
Institutes of Health, Bethesda, Maryland, USA
| | - Shun Kishimoto
- Radiation Biology Branch, Center for Cancer Research, National
Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
47
|
Abdelfattah AS, Ahuja S, Akkin T, Allu SR, Brake J, Boas DA, Buckley EM, Campbell RE, Chen AI, Cheng X, Čižmár T, Costantini I, De Vittorio M, Devor A, Doran PR, El Khatib M, Emiliani V, Fomin-Thunemann N, Fainman Y, Fernandez-Alfonso T, Ferri CGL, Gilad A, Han X, Harris A, Hillman EMC, Hochgeschwender U, Holt MG, Ji N, Kılıç K, Lake EMR, Li L, Li T, Mächler P, Miller EW, Mesquita RC, Nadella KMNS, Nägerl UV, Nasu Y, Nimmerjahn A, Ondráčková P, Pavone FS, Perez Campos C, Peterka DS, Pisano F, Pisanello F, Puppo F, Sabatini BL, Sadegh S, Sakadzic S, Shoham S, Shroff SN, Silver RA, Sims RR, Smith SL, Srinivasan VJ, Thunemann M, Tian L, Tian L, Troxler T, Valera A, Vaziri A, Vinogradov SA, Vitale F, Wang LV, Uhlířová H, Xu C, Yang C, Yang MH, Yellen G, Yizhar O, Zhao Y. Neurophotonic tools for microscopic measurements and manipulation: status report. NEUROPHOTONICS 2022; 9:013001. [PMID: 35493335 PMCID: PMC9047450 DOI: 10.1117/1.nph.9.s1.013001] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/30/2023]
Abstract
Neurophotonics was launched in 2014 coinciding with the launch of the BRAIN Initiative focused on development of technologies for advancement of neuroscience. For the last seven years, Neurophotonics' agenda has been well aligned with this focus on neurotechnologies featuring new optical methods and tools applicable to brain studies. While the BRAIN Initiative 2.0 is pivoting towards applications of these novel tools in the quest to understand the brain, this status report reviews an extensive and diverse toolkit of novel methods to explore brain function that have emerged from the BRAIN Initiative and related large-scale efforts for measurement and manipulation of brain structure and function. Here, we focus on neurophotonic tools mostly applicable to animal studies. A companion report, scheduled to appear later this year, will cover diffuse optical imaging methods applicable to noninvasive human studies. For each domain, we outline the current state-of-the-art of the respective technologies, identify the areas where innovation is needed, and provide an outlook for the future directions.
Collapse
Affiliation(s)
- Ahmed S. Abdelfattah
- Brown University, Department of Neuroscience, Providence, Rhode Island, United States
| | - Sapna Ahuja
- University of Pennsylvania, Perelman School of Medicine, Department of Biochemistry and Biophysics, Philadelphia, Pennsylvania, United States
- University of Pennsylvania, School of Arts and Sciences, Department of Chemistry, Philadelphia, Pennsylvania, United States
| | - Taner Akkin
- University of Minnesota, Department of Biomedical Engineering, Minneapolis, Minnesota, United States
| | - Srinivasa Rao Allu
- University of Pennsylvania, Perelman School of Medicine, Department of Biochemistry and Biophysics, Philadelphia, Pennsylvania, United States
- University of Pennsylvania, School of Arts and Sciences, Department of Chemistry, Philadelphia, Pennsylvania, United States
| | - Joshua Brake
- Harvey Mudd College, Department of Engineering, Claremont, California, United States
| | - David A. Boas
- Boston University, Department of Biomedical Engineering, Boston, Massachusetts, United States
| | - Erin M. Buckley
- Georgia Institute of Technology and Emory University, Wallace H. Coulter Department of Biomedical Engineering, Atlanta, Georgia, United States
- Emory University, Department of Pediatrics, Atlanta, Georgia, United States
| | - Robert E. Campbell
- University of Tokyo, Department of Chemistry, Tokyo, Japan
- University of Alberta, Department of Chemistry, Edmonton, Alberta, Canada
| | - Anderson I. Chen
- Boston University, Department of Biomedical Engineering, Boston, Massachusetts, United States
| | - Xiaojun Cheng
- Boston University, Department of Biomedical Engineering, Boston, Massachusetts, United States
| | - Tomáš Čižmár
- Institute of Scientific Instruments of the Czech Academy of Sciences, Brno, Czech Republic
| | - Irene Costantini
- University of Florence, European Laboratory for Non-Linear Spectroscopy, Department of Biology, Florence, Italy
- National Institute of Optics, National Research Council, Rome, Italy
| | - Massimo De Vittorio
- Istituto Italiano di Tecnologia, Center for Biomolecular Nanotechnologies, Arnesano, Italy
| | - Anna Devor
- Boston University, Department of Biomedical Engineering, Boston, Massachusetts, United States
- Massachusetts General Hospital, Harvard Medical School, Athinoula A. Martinos Center for Biomedical Imaging, Charlestown, Massachusetts, United States
| | - Patrick R. Doran
- Boston University, Department of Biomedical Engineering, Boston, Massachusetts, United States
| | - Mirna El Khatib
- University of Pennsylvania, Perelman School of Medicine, Department of Biochemistry and Biophysics, Philadelphia, Pennsylvania, United States
- University of Pennsylvania, School of Arts and Sciences, Department of Chemistry, Philadelphia, Pennsylvania, United States
| | | | - Natalie Fomin-Thunemann
- Boston University, Department of Biomedical Engineering, Boston, Massachusetts, United States
| | - Yeshaiahu Fainman
- University of California San Diego, Department of Electrical and Computer Engineering, La Jolla, California, United States
| | - Tomas Fernandez-Alfonso
- University College London, Department of Neuroscience, Physiology and Pharmacology, London, United Kingdom
| | - Christopher G. L. Ferri
- University of California San Diego, Departments of Neurosciences, La Jolla, California, United States
| | - Ariel Gilad
- The Hebrew University of Jerusalem, Institute for Medical Research Israel–Canada, Department of Medical Neurobiology, Faculty of Medicine, Jerusalem, Israel
| | - Xue Han
- Boston University, Department of Biomedical Engineering, Boston, Massachusetts, United States
| | - Andrew Harris
- Weizmann Institute of Science, Department of Brain Sciences, Rehovot, Israel
| | | | - Ute Hochgeschwender
- Central Michigan University, Department of Neuroscience, Mount Pleasant, Michigan, United States
| | - Matthew G. Holt
- University of Porto, Instituto de Investigação e Inovação em Saúde (i3S), Porto, Portugal
| | - Na Ji
- University of California Berkeley, Department of Physics, Berkeley, California, United States
| | - Kıvılcım Kılıç
- Boston University, Department of Biomedical Engineering, Boston, Massachusetts, United States
| | - Evelyn M. R. Lake
- Yale School of Medicine, Department of Radiology and Biomedical Imaging, New Haven, Connecticut, United States
| | - Lei Li
- California Institute of Technology, Andrew and Peggy Cherng Department of Medical Engineering, Department of Electrical Engineering, Pasadena, California, United States
| | - Tianqi Li
- University of Minnesota, Department of Biomedical Engineering, Minneapolis, Minnesota, United States
| | - Philipp Mächler
- Boston University, Department of Biomedical Engineering, Boston, Massachusetts, United States
| | - Evan W. Miller
- University of California Berkeley, Departments of Chemistry and Molecular & Cell Biology and Helen Wills Neuroscience Institute, Berkeley, California, United States
| | | | | | - U. Valentin Nägerl
- Interdisciplinary Institute for Neuroscience University of Bordeaux & CNRS, Bordeaux, France
| | - Yusuke Nasu
- University of Tokyo, Department of Chemistry, Tokyo, Japan
| | - Axel Nimmerjahn
- Salk Institute for Biological Studies, Waitt Advanced Biophotonics Center, La Jolla, California, United States
| | - Petra Ondráčková
- Institute of Scientific Instruments of the Czech Academy of Sciences, Brno, Czech Republic
| | - Francesco S. Pavone
- National Institute of Optics, National Research Council, Rome, Italy
- University of Florence, European Laboratory for Non-Linear Spectroscopy, Department of Physics, Florence, Italy
| | - Citlali Perez Campos
- Columbia University, Zuckerman Mind Brain Behavior Institute, New York, United States
| | - Darcy S. Peterka
- Columbia University, Zuckerman Mind Brain Behavior Institute, New York, United States
| | - Filippo Pisano
- Istituto Italiano di Tecnologia, Center for Biomolecular Nanotechnologies, Arnesano, Italy
| | - Ferruccio Pisanello
- Istituto Italiano di Tecnologia, Center for Biomolecular Nanotechnologies, Arnesano, Italy
| | - Francesca Puppo
- University of California San Diego, Departments of Neurosciences, La Jolla, California, United States
| | - Bernardo L. Sabatini
- Harvard Medical School, Howard Hughes Medical Institute, Department of Neurobiology, Boston, Massachusetts, United States
| | - Sanaz Sadegh
- University of California San Diego, Departments of Neurosciences, La Jolla, California, United States
| | - Sava Sakadzic
- Massachusetts General Hospital, Harvard Medical School, Athinoula A. Martinos Center for Biomedical Imaging, Charlestown, Massachusetts, United States
| | - Shy Shoham
- New York University Grossman School of Medicine, Tech4Health and Neuroscience Institutes, New York, New York, United States
| | - Sanaya N. Shroff
- Boston University, Department of Biomedical Engineering, Boston, Massachusetts, United States
| | - R. Angus Silver
- University College London, Department of Neuroscience, Physiology and Pharmacology, London, United Kingdom
| | - Ruth R. Sims
- Sorbonne University, INSERM, CNRS, Institut de la Vision, Paris, France
| | - Spencer L. Smith
- University of California Santa Barbara, Department of Electrical and Computer Engineering, Santa Barbara, California, United States
| | - Vivek J. Srinivasan
- New York University Langone Health, Departments of Ophthalmology and Radiology, New York, New York, United States
| | - Martin Thunemann
- Boston University, Department of Biomedical Engineering, Boston, Massachusetts, United States
| | - Lei Tian
- Boston University, Departments of Electrical Engineering and Biomedical Engineering, Boston, Massachusetts, United States
| | - Lin Tian
- University of California Davis, Department of Biochemistry and Molecular Medicine, Davis, California, United States
| | - Thomas Troxler
- University of Pennsylvania, Perelman School of Medicine, Department of Biochemistry and Biophysics, Philadelphia, Pennsylvania, United States
- University of Pennsylvania, School of Arts and Sciences, Department of Chemistry, Philadelphia, Pennsylvania, United States
| | - Antoine Valera
- University College London, Department of Neuroscience, Physiology and Pharmacology, London, United Kingdom
| | - Alipasha Vaziri
- Rockefeller University, Laboratory of Neurotechnology and Biophysics, New York, New York, United States
- The Rockefeller University, The Kavli Neural Systems Institute, New York, New York, United States
| | - Sergei A. Vinogradov
- University of Pennsylvania, Perelman School of Medicine, Department of Biochemistry and Biophysics, Philadelphia, Pennsylvania, United States
- University of Pennsylvania, School of Arts and Sciences, Department of Chemistry, Philadelphia, Pennsylvania, United States
| | - Flavia Vitale
- Center for Neuroengineering and Therapeutics, Departments of Neurology, Bioengineering, Physical Medicine and Rehabilitation, Philadelphia, Pennsylvania, United States
| | - Lihong V. Wang
- California Institute of Technology, Andrew and Peggy Cherng Department of Medical Engineering, Department of Electrical Engineering, Pasadena, California, United States
| | - Hana Uhlířová
- Institute of Scientific Instruments of the Czech Academy of Sciences, Brno, Czech Republic
| | - Chris Xu
- Cornell University, School of Applied and Engineering Physics, Ithaca, New York, United States
| | - Changhuei Yang
- California Institute of Technology, Departments of Electrical Engineering, Bioengineering and Medical Engineering, Pasadena, California, United States
| | - Mu-Han Yang
- University of California San Diego, Department of Electrical and Computer Engineering, La Jolla, California, United States
| | - Gary Yellen
- Harvard Medical School, Department of Neurobiology, Boston, Massachusetts, United States
| | - Ofer Yizhar
- Weizmann Institute of Science, Department of Brain Sciences, Rehovot, Israel
| | - Yongxin Zhao
- Carnegie Mellon University, Department of Biological Sciences, Pittsburgh, Pennsylvania, United States
| |
Collapse
|
48
|
Schilling K, Zhai Y, Zhou Z, Zhou B, Brown E, Zhang X. High-resolution imaging of the osteogenic and angiogenic interface at the site of murine cranial bone defect repair via multiphoton microscopy. eLife 2022; 11:83146. [PMID: 36326085 PMCID: PMC9678361 DOI: 10.7554/elife.83146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 10/31/2022] [Indexed: 11/05/2022] Open
Abstract
The spatiotemporal blood vessel formation and specification at the osteogenic and angiogenic interface of murine cranial bone defect repair were examined utilizing a high-resolution multiphoton-based imaging platform in conjunction with advanced optical techniques that allow interrogation of the oxygen microenvironment and cellular energy metabolism in living animals. Our study demonstrates the dynamic changes of vessel types, that is, arterial, venous, and capillary vessel networks at the superior and dura periosteum of cranial bone defect, suggesting a differential coupling of the vessel type with osteoblast expansion and bone tissue deposition/remodeling during repair. Employing transgenic reporter mouse models that label distinct types of vessels at the site of repair, we further show that oxygen distributions in capillary vessels at the healing site are heterogeneous as well as time- and location-dependent. The endothelial cells coupling to osteoblasts prefer glycolysis and are less sensitive to microenvironmental oxygen changes than osteoblasts. In comparison, osteoblasts utilize relatively more OxPhos and potentially consume more oxygen at the site of repair. Taken together, our study highlights the dynamics and functional significance of blood vessel types at the site of defect repair, opening up opportunities for further delineating the oxygen and metabolic microenvironment at the interface of bone tissue regeneration.
Collapse
Affiliation(s)
- Kevin Schilling
- Center for Musculoskeletal Research, University of Rochester, School of Medicine and DentistryRochesterUnited States,Department of Biomedical Engineering, University of RochesterRochesterUnited States
| | - Yuankn Zhai
- Center for Musculoskeletal Research, University of Rochester, School of Medicine and DentistryRochesterUnited States
| | - Zhuang Zhou
- Center for Musculoskeletal Research, University of Rochester, School of Medicine and DentistryRochesterUnited States
| | - Bin Zhou
- Shanghai Institutes for Biological SciencesShanghaiChina
| | - Edward Brown
- Department of Biomedical Engineering, University of RochesterRochesterUnited States
| | - Xinping Zhang
- Center for Musculoskeletal Research, University of Rochester, School of Medicine and DentistryRochesterUnited States
| |
Collapse
|
49
|
Solomatina AI, Chelushkin PS, Su SH, Wu CH, Chou PT, Tunik SP. Combined fluorophore and phosphor conjugation: a new design concept for simultaneous and spatially localized dual lifetime intracellular sensing of oxygen and pH. Chem Commun (Camb) 2021; 58:419-422. [PMID: 34897308 DOI: 10.1039/d1cc06132a] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
In this communication, we propose a new strategy for double-parametric biosensing and present a dual pH/O2 lifetime sensor based on the covalent conjugation of fluorescein (pH sensor) and an orthometalated iridium complex (O2 sensor) to human serum albumin (HSA). The resulting conjugate demonstrates biocompatibility, low toxicity, and fast cellular uptake, and displays independent lifetime responses towards variations in media acidity and oxygen concentration that makes it suitable for application as an effective pH/O2 probe in luminescence microscopy using the FLIM/PLIM detection mode. The concept applicability has been exemplified using the dual spatially and temporally localized intracellular sensing of pH and O2 concentration in living cells.
Collapse
Affiliation(s)
- Anastasia I Solomatina
- Institute of Chemistry, St. Petersburg State University, St. Petersburg, 198504, Russia.
| | - Pavel S Chelushkin
- Institute of Chemistry, St. Petersburg State University, St. Petersburg, 198504, Russia.
| | - Shih-Hao Su
- Department of Chemistry, National Taiwan University, Taipei, 10617, Taiwan, Republic of China.
| | - Cheng-Ham Wu
- Department of Chemistry, National Taiwan University, Taipei, 10617, Taiwan, Republic of China.
| | - Pi-Tai Chou
- Department of Chemistry, National Taiwan University, Taipei, 10617, Taiwan, Republic of China.
| | - Sergey P Tunik
- Institute of Chemistry, St. Petersburg State University, St. Petersburg, 198504, Russia.
| |
Collapse
|
50
|
Bochkarev LN, Parshina YP, Gracheva YV, Kovylina TA, Lermontova SA, Klapshina LG, Konev AN, Lopatin MA, Lukina MM, Komarova AD, Shcheslavskiy VI, Shirmanova MV. Red Light-Emitting Water-Soluble Luminescent Iridium-Containing Polynorbornenes: Synthesis, Characterization and Oxygen Sensing Properties in Biological Tissues In Vivo. Molecules 2021; 26:6349. [PMID: 34770757 PMCID: PMC8587708 DOI: 10.3390/molecules26216349] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 10/12/2021] [Accepted: 10/18/2021] [Indexed: 11/16/2022] Open
Abstract
New water-soluble polynorbornenes P1-P4 containing oligoether, amino acid groups and luminophoric complexes of iridium(III) were synthesized by ring-opening metathesis polymerization. The polymeric products in organic solvents and in water demonstrate intense photoluminescence in the red spectral region. The polymers P1 and P3 with 1-phenylisoquinoline cyclometalating ligands in iridium fragments reveal 4-6 fold higher emission quantum yields in solutions than those of P2 and P4 that contain iridium complexes with 1-(thien-2-yl)isoquinoline cyclometalating ligands. The emission parameters of P1-P4 in degassed solutions essentially differ from those in the aerated solutions showing oxygen-dependent quenching of phosphorescence. Biological testing of P1 and P3 demonstrates that the polymers do not penetrate into live cultured cancer cells and normal skin fibroblasts and do not possess cytotoxicity within the concentrations and time ranges reasonable for biological studies. In vivo, the polymers display longer phosphorescence lifetimes in mouse tumors than in muscle, as measured using phosphorescence lifetime imaging (PLIM), which correlates with tumor hypoxia. Therefore, preliminary evaluation of the synthesized polymers shows their suitability for noninvasive in vivo assessments of oxygen levels in biological tissues.
Collapse
Affiliation(s)
- Leonid N. Bochkarev
- Razuvaev Institute of Organometallic Chemistry, Russian Academy of Sciences, Tropinina, 49, 603950 Nizhny Novgorod, Russia; (Y.P.P.); (Y.V.G.); (T.A.K.); (S.A.L.); (L.G.K.); (A.N.K.); (M.A.L.)
| | - Yulia P. Parshina
- Razuvaev Institute of Organometallic Chemistry, Russian Academy of Sciences, Tropinina, 49, 603950 Nizhny Novgorod, Russia; (Y.P.P.); (Y.V.G.); (T.A.K.); (S.A.L.); (L.G.K.); (A.N.K.); (M.A.L.)
| | - Yana V. Gracheva
- Razuvaev Institute of Organometallic Chemistry, Russian Academy of Sciences, Tropinina, 49, 603950 Nizhny Novgorod, Russia; (Y.P.P.); (Y.V.G.); (T.A.K.); (S.A.L.); (L.G.K.); (A.N.K.); (M.A.L.)
| | - Tatyana A. Kovylina
- Razuvaev Institute of Organometallic Chemistry, Russian Academy of Sciences, Tropinina, 49, 603950 Nizhny Novgorod, Russia; (Y.P.P.); (Y.V.G.); (T.A.K.); (S.A.L.); (L.G.K.); (A.N.K.); (M.A.L.)
| | - Svetlana A. Lermontova
- Razuvaev Institute of Organometallic Chemistry, Russian Academy of Sciences, Tropinina, 49, 603950 Nizhny Novgorod, Russia; (Y.P.P.); (Y.V.G.); (T.A.K.); (S.A.L.); (L.G.K.); (A.N.K.); (M.A.L.)
| | - Larisa G. Klapshina
- Razuvaev Institute of Organometallic Chemistry, Russian Academy of Sciences, Tropinina, 49, 603950 Nizhny Novgorod, Russia; (Y.P.P.); (Y.V.G.); (T.A.K.); (S.A.L.); (L.G.K.); (A.N.K.); (M.A.L.)
| | - Aleksey N. Konev
- Razuvaev Institute of Organometallic Chemistry, Russian Academy of Sciences, Tropinina, 49, 603950 Nizhny Novgorod, Russia; (Y.P.P.); (Y.V.G.); (T.A.K.); (S.A.L.); (L.G.K.); (A.N.K.); (M.A.L.)
| | - Mikhail A. Lopatin
- Razuvaev Institute of Organometallic Chemistry, Russian Academy of Sciences, Tropinina, 49, 603950 Nizhny Novgorod, Russia; (Y.P.P.); (Y.V.G.); (T.A.K.); (S.A.L.); (L.G.K.); (A.N.K.); (M.A.L.)
| | - Maria M. Lukina
- Institute of Experimental Oncology and Biomedical Technologies, Privolzhsky Research Medical University, Minin and Pozharsky Sq. 10/1, 603005 Nizhny Novgorod, Russia; (M.M.L.); (A.D.K.); (V.I.S.); (M.V.S.)
| | - Anastasia D. Komarova
- Institute of Experimental Oncology and Biomedical Technologies, Privolzhsky Research Medical University, Minin and Pozharsky Sq. 10/1, 603005 Nizhny Novgorod, Russia; (M.M.L.); (A.D.K.); (V.I.S.); (M.V.S.)
| | - Vladislav I. Shcheslavskiy
- Institute of Experimental Oncology and Biomedical Technologies, Privolzhsky Research Medical University, Minin and Pozharsky Sq. 10/1, 603005 Nizhny Novgorod, Russia; (M.M.L.); (A.D.K.); (V.I.S.); (M.V.S.)
- Becker&Hickl GmbH, Nunsdorfer Ring 7-9, 12277 Berlin, Germany
| | - Marina V. Shirmanova
- Institute of Experimental Oncology and Biomedical Technologies, Privolzhsky Research Medical University, Minin and Pozharsky Sq. 10/1, 603005 Nizhny Novgorod, Russia; (M.M.L.); (A.D.K.); (V.I.S.); (M.V.S.)
| |
Collapse
|