1
|
Stamenkovic S, Schmid F, Gurler G, Abolmaali F, Weitermann NA, Takasaki KT, Bonney SK, Sosa MJ, Bennett HC, Kim Y, Waters J, Shih AY. Impaired capillary-venous drainage contributes to gliosis and demyelination in white matter during aging. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.02.11.579849. [PMID: 38405879 PMCID: PMC10888936 DOI: 10.1101/2024.02.11.579849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/27/2024]
Abstract
The progressive loss of cerebral white matter during aging contributes to cognitive decline, but whether reduced blood flow is a cause or consequence remains debated. Using deep multi-photon imaging in mice, we examined microvascular networks perfusing myelinated tissues in cortical layer 6 and corpus callosum. We identified sparse, wide-reaching venules, termed principal cortical venules, that exclusively drain deep tissues and resemble vasculature at the human cortex and U-fiber interface. Aging involved selective constriction and rarefaction of capillaries in deep branches of principal cortical venules. This resulted in mild hypoperfusion that was associated with microgliosis, astrogliosis and demyelination in deep tissues, but not upper cortex. Inducing a comparable hypoperfusion in adult mice using carotid artery stenosis triggered a similar tissue pathology specific to layer 6 and corpus callosum. Thus, impaired capillary-venous drainage is a contributor to hypoperfusion and a potential therapeutic target for preserving blood flow to white matter during aging.
Collapse
|
2
|
Saputra F, Tseng TM, Casuga FP, Lai YH, Hung CH, Hsiao CD. Application of a ImageJ-Based Method to Measure Blood Flow in Adult Zebrafish and Its Applications for Toxicological and Pharmacological Assessments. BIOLOGY 2025; 14:51. [PMID: 39857282 PMCID: PMC11763070 DOI: 10.3390/biology14010051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 12/26/2024] [Accepted: 01/08/2025] [Indexed: 01/27/2025]
Abstract
Blood flow is an important physiological endpoint to measure cardiovascular performance in animals. Because of their innate transparent bodies, zebrafish is an excellent animal model for assessing in vivo cardiovascular performance. Previously, various helpful methods for measuring blood flow in zebrafish larvae were discovered and developed. However, an optimized method to measure blood flow in adult zebrafish has not been reported. In this paper, the tail fin region was selected as target for blood flow measurements using the Trackmate method, provided by ImageJ platform. Based on power statistic calculations, the aortic vessel at the tail base was selected, and other parameters, such as ambient temperature, were investigated for method standardization, in order to minimize experimental variation. The method was also validated using fenpropathrin and ponatinib, which showed some cardiac alterations in a previous zebrafish study. We also checked the versatility of this method by following the same setup in black tetra and medaka and found that this method performed well. However, our results show that heavy pigmentation, like that found in tiger barb, and overlapping vessels, like those in parrot fish, make it hard for this method to perform well. Overall, an optimized protocol was used for the first time to measure blood flow velocity in adult wild-type zebrafish without the aid of transgenic lines or fluorescent dye.
Collapse
Affiliation(s)
- Ferry Saputra
- Department of Chemistry, Chung Yuan Christian University, Taoyuan 320314, Taiwan;
- Department of Orthopaedics, E-Da Hospital/E-Da Dachang Hospital, Kaohsiung 82445, Taiwan
| | - Tzu-Ming Tseng
- School of Medicine for International Students, College of Medicine, I-Shou University, Kaohsiung 82445, Taiwan;
- Department of Bioscience Technology, Chung Yuan Christian University, Taoyuan 320314, Taiwan
- Institute of Biotechnology and Chemical Engineering, I-Shou University, Kaohsiung 84001, Taiwan
| | - Franelyne P. Casuga
- Department of Pharmacy, Research Center for the Natural and Applied Science, University of Santo Tomas, Manila 1008, Philippines;
| | - Yu-Heng Lai
- Department of Chemistry, Chinese Culture University, Taipei 11114, Taiwan;
| | - Chih-Hsin Hung
- Institute of Biotechnology and Chemical Engineering, I-Shou University, Kaohsiung 84001, Taiwan
| | - Chung-Der Hsiao
- Department of Chemistry, Chung Yuan Christian University, Taoyuan 320314, Taiwan;
- Department of Bioscience Technology, Chung Yuan Christian University, Taoyuan 320314, Taiwan
- Department of Chemistry, Chinese Culture University, Taipei 11114, Taiwan;
- Research Center for Aquatic Toxicology and Pharmacology, Chung Yuan Christian University, Taoyuan 320314, Taiwan
| |
Collapse
|
3
|
Evans LE, Gray AL, Walsh KR, Danby TGE, Pritchard HAT, Allan SM, Gurney AM, Greenstein AS, Schiessl I. Combining In Vivo Two-Photon and Laser Speckle Microscopy With the Ex Vivo Capillary-Parenchymal Arteriole Preparation as a Novel Approach to Study Neurovascular Coupling. Microcirculation 2025; 32:e70001. [PMID: 39777814 PMCID: PMC11706670 DOI: 10.1111/micc.70001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 12/09/2024] [Accepted: 12/23/2024] [Indexed: 01/11/2025]
Abstract
OBJECTIVE Cerebral blood flow (CBF) decline is increasingly recognized as an area of importance for targeting neurodegenerative disorders, yet full understanding of the mechanisms that underlie CBF changes are lacking. Animal models are crucial for expanding our knowledge as methods for studying global CBF and neurovascular coupling in humans are limited and require expensive specialized scanners. METHODS Use of appropriate animal models can increase our understanding of cerebrovascular function, so we have combined chronic cranial windows with in vivo two-photon and laser speckle microscopy and ex vivo capillary-parenchymal arteriole (CaPA) preparations. Chronic cranial windows allow for longitudinal direct observation of the cerebral microvasculature and surrounding parenchyma while the CaPA preparation can assess capillary and arteriole function in isolation of the neuronal tissue. RESULTS Here, we found that extra-dural cranial windows and related imaging protocols do not affect vascular function in the CaPA preparation. Cortical vessels from animals that have undergone imaging can therefore be taken to discover physiological alterations in the cerebral vasculature that contribute to any observed in vivo changes. CONCLUSION This approach will enhance neurodegenerative research with the benefit of limiting animal usage.
Collapse
Affiliation(s)
- Lowri E. Evans
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance NHS Foundation TrustUniversity of ManchesterManchesterUK
- Division of Cardiovascular Sciences, School of Biological Sciences, Faculty of Biology, Medicine and HealthUniversity of ManchesterManchesterUK
| | - Anna L. Gray
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance NHS Foundation TrustUniversity of ManchesterManchesterUK
- Division of Infection, Immunity & Respiratory Medicine, Faculty of Biology, Medicine and HealthUniversity of ManchesterManchesterUK
- Wellcome Centre for Cell‐Matrix Research, Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology, Medicine and Health, Manchester Academic Health Science CentreUniversity of ManchesterManchesterUK
| | - Katy R. Walsh
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance NHS Foundation TrustUniversity of ManchesterManchesterUK
- Division of Cardiovascular Sciences, School of Biological Sciences, Faculty of Biology, Medicine and HealthUniversity of ManchesterManchesterUK
| | - Thea G. E. Danby
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance NHS Foundation TrustUniversity of ManchesterManchesterUK
- Division of Cardiovascular Sciences, School of Biological Sciences, Faculty of Biology, Medicine and HealthUniversity of ManchesterManchesterUK
| | - Harry A. T. Pritchard
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance NHS Foundation TrustUniversity of ManchesterManchesterUK
- Division of Cardiovascular Sciences, School of Biological Sciences, Faculty of Biology, Medicine and HealthUniversity of ManchesterManchesterUK
| | - Stuart M. Allan
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance NHS Foundation TrustUniversity of ManchesterManchesterUK
- Division of Neuroscience, School of Biological Sciences, Faculty of Biology, Medicine and HealthUniversity of ManchesterManchesterUK
| | - Alison M. Gurney
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance NHS Foundation TrustUniversity of ManchesterManchesterUK
- Division of Pharmacy & Optometry, School of Health Sciences, Faculty of Biology, Medicine and HealthUniversity of ManchesterManchesterUK
| | - Adam S. Greenstein
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance NHS Foundation TrustUniversity of ManchesterManchesterUK
- Division of Cardiovascular Sciences, School of Biological Sciences, Faculty of Biology, Medicine and HealthUniversity of ManchesterManchesterUK
| | - Ingo Schiessl
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance NHS Foundation TrustUniversity of ManchesterManchesterUK
- Division of Neuroscience, School of Biological Sciences, Faculty of Biology, Medicine and HealthUniversity of ManchesterManchesterUK
| |
Collapse
|
4
|
Battini S, Cantarutti N, Kotsalos C, Roussel Y, Cattabiani A, Arnaudon A, Favreau C, Antonel S, Markram H, Keller D. Modeling of Blood Flow Dynamics in Rat Somatosensory Cortex. Biomedicines 2024; 13:72. [PMID: 39857656 PMCID: PMC11761867 DOI: 10.3390/biomedicines13010072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Revised: 12/11/2024] [Accepted: 12/24/2024] [Indexed: 01/27/2025] Open
Abstract
Background: The cerebral microvasculature forms a dense network of interconnected blood vessels where flow is modulated partly by astrocytes. Increased neuronal activity stimulates astrocytes to release vasoactive substances at the endfeet, altering the diameters of connected vessels. Methods: Our study simulated the coupling between blood flow variations and vessel diameter changes driven by astrocytic activity in the rat somatosensory cortex. We developed a framework with three key components: coupling between the vasculature and synthesized astrocytic morphologies, a fluid dynamics model to compute flow in each vascular segment, and a stochastic process replicating the effect of astrocytic endfeet on vessel radii. Results: The model was validated against experimental flow values from the literature across cortical depths. We found that local vasodilation from astrocyte activity increased blood flow, especially in capillaries, exhibiting a layer-specific response in deeper cortical layers. Additionally, the highest blood flow variability occurred in capillaries, emphasizing their role in cerebral perfusion regulation. We discovered that astrocytic activity impacted blood flow dynamics in a localized, clustered manner, with most vascular segments influenced by two to three neighboring endfeet. Conclusions: These insights enhance our understanding of neurovascular coupling and guide future research on blood flow-related diseases.
Collapse
Affiliation(s)
- Stéphanie Battini
- Blue Brain Project, École Polytechnique Fédérale de Lausanne (EPFL), Campus Biotech, 1202 Geneva, Switzerland
| | | | | | | | | | | | | | | | | | | |
Collapse
|
5
|
Gomez-Garcia MJ, Abdelkarim M, Cramb DT, Childs SJ, Rinker KD, Labouta HI. Blood vessel wall shear stress determines regions of liposome accumulation in angiogenic vasculature. Drug Deliv Transl Res 2024; 14:3608-3620. [PMID: 39042244 DOI: 10.1007/s13346-024-01671-1] [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] [Accepted: 07/09/2024] [Indexed: 07/24/2024]
Abstract
Nanoparticles used for drug delivery often require intravenous administration exposing them to fluid forces within the vasculature, yet the impact of blood flow on nanoparticle delivery remains incompletely understood. Here, we utilized transgenic zebrafish embryos to investigate the relationship between the accumulation of fluorescently labeled PEGylated liposomes and various hemodynamic factors (such as flow velocity, wall shear stress (WSS), and flow pattern) across a wide range of angiogenic blood vessels. We reconstructed 3D models of vascular structures from confocal images and used computational fluid dynamics to calculate local WSS, velocities, and define flow patterns. The spatial distribution of fluorescently labeled liposomes was subsequently mapped within the same 3D space and correlated with local hemodynamic parameters. Through the integration of computational fluid dynamics and in vivo experimentation, we show that liposomes accumulated in vessel regions with WSS between 0.1-0.8 Pa, displaying an inverse linear correlation (R2 > 0.85) between time-averaged wall shear stress and liposome localization in vivo. Interestingly, flow pattern did not appear to impact liposome accumulation. Collectively, our findings suggest the potential of stealth liposomes for passive targeting of low-flow vasculature, including capillaries and intricate angiogenic vasculature resembling that of tumor vessel networks.
Collapse
Affiliation(s)
- M Juliana Gomez-Garcia
- Biomedical Engineering, Faculty of Engineering, University of Toronto, Toronto, ON, Canada
- Biomedical Engineering, Schulich School of Engineering, University of Calgary, Calgary, AB, Canada
| | - Mahmoud Abdelkarim
- Biomedical Engineering, Faculty of Engineering, University of Toronto, Toronto, ON, Canada
- Keenan Research Centre for Biomedical Science, Unity Health Toronto, Toronto, ON, Canada
| | - David T Cramb
- Department of Chemistry and Biology, Faculty of Science, Toronto Metropolitan University, Toronto, ON, Canada
| | - Sarah J Childs
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Kristina D Rinker
- Department of Chemical and Petroleum Engineering, Schulich School of Engineering, University of Calgary, Calgary, AB, Canada.
| | - Hagar I Labouta
- Biomedical Engineering, Faculty of Engineering, University of Toronto, Toronto, ON, Canada.
- Keenan Research Centre for Biomedical Science, Unity Health Toronto, Toronto, ON, Canada.
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
6
|
Nyúl-Tóth Á, Negri S, Sanford M, Jiang R, Patai R, Budda M, Petersen B, Pinckard J, Chandragiri SS, Shi H, Reyff Z, Ballard C, Gulej R, Csik B, Ferrier J, Balasubramanian P, Yabluchanskiy A, Cleuren A, Conley S, Ungvari Z, Csiszar A, Tarantini S. Novel intravital approaches to quantify deep vascular structure and perfusion in the aging mouse brain using ultrasound localization microscopy (ULM). J Cereb Blood Flow Metab 2024; 44:1378-1396. [PMID: 38867576 PMCID: PMC11542130 DOI: 10.1177/0271678x241260526] [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: 10/27/2023] [Revised: 04/15/2024] [Accepted: 05/16/2024] [Indexed: 06/14/2024]
Abstract
Intra-vital visualization of deep cerebrovascular structures and blood flow in the aging brain has been a difficult challenge in the field of neurovascular research, especially when considering the key role played by the cerebrovasculature in the pathogenesis of both vascular cognitive impairment and dementia (VCID) and Alzheimer's disease (AD). Traditional imaging methods face difficulties with the thicker skull of older brains, making high-resolution imaging and cerebral blood flow (CBF) assessment challenging. However, functional ultrasound (fUS) imaging, an emerging non-invasive technique, provides real-time CBF insights with notable spatial-temporal resolution. This study introduces an enhanced longitudinal fUS method for aging brains. Using elderly (24-month C57BL/6) mice, we detail replacing the skull with a polymethylpentene window for consistent fUS imaging over extended periods. Ultrasound localization mapping (ULM), involving the injection of a microbubble (<<10 μm) suspension allows for recording of high-resolution microvascular vessels and flows. ULM relies on the localization and tracking of single circulating microbubbles in the blood flow. A FIJI-based analysis interprets these high-quality ULM visuals. Testing on older mouse brains, our method successfully unveils intricate vascular specifics even in-depth, showcasing its utility for longitudinal studies that require ongoing evaluations of CBF and vascular aspects in aging-focused research.
Collapse
Affiliation(s)
- Ádám Nyúl-Tóth
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, Budapest, Hungary
| | - Sharon Negri
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK, USA
| | - Madison Sanford
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK, USA
| | - Raymond Jiang
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Roland Patai
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Madeline Budda
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Department of Cellular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Benjamin Petersen
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Jessica Pinckard
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Department of Cellular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Siva Sai Chandragiri
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Helen Shi
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Zeke Reyff
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Cade Ballard
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Rafal Gulej
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Boglarka Csik
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK, USA
| | | | - Priya Balasubramanian
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Andriy Yabluchanskiy
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK, USA
| | - Audrey Cleuren
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Shannon Conley
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Department of Cellular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Zoltan Ungvari
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, Budapest, Hungary
- Department of Public Health, Semmelweis University, Semmelweis University, Budapest, Hungary
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Anna Csiszar
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, Budapest, Hungary
| | - Stefano Tarantini
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK, USA
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| |
Collapse
|
7
|
Bonney SK, Nielson CD, Sosa MJ, Bonnar O, Shih AY. Capillary regression leads to sustained local hypoperfusion by inducing constriction of upstream transitional vessels. Proc Natl Acad Sci U S A 2024; 121:e2321021121. [PMID: 39236241 PMCID: PMC11406265 DOI: 10.1073/pnas.2321021121] [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: 12/01/2023] [Accepted: 08/07/2024] [Indexed: 09/07/2024] Open
Abstract
In the brain, a microvascular sensory web coordinates oxygen delivery to regions of neuronal activity. This involves a dense network of capillaries that send conductive signals upstream to feeding arterioles to promote vasodilation and blood flow. Although this process is critical to the metabolic supply of healthy brain tissue, it may also be a point of vulnerability in disease. Deterioration of capillary networks is a feature of many neurological disorders and injuries and how this web is engaged during vascular damage remains unknown. We performed in vivo two-photon microscopy on young adult mural cell reporter mice and induced focal capillary injuries using precise two-photon laser irradiation of single capillaries. We found that ~59% of the injuries resulted in regression of the capillary segment 7 to 14 d following injury, and the remaining repaired to reestablish blood flow within 7 d. Injuries that resulted in capillary regression induced sustained vasoconstriction in the upstream arteriole-capillary transition (ACT) zone at least 21 days postinjury in both awake and anesthetized mice. The degree of vasomotor dynamics was chronically attenuated in the ACT zone consequently reducing blood flow in the ACT zone and in secondary, uninjured downstream capillaries. These findings demonstrate how focal capillary injury and regression can impair the microvascular sensory web and contribute to cerebral hypoperfusion.
Collapse
Affiliation(s)
- Stephanie K. Bonney
- Center for Developmental Biology and Regenerative Medicine, Seattle Children’s Research Institute, Seattle, WA98101
| | - Cara D. Nielson
- Center for Developmental Biology and Regenerative Medicine, Seattle Children’s Research Institute, Seattle, WA98101
- Graduate Program in Neuroscience, University of Washington, Seattle, WA98195
| | - Maria J. Sosa
- Center for Developmental Biology and Regenerative Medicine, Seattle Children’s Research Institute, Seattle, WA98101
| | - Orla Bonnar
- MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital and Harvard Medical School, Boston, MA02129
| | - Andy Y. Shih
- Center for Developmental Biology and Regenerative Medicine, Seattle Children’s Research Institute, Seattle, WA98101
- Department of Pediatrics, University of Washington, Seattle, WA98195
- Department of Bioengineering, University of Washington, Seattle, WA98195
| |
Collapse
|
8
|
Bahari F, Dzhala V, Balena T, Lillis KP, Staley KJ. Intraventricular haemorrhage in premature infants: the role of immature neuronal salt and water transport. Brain 2024; 147:3216-3233. [PMID: 38815055 PMCID: PMC11370806 DOI: 10.1093/brain/awae161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 04/25/2024] [Accepted: 04/28/2024] [Indexed: 06/01/2024] Open
Abstract
Intraventricular haemorrhage is a common complication of premature birth. Survivors are often left with cerebral palsy, intellectual disability and/or hydrocephalus. Animal models suggest that brain tissue shrinkage, with subsequent vascular stretch and tear, is an important step in the pathophysiology, but the cause of this shrinkage is unknown. Clinical risk factors for intraventricular haemorrhage are biomarkers of hypoxic-ischaemic stress, which causes mature neurons to swell. However, immature neuronal volume might shift in the opposite direction in these conditions. This is because immature neurons express the chloride, salt and water transporter NKCC1, which subserves regulatory volume increases in non-neural cells, whereas mature neurons express KCC2, which subserves regulatory volume decreases. When hypoxic-ischaemic conditions reduce active ion transport and increase the cytoplasmic membrane permeability, the effects of these transporters are diminished. Consequentially, mature neurons swell (cytotoxic oedema), whereas immature neurons might shrink. After hypoxic-ischaemic stress, in vivo and in vitro multi-photon imaging of perinatal transgenic mice demonstrated shrinkage of viable immature neurons, bulk tissue shrinkage and blood vessel displacement. Neuronal shrinkage was correlated with age-dependent membrane salt and water transporter expression using immunohistochemistry. Shrinkage of immature neurons was prevented by prior genetic or pharmacological inhibition of NKCC1 transport. These findings open new avenues of investigation for the detection of acute brain injury by neuroimaging, in addition to prevention of neuronal shrinkage and the ensuing intraventricular haemorrhage, in premature infants.
Collapse
Affiliation(s)
- Fatemeh Bahari
- Department of Neurology, Massachusetts General Hospital, Boston, MA 02114, USA
- Department of Neurology, Harvard Medical School, Boston, MA 02115, USA
| | - Volodymyr Dzhala
- Department of Neurology, Massachusetts General Hospital, Boston, MA 02114, USA
- Department of Neurology, Harvard Medical School, Boston, MA 02115, USA
| | - Trevor Balena
- Department of Neurology, Massachusetts General Hospital, Boston, MA 02114, USA
- Department of Neurology, Harvard Medical School, Boston, MA 02115, USA
| | - Kyle P Lillis
- Department of Neurology, Massachusetts General Hospital, Boston, MA 02114, USA
- Department of Neurology, Harvard Medical School, Boston, MA 02115, USA
| | - Kevin J Staley
- Department of Neurology, Massachusetts General Hospital, Boston, MA 02114, USA
- Department of Neurology, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
9
|
Ghigo N, Ramos-Palacios G, Bourquin C, Xing P, Wu A, Cortés N, Ladret H, Ikan L, Casanova C, Porée J, Sadikot A, Provost J. Dynamic Ultrasound Localization Microscopy Without ECG-Gating. ULTRASOUND IN MEDICINE & BIOLOGY 2024; 50:1436-1448. [PMID: 38969526 DOI: 10.1016/j.ultrasmedbio.2024.05.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 05/04/2024] [Accepted: 05/22/2024] [Indexed: 07/07/2024]
Abstract
OBJECTIVE Dynamic Ultrasound Localization Microscopy (DULM) has first been developed for non-invasive Pulsatility measurements in the rodent brain. DULM relies on the localization and tracking of microbubbles (MBs) injected into the bloodstream, to obtain highly resolved velocity and density cine-loops. Previous DULM techniques required ECG-gating, limiting its application to specific datasets, and increasing acquisition time. The objective of this study is to eliminate the need for ECG-gating in DULM experiments by introducing a motion-matching method for time registration. METHODS We developed a motion-matching algorithm based on tissue Doppler that leverages the cyclic tissue motion within the brain. Tissue Doppler was estimated for each group of frames in the acquisitions, at multiple locations identified as local maxima in the skin above the skull. Subsequently, each group of frames was time-registered to a reference group by delaying it based on the maximum correlation value between their respective tissue Doppler signals. This synchronization ensured that each group of frames aligned with the brain tissue motion of the reference group, and consequently, with its cardiac cycle. As a result, velocities of MBs could be averaged to retrieve flow velocity variations over time. RESULTS Initially validated in ECG-gated acquisitions in a rat model (n = 1), the proposed method was successfully applied in a mice model in 2D (n = 3) and in a feline model in 3D (n = 1). Performing time-registration with the proposed motion-matching method or by using ECG-gating leads to similar results. For the first time, dynamic velocity and density cine-loops were extracted without the need for any information on the animal ECG, and complex dynamic markers such as the Pulsatility index were estimated. CONCLUSION Results suggest that DULM can be performed without external gating, enabling the use of DULM on any ULM dataset where enough MBs are detectable. Time registration by motion-matching represents a significant advancement in DULM techniques, making DULM more accessible by simplifying its experimental complexity.
Collapse
Affiliation(s)
- Nin Ghigo
- Department of Engineering Physics, Polytechnique Montréal, Montréal, Quebec, Canada.
| | | | - Chloé Bourquin
- Department of Engineering Physics, Polytechnique Montréal, Montréal, Quebec, Canada
| | - Paul Xing
- Department of Engineering Physics, Polytechnique Montréal, Montréal, Quebec, Canada
| | - Alice Wu
- Department of Engineering Physics, Polytechnique Montréal, Montréal, Quebec, Canada
| | - Nelson Cortés
- School of Optometry, University of Montreal, Montréal, Quebec, Canada
| | - Hugo Ladret
- School of Optometry, University of Montreal, Montréal, Quebec, Canada; Institut de Neurosciences de la Timone, UMR 7289, CNRS and Aix-Marseille Université, Marseille, France
| | - Lamyae Ikan
- School of Optometry, University of Montreal, Montréal, Quebec, Canada
| | | | - Jonathan Porée
- Department of Engineering Physics, Polytechnique Montréal, Montréal, Quebec, Canada
| | - Abbas Sadikot
- Montreal Neurological Institute, McGill University, Montréal, Quebec, Canada
| | - Jean Provost
- Department of Engineering Physics, Polytechnique Montréal, Montréal, Quebec, Canada; Montreal Heart Institute, Montréal, Quebec, Canada
| |
Collapse
|
10
|
Seyedhassantehrani N, Burns CS, Verrinder R, Okafor V, Abbasizadeh N, Spencer JA. Intravital two-photon microscopy of the native mouse thymus. PLoS One 2024; 19:e0307962. [PMID: 39088574 PMCID: PMC11293686 DOI: 10.1371/journal.pone.0307962] [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: 02/27/2024] [Accepted: 07/15/2024] [Indexed: 08/03/2024] Open
Abstract
The thymus, a key organ in the adaptive immune system, is sensitive to a variety of insults including cytotoxic preconditioning, which leads to atrophy, compression of the blood vascular system, and alterations in hemodynamics. Although the thymus has innate regenerative capabilities, the production of T cells relies on the trafficking of lymphoid progenitors from the bone marrow through the altered thymic blood vascular system. Our understanding of thymic blood vascular hemodynamics is limited due to technical challenges associated with accessing the native thymus in live mice. To overcome this challenge, we developed an intravital two-photon imaging method to visualize the native thymus in vivo and investigated functional changes to the vascular system following sublethal irradiation. We quantified blood flow velocity and shear rate in cortical blood vessels and identified a subtle but significant increase in vessel leakage and diameter ~24 hrs post-sublethal irradiation. Ex vivo whole organ imaging of optically cleared thymus lobes confirmed a disruption of the thymus vascular structure, resulting in an increase in blood vessel diameter and vessel area, and concurrent thymic atrophy. This novel two-photon intravital imaging method enables a new paradigm for directly investigating the thymic microenvironment in vivo.
Collapse
Affiliation(s)
- Negar Seyedhassantehrani
- Quantitative and Systems Biology Graduate Program, University of California Merced, Merced, California, United States of America
- NSF-CREST Center for Cellular and Biomolecular Machines, University of California Merced, Merced, California, United States of America
| | - Christian S. Burns
- Quantitative and Systems Biology Graduate Program, University of California Merced, Merced, California, United States of America
- NSF-CREST Center for Cellular and Biomolecular Machines, University of California Merced, Merced, California, United States of America
| | - Ruth Verrinder
- NSF-CREST Center for Cellular and Biomolecular Machines, University of California Merced, Merced, California, United States of America
- Department of Bioengineering, University of California Merced, Merced, California, United States of America
| | - Victoria Okafor
- NSF-CREST Center for Cellular and Biomolecular Machines, University of California Merced, Merced, California, United States of America
- Department of Bioengineering, University of California Merced, Merced, California, United States of America
| | - Nastaran Abbasizadeh
- Quantitative and Systems Biology Graduate Program, University of California Merced, Merced, California, United States of America
- NSF-CREST Center for Cellular and Biomolecular Machines, University of California Merced, Merced, California, United States of America
| | - Joel A. Spencer
- Quantitative and Systems Biology Graduate Program, University of California Merced, Merced, California, United States of America
- NSF-CREST Center for Cellular and Biomolecular Machines, University of California Merced, Merced, California, United States of America
- Department of Bioengineering, University of California Merced, Merced, California, United States of America
- Health Science Research Institute, University of California Merced, Merced, California, United States of America
| |
Collapse
|
11
|
Abbasizadeh N, Burns CS, Verrinder R, Ghazali F, Seyedhassantehrani N, Spencer JA. Age and dose dependent changes to the bone and bone marrow microenvironment after cytotoxic conditioning with busulfan. Front Cell Dev Biol 2024; 12:1441381. [PMID: 39139448 PMCID: PMC11319712 DOI: 10.3389/fcell.2024.1441381] [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: 05/31/2024] [Accepted: 07/09/2024] [Indexed: 08/15/2024] Open
Abstract
Preparative regimens before Hematopoietic Cell Transplantation (HCT) damage the bone marrow (BM) microenvironment, potentially leading to secondary morbidity and even mortality. The precise effects of cytotoxic preconditioning on bone and BM remodeling, regeneration, and subsequent hematopoietic recovery over time remain unclear. Moreover, the influence of recipient age and cytotoxic dose have not been fully described. In this study, we longitudinally investigated bone and BM remodeling after busulfan treatment with low intensity (LI) and high intensity (HI) regimens as a function of animal age. As expected, higher donor chimerism was observed in young mice in both LI and HI regimens compared to adult mice. Noticeably in adult mice, significant engraftment was only observed in the HI group. The integrity of the blood-bone marrow barrier in calvarial BM blood vessels was lost after busulfan treatment in the young mice and remained altered even 6 weeks after HCT. In adult mice, the severity of vascular leakage appeared to be dose-dependent, being more pronounced in HI compared to LI recipients. Interestingly, no noticeable change in blood flow velocity was observed following busulfan treatment. Ex vivo imaging of the long bones revealed a reduction in the frequency and an increase in the diameter and density of the blood vessels shortly after treatment, a phenomenon that largely recovered in young mice but persisted in older mice after 6 weeks. Furthermore, analysis of bone remodeling indicated a significant alteration in bone turnover at 6 weeks compared to earlier timepoints in both young and adult mice. Overall, our results reveal new aspects of bone and BM remodeling, as well as hematopoietic recovery, which is dependent on the cytotoxic dose and recipient age.
Collapse
Affiliation(s)
- Nastaran Abbasizadeh
- Department of Bioengineering, University of California, Merced, Merced, CA, United States
- Center for Cellular and Biomolecular Machines, University of California, Merced, Merced, CA, United States
| | - Christian S. Burns
- Department of Bioengineering, University of California, Merced, Merced, CA, United States
- Center for Cellular and Biomolecular Machines, University of California, Merced, Merced, CA, United States
| | - Ruth Verrinder
- Department of Bioengineering, University of California, Merced, Merced, CA, United States
- Center for Cellular and Biomolecular Machines, University of California, Merced, Merced, CA, United States
| | - Farhad Ghazali
- Department of Bioengineering, University of California, Merced, Merced, CA, United States
| | - Negar Seyedhassantehrani
- Department of Bioengineering, University of California, Merced, Merced, CA, United States
- Center for Cellular and Biomolecular Machines, University of California, Merced, Merced, CA, United States
| | - Joel A. Spencer
- Department of Bioengineering, University of California, Merced, Merced, CA, United States
- Center for Cellular and Biomolecular Machines, University of California, Merced, Merced, CA, United States
- Health Sciences Research Institute, University of California, Merced, Merced, CA, United States
| |
Collapse
|
12
|
Mester JR, Rozak MW, Dorr A, Goubran M, Sled JG, Stefanovic B. Network response of brain microvasculature to neuronal stimulation. Neuroimage 2024; 287:120512. [PMID: 38199427 DOI: 10.1016/j.neuroimage.2024.120512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 12/21/2023] [Accepted: 01/04/2024] [Indexed: 01/12/2024] Open
Abstract
Neurovascular coupling (NVC), or the adjustment of blood flow in response to local increases in neuronal activity is a hallmark of healthy brain function, and the physiological foundation for functional magnetic resonance imaging (fMRI). However, it remains only partly understood due to the high complexity of the structure and function of the cerebrovascular network. Here we set out to understand NVC at the network level, i.e. map cerebrovascular network reactivity to activation of neighbouring neurons within a 500×500×500 μm3 cortical volume (∼30 high-resolution 3-nL fMRI voxels). Using 3D two-photon fluorescence microscopy data, we quantified blood volume and flow changes in the brain vessels in response to spatially targeted optogenetic activation of cortical pyramidal neurons. We registered the vessels in a series of image stacks acquired before and after stimulations and applied a deep learning pipeline to segment the microvascular network from each time frame acquired. We then performed image analysis to extract the microvascular graphs, and graph analysis to identify the branch order of each vessel in the network, enabling the stratification of vessels by their branch order, designating branches 1-3 as precapillary arterioles and branches 4+ as capillaries. Forty-five percent of all vessels showed significant calibre changes; with 85 % of responses being dilations. The largest absolute CBV change was in the capillaries; the smallest, in the venules. Capillary CBV change was also the largest fraction of the total CBV change, but normalized to the baseline volume, arterioles and precapillary arterioles showed the biggest relative CBV change. From linescans along arteriole-venule microvascular paths, we measured red blood cell velocities and hematocrit, allowing for estimation of pressure and local resistance along these paths. While diameter changes following neuronal activation gradually declined along the paths; the pressure drops from arterioles to venules increased despite decreasing resistance: blood flow thus increased more than local resistance decreases would predict. By leveraging functional volumetric imaging and high throughput deep learning-based analysis, our study revealed distinct hemodynamic responses across the vessel types comprising the microvascular network. Our findings underscore the need for large, dense sampling of brain vessels for characterization of neurovascular coupling at the network level in health and disease.
Collapse
Affiliation(s)
- James R Mester
- University of Toronto, Department of Medical Biophysics, Toronto, Ontario, Canada; Physical Sciences, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - Matthew W Rozak
- University of Toronto, Department of Medical Biophysics, Toronto, Ontario, Canada; Physical Sciences, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - Adrienne Dorr
- Physical Sciences, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - Maged Goubran
- University of Toronto, Department of Medical Biophysics, Toronto, Ontario, Canada; Physical Sciences, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - John G Sled
- University of Toronto, Department of Medical Biophysics, Toronto, Ontario, Canada; Mouse Imaging Centre, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Bojana Stefanovic
- University of Toronto, Department of Medical Biophysics, Toronto, Ontario, Canada; Physical Sciences, Sunnybrook Research Institute, Toronto, Ontario, Canada.
| |
Collapse
|
13
|
Bonney SK, Nielson CD, Sosa MJ, Shih AY. Capillary regression leads to sustained local hypoperfusion by inducing constriction of upstream transitional vessels. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.28.564529. [PMID: 37961686 PMCID: PMC10635020 DOI: 10.1101/2023.10.28.564529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
In the brain, a microvascular sensory web coordinates oxygen delivery to regions of neuronal activity. This involves a dense network of capillaries that send conductive signals upstream to feeding arterioles to promote vasodilation and blood flow. Although this process is critical to the metabolic supply of healthy brain tissue, it may also be a point of vulnerability in disease. Deterioration of capillary networks is a hallmark of many neurological disorders and how this web is engaged during vascular damage remains unknown. We performed in vivo two-photon microscopy on young adult mural cell reporter mice and induced focal capillary injuries using precise two-photon laser irradiation of single capillaries. We found that ∼63% of the injuries resulted in regression of the capillary segment 7-14 days following injury, and the remaining repaired to re-establish blood flow within 7 days. Injuries that resulted in capillary regression induced sustained vasoconstriction in the upstream arteriole-capillary transition (ACT) zone at least 21 days post-injury in both awake and anesthetized mice. This abnormal vasoconstriction involved attenuation of vasomotor dynamics and uncoupling from mural cell calcium signaling following capillary regression. Consequently, blood flow was reduced in the ACT zone and in secondary, uninjured downstream capillaries. These findings demonstrate how capillary injury and regression, as often seen in age-related neurological disease, can impair the microvascular sensory web and contribute to cerebral hypoperfusion. SIGNIFICANCE Deterioration of the capillary network is a characteristic of many neurological diseases and can exacerbate neuronal dysfunction and degeneration due to poor blood perfusion. Here we show that focal capillary injuries can induce vessel regression and elicit sustained vasoconstriction in upstream transitional vessels that branch from cortical penetrating arterioles. This reduces blood flow to broader, uninjured regions of the same microvascular network. These findings suggest that widespread and cumulative damage to brain capillaries in neurological disease may broadly affect blood supply and contribute to hypoperfusion through their remote actions.
Collapse
|
14
|
Meng L, Huang M, Feng S, Wang Y, Lu J, Li P. Optical Flow-Based Full-Field Quantitative Blood-Flow Velocimetry Using Temporal Direction Filtering and Peak Interpolation. Int J Mol Sci 2023; 24:12048. [PMID: 37569421 PMCID: PMC10419297 DOI: 10.3390/ijms241512048] [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: 06/22/2023] [Revised: 07/15/2023] [Accepted: 07/25/2023] [Indexed: 08/13/2023] Open
Abstract
The quantitative measurement of the microvascular blood-flow velocity is critical to the early diagnosis of microvascular dysfunction, yet there are several challenges with the current quantitative flow velocity imaging techniques for the microvasculature. Optical flow analysis allows for the quantitative imaging of the blood-flow velocity with a high spatial resolution, using the variation in pixel brightness between consecutive frames to trace the motion of red blood cells. However, the traditional optical flow algorithm usually suffers from strong noise from the background tissue, and a significant underestimation of the blood-flow speed in blood vessels, due to the errors in detecting the feature points in optical images. Here, we propose a temporal direction filtering and peak interpolation optical flow method (TPIOF) to suppress the background noise, and improve the accuracy of the blood-flow velocity estimation. In vitro phantom experiments and in vivo animal experiments were performed to validate the improvements in our new method.
Collapse
Affiliation(s)
- Liangwei Meng
- Britton Chance Center for Biomedical Photonics and MoE Key Laboratory for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan 430074, China; (L.M.); (M.H.); (Y.W.); (J.L.)
- Research Unit of Multimodal Cross Scale Neural Signal Detection and Imaging, Chinese Academy of Medical Science, HUST-Suzhou Institute for Brainsmatics, Jiangsu Industrial Technology Reserch Institute (JITRI), Suzhou 215100, China
| | - Mange Huang
- Britton Chance Center for Biomedical Photonics and MoE Key Laboratory for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan 430074, China; (L.M.); (M.H.); (Y.W.); (J.L.)
- Research Unit of Multimodal Cross Scale Neural Signal Detection and Imaging, Chinese Academy of Medical Science, HUST-Suzhou Institute for Brainsmatics, Jiangsu Industrial Technology Reserch Institute (JITRI), Suzhou 215100, China
| | - Shijie Feng
- Britton Chance Center for Biomedical Photonics and MoE Key Laboratory for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan 430074, China; (L.M.); (M.H.); (Y.W.); (J.L.)
- Research Unit of Multimodal Cross Scale Neural Signal Detection and Imaging, Chinese Academy of Medical Science, HUST-Suzhou Institute for Brainsmatics, Jiangsu Industrial Technology Reserch Institute (JITRI), Suzhou 215100, China
| | - Yiqian Wang
- Britton Chance Center for Biomedical Photonics and MoE Key Laboratory for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan 430074, China; (L.M.); (M.H.); (Y.W.); (J.L.)
- Research Unit of Multimodal Cross Scale Neural Signal Detection and Imaging, Chinese Academy of Medical Science, HUST-Suzhou Institute for Brainsmatics, Jiangsu Industrial Technology Reserch Institute (JITRI), Suzhou 215100, China
| | - Jinling Lu
- Britton Chance Center for Biomedical Photonics and MoE Key Laboratory for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan 430074, China; (L.M.); (M.H.); (Y.W.); (J.L.)
- Research Unit of Multimodal Cross Scale Neural Signal Detection and Imaging, Chinese Academy of Medical Science, HUST-Suzhou Institute for Brainsmatics, Jiangsu Industrial Technology Reserch Institute (JITRI), Suzhou 215100, China
| | - Pengcheng Li
- Britton Chance Center for Biomedical Photonics and MoE Key Laboratory for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan 430074, China; (L.M.); (M.H.); (Y.W.); (J.L.)
- Research Unit of Multimodal Cross Scale Neural Signal Detection and Imaging, Chinese Academy of Medical Science, HUST-Suzhou Institute for Brainsmatics, Jiangsu Industrial Technology Reserch Institute (JITRI), Suzhou 215100, China
- Department of Biomedical Engineering, Hainan University, Haikou 570228, China
| |
Collapse
|
15
|
Wang X, Liisberg MB, Vonlehmden GL, Fu X, Cerretani C, Li L, Johnson LA, Vosch T, Richards CI. DNA-AgNC Loaded Liposomes for Measuring Cerebral Blood Flow Using Two-Photon Fluorescence Correlation Spectroscopy. ACS NANO 2023; 17:12862-12874. [PMID: 37341451 PMCID: PMC11065323 DOI: 10.1021/acsnano.3c04489] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/22/2023]
Abstract
Unraveling the transport of drugs and nanocarriers in cerebrovascular networks is important for pharmacokinetic and hemodynamic studies but is challenging due to the complexity of sensing individual particles within the circulatory system of a live animal. Here, we demonstrate that a DNA-stabilized silver nanocluster (DNA-Ag16NC) that emits in the first near-infrared window upon two-photon excitation in the second NIR window can be used for multiphoton in vivo fluorescence correlation spectroscopy for the measurement of cerebral blood flow rates in live mice with high spatial and temporal resolution. To ensure bright and stable emission during in vivo experiments, we loaded DNA-Ag16NCs into liposomes, which served the dual purposes of concentrating the fluorescent label and protecting it from degradation. DNA-Ag16NC-loaded liposomes enabled the quantification of cerebral blood flow velocities within individual vessels of a living mouse.
Collapse
Affiliation(s)
- Xiaojin Wang
- Department of Chemistry, University of Kentucky, Lexington, Kentucky 40506, United States
| | - Mikkel B. Liisberg
- Department of Chemistry, University of Copenhagen, Universitetsparken 5, DK-2100 Copenhagen, Denmark
| | - Georgia L. Vonlehmden
- Department of Physiology, University of Kentucky, Lexington, Kentucky 40536, United States
| | - Xu Fu
- Light Microscopy Core, University of Kentucky, Lexington, Kentucky 40536, United States
| | - Cecilia Cerretani
- Department of Chemistry, University of Copenhagen, Universitetsparken 5, DK-2100 Copenhagen, Denmark
| | - Lan Li
- Department of Chemistry, University of Kentucky, Lexington, Kentucky 40506, United States
| | - Lance A. Johnson
- Department of Physiology, University of Kentucky, Lexington, Kentucky 40536, United States
- Sanders Brown Center on Aging, University of Kentucky, Lexington, Kentucky 40508, United States
| | - Tom Vosch
- Department of Chemistry, University of Copenhagen, Universitetsparken 5, DK-2100 Copenhagen, Denmark
- Nanoscience Center, University of Copenhagen, Universitetsparken 5, DK-2100 Copenhagen, Denmark
| | | |
Collapse
|
16
|
Guo Y, Wang L, Luo Z, Zhu Y, Gao X, Weng X, Wang Y, Yan W, Qu J. Dynamic Volumetric Imaging of Mouse Cerebral Blood Vessels In Vivo with an Ultralong Anti-Diffracting Beam. Molecules 2023; 28:4936. [PMID: 37446598 DOI: 10.3390/molecules28134936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Revised: 06/19/2023] [Accepted: 06/21/2023] [Indexed: 07/15/2023] Open
Abstract
Volumetric imaging of a mouse brain in vivo with one-photon and two-photon ultralong anti-diffracting (UAD) beam illumination was performed. The three-dimensional (3D) structure of blood vessels in the mouse brain were mapped to a two-dimensional (2D) image. The speed of volumetric imaging was significantly improved due to the long focal length of the UAD beam. Comparing one-photon and two-photon UAD beam volumetric imaging, we found that the imaging depth of two-photon volumetric imaging (80 μm) is better than that of one-photon volumetric imaging (60 μm), and the signal-to-background ratio (SBR) of two-photon volumetric imaging is two times that of one-photon volumetric imaging. Therefore, we used two-photon UAD volumetric imaging to perform dynamic volumetric imaging of mouse brain blood vessels in vivo, and obtained the blood flow velocity.
Collapse
Affiliation(s)
- Yong Guo
- State Key Laboratory of Radio Frequency Heterogeneous Integration (Shenzhen University), College of Physics and Optoelectronic Engineering, Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, Shenzhen University, Shenzhen 518060, China
| | - Luwei Wang
- State Key Laboratory of Radio Frequency Heterogeneous Integration (Shenzhen University), College of Physics and Optoelectronic Engineering, Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, Shenzhen University, Shenzhen 518060, China
| | - Ziyi Luo
- State Key Laboratory of Radio Frequency Heterogeneous Integration (Shenzhen University), College of Physics and Optoelectronic Engineering, Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, Shenzhen University, Shenzhen 518060, China
| | - Yinru Zhu
- State Key Laboratory of Radio Frequency Heterogeneous Integration (Shenzhen University), College of Physics and Optoelectronic Engineering, Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, Shenzhen University, Shenzhen 518060, China
| | - Xinwei Gao
- State Key Laboratory of Radio Frequency Heterogeneous Integration (Shenzhen University), College of Physics and Optoelectronic Engineering, Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, Shenzhen University, Shenzhen 518060, China
| | - Xiaoyu Weng
- State Key Laboratory of Radio Frequency Heterogeneous Integration (Shenzhen University), College of Physics and Optoelectronic Engineering, Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, Shenzhen University, Shenzhen 518060, China
| | - Yiping Wang
- State Key Laboratory of Radio Frequency Heterogeneous Integration (Shenzhen University), College of Physics and Optoelectronic Engineering, Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, Shenzhen University, Shenzhen 518060, China
| | - Wei Yan
- State Key Laboratory of Radio Frequency Heterogeneous Integration (Shenzhen University), College of Physics and Optoelectronic Engineering, Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, Shenzhen University, Shenzhen 518060, China
| | - Junle Qu
- State Key Laboratory of Radio Frequency Heterogeneous Integration (Shenzhen University), College of Physics and Optoelectronic Engineering, Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, Shenzhen University, Shenzhen 518060, China
| |
Collapse
|
17
|
Zhu L, Wang M, Liu Y, Fu P, Zhang W, Zhang H, Roe AW, Xi W. Single-microvessel occlusion produces lamina-specific microvascular flow vasodynamics and signs of neurodegenerative change. Cell Rep 2023; 42:112469. [PMID: 37141094 DOI: 10.1016/j.celrep.2023.112469] [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: 04/15/2022] [Revised: 01/12/2023] [Accepted: 04/18/2023] [Indexed: 05/05/2023] Open
Abstract
Recent studies have highlighted the importance of understanding the architecture and function of microvasculature, and dysfunction of these microvessels may underlie neurodegenerative disease. Here, we utilize a high-precision ultrafast laser-induced photothrombosis (PLP) method to occlude single capillaries and then quantitatively study the effects on vasodynamics and surrounding neurons. Analysis of the microvascular architecture and hemodynamics after single-capillary occlusion reveals distinct changes upstream vs. downstream branches, which shows rapid regional flow redistribution and local downstream blood-brain barrier (BBB) leakage. Focal ischemia via capillary occlusions surrounding labeled target neurons induces dramatic and rapid lamina-specific changes in neuronal dendritic architecture. Further, we find that micro-occlusion at two different depths within the same vascular arbor results in distinct effects on flow profiles in layers 2/3 vs layer 4. The current results reveal laminar-scale regulation distinctions in microinfarct response and raise the possibility that relatively greater impacts on microvascular function contribute to cognitive decline in neurodegenerative disease.
Collapse
Affiliation(s)
- Liang Zhu
- Interdisciplinary Institute of Neuroscience and Technology (ZIINT), The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310020, China; Interdisciplinary Institute of Neuroscience and Technology (ZIINT), College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou 310027, China
| | - Mengqi Wang
- Interdisciplinary Institute of Neuroscience and Technology (ZIINT), The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310020, China
| | - Yin Liu
- Interdisciplinary Institute of Neuroscience and Technology (ZIINT), The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310020, China
| | - Peng Fu
- Interdisciplinary Institute of Neuroscience and Technology (ZIINT), The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310020, China
| | - Weijie Zhang
- Interdisciplinary Institute of Neuroscience and Technology (ZIINT), College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou 310027, China
| | - Hequn Zhang
- Interdisciplinary Institute of Neuroscience and Technology (ZIINT), The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310020, China
| | - Anna Wang Roe
- Interdisciplinary Institute of Neuroscience and Technology (ZIINT), The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310020, China; MOE Frontier Science Center for Brain Research and Brain Machine Integration, Zhejiang University, Hangzhou 310058, China; Key Laboratory of Biomedical Engineering of the Ministry of Education, College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou 310027, China.
| | - Wang Xi
- Interdisciplinary Institute of Neuroscience and Technology (ZIINT), The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310020, China; MOE Frontier Science Center for Brain Research and Brain Machine Integration, Zhejiang University, Hangzhou 310058, China; Key Laboratory of Biomedical Engineering of the Ministry of Education, College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou 310027, China.
| |
Collapse
|
18
|
Qiu B, Zhao Z, Wang N, Feng Z, Chen XJ, Chen W, Sun W, Ge WP, Wang Y. A systematic observation of vasodynamics from different segments along the cerebral vasculature in the penumbra zone of awake mice following cerebral ischemia and recanalization. J Cereb Blood Flow Metab 2023; 43:665-679. [PMID: 36524693 PMCID: PMC10108196 DOI: 10.1177/0271678x221146128] [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: 08/18/2022] [Revised: 11/09/2022] [Accepted: 11/14/2022] [Indexed: 12/23/2022]
Abstract
Different segments of the cerebral vascular network may react distinctly to brain ischemia and recanalization. However, there are limited systematic observations of these vascular responses in mice under a physiological state following ischemic stroke. Herein, we aimed to investigate the vasodynamics among several segments along the cerebral vessels in awake mice following cerebral ischemia/recanalization via two-photon imaging. Plasma in the blood vessels were labelled with fluorescein isothiocyanate dextran. Smooth muscle cells and pericytes were labelled via a genetic mouse line (PDGFRβ-tdTomato). We observed a no-reflow phenomenon in downstream microcirculation, and the vasodynamics of different segments of larger cerebral vessels varied in the penumbra area following cerebral ischemia-reperfusion. Despite obtaining reperfusion from the middle cerebral artery, there were significant constrictions of the downstream blood vessels in the ischemic penumbra zone. Interestingly, we observed an extensive constriction of the capillaries 3 hours following recanalization, both at the site covered by pericyte soma and by the pericyte process alone. In addition, we did not observe a significant positive correlation between the changed capillary diameter and pericyte coverage along the capillary. Taken together, abnormal constrictions and vasodynamics of cerebral large and small vessels may directly contribute to microcirculation failure following recanalization in ischemic stroke.
Collapse
Affiliation(s)
- Baoshan Qiu
- Department of Neurology, Beijing Tiantan
Hospital, Capital Medical University, Beijing, China
- Chinese Institute for Brain Research,
Beijing, China
| | - Zichen Zhao
- Department of Neurology, Beijing Tiantan
Hospital, Capital Medical University, Beijing, China
- Chinese Institute for Brain Research,
Beijing, China
| | - Nan Wang
- Department of Neurology, Beijing Tiantan
Hospital, Capital Medical University, Beijing, China
- Chinese Institute for Brain Research,
Beijing, China
| | - Ziyan Feng
- Chinese Institute for Brain Research,
Beijing, China
| | - Xing-jun Chen
- Chinese Institute for Brain Research,
Beijing, China
- Academy for Advanced Interdisciplinary
Studies (AAIS), Peking University, Beijing, China
| | - Weiqi Chen
- Department of Neurology, Beijing Tiantan
Hospital, Capital Medical University, Beijing, China
| | - Wenzhi Sun
- Chinese Institute for Brain Research,
Beijing, China
- School of Basic Medical Sciences, Capital
Medical University, Beijing, China
| | - Woo-ping Ge
- Chinese Institute for Brain Research,
Beijing, China
| | - Yilong Wang
- Department of Neurology, Beijing Tiantan
Hospital, Capital Medical University, Beijing, China
- Chinese Institute for Brain Research,
Beijing, China
- China National Clinical Research Centre for
Neurological Diseases, Beijing, China
- Advanced Innovation Centre for Human Brain
Protection, Capital Medical University, Beijing, China
- National Centre for Neurological Diseases,
Beijing, China
| |
Collapse
|
19
|
Zhu L, Wang M, Fu P, Liu Y, Zhang H, Roe AW, Xi W. Precision 1070 nm Ultrafast Laser-Induced Photothrombosis of Depth-Targeted Vessels In Vivo. SMALL METHODS 2023; 7:e2200917. [PMID: 36286988 DOI: 10.1002/smtd.202200917] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 09/09/2022] [Indexed: 06/16/2023]
Abstract
The cerebrovasculature plays an essential role in neurovascular and homeostatic functions in health and disease conditions. Many efforts have been made for developing vascular thrombosis methods to study vascular dysfunction in vivo, while technical challenges remain, such as accuracy and depth-selectivity to target a single vessel in the cerebral cortex. Herein, this paper first demonstrates the evaluation and quantification of the feasibility and effects of Rose Bengal (RB)-induced photothrombosis with 720-1070 nm ultrafast lasers in a raster scan. A flexible and reproducible approach is then proposed to employ a 1070 nm ultrafast laser with a spiral scan for producing RB-induced occlusion, which is described as precision ultrafast laser-induced photothrombosis (PLP). Combine with two-photon microscopy imaging, this PLP displays highly precise and fast occlusion induction of various vessel types, sizes, and depths, which enhances the precision and power of the photothrombosis protocol. Overall, the PLP method provides a real-time, practical, precise, and depth-selected single-vessel photothrombosis technology in the cerebral cortex with commercially available optical equipment, which is crucial for exploring brain vascular function with high spatial-temporal resolution in the brain.
Collapse
Affiliation(s)
- Liang Zhu
- Interdisciplinary Institute of Neuroscience and Technology (ZIINT), the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310020, China
- Interdisciplinary Institute of Neuroscience and Technology (ZIINT), College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou, 310027, China
| | - Mengqi Wang
- Interdisciplinary Institute of Neuroscience and Technology (ZIINT), the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310020, China
| | - Peng Fu
- Interdisciplinary Institute of Neuroscience and Technology (ZIINT), the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310020, China
| | - Yin Liu
- Interdisciplinary Institute of Neuroscience and Technology (ZIINT), the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310020, China
| | - Hequn Zhang
- Interdisciplinary Institute of Neuroscience and Technology (ZIINT), the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310020, China
| | - Anna Wang Roe
- Interdisciplinary Institute of Neuroscience and Technology (ZIINT), the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310020, China
- MOE Frontier Science Center for Brain Research and Brain Machine Integration, Zhejiang University, Hangzhou, 310058, China
- Key Laboratory of Biomedical Engineering of Ministry of Education, College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou, 310027, China
| | - Wang Xi
- Interdisciplinary Institute of Neuroscience and Technology (ZIINT), the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310020, China
- MOE Frontier Science Center for Brain Research and Brain Machine Integration, Zhejiang University, Hangzhou, 310058, China
- Key Laboratory of Biomedical Engineering of Ministry of Education, College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou, 310027, China
| |
Collapse
|
20
|
Torrisi F, Stella G, Guarino FM, Bucolo M. Cell counting and velocity algorithms for hydrodynamic study of unsteady biological flows in micro-channels. BIOMICROFLUIDICS 2023; 17:014105. [PMID: 36714795 PMCID: PMC9878589 DOI: 10.1063/5.0138587] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 12/20/2022] [Indexed: 05/20/2023]
Abstract
In this paper, the combination of two algorithms, a cell counting algorithm and a velocity algorithm based on a Digital Particle Image Velocimetry (DPIV) method, is presented to study the collective behavior of micro-particles in response to hydrodynamic stimuli. A wide experimental campaign was conducted using micro-particles of different natures and diameters (from 5 to 16 μ m ), such as living cells and silica beads. The biological fluids were injected at the inlet of a micro-channel with an external oscillating flow, and the process was monitored in an investigated area, simultaneously, through a CCD camera and a photo-detector. The proposed data analysis procedure is based on the DPIV-based algorithm to extrapolate the micro-particles velocities and a custom counting algorithm to obtain the instantaneous micro-particles number. The counting algorithm was easily integrated with the DPIV-based algorithm, to automatically run the analysis to different videos and to post-process the results in time and frequency domain. The performed experiments highlight the difference in the micro-particles hydrodynamic responses to external stimuli and the possibility to associate them with the micro-particles physical properties. Furthermore, in order to overcome the hardware and software requirements for the development of a real-time approach, it was also investigated the possibility to detect the flows by photo-detector signals as an alternative to camera acquisition. The photo-detector signals were compared with the velocity trends as a proof of concept for further simplification and speed-up of the data acquisition and analysis. The algorithm flexibility underlines the potential of the proposed methodology to be suitable for real-time detection in embedded systems.
Collapse
Affiliation(s)
- Federica Torrisi
- Department of Electrical, Electronic and Computer Engineering, University of Catania, 95125 Catania, Italy
| | - Giovanna Stella
- Department of Electrical, Electronic and Computer Engineering, University of Catania, 95125 Catania, Italy
| | - Francesca M. Guarino
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95125 Catania, Italy
| | - Maide Bucolo
- Department of Electrical, Electronic and Computer Engineering, University of Catania, 95125 Catania, Italy
| |
Collapse
|
21
|
Cheng H, Chen X, Zhong J, Li J, Qiu P, Wang K. Label-free measurement of wall shear stress in the brain venule and arteriole using dual-wavelength third-harmonic-generation line-scanning imaging. OPTICS LETTERS 2022; 47:5618-5621. [PMID: 37219285 DOI: 10.1364/ol.472136] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 10/06/2022] [Indexed: 05/24/2023]
Abstract
Wall shear stress (WSS) is of fundamental physiological and pathological significance. Current measurement technologies suffer from poor spatial resolution or cannot measure instantaneous values in a label-free manner. Here we demonstrate dual-wavelength third-harmonic-generation (THG) line-scanning imaging, for instantaneous wall shear rate and WSS measurement in vivo. We used the soliton self-frequency shift to generate dual-wavelength femtosecond pulses. Simultaneous acquisition of dual-wavelength THG line-scanning signals extract blood flow velocities at adjacent radial positions for instantaneous wall shear rate and WSS measurement. Our results show the oscillating behavior of WSS in brain venules and arterioles at micron spatial resolution in a label-free manner.
Collapse
|
22
|
Zhang H, Fu P, Liu Y, Zheng Z, Zhu L, Wang M, Abdellah M, He M, Qian J, Roe AW, Xi W. Large-depth three-photon fluorescence microscopy imaging of cortical microvasculature on nonhuman primates with bright AIE probe In vivo. Biomaterials 2022; 289:121809. [PMID: 36166895 DOI: 10.1016/j.biomaterials.2022.121809] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Revised: 09/08/2022] [Accepted: 09/13/2022] [Indexed: 11/02/2022]
Abstract
Multiphoton microscopy has been a powerful tool in brain research, three-photon fluorescence microscopy is increasingly becoming an emerging technique for neurological research of the cortex in depth. Nonhuman primates play important roles in the study of brain science because of their neural and vascular similarity to humans. However, there are few research results of three-photon fluorescence microscopy on the brain of nonhuman primates due to the lack of optimized imaging systems and excellent fluorescent probes. Here we introduced a bright aggregation-induced emission (AIE) probe with excellent three-photon fluorescence efficiency as well as facile synthesis process and we validated its biocompatibility in the macaque monkey. We achieved a large-depth vascular imaging of approximately 1 mm in the cerebral cortex of macaque monkey with our lab-modified three-photon fluorescence microscopy system and the AIE probe. Functional measurement of blood velocity in deep cortex capillaries was also performed. Furthermore, the comparison of cortical deep vascular structure parameters across species was presented on the monkey and mouse cortex. This work is the first in vivo three-photon fluorescence microscopic imaging research on the macaque monkey cortex reaching the imaging depth of ∼1 mm with the bright AIE probe. The results demonstrate the potential of three-photon microscopy as primate-compatible method for imaging fine vascular networks and will advance our understanding of vascular function in normal and disease in humans.
Collapse
Affiliation(s)
- Hequn Zhang
- Interdisciplinary Institute of Neuroscience and Technology (ZIINT), Department of Anesthesiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310020, China; MOE Frontier Science Center for Brain Research and Brain Machine Integration, Zhejiang University, Hangzhou, 310058, China; State Key Laboratory of Modern Optical Instrumentations, Centre for Optical and Electromagnetic Research, College of Optical Science and Engineering, International Research Center for Advanced Photonics, Zhejiang University, Hangzhou, 310058, China
| | - Peng Fu
- Interdisciplinary Institute of Neuroscience and Technology (ZIINT), Department of Anesthesiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310020, China; MOE Frontier Science Center for Brain Research and Brain Machine Integration, Zhejiang University, Hangzhou, 310058, China
| | - Yin Liu
- Interdisciplinary Institute of Neuroscience and Technology (ZIINT), Department of Anesthesiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310020, China; MOE Frontier Science Center for Brain Research and Brain Machine Integration, Zhejiang University, Hangzhou, 310058, China
| | - Zheng Zheng
- School of Chemistry and Chemical Engineering, Hefei University of Technology, Hefei, 230009, China
| | - Liang Zhu
- Interdisciplinary Institute of Neuroscience and Technology (ZIINT), College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou, 310027, China
| | - Mengqi Wang
- Interdisciplinary Institute of Neuroscience and Technology (ZIINT), Department of Anesthesiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310020, China; MOE Frontier Science Center for Brain Research and Brain Machine Integration, Zhejiang University, Hangzhou, 310058, China
| | - Marwan Abdellah
- Blue Brain Project (BBP), École Polytechnique Fédérale de Lausanne (EPFL), Campus Biotech, 1202, Geneva, Switzerland
| | - Mubin He
- State Key Laboratory of Modern Optical Instrumentations, Centre for Optical and Electromagnetic Research, College of Optical Science and Engineering, International Research Center for Advanced Photonics, Zhejiang University, Hangzhou, 310058, China
| | - Jun Qian
- State Key Laboratory of Modern Optical Instrumentations, Centre for Optical and Electromagnetic Research, College of Optical Science and Engineering, International Research Center for Advanced Photonics, Zhejiang University, Hangzhou, 310058, China.
| | - Anna Wang Roe
- Interdisciplinary Institute of Neuroscience and Technology (ZIINT), Department of Anesthesiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310020, China; MOE Frontier Science Center for Brain Research and Brain Machine Integration, Zhejiang University, Hangzhou, 310058, China
| | - Wang Xi
- Interdisciplinary Institute of Neuroscience and Technology (ZIINT), Department of Anesthesiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310020, China; MOE Frontier Science Center for Brain Research and Brain Machine Integration, Zhejiang University, Hangzhou, 310058, China; Key Laboratory of Biomedical Engineering of Ministry of Education, College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou, 310027, China
| |
Collapse
|
23
|
Zhang H, Zhu L, Gao DS, Liu Y, Zhang J, Yan M, Qian J, Xi W. Imaging the Deep Spinal Cord Microvascular Structure and Function with High-Speed NIR-II Fluorescence Microscopy. SMALL METHODS 2022; 6:e2200155. [PMID: 35599368 DOI: 10.1002/smtd.202200155] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 04/23/2022] [Indexed: 06/15/2023]
Abstract
The spinal cord (SC) is crucial for a myriad of somatosensory, autonomic signal processing, and transductions. Understanding the SC vascular structure and function thus plays an integral part in neuroscience and clinical research. However, the dense layers of myelinated ascending axons on the dorsal side inconveniently grant the SC tissue with high optical scattering property, which significantly hinders the imaging depth of the SC vasculature in vivo. Commonly used antiscattering techniques such as multiphoton fluorescence microscopy have low imaging speed and cannot capture the rapid vascular particle flow without significant motion blur. Here, advantage of the high penetration of near-infrared (NIR)-II fluorescence is taken to demonstrate a deep SC vascular structural image stack up to 350 µm, comparable to two-photon microscopy. Furthermore, the red blood cells are labelled with the clinically approved NIR dye indocyanine. The combination of a fast NIR camera and indocyanine green-red blood cells (RBCs) makes it possible to attain high-speed 100 frame-per-second NIR-II imaging to identify the corresponding changes in RBC velocity during the external hind leg stimulus. For the first time, it is established that the NIR-II region would be a promising spectral window for SC imaging. NIR-II fluorescence microscopy has excellent potential for clinical and basic science research on SC.
Collapse
Affiliation(s)
- Hequn Zhang
- Interdisciplinary Institute of Neuroscience and Technology (ZIINT), Department of Anesthesiology, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310020, China
- MOE Frontier Science Center for Brain Research and Brain Machine Integration, Zhejiang University, Hangzhou, 310058, China
| | - Liang Zhu
- Interdisciplinary Institute of Neuroscience and Technology (ZIINT), Department of Anesthesiology, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310020, China
- MOE Frontier Science Center for Brain Research and Brain Machine Integration, Zhejiang University, Hangzhou, 310058, China
- Interdisciplinary Institute of Neuroscience and Technology (ZIINT), College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou, 310027, China
| | - Dave Schwinn Gao
- Key Laboratory of The Diagnosis and Treatment of Severe Trauma and Burn of Zhejiang Province, Department of Anesthesiology, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Yin Liu
- Interdisciplinary Institute of Neuroscience and Technology (ZIINT), Department of Anesthesiology, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310020, China
- MOE Frontier Science Center for Brain Research and Brain Machine Integration, Zhejiang University, Hangzhou, 310058, China
| | - Jun Zhang
- Department of Spine Surgery, Zhejiang Provincial People's Hospital, Hangzhou Medical School People's Hospital, Shangtang Road 158th, Hangzhou, Zhejiang Province, 310014, China
| | - Min Yan
- Key Laboratory of The Diagnosis and Treatment of Severe Trauma and Burn of Zhejiang Province, Department of Anesthesiology, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Jun Qian
- State Key Laboratory of Modern Optical Instrumentations, Centre for Optical and Electromagnetic Research, College of Optical Science and Engineering, Zhejiang University, Hangzhou, 310058, China
| | - Wang Xi
- Interdisciplinary Institute of Neuroscience and Technology (ZIINT), Department of Anesthesiology, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310020, China
- MOE Frontier Science Center for Brain Research and Brain Machine Integration, Zhejiang University, Hangzhou, 310058, China
- Key Laboratory of Biomedical Engineering of Ministry of Education, College of Biomedical Engineering and instrument Science, Zhejiang University, Hangzhou, 310027, China
| |
Collapse
|
24
|
A FACED lift for cerebral blood flow imaging. Proc Natl Acad Sci U S A 2022; 119:e2207474119. [PMID: 35867770 PMCID: PMC9282448 DOI: 10.1073/pnas.2207474119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
25
|
Coelho-Santos V, Tieu T, Shih AY. Reinforced thinned-skull window for repeated imaging of the neonatal mouse brain. NEUROPHOTONICS 2022; 9:031918. [PMID: 35673538 PMCID: PMC9163199 DOI: 10.1117/1.nph.9.3.031918] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 05/03/2022] [Indexed: 06/15/2023]
Abstract
Significance: Two-photon microscopy is a powerful tool for in vivo imaging of the mammalian brain at cellular to subcellular resolution. However, resources that describe methods for imaging live newborn mice have remained sparse. Aim: We describe a non-invasive cranial window procedure for longitudinal imaging of neonatal mice. Approach: We demonstrate construction of the cranial window by iterative shaving of the calvarium of P0 to P12 mouse pups. We use the edge of a syringe needle and scalpel blades to thin the bone to ∼ 15 - μ m thickness. The window is then reinforced with cyanoacrylate glue and a coverslip to promote stability and optical access for at least a week. The head cap also includes a light-weight aluminum flange for head-fixation during imaging. Results: The resulting chronic thinned-skull window enables in vivo imaging to a typical cortical depth of ∼ 200 μ m without disruption of the intracranial environment. We highlight techniques to measure vascular structure and blood flow during development, including use of intravenous tracers and transgenic mice to label the blood plasma and vascular cell types, respectively. Conclusions: This protocol enables direct visualization of the developing neurogliovascular unit in the live neonatal brain during both normal and pathological states.
Collapse
Affiliation(s)
- Vanessa Coelho-Santos
- Seattle Children’s Research Institute, Center for Developmental Biology and Regenerative Medicine, Seattle, Washington, United States
- University of Washington, Department of Pediatrics, Seattle, Washington, United States
| | - Taryn Tieu
- Seattle Children’s Research Institute, Center for Developmental Biology and Regenerative Medicine, Seattle, Washington, United States
| | - Andy Y. Shih
- Seattle Children’s Research Institute, Center for Developmental Biology and Regenerative Medicine, Seattle, Washington, United States
- University of Washington, Department of Pediatrics, Seattle, Washington, United States
- University of Washington, Department of Bioengineering, Seattle, Washington, United States
| |
Collapse
|
26
|
Ultrafast two-photon fluorescence imaging of cerebral blood circulation in the mouse brain in vivo. Proc Natl Acad Sci U S A 2022; 119:e2117346119. [PMID: 35648820 PMCID: PMC9191662 DOI: 10.1073/pnas.2117346119] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
SignificanceCharacterizing blood flow by tracking individual red blood cells as they move through vessels is essential for understanding vascular function. With high spatial resolution, two-photon fluorescence microscopy is the method of choice for imaging blood flow at the cellular level. However, its application is limited to a low flow speed regimen in anesthetized animals by its slow focus scanning mechanism. Using an ultrafast scanning module, we demonstrated two-photon fluorescence imaging of blood flow at 1,000 two-dimensional frames and 1,000,000 one-dimensional line scans per second in the brains of awake mice. These ultrafast measurements enabled us to study hemodynamic and fluid mechanical regimens beyond the reach of conventional methods.
Collapse
|
27
|
Xu Z, Zhang Z, Deng X, Li J, Jiang Y, Law WC, Yang C, Zhang W, Chen X, Wang K, Wang D, Xu G. Deep-Brain Three-Photon Imaging Enabled by Aggregation-Induced Emission Luminogens with Near-Infrared-III Excitation. ACS NANO 2022; 16:6712-6724. [PMID: 35293713 DOI: 10.1021/acsnano.2c01349] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Understanding the morphology and hemodynamics of cerebral vasculature at large penetration depths and microscale resolution is fundamentally important to decipher brain diseases. Among the various imaging technologies, three-photon (3P) microscopy is of significance by virtue of its deep-penetrating capability and submicron resolution, which especially benefits in vivo vascular imaging. Aggregation-induced emission luminogens (AIEgens) have been recognized to be extraordinarily powerful as 3P probes. However, systematic studies on the structure-performance relationship of 3P AIEgens have been seldom reported. Herein, a series of AIEgens has been designed and synthesized. By intentionally introducing benzene rings onto electron donors (D) and acceptors (A), the molecular distortion, conjugation strength, and the D-A relationship can be facilely manipulated. Upon encapsulation with DSPE-PEG2000, the optimized AIEgens are successfully applied for 3P microscopy with emission in the far-red/near-infrared-I (NIR-I, 700-950 nm) region under the near-infrared-III (NIR-III, 1600-1870 nm) excitation. Impressively, using mice with an opened skull, vasculature within 1700 μm and a microvessel with a diameter of 2.2 μm in deep mouse brain were clearly visualized. In addition, the hemodynamics of blood vessels were well-characterized. Thus, this work not only proposes a molecular design strategy of 3P AIEgens but also promotes the performance of 3P imaging in cerebral vasculature.
Collapse
Affiliation(s)
- Zhourui Xu
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen 518060, China
| | - Zhijun Zhang
- Center for AIE Research, Shenzhen Key Laboratory of Polymer Science and Technology, Guangdong Research Center for Interfacial Engineering of Functional Materials, College of Material Science and Engineering, Shenzhen University, Shenzhen 518060, China
| | - Xiangquan Deng
- Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, China
| | - Jiangao Li
- Center for AIE Research, Shenzhen Key Laboratory of Polymer Science and Technology, Guangdong Research Center for Interfacial Engineering of Functional Materials, College of Material Science and Engineering, Shenzhen University, Shenzhen 518060, China
| | - Yihang Jiang
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen 518060, China
| | - Wing-Cheung Law
- Department of Industrial and Systems Engineering, The Hong Kong Polytechnic University, Hong Kong
| | - Chengbin Yang
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen 518060, China
| | - Wanjian Zhang
- Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, China
| | - Xiaolin Chen
- Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, China
| | - Ke Wang
- Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, China
| | - Dong Wang
- Center for AIE Research, Shenzhen Key Laboratory of Polymer Science and Technology, Guangdong Research Center for Interfacial Engineering of Functional Materials, College of Material Science and Engineering, Shenzhen University, Shenzhen 518060, China
| | - Gaixia Xu
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen 518060, China
| |
Collapse
|
28
|
Chicana B, Abbasizadeh N, Burns C, Taglinao H, Spencer JA, Manilay JO. Deletion of Vhl in Dmp1-Expressing Cells Causes Microenvironmental Impairment of B Cell Lymphopoiesis. Front Immunol 2022; 13:780945. [PMID: 35250971 PMCID: PMC8889104 DOI: 10.3389/fimmu.2022.780945] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 01/11/2022] [Indexed: 12/25/2022] Open
Abstract
The contributions of skeletal cells to the processes of B cell development in the bone marrow (BM) have not been completely described. The von-Hippel Lindau protein (VHL) plays a key role in cellular responses to hypoxia. Previous work showed that Dmp1-Cre;Vhl conditional knockout mice (VhlcKO), which deletes Vhl in subsets of mesenchymal stem cells, late osteoblasts and osteocytes, display dysregulated bone growth and reduction in B cells. Here, we investigated the mechanisms underlying the B cell defects using flow cytometry and high-resolution imaging. In the VhlcKO BM, B cell progenitors were increased in frequency and number, whereas Hardy Fractions B-F were decreased. VhlcKO Fractions B-C cells showed increased apoptosis and quiescence. Reciprocal BM chimeras confirmed a B cell-extrinsic source of the VhlcKO B cell defects. In support of this, VhlcKO BM supernatant contained reduced CXCL12 and elevated EPO levels. Intravital and ex vivo imaging revealed VhlcKO BM blood vessels with increased diameter, volume, and a diminished blood-BM barrier. Staining of VhlcKO B cells with an intracellular hypoxic marker indicated the natural existence of distinct B cell microenvironments that differ in local oxygen tensions and that the B cell developmental defects in VhlcKO BM are not initiated by hypoxia. Our studies identify novel mechanisms linking altered bone homeostasis with drastic BM microenvironmental changes that dysregulate B cell development.
Collapse
Affiliation(s)
- Betsabel Chicana
- Department of Molecular and Cell Biology, School of Natural Sciences, University of California, Merced, Merced, CA, United States.,Quantitative and Systems Biology Graduate Program, University of California, Merced, Merced, CA, United States
| | - Nastaran Abbasizadeh
- Quantitative and Systems Biology Graduate Program, University of California, Merced, Merced, CA, United States.,Department of Bioengineering, School of Engineering, University of California, Merced, Merced, CA, United States
| | - Christian Burns
- Quantitative and Systems Biology Graduate Program, University of California, Merced, Merced, CA, United States.,Department of Bioengineering, School of Engineering, University of California, Merced, Merced, CA, United States
| | - Hanna Taglinao
- Department of Molecular and Cell Biology, School of Natural Sciences, University of California, Merced, Merced, CA, United States
| | - Joel A Spencer
- Quantitative and Systems Biology Graduate Program, University of California, Merced, Merced, CA, United States.,Department of Bioengineering, School of Engineering, University of California, Merced, Merced, CA, United States.,Bioengineering Graduate Program, University of California, Merced, Merced, CA, United States
| | - Jennifer O Manilay
- Department of Molecular and Cell Biology, School of Natural Sciences, University of California, Merced, Merced, CA, United States.,Quantitative and Systems Biology Graduate Program, University of California, Merced, Merced, CA, United States
| |
Collapse
|
29
|
Chaigneau E, Charpak S. Measurement of Blood Velocity With Laser Scanning Microscopy: Modeling and Comparison of Line-Scan Image-Processing Algorithms. Front Physiol 2022; 13:848002. [PMID: 35464098 PMCID: PMC9022085 DOI: 10.3389/fphys.2022.848002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Accepted: 02/22/2022] [Indexed: 11/13/2022] Open
Abstract
Laser scanning microscopy is widely used to measure blood hemodynamics with line-scans in physiological and pathological vessels. With scans of broken lines, i.e., lines made of several segments with different orientations, it also allows simultaneous monitoring of vessel diameter dynamics or the activity of specific cells. Analysis of red blood cell (RBC) velocity from line-scans requires specific image-processing algorithms, as angle measurements, Line-Scanning Particle Image Velocimetry (LSPIV) or Fourier transformation of line-scan images. The conditions under which these image-processing algorithms give accurate measurements have not been fully characterized although the accuracy of measurements vary according to specific experimental parameters: the vessel type, the RBC velocity, the scanning parameters, and the image signal to noise ratio. Here, we developed mathematical models for the three previously mentioned line-scan image-processing algorithms. Our models predict the experimental conditions in which RBC velocity measurements are accurate. We illustrate the case of different vessel types and give the parameter space available for each of them. Last, we developed a software generating artificial line-scan images and used it to validate our models.
Collapse
Affiliation(s)
- Emmanuelle Chaigneau
- Institut de la Vision, INSERM U968, Paris, France
- Institut de la Vision, CNRS UMR 7210, Paris, France
- Institut de la Vision, Sorbonne Université, Paris, France
- *Correspondence: Emmanuelle Chaigneau,
| | - Serge Charpak
- Institut de la Vision, INSERM U968, Paris, France
- Institut de la Vision, CNRS UMR 7210, Paris, France
- Institut de la Vision, Sorbonne Université, Paris, France
- Serge Charpak,
| |
Collapse
|
30
|
Choe YG, Yoon JH, Joo J, Kim B, Hong SP, Koh GY, Lee DS, Oh WY, Jeong Y. Pericyte Loss Leads to Capillary Stalling Through Increased Leukocyte-Endothelial Cell Interaction in the Brain. Front Cell Neurosci 2022; 16:848764. [PMID: 35360491 PMCID: PMC8962364 DOI: 10.3389/fncel.2022.848764] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 02/14/2022] [Indexed: 12/13/2022] Open
Abstract
The neurovascular unit is a functional unit composed of neurons, glial cells, pericytes, and endothelial cells which sustain brain activity. While pericyte is a key component of the neurovascular unit, its role in cerebral blood flow regulation remains elusive. Recently, capillary stalling, which means the transient interruption of microcirculation in capillaries, has been shown to have an outsized impact on microcirculatory changes in several neurological diseases. In this study, we investigated capillary stalling and its possible causes, such as the cerebral endothelial glycocalyx and leukocyte adhesion molecules after depleting pericytes postnatally in mice. Moreover, we investigated hypoxia and gliosis as consequences of capillary stalling. Although there were no differences in the capillary structure and RBC flow, longitudinal optical coherence tomography angiography showed an increased number of stalled segments in capillaries after pericyte loss. Furthermore, the extent of the cerebral endothelial glycocalyx was decreased with increased expression of leukocyte adhesion molecules, suggesting enhanced interaction between leukocytes and endothelial cells. Finally, pericyte loss induced cerebral hypoxia and gliosis. Cumulatively, the results suggest that pericyte loss induces capillary stalling through increased interaction between leukocytes and endothelial cells in the brain.
Collapse
|
31
|
Michau A, Lafont C, Bargi-Souza P, Kemkem Y, Guillou A, Ravier MA, Bertrand G, Varrault A, Fiordelisio T, Hodson DJ, Mollard P, Schaeffer M. Metabolic Stress Impairs Pericyte Response to Optogenetic Stimulation in Pancreatic Islets. Front Endocrinol (Lausanne) 2022; 13:918733. [PMID: 35813647 PMCID: PMC9259887 DOI: 10.3389/fendo.2022.918733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 05/23/2022] [Indexed: 11/13/2022] Open
Abstract
Pancreatic islets are highly vascularized micro-organs ensuring whole body glucose homeostasis. Islet vascular cells play an integral part in sustaining adequate insulin release by beta cells. In particular, recent studies have demonstrated that islet pericytes regulate local blood flow velocity and are required for maintenance of beta cell maturity and function. In addition, increased metabolic demand accompanying obesity alters islet pericyte morphology. Here, we sought to explore the effects of metabolic stress on islet pericyte functional response to stimulation in a mouse model of type 2 diabetes, directly in the pancreas in vivo . We found that high fat diet induced islet pericyte hypertrophy without alterations in basal local blood flow. However, optogenetic stimulation of pericyte activity revealed impaired islet vascular responses, despite increased expression of genes encoding proteins directly or indirectly involved in cell contraction. These findings suggest that metabolic stress impinges upon islet pericyte function, which may contribute to beta cell failure during T2D.
Collapse
Affiliation(s)
- Aurélien Michau
- Institute of Functional Genomics, Univ. Montpellier, CNRS, INSERM, Montpellier, France
| | - Chrystel Lafont
- Institute of Functional Genomics, Univ. Montpellier, CNRS, INSERM, Montpellier, France
| | - Paula Bargi-Souza
- Institute of Functional Genomics, Univ. Montpellier, CNRS, INSERM, Montpellier, France
- Department of Physiology and Biophysics of the Institute of Biological Sciences, Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Yasmine Kemkem
- Institute of Functional Genomics, Univ. Montpellier, CNRS, INSERM, Montpellier, France
| | - Anne Guillou
- Institute of Functional Genomics, Univ. Montpellier, CNRS, INSERM, Montpellier, France
| | - Magalie A. Ravier
- Institute of Functional Genomics, Univ. Montpellier, CNRS, INSERM, Montpellier, France
| | - Gyslaine Bertrand
- Institute of Functional Genomics, Univ. Montpellier, CNRS, INSERM, Montpellier, France
| | - Annie Varrault
- Institute of Functional Genomics, Univ. Montpellier, CNRS, INSERM, Montpellier, France
| | - Tatiana Fiordelisio
- Institute of Functional Genomics, Univ. Montpellier, CNRS, INSERM, Montpellier, France
- Laboratorio de Neuroendocrinología Comparada, Laboratorio Nacional de Soluciones Biomiméticas para Diagnóstico y Terapia LaNSBioDyT, Facultad de Ciencias, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - David J. Hodson
- Oxford Centre for Diabetes, Endocrinology and Metabolism (OCDEM), National Institute for Health and Care Research (NIHR) Oxford Biomedical Research Centre, Churchill Hospital, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Patrice Mollard
- Institute of Functional Genomics, Univ. Montpellier, CNRS, INSERM, Montpellier, France
| | - Marie Schaeffer
- Institute of Functional Genomics, Univ. Montpellier, CNRS, INSERM, Montpellier, France
- Centre de Biologie Structurale, CNRS UMR 5048, INSERM U1054, Univ Montpellier, Montpellier, France
- *Correspondence: Marie Schaeffer,
| |
Collapse
|
32
|
Bazzigaluppi P, Mester J, Joo IL, Weisspapir I, Dorr A, Koletar MM, Beckett TL, Khosravani H, Carlen P, Stefanovic B. Frequency selective neuronal modulation triggers spreading depolarizations in the rat endothelin-1 model of stroke. J Cereb Blood Flow Metab 2021; 41:2756-2768. [PMID: 33969731 PMCID: PMC8504421 DOI: 10.1177/0271678x211013656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Ischemia is one of the most common causes of acquired brain injury. Central to its noxious sequelae are spreading depolarizations (SDs), waves of persistent depolarizations which start at the location of the flow obstruction and expand outwards leading to excitotoxic damage. The majority of acute stage of stroke studies to date have focused on the phenomenology of SDs and their association with brain damage. In the current work, we investigated the role of peri-injection zone pyramidal neurons in triggering SDs by optogenetic stimulation in an endothelin-1 rat model of focal ischemia. Our concurrent two photon fluorescence microscopy data and local field potential recordings indicated that a ≥ 60% drop in cortical arteriolar red blood cell velocity was associated with SDs at the ET-1 injection site. SDs were also observed in the peri-injection zone, which subsequently exhibited elevated neuronal activity in the low-frequency bands. Critically, SDs were triggered by low- but not high-frequency optogenetic stimulation of peri-injection zone pyramidal neurons. Our findings depict a complex etiology of SDs post focal ischemia and reveal that effects of neuronal modulation exhibit spectral and spatial selectivity.
Collapse
Affiliation(s)
- Paolo Bazzigaluppi
- Sunnybrook Research Institute, Physical Sciences, Toronto, ON, Canada
- Paolo Bazzigaluppi, Sunnybrook Research Institute, 2075 Bayview Ave., S646, Toronto, ON M4N 3M5, Canada.
| | - James Mester
- Sunnybrook Research Institute, Physical Sciences, Toronto, ON, Canada
- Department of Medical Biophysics, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Illsung L Joo
- Sunnybrook Research Institute, Physical Sciences, Toronto, ON, Canada
| | - Iliya Weisspapir
- Sunnybrook Research Institute, Physical Sciences, Toronto, ON, Canada
| | - Adrienne Dorr
- Sunnybrook Research Institute, Physical Sciences, Toronto, ON, Canada
| | | | - Tina L Beckett
- Sunnybrook Research Institute, Physical Sciences, Toronto, ON, Canada
| | - Houman Khosravani
- Division of Neurology and Interdepartmental Division of Critical Care, Department of Medicine, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON, Canada
- Interdepartmental Division of Critical Care, Department of Medicine, University of Toronto, Toronto, ON, Canada
| | - Peter Carlen
- Krembil Research Institute, University of Toronto, Toronto, ON, Canada
| | - Bojana Stefanovic
- Sunnybrook Research Institute, Physical Sciences, Toronto, ON, Canada
- Department of Medical Biophysics, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
33
|
Wu JW, Jung Y, Yeh SCA, Seo Y, Runnels JM, Burns CS, Mizoguchi T, Ito K, Spencer JA, Lin CP. Intravital fluorescence microscopy with negative contrast. PLoS One 2021; 16:e0255204. [PMID: 34351959 PMCID: PMC8341626 DOI: 10.1371/journal.pone.0255204] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 07/13/2021] [Indexed: 11/23/2022] Open
Abstract
Advances in intravital microscopy (IVM) have enabled the studies of cellular organization and dynamics in the native microenvironment of intact organisms with minimal perturbation. The abilities to track specific cell populations and monitor their interactions have opened up new horizons for visualizing cell biology in vivo, yet the success of standard fluorescence cell labeling approaches for IVM comes with a "dark side" in that unlabeled cells are invisible, leaving labeled cells or structures to appear isolated in space, devoid of their surroundings and lacking proper biological context. Here we describe a novel method for "filling in the void" by harnessing the ubiquity of extracellular (interstitial) fluid and its ease of fluorescence labelling by commonly used vascular and lymphatic tracers. We show that during routine labeling of the vasculature and lymphatics for IVM, commonly used fluorescent tracers readily perfuse the interstitial spaces of the bone marrow (BM) and the lymph node (LN), outlining the unlabeled cells and forming negative contrast images that complement standard (positive) cell labeling approaches. The method is simple yet powerful, offering a comprehensive view of the cellular landscape such as cell density and spatial distribution, as well as dynamic processes such as cell motility and transmigration across the vascular endothelium. The extracellular localization of the dye and the interstitial flow provide favorable conditions for prolonged Intravital time lapse imaging with minimal toxicity and photobleaching.
Collapse
Affiliation(s)
- Juwell W. Wu
- Center for Systems Biology and Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Yookyung Jung
- Center for Systems Biology and Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
- Center for Molecular Spectroscopy and Dynamics, Institute for Basic Science, Seoul, Republic of Korea
| | - Shu-Chi A. Yeh
- Center for Systems Biology and Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Yongwan Seo
- Department of Chemical and Biomolecular Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - Judith M. Runnels
- Center for Systems Biology and Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Christian S. Burns
- Department of Bioengineering, University of California Merced, Merced, California, United States of America
- NSF-CREST Center for Cellular and Biomolecular Machines and the Health Science Research Institute, University of California Merced, Merced, California, United States of America
| | - Toshihide Mizoguchi
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Departments of Cell Biology and Medicine, Albert Einstein College of Medicine, Bronx, New York, United States of America
- Oral Health Science Center, Tokyo Dental College, Tokyo, Japan
| | - Keisuke Ito
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Departments of Cell Biology and Medicine, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Joel A. Spencer
- Department of Bioengineering, University of California Merced, Merced, California, United States of America
- NSF-CREST Center for Cellular and Biomolecular Machines and the Health Science Research Institute, University of California Merced, Merced, California, United States of America
| | - Charles P. Lin
- Center for Systems Biology and Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
| |
Collapse
|
34
|
Glück C, Ferrari KD, Binini N, Keller A, Saab AS, Stobart JL, Weber B. Distinct signatures of calcium activity in brain mural cells. eLife 2021; 10:e70591. [PMID: 34227466 PMCID: PMC8294852 DOI: 10.7554/elife.70591] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Accepted: 06/15/2021] [Indexed: 12/15/2022] Open
Abstract
Pericytes have been implicated in various neuropathologies, yet little is known about their function and signaling pathways in health. Here, we characterized calcium dynamics of cortical mural cells in anesthetized or awake Pdgfrb-CreERT2;Rosa26< LSL-GCaMP6s > mice and in acute brain slices. Smooth muscle cells (SMCs) and ensheathing pericytes (EPs), also named as terminal vascular SMCs, revealed similar calcium dynamics in vivo. In contrast, calcium signals in capillary pericytes (CPs) were irregular, higher in frequency, and occurred in cellular microdomains. In the absence of the vessel constricting agent U46619 in acute slices, SMCs and EPs revealed only sparse calcium signals, whereas CPs retained their spontaneous calcium activity. Interestingly, chemogenetic activation of neurons in vivo and acute elevations of extracellular potassium in brain slices strongly decreased calcium activity in CPs. We propose that neuronal activation and an extracellular increase in potassium suppress calcium activity in CPs, likely mediated by Kir2.2 and KATP channels.
Collapse
Affiliation(s)
- Chaim Glück
- Institute of Pharmacology and Toxicology, University of ZurichZürichSwitzerland
- Neuroscience Center Zurich, University and ETH ZurichZurichSwitzerland
| | - Kim David Ferrari
- Institute of Pharmacology and Toxicology, University of ZurichZürichSwitzerland
- Neuroscience Center Zurich, University and ETH ZurichZurichSwitzerland
| | - Noemi Binini
- Institute of Pharmacology and Toxicology, University of ZurichZürichSwitzerland
- Neuroscience Center Zurich, University and ETH ZurichZurichSwitzerland
| | - Annika Keller
- Neuroscience Center Zurich, University and ETH ZurichZurichSwitzerland
- Department of Neurosurgery, University of ZurichSchlierenSwitzerland
| | - Aiman S Saab
- Institute of Pharmacology and Toxicology, University of ZurichZürichSwitzerland
- Neuroscience Center Zurich, University and ETH ZurichZurichSwitzerland
| | - Jillian L Stobart
- Institute of Pharmacology and Toxicology, University of ZurichZürichSwitzerland
- Rady Faculty of Health Sciences, College of PharmacyWinnipegCanada
| | - Bruno Weber
- Institute of Pharmacology and Toxicology, University of ZurichZürichSwitzerland
- Neuroscience Center Zurich, University and ETH ZurichZurichSwitzerland
| |
Collapse
|
35
|
Abstract
The distribution of blood throughout the brain is facilitated by highly interconnected capillary networks. However, the steps involved in the construction of these networks has remained unclear. We used in vivo two-photon imaging through noninvasive cranial windows to study the engineering of capillary networks in the cerebral cortex of mouse neonates. We find that angiogenic activity originates at ascending venules, which undergo a burst of sprouting in the second postnatal week. This sprouting activity first establishes long paths to connect venules to blood input from neighboring arterioles, and then expands capillary interconnectivity with a multitude of short-range connections. Our study provides an experimental foundation to understand how capillary networks are shaped in the living mammalian brain during postnatal development. Capillary networks are essential for distribution of blood flow through the brain, and numerous other homeostatic functions, including neurovascular signal conduction and blood–brain barrier integrity. Accordingly, the impairment of capillary architecture and function lies at the root of many brain diseases. Visualizing how brain capillary networks develop in vivo can reveal innate programs for cerebrovascular growth and repair. Here, we use longitudinal two-photon imaging through noninvasive thinned skull windows to study a burst of angiogenic activity during cerebrovascular development in mouse neonates. We find that angiogenesis leading to the formation of capillary networks originated exclusively from cortical ascending venules. Two angiogenic sprouting activities were observed: 1) early, long-range sprouts that directly connected venules to upstream arteriolar input, establishing the backbone of the capillary bed, and 2) short-range sprouts that contributed to expansion of anastomotic connectivity within the capillary bed. All nascent sprouts were prefabricated with an intact endothelial lumen and pericyte coverage, ensuring their immediate perfusion and stability upon connection to their target vessels. The bulk of this capillary expansion spanned only 2 to 3 d and contributed to an increase of blood flow during a critical period in cortical development.
Collapse
|
36
|
Mester JR, Bazzigaluppi P, Dorr A, Beckett T, Burke M, McLaurin J, Sled JG, Stefanovic B. Attenuation of tonic inhibition prevents chronic neurovascular impairments in a Thy1-ChR2 mouse model of repeated, mild traumatic brain injury. Am J Cancer Res 2021; 11:7685-7699. [PMID: 34335958 PMCID: PMC8315057 DOI: 10.7150/thno.60190] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Accepted: 06/04/2021] [Indexed: 12/24/2022] Open
Abstract
Rationale: Mild traumatic brain injury (mTBI), the most common type of brain trauma, frequently leads to chronic cognitive and neurobehavioral deficits. Intervening effectively is impeded by our poor understanding of its pathophysiological sequelae. Methods: To elucidate the long-term neurovascular sequelae of mTBI, we combined optogenetics, two-photon fluorescence microscopy, and intracortical electrophysiological recordings in mice to selectively stimulate peri-contusional neurons weeks following repeated closed-head injury and probe individual vessel's function and local neuronal reactivity. Results: Compared to sham-operated animals, mTBI mice showed doubled cortical venular speeds (115 ± 25%) and strongly elevated cortical venular reactivity (53 ± 17%). Concomitantly, the pericontusional neurons exhibited attenuated spontaneous activity (-57 ± 79%) and decreased reactivity (-47 ± 28%). Post-mortem immunofluorescence revealed signs of peri-contusional senescence and DNA damage, in the absence of neuronal loss or gliosis. Alteration of neuronal and vascular functioning was largely prevented by chronic, low dose, systemic administration of a GABA-A receptor inverse agonist (L-655,708), commencing 3 days following the third impact. Conclusions: Our findings indicate that repeated mTBI leads to dramatic changes in the neurovascular unit function and that attenuation of tonic inhibition can prevent these alterations. The sustained disruption of the neurovascular function may underlie the concussed brain's long-term susceptibility to injury, and calls for development of better functional assays as well as of neurovascularly targeted interventions.
Collapse
|
37
|
Regeneration of the neurogliovascular unit visualized in vivo by transcranial live-cell imaging. J Neurosci Methods 2020; 343:108808. [DOI: 10.1016/j.jneumeth.2020.108808] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 06/02/2020] [Accepted: 06/11/2020] [Indexed: 12/15/2022]
|
38
|
Nelson AR, Sagare MA, Wang Y, Kisler K, Zhao Z, Zlokovic BV. Channelrhodopsin Excitation Contracts Brain Pericytes and Reduces Blood Flow in the Aging Mouse Brain in vivo. Front Aging Neurosci 2020; 12:108. [PMID: 32410982 PMCID: PMC7201096 DOI: 10.3389/fnagi.2020.00108] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Accepted: 03/30/2020] [Indexed: 01/02/2023] Open
Abstract
Brains depend on blood flow for the delivery of oxygen and nutrients essential for proper neuronal and synaptic functioning. French physiologist Rouget was the first to describe pericytes in 1873 as regularly arranged longitudinal amoeboid cells on capillaries that have a muscular coat, implying that these are contractile cells that regulate blood flow. Although there have been >30 publications from different groups, including our group, demonstrating that pericytes are contractile cells that can regulate hemodynamic responses in the brain, the role of pericytes in controlling cerebral blood flow (CBF) has not been confirmed by all studies. Moreover, recent studies using different optogenetic models to express light-sensitive channelrhodopsin-2 (ChR2) cation channels in pericytes were not conclusive; one, suggesting that pericytes expressing ChR2 do not contract after light stimulus, and the other, demonstrating contraction of pericytes expressing ChR2 after light stimulus. Since two-photon optogenetics provides a powerful tool to study mechanisms of blood flow regulation at the level of brain capillaries, we re-examined the contractility of brain pericytes in vivo using a new optogenetic model developed by crossing our new inducible pericyte-specific CreER mouse line with ChR2 mice. We induced expression of ChR2 in pericytes with tamoxifen, excited ChR2 by 488 nm light, and monitored pericyte contractility, brain capillary diameter changes, and red blood cell (RBC) velocity in aged mice by in vivo two-photon microscopy. Excitation of ChR2 resulted in pericyte contraction followed by constriction of the underlying capillary leading to approximately an 8% decrease (p = 0.006) in capillary diameter. ChR2 excitation in pericytes substantially reduced capillary RBC flow by 42% (p = 0.03) during the stimulation period compared to the velocity before stimulation. Our data suggests that pericytes contract in vivo and regulate capillary blood flow in the aging mouse brain. By extension, this might have implications for neurological disorders of the aging human brain associated with neurovascular dysfunction and pericyte loss such as stroke and Alzheimer's disease.
Collapse
Affiliation(s)
| | | | | | | | | | - Berislav V. Zlokovic
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, CA, United States
| |
Collapse
|
39
|
Han X, Chai Z, Ping X, Song LJ, Ma C, Ruan Y, Jin X. In vivo Two-Photon Imaging Reveals Acute Cerebral Vascular Spasm and Microthrombosis After Mild Traumatic Brain Injury in Mice. Front Neurosci 2020; 14:210. [PMID: 32210758 PMCID: PMC7077429 DOI: 10.3389/fnins.2020.00210] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2019] [Accepted: 02/25/2020] [Indexed: 12/23/2022] Open
Abstract
Mild traumatic brain injury (mTBI), or concussion, is reported to interfere with cerebral blood flow and microcirculation in patients, but our current understanding is quite limited and the results are often controversial. Here we used longitudinal in vivo two-photon imaging to investigate dynamic changes in cerebral vessels and velocities of red blood cells (RBC) following mTBI. Closed-head mTBI induced using a controlled cortical impact device resulted in a significant reduction of dwell time in a Rotarod test but no significant change in water maze test. Cerebral blood vessels were repeatedly imaged through a thinned skull window at baseline, 0.5, 1, 6 h, and 1 day following mTBI. In both arterioles and capillaries, their diameters and RBC velocities were significantly decreased at 0.5, 1, and 6 h after injury, and recovered in 1 day post-mTBI. In contrast, decreases in the diameter and RBC velocity of venules occurred only in 0.5–1 h after mTBI. We also observed formation and clearance of transient microthrombi in capillaries within 1 h post-mTBI. We concluded that in vivo two-photon imaging is useful for studying earlier alteration of vascular dynamics after mTBI and that mTBI induced reduction of cerebral blood flow, vasospasm, and formation of microthrombi in the acute stage following injury. These changes may contribute to early brain functional deficits of mTBI.
Collapse
Affiliation(s)
- Xinjia Han
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, United States.,Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, IN, United States.,Department of Obstetrics and Gynecology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China.,GHM Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou, China
| | - Zhi Chai
- Neurobiology Research Center, Shanxi Key Laboratory of Innovative Drugs for Serious Illness, College of Basic Medicine, Shaanxi University of Chinese Medicine, Jinzhong, China
| | - Xingjie Ping
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, United States.,Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Li-Juan Song
- Neurobiology Research Center, Shanxi Key Laboratory of Innovative Drugs for Serious Illness, College of Basic Medicine, Shaanxi University of Chinese Medicine, Jinzhong, China
| | - Cungen Ma
- Neurobiology Research Center, Shanxi Key Laboratory of Innovative Drugs for Serious Illness, College of Basic Medicine, Shaanxi University of Chinese Medicine, Jinzhong, China
| | - Yiwen Ruan
- GHM Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou, China.,Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Xiaoming Jin
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, United States.,Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, IN, United States
| |
Collapse
|
40
|
Brondi M, Moroni M, Vecchia D, Molano-Mazón M, Panzeri S, Fellin T. High-Accuracy Detection of Neuronal Ensemble Activity in Two-Photon Functional Microscopy Using Smart Line Scanning. Cell Rep 2020; 30:2567-2580.e6. [PMID: 32101736 PMCID: PMC7043026 DOI: 10.1016/j.celrep.2020.01.105] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 01/10/2020] [Accepted: 01/29/2020] [Indexed: 11/07/2022] Open
Abstract
Two-photon functional imaging using genetically encoded calcium indicators (GECIs) is one prominent tool to map neural activity. Under optimized experimental conditions, GECIs detect single action potentials in individual cells with high accuracy. However, using current approaches, these optimized conditions are never met when imaging large ensembles of neurons. Here, we developed a method that substantially increases the signal-to-noise ratio (SNR) of population imaging of GECIs by using galvanometric mirrors and fast smart line scan (SLS) trajectories. We validated our approach in anesthetized and awake mice on deep and dense GCaMP6 staining in the mouse barrel cortex during spontaneous and sensory-evoked activity. Compared to raster population imaging, SLS led to increased SNR, higher probability of detecting calcium events, and more precise identification of functional neuronal ensembles. SLS provides a cheap and easily implementable tool for high-accuracy population imaging of neural GCaMP6 signals by using galvanometric-based two-photon microscopes.
Collapse
Affiliation(s)
- Marco Brondi
- Optical Approaches to Brain Function Laboratory, Istituto Italiano di Tecnologia, Genova, Italy; Neural Coding Laboratory, Istituto Italiano di Tecnologia, Genova and Rovereto, Italy
| | - Monica Moroni
- Neural Coding Laboratory, Istituto Italiano di Tecnologia, Genova and Rovereto, Italy; Neural Computation Laboratory, Center for Neuroscience and Cognitive Systems @UniTn, Istituto Italiano di Tecnologia, Rovereto, Italy; Center for Mind and Brain Sciences (CIMeC), University of Trento, Trento, Italy
| | - Dania Vecchia
- Optical Approaches to Brain Function Laboratory, Istituto Italiano di Tecnologia, Genova, Italy; Neural Coding Laboratory, Istituto Italiano di Tecnologia, Genova and Rovereto, Italy
| | - Manuel Molano-Mazón
- Neural Coding Laboratory, Istituto Italiano di Tecnologia, Genova and Rovereto, Italy; Neural Computation Laboratory, Center for Neuroscience and Cognitive Systems @UniTn, Istituto Italiano di Tecnologia, Rovereto, Italy
| | - Stefano Panzeri
- Neural Coding Laboratory, Istituto Italiano di Tecnologia, Genova and Rovereto, Italy; Neural Computation Laboratory, Center for Neuroscience and Cognitive Systems @UniTn, Istituto Italiano di Tecnologia, Rovereto, Italy
| | - Tommaso Fellin
- Optical Approaches to Brain Function Laboratory, Istituto Italiano di Tecnologia, Genova, Italy; Neural Coding Laboratory, Istituto Italiano di Tecnologia, Genova and Rovereto, Italy.
| |
Collapse
|
41
|
Arango-Lievano M, Boussadia B, De Terdonck LDT, Gault C, Fontanaud P, Lafont C, Mollard P, Marchi N, Jeanneteau F. Topographic Reorganization of Cerebrovascular Mural Cells under Seizure Conditions. Cell Rep 2019; 23:1045-1059. [PMID: 29694884 DOI: 10.1016/j.celrep.2018.03.110] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Revised: 01/04/2018] [Accepted: 03/22/2018] [Indexed: 12/27/2022] Open
Abstract
Reorganization of the neurovascular unit has been suggested in the epileptic brain, although the dynamics and functional significance remain unclear. Here, we tracked the in vivo dynamics of perivascular mural cells as a function of electroencephalogram (EEG) activity following status epilepticus. We segmented the cortical vascular bed to provide a size- and type-specific analysis of mural cell plasticity topologically. We find that mural cells are added and removed from veins, arterioles, and capillaries after seizure induction. Loss of mural cells is proportional to seizure severity and vascular pathology (e.g., rigidity, perfusion, and permeability). Treatment with platelet-derived growth factor subunits BB (PDGF-BB) reduced mural cell loss, vascular pathology, and epileptiform EEG activity. We propose that perivascular mural cells play a pivotal role in seizures and are potential targets for reducing pathophysiology.
Collapse
Affiliation(s)
- Margarita Arango-Lievano
- Departments of Neuroscience & Physiology, Laboratory of Stress Hormones & Plasticity, Institut de Génomique Fonctionnelle, INSERM, CNRS, University of Montpellier, 34093 Montpellier, France.
| | - Badreddine Boussadia
- Department of Neuroscience, Laboratory of Cerebrovascular Mechanisms of Brain Disorders, Institut de Génomique Fonctionnelle, INSERM, CNRS, University of Montpellier, 34093 Montpellier, France
| | - Lucile Du Trieu De Terdonck
- Departments of Neuroscience & Physiology, Laboratory of Stress Hormones & Plasticity, Institut de Génomique Fonctionnelle, INSERM, CNRS, University of Montpellier, 34093 Montpellier, France
| | - Camille Gault
- Departments of Neuroscience & Physiology, Laboratory of Stress Hormones & Plasticity, Institut de Génomique Fonctionnelle, INSERM, CNRS, University of Montpellier, 34093 Montpellier, France
| | - Pierre Fontanaud
- Department of Physiology, Laboratory of Networks and Rhythms in Endocrine Glands, Institut de Génomique Fonctionnelle, INSERM, CNRS, University of Montpellier, 34093 Montpellier, France
| | - Chrystel Lafont
- Department of Physiology, Laboratory of Networks and Rhythms in Endocrine Glands, Institut de Génomique Fonctionnelle, INSERM, CNRS, University of Montpellier, 34093 Montpellier, France
| | - Patrice Mollard
- Department of Physiology, Laboratory of Networks and Rhythms in Endocrine Glands, Institut de Génomique Fonctionnelle, INSERM, CNRS, University of Montpellier, 34093 Montpellier, France
| | - Nicola Marchi
- Department of Neuroscience, Laboratory of Cerebrovascular Mechanisms of Brain Disorders, Institut de Génomique Fonctionnelle, INSERM, CNRS, University of Montpellier, 34093 Montpellier, France.
| | - Freddy Jeanneteau
- Departments of Neuroscience & Physiology, Laboratory of Stress Hormones & Plasticity, Institut de Génomique Fonctionnelle, INSERM, CNRS, University of Montpellier, 34093 Montpellier, France.
| |
Collapse
|
42
|
Chaigneau E, Roche M, Charpak S. Unbiased Analysis Method for Measurement of Red Blood Cell Size and Velocity With Laser Scanning Microscopy. Front Neurosci 2019; 13:644. [PMID: 31316334 PMCID: PMC6610068 DOI: 10.3389/fnins.2019.00644] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Accepted: 06/05/2019] [Indexed: 12/28/2022] Open
Abstract
Two-photon laser scanning microscopy is widely used to measure blood hemodynamics in brain blood vessels. Still, the algorithms used so far to extract red blood cell (RBC) size and velocity from line-scan acquisitions have ignored the extent to which scanning speed influences the measurements. Here, we used a theoretical approach that takes into account the velocity and direction of both scanning mirrors and RBCs during acquisition to provide an algorithm that measures the real RBC size and velocity. We validate our approach in brain vessels of anesthetized mice, and demonstrate that it corrects online measurement errors that can reach several 10s of percent as well as data previously acquired. To conclude, our analysis allows unbiased comparisons of blood hemodynamic parameters from brain capillaries and large vessels in control and pathological animal models.
Collapse
Affiliation(s)
| | | | - Serge Charpak
- INSERM U1128, Laboratory of Neurophysiology and New Microscopy, Université Paris Descartes, Paris, France
| |
Collapse
|
43
|
Joseph A, Guevara-Torres A, Schallek J. Imaging single-cell blood flow in the smallest to largest vessels in the living retina. eLife 2019; 8:e45077. [PMID: 31084705 PMCID: PMC6516827 DOI: 10.7554/elife.45077] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Accepted: 04/15/2019] [Indexed: 01/15/2023] Open
Abstract
Tissue light scatter limits the visualization of the microvascular network deep inside the living mammal. The transparency of the mammalian eye provides a noninvasive view of the microvessels of the retina, a part of the central nervous system. Despite its clarity, imperfections in the optics of the eye blur microscopic retinal capillaries, and single blood cells flowing within. This limits early evaluation of microvascular diseases that originate in capillaries. To break this barrier, we use 15 kHz adaptive optics imaging to noninvasively measure single-cell blood flow, in one of the most widely used research animals: the C57BL/6J mouse. Measured flow ranged four orders of magnitude (0.0002-1.55 µL min-1) across the full spectrum of retinal vessel diameters (3.2-45.8 µm), without requiring surgery or contrast dye. Here, we describe the ultrafast imaging, analysis pipeline and automated measurement of millions of blood cell speeds.
Collapse
Affiliation(s)
- Aby Joseph
- Institute of OpticsUniversity of RochesterNew YorkUnited States
- Center for Visual ScienceUniversity of RochesterNew YorkUnited States
| | - Andres Guevara-Torres
- Institute of OpticsUniversity of RochesterNew YorkUnited States
- Center for Visual ScienceUniversity of RochesterNew YorkUnited States
| | - Jesse Schallek
- Center for Visual ScienceUniversity of RochesterNew YorkUnited States
- Flaum Eye InstituteUniversity of RochesterNew YorkUnited States
- Department of NeuroscienceUniversity of RochesterNew YorkUnited States
| |
Collapse
|
44
|
In vivo neurovascular response to focused photoactivation of Channelrhodopsin-2. Neuroimage 2019; 192:135-144. [DOI: 10.1016/j.neuroimage.2019.01.036] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Revised: 12/31/2018] [Accepted: 01/14/2019] [Indexed: 01/03/2023] Open
|
45
|
Koizumi K, Hattori Y, Ahn SJ, Buendia I, Ciacciarelli A, Uekawa K, Wang G, Hiller A, Zhao L, Voss HU, Paul SM, Schaffer C, Park L, Iadecola C. Apoε4 disrupts neurovascular regulation and undermines white matter integrity and cognitive function. Nat Commun 2018; 9:3816. [PMID: 30232327 PMCID: PMC6145902 DOI: 10.1038/s41467-018-06301-2] [Citation(s) in RCA: 117] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Accepted: 08/14/2018] [Indexed: 12/12/2022] Open
Abstract
The ApoE4 allele is associated with increased risk of small vessel disease, which is a cause of vascular cognitive impairment. Here, we report that mice with targeted replacement (TR) of the ApoE gene with human ApoE4 have reduced neocortical cerebral blood flow compared to ApoE3-TR mice, an effect due to reduced vascular density rather than slowing of microvascular red blood cell flow. Furthermore, homeostatic mechanisms matching local delivery of blood flow to brain activity are impaired in ApoE4-TR mice. In a model of cerebral hypoperfusion, these cerebrovascular alterations exacerbate damage to the white matter of the corpus callosum and worsen cognitive dysfunction. Using 3-photon microscopy we found that the increased white matter damage is linked to an enhanced reduction of microvascular flow resulting in local hypoxia. Such alterations may be responsible for the increased susceptibility to hypoxic-ischemic lesions in the subcortical white matter of individuals carrying the ApoE4 allele. ApoE4 is a risk factor for small vessel disease, which can lead to cognitive impairment. Here the authors assess the microvasculature of the corpus callosum using 3-photon microscopy and find that mice expressing the ApoE4 allele are more susceptible than wild-type to white matter injury and cognitive impairment in a model of hypoperfusion-induced hypoxia.
Collapse
Affiliation(s)
- Kenzo Koizumi
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, 10065, NY, USA
| | - Yorito Hattori
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, 10065, NY, USA
| | - Sung Ji Ahn
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, 10065, NY, USA
| | - Izaskun Buendia
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, 10065, NY, USA
| | - Antonio Ciacciarelli
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, 10065, NY, USA
| | - Ken Uekawa
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, 10065, NY, USA
| | - Gang Wang
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, 10065, NY, USA
| | - Abigail Hiller
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, 10065, NY, USA
| | - Lingzhi Zhao
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, 10065, NY, USA
| | - Henning U Voss
- Department of Radiology, Weill Cornell Medicine, New York, 10065, NY, USA
| | - Steven M Paul
- Department of Neurology and Psychiatry, Washington University in St. Louis, St. Louis, 63110, MO, USA
| | - Chris Schaffer
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, 10065, NY, USA.,Meinig School of Biomedical Engineering, Cornell University, Ithaca, 14853, NY, USA
| | - Laibaik Park
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, 10065, NY, USA.
| | - Costantino Iadecola
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, 10065, NY, USA.
| |
Collapse
|
46
|
Lasch M, Nekolla K, Klemm AH, Buchheim JI, Pohl U, Dietzel S, Deindl E. Estimating hemodynamic shear stress in murine peripheral collateral arteries by two-photon line scanning. Mol Cell Biochem 2018; 453:41-51. [PMID: 30128948 DOI: 10.1007/s11010-018-3430-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Accepted: 08/16/2018] [Indexed: 12/20/2022]
Abstract
Changes in wall shear stress of blood vessels are assumed to be an important component of many physiological and pathophysiological processes. However, due to technical limitations experimental in vivo data are rarely available. Here, we investigated two-photon excitation fluorescence microscopy as an option to measure vessel diameter as well as blood flow velocities in a murine hindlimb model of arteriogenesis (collateral artery growth). Using line scanning at high frequencies, we measured the movement of blood cells along the vessel axis. We found that peak systolic blood flow velocity averaged 9 mm/s and vessel diameter 42 µm in resting collaterals. Induction of arteriogenesis by femoral artery ligation resulted in a significant increase in centerline peak systolic velocity after 1 day with an average of 51 mm/s, whereas the averaged luminal diameter of collaterals (52 µm) changed much less. Thereof calculations revealed a significant fourfold increase in hemodynamic wall shear rate. Our results indicate that two-photon line scanning is a suitable tool to estimate wall shear stress e.g., in experimental animal models, such as of arteriogenesis, which may not only help to understand the relevance of mechanical forces in vivo, but also to adjust wall shear stress in ex vivo investigations on isolated vessels as well as cell culture experiments.
Collapse
Affiliation(s)
- Manuel Lasch
- Walter-Brendel-Centre of Experimental Medicine, University Hospital, LMU Munich, Marchioninistr.15, 81377, Munich, Germany.,Department of Otorhinolaryngology, Head & Neck Surgery, Klinikum der Universität München, Ludwig- Maximilians-Universität München, Munich, Germany
| | - Katharina Nekolla
- Walter-Brendel-Centre of Experimental Medicine, University Hospital, LMU Munich, Marchioninistr.15, 81377, Munich, Germany
| | - Anna H Klemm
- Walter-Brendel-Centre of Experimental Medicine, University Hospital, LMU Munich, Marchioninistr.15, 81377, Munich, Germany.,Core Facility Bioimaging at the Biomedical Center, LMU Munich, Planegg-Martinsried, Germany
| | - Judith-Irina Buchheim
- Walter-Brendel-Centre of Experimental Medicine, University Hospital, LMU Munich, Marchioninistr.15, 81377, Munich, Germany.,Department of Anesthesiology, Laboratory for Stress and Immunity, Hospital of the University of the LMU Munich, Munich, Germany
| | - Ulrich Pohl
- Walter-Brendel-Centre of Experimental Medicine, University Hospital, LMU Munich, Marchioninistr.15, 81377, Munich, Germany.,Core Facility Bioimaging at the Biomedical Center, LMU Munich, Planegg-Martinsried, Germany.,German Center for Cardiovascular Research, Partner Site Munich Heart Alliance, Munich, Germany
| | - Steffen Dietzel
- Walter-Brendel-Centre of Experimental Medicine, University Hospital, LMU Munich, Marchioninistr.15, 81377, Munich, Germany.,Core Facility Bioimaging at the Biomedical Center, LMU Munich, Planegg-Martinsried, Germany
| | - Elisabeth Deindl
- Walter-Brendel-Centre of Experimental Medicine, University Hospital, LMU Munich, Marchioninistr.15, 81377, Munich, Germany.
| |
Collapse
|
47
|
Gu B, Wang X, Twa MD, Tam J, Girkin CA, Zhang Y. Noninvasive in vivo characterization of erythrocyte motion in human retinal capillaries using high-speed adaptive optics near-confocal imaging. BIOMEDICAL OPTICS EXPRESS 2018; 9:3653-3677. [PMID: 30338146 PMCID: PMC6191635 DOI: 10.1364/boe.9.003653] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Revised: 07/02/2018] [Accepted: 07/02/2018] [Indexed: 05/18/2023]
Abstract
The flow of erythrocytes in parafoveal capillaries was imaged in the living human eye with an adaptive optics near-confocal ophthalmoscope at a frame rate of 800 Hz with a low coherence near-infrared (NIR) light source. Spatiotemporal traces of the erythrocyte movement were extracted from consecutive images. Erythrocyte velocity was measured using custom software based on the Radon transform. The impact of imaging speed on velocity measurement was estimated using images of frame rates of 200, 400, and 800 Hz. The NIR light allowed for long imaging periods without visually stimulating the retina and disturbing the natural rheological state. High speed near-confocal imaging enabled direct and accurate measurement of erythrocyte velocity, and revealed a distinctively cardiac-dependent pulsatile velocity waveform of the erythrocyte flow in retinal capillaries, disclosed the impact of the leukocytes on erythrocyte motion, and provided new metrics for precise assessment of erythrocyte movement. The approach may facilitate new investigations on the pathophysiology of retinal microcirculation with applications for ocular and systemic diseases.
Collapse
Affiliation(s)
- Boyu Gu
- Department of Ophthalmology and Visual Sciences, School of Medicine, University of Alabama at Birmingham, 1670 University Boulevard, Birmingham, AL 35294, USA
| | - Xiaolin Wang
- Department of Ophthalmology and Visual Sciences, School of Medicine, University of Alabama at Birmingham, 1670 University Boulevard, Birmingham, AL 35294, USA
| | - Michael D. Twa
- Department of Optometry and Vision Science, School of Optometry, University of Alabama at Birmingham, 1716 University Boulevard, Birmingham, AL 35294, USA
| | - Johnny Tam
- National Eye Institute, National Institutes of Health, 10 Center Drive, Bethesda, MD 20892, USA
| | - Christopher A. Girkin
- Department of Ophthalmology and Visual Sciences, School of Medicine, University of Alabama at Birmingham, 1670 University Boulevard, Birmingham, AL 35294, USA
| | - Yuhua Zhang
- Department of Ophthalmology and Visual Sciences, School of Medicine, University of Alabama at Birmingham, 1670 University Boulevard, Birmingham, AL 35294, USA
| |
Collapse
|
48
|
Kisler K, Lazic D, Sweeney MD, Plunkett S, Khatib ME, Vinogradov SA, Boas DA, Sakadžić S, Zlokovic BV. In vivo imaging and analysis of cerebrovascular hemodynamic responses and tissue oxygenation in the mouse brain. Nat Protoc 2018; 13:1377-1402. [PMID: 29844521 PMCID: PMC6402338 DOI: 10.1038/nprot.2018.034] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Cerebrovascular dysfunction has an important role in the pathogenesis of multiple brain disorders. Measurement of hemodynamic responses in vivo can be challenging, particularly as techniques are often not described in sufficient detail and vary between laboratories. We present a set of standardized in vivo protocols that describe high-resolution two-photon microscopy and intrinsic optical signal (IOS) imaging to evaluate capillary and arteriolar responses to a stimulus, regional hemodynamic responses, and oxygen delivery to the brain. The protocol also describes how to measure intrinsic NADH fluorescence to understand how blood O2 supply meets the metabolic demands of activated brain tissue, and to perform resting-state absolute oxygen partial pressure (pO2) measurements of brain tissue. These methods can detect cerebrovascular changes at far higher resolution than MRI techniques, although the optical nature of these techniques limits their achievable imaging depths. Each individual procedure requires 1-2 h to complete, with two to three procedures typically performed per animal at a time. These protocols are broadly applicable in studies of cerebrovascular function in healthy and diseased brain in any of the existing mouse models of neurological and vascular disorders. All these procedures can be accomplished by a competent graduate student or experienced technician, except the two-photon measurement of absolute pO2 level, which is better suited to a more experienced, postdoctoral-level researcher.
Collapse
Affiliation(s)
- Kassandra Kisler
- Department of Physiology and Neuroscience and the Zilkha Neurogenetic Institute, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90089
| | - Divna Lazic
- Department of Physiology and Neuroscience and the Zilkha Neurogenetic Institute, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90089
- Department of Neurobiology, Institute for Biological Research, University of Belgrade, Belgrade, Republic of Serbia
| | - Melanie D. Sweeney
- Department of Physiology and Neuroscience and the Zilkha Neurogenetic Institute, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90089
| | - Shane Plunkett
- Departments of Biochemistry and Biophysics and of Chemistry, University of Pennsylvania, Philadelphia, PA 19104
| | - Mirna El Khatib
- Departments of Biochemistry and Biophysics and of Chemistry, University of Pennsylvania, Philadelphia, PA 19104
| | - Sergei A. Vinogradov
- Departments of Biochemistry and Biophysics and of Chemistry, University of Pennsylvania, Philadelphia, PA 19104
| | - David A. Boas
- Optics Division, MGH/HMS/MIT Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, 149 13th Street, Charlestown, MA 02129
- Department of Biomedical Engineering, Boston University, 44 Cummington Mall, Boston, MA 02215
| | - Sava Sakadžić
- Optics Division, MGH/HMS/MIT Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, 149 13th Street, Charlestown, MA 02129
| | - Berislav V. Zlokovic
- Department of Physiology and Neuroscience and the Zilkha Neurogenetic Institute, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90089
| |
Collapse
|
49
|
Liu S, Wei C, Kang N, He Q, Liang J, Wang H, Chang L, Chen D, Zhang Q, Chang C, Zhang J, Ren H, Wu Y. Chinese medicine Tongxinluo capsule alleviates cerebral microcirculatory disturbances in ischemic stroke by modulating vascular endothelial function and inhibiting leukocyte–endothelial cell interactions in mice: A two‐photon laser scanning microscopy study. Microcirculation 2018; 25. [DOI: 10.1111/micc.12437] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Accepted: 12/08/2017] [Indexed: 12/14/2022]
Abstract
AbstractObjectiveThe aim of this study was to examine the effect of TXL, a Chinese medicine prescription, on cerebral microcirculatory disturbances after pMCAO in mice using TPLSM and further explore the underlying mechanisms.MethodsAdlut male C57BL/6J mice were subjected to pMCAO and orally administered with TXL (3.0, 1.5 and 0.75 g/kg/d) at 1, 3, and 21 hours after pMCAO. The following parameters were examined at 6 and 24 hours after pMCAO: neurological deficits, infarct volume, BBB permeability, cerebral microvessel structure, brain microcirculation (TPLSM imaging), vasoactive factors, and adhesion molecules.ResultsTXL improved neurological deficits, reduced infarct volume, attenuated BBB disruption, protected cerebral microvessel structure, increased cerebral capillary flow velocity and volume flux, and inhibited leukocyte–endothelial cell interactions at 6 or 24 hours after pMCAO. The therapeutic efficacy was exerted in a dose‐dependent manner. Further study revealed that TXL (high dose) regulated the expression of PGI2, TXA2, and ET‐1, and suppressed ICAM‐1 and P‐selectin.ConclusionsTXL alleviates cerebral microcirculatory disturbances against ischemic injury by modulating endothelial function and inhibiting leukocyte–endothelial cell interactions. These effects are associated with regulating the expression of PGI2, TXA2, and ET‐1, and suppressing ICAM‐1 and P‐selectin expression.
Collapse
Affiliation(s)
- Shen Liu
- Dongzhimen Hospital Beijing University of Chinese Medicine Beijing China
- National Key Laboratory of Collateral Disease Research and Innovative Chinese Medicine Hebei Yiling Chinese Medicine Research Institute Shijiazhuang China
| | - Cong Wei
- National Key Laboratory of Collateral Disease Research and Innovative Chinese Medicine Hebei Yiling Chinese Medicine Research Institute Shijiazhuang China
| | - Ning Kang
- National Key Laboratory of Collateral Disease Research and Innovative Chinese Medicine Hebei Yiling Chinese Medicine Research Institute Shijiazhuang China
| | - Qilong He
- National Key Laboratory of Collateral Disease Research and Innovative Chinese Medicine Hebei Yiling Chinese Medicine Research Institute Shijiazhuang China
| | - Junqing Liang
- National Key Laboratory of Collateral Disease Research and Innovative Chinese Medicine Hebei Yiling Chinese Medicine Research Institute Shijiazhuang China
| | - Hongtao Wang
- National Key Laboratory of Collateral Disease Research and Innovative Chinese Medicine Hebei Yiling Chinese Medicine Research Institute Shijiazhuang China
| | - Liping Chang
- National Key Laboratory of Collateral Disease Research and Innovative Chinese Medicine Hebei Yiling Chinese Medicine Research Institute Shijiazhuang China
| | - Daohong Chen
- National Key Laboratory of Collateral Disease Research and Innovative Chinese Medicine Hebei Yiling Chinese Medicine Research Institute Shijiazhuang China
| | - Qiuyan Zhang
- National Key Laboratory of Collateral Disease Research and Innovative Chinese Medicine Hebei Yiling Chinese Medicine Research Institute Shijiazhuang China
| | - Chengcheng Chang
- National Key Laboratory of Collateral Disease Research and Innovative Chinese Medicine Hebei Yiling Chinese Medicine Research Institute Shijiazhuang China
| | - Junfang Zhang
- National Key Laboratory of Collateral Disease Research and Innovative Chinese Medicine Hebei Yiling Chinese Medicine Research Institute Shijiazhuang China
| | - Hong Ren
- Graduate School Hebei Medical University Shijiazhuang China
| | - Yiling Wu
- Dongzhimen Hospital Beijing University of Chinese Medicine Beijing China
- National Key Laboratory of Collateral Disease Research and Innovative Chinese Medicine Hebei Yiling Chinese Medicine Research Institute Shijiazhuang China
| |
Collapse
|
50
|
Chen C, Zhang YP, Sun Y, Xiong W, Shields LBE, Shields CB, Jin X, Xu XM. An In Vivo Duo-color Method for Imaging Vascular Dynamics Following Contusive Spinal Cord Injury. J Vis Exp 2017:56565. [PMID: 29364252 PMCID: PMC5908407 DOI: 10.3791/56565] [Citation(s) in RCA: 10] [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] [Indexed: 01/22/2023] Open
Abstract
Spinal cord injury (SCI) causes significant vascular disruption at the site of injury. Vascular pathology occurs immediately after SCI and continues throughout the acute injury phase. In fact, endothelial cells appear to be the first to die after a contusive SCI. The early vascular events, including increased permeability of the blood-spinal cord barrier (BSCB), induce vasogenic edema and contribute to detrimental secondary injury events caused by complex injury mechanisms. Targeting the vascular disruption, therefore, could be a key strategy to reduce secondary injury cascades that contribute to histological and functional impairments after SCI. Previous studies were mostly performed on postmortem samples and were unable to capture the dynamic changes of the vascular network. In this study, we have developed an in vivo duo-color two-photon imaging method to monitor acute vascular dynamic changes following contusive SCI. This approach allows detecting blood flow, vessel diameter, and other vascular pathologies at various sites of the same rat pre- and post-injury. Overall, this method provides an excellent venue for investigating vascular dynamics.
Collapse
Affiliation(s)
- Chen Chen
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, and Department of Neurological Surgery, Indiana University School of Medicine; Program in Medical Neuroscience, Stark Neurosciences Research Institute, Indiana University School of Medicine
| | | | - Yan Sun
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, and Department of Neurological Surgery, Indiana University School of Medicine; Department of Human Anatomy & Histoembryology, School of Basic Medical Sciences, Fudan University
| | - Wenhui Xiong
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, and Department of Neurological Surgery, Indiana University School of Medicine
| | | | - Christopher B Shields
- Norton Neuroscience Institute, Norton Healthcare; Department of Neurological Surgery, University of Louisville School of Medicine
| | - Xiaoming Jin
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, and Department of Neurological Surgery, Indiana University School of Medicine
| | - Xiao-Ming Xu
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, and Department of Neurological Surgery, Indiana University School of Medicine;
| |
Collapse
|