1
|
Qi L, Wang C, Deng L, Pan JJ, Suo Q, Wu S, Cai L, Shi X, Sun J, Wang Y, Tang Y, Qiu W, Yang GY, Wang J, Zhang Z. Low-intensity focused ultrasound stimulation promotes stroke recovery via astrocytic HMGB1 and CAMK2N1 in mice. Stroke Vasc Neurol 2024; 9:505-518. [PMID: 38191183 DOI: 10.1136/svn-2023-002614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 11/28/2023] [Indexed: 01/10/2024] Open
Abstract
BACKGROUND Low-intensity focused ultrasound stimulation (LIFUS) has been developed to enhance neurological repair and remodelling during the late acute stage of ischaemic stroke in rodents. However, the cellular and molecular mechanisms of neurological repair and remodelling after LIFUS in ischaemic stroke are unclear. METHODS Ultrasound stimulation was treated in adult male mice 7 days after transient middle cerebral artery occlusion. Angiogenesis was measured by laser speckle imaging and histological analyses. Electromyography and fibre photometry records were used for synaptogenesis. Brain atrophy volume and neurobehaviour were assessed 0-14 days after ischaemia. iTRAQ proteomic analysis was performed to explore the differentially expressed protein. scRNA-seq was used for subcluster analysis of astrocytes. Fluorescence in situ hybridisation and Western blot detected the expression of HMGB1 and CAMK2N1. RESULTS Optimal ultrasound stimulation increased cerebral blood flow, and improved neurobehavioural outcomes in ischaemic mice (p<0.05). iTRAQ proteomic analysis revealed that the expression of HMGB1 increased and CAMK2N1 decreased in the ipsilateral hemisphere of the brain at 14 days after focal cerebral ischaemia with ultrasound treatment (p<0.05). scRNA-seq revealed that this expression pattern belonged to a subcluster of astrocytes after LIFUS in the ischaemic brain. LIFUS upregulated HMGB1 expression, accompanied by VEGFA elevation compared with the control group (p<0.05). Inhibition of HMGB1 expression in astrocytes decreased microvessels counts and cerebral blood flow (p<0.05). LIFUS reduced CAMK2N1 expression level, accompanied by increased extracellular calcium ions and glutamatergic synapses (p<0.05). CAMK2N1 overexpression in astrocytes decreased dendritic spines, and aggravated neurobehavioural outcomes (p<0.05). CONCLUSION Our results demonstrated that LIFUS promoted angiogenesis and synaptogenesis after focal cerebral ischaemia by upregulating HMGB1 and downregulating CAMK2N1 in a subcluster of astrocytes, suggesting that LIFUS activated specific astrocyte subcluster could be a key target for ischaemic brain therapy.
Collapse
MESH Headings
- Animals
- Astrocytes/metabolism
- Astrocytes/pathology
- Astrocytes/enzymology
- Male
- Recovery of Function
- Disease Models, Animal
- Infarction, Middle Cerebral Artery/metabolism
- Infarction, Middle Cerebral Artery/pathology
- Infarction, Middle Cerebral Artery/therapy
- Infarction, Middle Cerebral Artery/physiopathology
- Infarction, Middle Cerebral Artery/genetics
- Infarction, Middle Cerebral Artery/enzymology
- Mice, Inbred C57BL
- HMGB1 Protein/metabolism
- HMGB1 Protein/genetics
- Neovascularization, Physiologic
- Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism
- Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics
- Cerebrovascular Circulation
- Ultrasonic Therapy
- Signal Transduction
- Ischemic Stroke/metabolism
- Ischemic Stroke/physiopathology
- Ischemic Stroke/therapy
- Ischemic Stroke/pathology
- Behavior, Animal
- Time Factors
- Mice
- Proteomics
- Neurogenesis
Collapse
Affiliation(s)
- Lin Qi
- Shanghai Jiao Tong Affiliated Sixth People's Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, Shanghai, China
| | - Cheng Wang
- Shanghai Jiao Tong Affiliated Sixth People's Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, Shanghai, China
| | - Lidong Deng
- Shanghai Jiao Tong Affiliated Sixth People's Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, Shanghai, China
| | - Jia-Ji Pan
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical Collage, Fudan University, Shanghai, China
| | - Qian Suo
- Shanghai Jiao Tong Affiliated Sixth People's Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, Shanghai, China
| | - Shengju Wu
- Shanghai Jiao Tong Affiliated Sixth People's Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, Shanghai, China
| | - Lin Cai
- Department of Neurosurgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University, Shanghai, China
| | - Xudong Shi
- Paul C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Beijing, China
| | - Junfeng Sun
- Shanghai Jiao Tong Affiliated Sixth People's Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, Shanghai, China
| | - Yongting Wang
- Shanghai Jiao Tong Affiliated Sixth People's Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, Shanghai, China
| | - Yaohui Tang
- Shanghai Jiao Tong Affiliated Sixth People's Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, Shanghai, China
| | - Weibao Qiu
- Paul C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Beijing, China
| | - Guo-Yuan Yang
- Neuroscience and Neuroengineering Center, Shanghai Jiao Tong University School of Biomedical Engineering, Shanghai, China
- Shanghai Jiao Tong University Medical School Affiliated Ruijin Hospital, Shanghai, China
| | - Jixian Wang
- Shanghai Jiao Tong University Medical School Affiliated Ruijin Hospital, Shanghai, China
| | - Zhijun Zhang
- Shanghai Jiao Tong Affiliated Sixth People's Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, Shanghai, China
| |
Collapse
|
2
|
Yu PK, Mehnert A, Dickson JB, Qambari H, Balaratnasingam C, Cringle S, Darcey D, Yu DY. Quantitative study of spatial and temporal variation in retinal capillary network perfusion in rat eye by in vivo confocal imaging. Sci Rep 2023; 13:18923. [PMID: 37919331 PMCID: PMC10622421 DOI: 10.1038/s41598-023-44480-1] [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] [Received: 08/04/2023] [Accepted: 10/09/2023] [Indexed: 11/04/2023] Open
Abstract
Microvascular dysfunction is the underlying pathological process in many systemic diseases. However, investigation into its pathogenesis is impeded by the accessibility and complexity of the microvasculature within different organs, particularly for the central nervous system. The retina as an extension of the cerebrum provides a glimpse into the brain through which the microvasculature can be observed. Two major questions remain unanswered: How do the microvessels regulate spatial and temporal delivery to satisfy the varying cellular demands, and how can we quantify blood perfusion in the 3D capillary network? Here, quantitative measurements of red blood cell (RBC) speed in each vessel in the field were made in the in vivo rat retinal capillary network using an ultrafast confocal technique with fluorescently labelled RBCs. Retinal RBC speed and number were found to vary remarkably between microvessels ranging from 215 to 6641 microns per second with significant variations spatially and temporally. Overall, the RBC speed was significantly faster in the microvessels in the superficial retina than in the deep retina (estimated marginal means of 2405 ± 238.2 µm/s, 1641 ± 173.0 µm/s respectively). These observations point to a highly dynamic nature of microvasculature that is specific to its immediate cellular environment and is constantly changing.
Collapse
Affiliation(s)
- Paula Kun Yu
- Centre for Ophthalmology and Visual Science, The University of Western Australia, Perth, Australia
- Lions Eye Institute, 2 Verdun Street, Nedlands, WA, Australia
| | - Andrew Mehnert
- Centre for Ophthalmology and Visual Science, The University of Western Australia, Perth, Australia
- Lions Eye Institute, 2 Verdun Street, Nedlands, WA, Australia
| | | | - Hassanain Qambari
- Centre for Ophthalmology and Visual Science, The University of Western Australia, Perth, Australia
- Lions Eye Institute, 2 Verdun Street, Nedlands, WA, Australia
| | - Chandrakumar Balaratnasingam
- Centre for Ophthalmology and Visual Science, The University of Western Australia, Perth, Australia
- Lions Eye Institute, 2 Verdun Street, Nedlands, WA, Australia
- Department of Ophthalmology, Sir Charles Gairdner Hospital, Nedlands, WA, Australia
| | - Stephen Cringle
- Centre for Ophthalmology and Visual Science, The University of Western Australia, Perth, Australia
- Lions Eye Institute, 2 Verdun Street, Nedlands, WA, Australia
| | - Dean Darcey
- Lions Eye Institute, 2 Verdun Street, Nedlands, WA, Australia
| | - Dao-Yi Yu
- Centre for Ophthalmology and Visual Science, The University of Western Australia, Perth, Australia.
- Lions Eye Institute, 2 Verdun Street, Nedlands, WA, Australia.
| |
Collapse
|
3
|
Alarcon-Martinez L, Shiga Y, Villafranca-Baughman D, Cueva Vargas JL, Vidal Paredes IA, Quintero H, Fortune B, Danesh-Meyer H, Di Polo A. Neurovascular dysfunction in glaucoma. Prog Retin Eye Res 2023; 97:101217. [PMID: 37778617 DOI: 10.1016/j.preteyeres.2023.101217] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 09/23/2023] [Accepted: 09/25/2023] [Indexed: 10/03/2023]
Abstract
Retinal ganglion cells, the neurons that die in glaucoma, are endowed with a high metabolism requiring optimal provision of oxygen and nutrients to sustain their activity. The timely regulation of blood flow is, therefore, essential to supply firing neurons in active areas with the oxygen and glucose they need for energy. Many glaucoma patients suffer from vascular deficits including reduced blood flow, impaired autoregulation, neurovascular coupling dysfunction, and blood-retina/brain-barrier breakdown. These processes are tightly regulated by a community of cells known as the neurovascular unit comprising neurons, endothelial cells, pericytes, Müller cells, astrocytes, and microglia. In this review, the neurovascular unit takes center stage as we examine the ability of its members to regulate neurovascular interactions and how their function might be altered during glaucomatous stress. Pericytes receive special attention based on recent data demonstrating their key role in the regulation of neurovascular coupling in physiological and pathological conditions. Of particular interest is the discovery and characterization of tunneling nanotubes, thin actin-based conduits that connect distal pericytes, which play essential roles in the complex spatial and temporal distribution of blood within the retinal capillary network. We discuss cellular and molecular mechanisms of neurovascular interactions and their pathophysiological implications, while highlighting opportunities to develop strategies for vascular protection and regeneration to improve functional outcomes in glaucoma.
Collapse
Affiliation(s)
- Luis Alarcon-Martinez
- Department of Neuroscience, Université de Montréal, PO Box 6128, Station centre-ville, Montreal, QC, Canada; Neuroscience Division, Centre de recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), 900 Saint Denis Street, Montreal, QC, Canada; Centre for Eye Research Australia, University of Melbourne, Melbourne, Australia
| | - Yukihiro Shiga
- Department of Neuroscience, Université de Montréal, PO Box 6128, Station centre-ville, Montreal, QC, Canada; Neuroscience Division, Centre de recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), 900 Saint Denis Street, Montreal, QC, Canada
| | - Deborah Villafranca-Baughman
- Department of Neuroscience, Université de Montréal, PO Box 6128, Station centre-ville, Montreal, QC, Canada; Neuroscience Division, Centre de recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), 900 Saint Denis Street, Montreal, QC, Canada
| | - Jorge L Cueva Vargas
- Department of Neuroscience, Université de Montréal, PO Box 6128, Station centre-ville, Montreal, QC, Canada; Neuroscience Division, Centre de recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), 900 Saint Denis Street, Montreal, QC, Canada
| | - Isaac A Vidal Paredes
- Department of Neuroscience, Université de Montréal, PO Box 6128, Station centre-ville, Montreal, QC, Canada; Neuroscience Division, Centre de recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), 900 Saint Denis Street, Montreal, QC, Canada
| | - Heberto Quintero
- Department of Neuroscience, Université de Montréal, PO Box 6128, Station centre-ville, Montreal, QC, Canada; Neuroscience Division, Centre de recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), 900 Saint Denis Street, Montreal, QC, Canada
| | - Brad Fortune
- Discoveries in Sight Research Laboratories, Devers Eye Institute and Legacy Research Institute, Legacy Healthy, Portland, OR, USA
| | - Helen Danesh-Meyer
- Department of Ophthalmology, New Zealand National Eye Centre, Faculty of Medical and Health Sciences, University of Auckland, New Zealand
| | - Adriana Di Polo
- Department of Neuroscience, Université de Montréal, PO Box 6128, Station centre-ville, Montreal, QC, Canada; Neuroscience Division, Centre de recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), 900 Saint Denis Street, Montreal, QC, Canada.
| |
Collapse
|
4
|
Yu DY, Mehnert A, Balaratnasingam C, Yu PK, Hein M, An D, Cringle SJ. An assessment of microvascular hemodynamics in human macula. Sci Rep 2023; 13:7550. [PMID: 37160984 PMCID: PMC10169832 DOI: 10.1038/s41598-023-33490-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 04/13/2023] [Indexed: 05/11/2023] Open
Abstract
An adequate blood supply to meet the energy demands is essential for any tissue, particularly for high energy demand tissues such as the retina. A critical question is: How is the dynamic match between neuronal demands and blood supply achieved? We present a quantitative assessment of temporal and spatial variations in perfusion in the macular capillary network in 10 healthy human subjects using a non-invasive and label-free imaging technique. The assessment is based on the calculation of the coefficient of variation (CoV) of the perfusion signal from arterioles, venules and capillaries from a sequence of optical coherence tomography angiography images centred on the fovea. Significant heterogeneity of the spatial and temporal variation was found within arterioles, venules and capillary networks. The CoV values of the capillaries and smallest vessels were significantly higher than that in the larger vessels. Our results demonstrate the presence of significant heterogeneity of spatial and temporal variation within each element of the macular microvasculature, particularly in the capillaries and finer vessels. Our findings suggest that the dynamic match between neuronal demands and blood supply is achieved by frequent alteration of local blood flow evidenced by capillary perfusion variations both spatially and temporally in the macular region.
Collapse
Affiliation(s)
- Dao-Yi Yu
- Centre for Ophthalmology and Visual Science, The University of Western Australia, Perth, Australia.
- Lions Eye Institute, 2 Verdun St, Nedlands, Perth, WA, 6009, Australia.
| | - Andrew Mehnert
- Centre for Ophthalmology and Visual Science, The University of Western Australia, Perth, Australia
- Lions Eye Institute, 2 Verdun St, Nedlands, Perth, WA, 6009, Australia
| | - Chandrakumar Balaratnasingam
- Centre for Ophthalmology and Visual Science, The University of Western Australia, Perth, Australia
- Lions Eye Institute, 2 Verdun St, Nedlands, Perth, WA, 6009, Australia
- Sir Charles Gairdner Hospital, Perth, Australia
| | - Paula K Yu
- Centre for Ophthalmology and Visual Science, The University of Western Australia, Perth, Australia
- Lions Eye Institute, 2 Verdun St, Nedlands, Perth, WA, 6009, Australia
| | - Martin Hein
- Centre for Ophthalmology and Visual Science, The University of Western Australia, Perth, Australia
- Lions Eye Institute, 2 Verdun St, Nedlands, Perth, WA, 6009, Australia
| | - Dong An
- Centre for Ophthalmology and Visual Science, The University of Western Australia, Perth, Australia
- Lions Eye Institute, 2 Verdun St, Nedlands, Perth, WA, 6009, Australia
| | - Stephen J Cringle
- Centre for Ophthalmology and Visual Science, The University of Western Australia, Perth, Australia
- Lions Eye Institute, 2 Verdun St, Nedlands, Perth, WA, 6009, Australia
| |
Collapse
|
5
|
Klug NR, Sancho M, Gonzales AL, Heppner TJ, O’Brien RIC, Hill-Eubanks D, Nelson MT. Intraluminal pressure elevates intracellular calcium and contracts CNS pericytes: Role of voltage-dependent calcium channels. Proc Natl Acad Sci U S A 2023; 120:e2216421120. [PMID: 36802432 PMCID: PMC9992766 DOI: 10.1073/pnas.2216421120] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 01/24/2023] [Indexed: 02/23/2023] Open
Abstract
Arteriolar smooth muscle cells (SMCs) and capillary pericytes dynamically regulate blood flow in the central nervous system in the face of fluctuating perfusion pressures. Pressure-induced depolarization and Ca2+ elevation provide a mechanism for regulation of SMC contraction, but whether pericytes participate in pressure-induced changes in blood flow remains unknown. Here, utilizing a pressurized whole-retina preparation, we found that increases in intraluminal pressure in the physiological range induce contraction of both dynamically contractile pericytes in the arteriole-proximate transition zone and distal pericytes of the capillary bed. We found that the contractile response to pressure elevation was slower in distal pericytes than in transition zone pericytes and arteriolar SMCs. Pressure-evoked elevation of cytosolic Ca2+ and contractile responses in SMCs were dependent on voltage-dependent Ca2+ channel (VDCC) activity. In contrast, Ca2+ elevation and contractile responses were partially dependent on VDCC activity in transition zone pericytes and independent of VDCC activity in distal pericytes. In both transition zone and distal pericytes, membrane potential at low inlet pressure (20 mmHg) was approximately -40 mV and was depolarized to approximately -30 mV by an increase in pressure to 80 mmHg. The magnitude of whole-cell VDCC currents in freshly isolated pericytes was approximately half that measured in isolated SMCs. Collectively, these results indicate a loss of VDCC involvement in pressure-induced constriction along the arteriole-capillary continuum. They further suggest that alternative mechanisms and kinetics of Ca2+ elevation, contractility, and blood flow regulation exist in central nervous system capillary networks, distinguishing them from neighboring arterioles.
Collapse
Affiliation(s)
- Nicholas R. Klug
- Department of Pharmacology, Larner College of Medicine, University of Vermont, Burlington, VT05405
| | - Maria Sancho
- Department of Pharmacology, Larner College of Medicine, University of Vermont, Burlington, VT05405
| | - Albert L. Gonzales
- Department of Pharmacology, Larner College of Medicine, University of Vermont, Burlington, VT05405
- Department of Physiology and Cell Biology, School of Medicine, University of Nevada, Reno, NV89557
| | - Thomas J. Heppner
- Department of Pharmacology, Larner College of Medicine, University of Vermont, Burlington, VT05405
| | | | - David Hill-Eubanks
- Department of Pharmacology, Larner College of Medicine, University of Vermont, Burlington, VT05405
| | - Mark T. Nelson
- Department of Pharmacology, Larner College of Medicine, University of Vermont, Burlington, VT05405
- Division of Cardiovascular Sciences, University of Manchester, ManchesterM13 9PL, UK
| |
Collapse
|
6
|
Hirunpattarasilp C, Barkaway A, Davis H, Pfeiffer T, Sethi H, Attwell D. Hyperoxia evokes pericyte-mediated capillary constriction. J Cereb Blood Flow Metab 2022; 42:2032-2047. [PMID: 35786054 PMCID: PMC9580167 DOI: 10.1177/0271678x221111598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Oxygen supplementation is regularly prescribed to patients to treat or prevent hypoxia. However, excess oxygenation can lead to reduced cerebral blood flow (CBF) in healthy subjects and worsen the neurological outcome of critically ill patients. Most studies on the vascular effects of hyperoxia focus on arteries but there is no research on the effects on cerebral capillary pericytes, which are major regulators of CBF. Here, we used bright-field imaging of cerebral capillaries and modeling of CBF to show that hyperoxia (95% superfused O2) led to an increase in intracellular calcium level in pericytes and a significant capillary constriction, sufficient to cause an estimated 25% decrease in CBF. Although hyperoxia is reported to cause vascular smooth muscle cell contraction via generation of reactive oxygen species (ROS), endothelin-1 and 20-HETE, we found that increased cytosolic and mitochondrial ROS levels and endothelin release were not involved in the pericyte-mediated capillary constriction. However, a 20-HETE synthesis blocker greatly reduced the hyperoxia-evoked capillary constriction. Our findings establish pericytes as regulators of CBF in hyperoxia and 20-HETE synthesis as an oxygen sensor in CBF regulation. The results also provide a mechanism by which clinically administered oxygen can lead to a worse neurological outcome.
Collapse
Affiliation(s)
- Chanawee Hirunpattarasilp
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, London, UK.,Princess Srisavangavadhana College of Medicine, Chulabhorn Royal Academy, Bangkok, Thailand
| | - Anna Barkaway
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, London, UK.,Princess Srisavangavadhana College of Medicine, Chulabhorn Royal Academy, Bangkok, Thailand
| | - Harvey Davis
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, London, UK.,Princess Srisavangavadhana College of Medicine, Chulabhorn Royal Academy, Bangkok, Thailand
| | - Thomas Pfeiffer
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, London, UK
| | - Huma Sethi
- Division of Neurosurgery, UCL Queen Square Institute of Neurology, Queen Square, London, UK
| | - David Attwell
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, London, UK
| |
Collapse
|
7
|
Nippert AR, Chiang PP, Del Franco AP, Newman EA. Astrocyte regulation of cerebral blood flow during hypoglycemia. J Cereb Blood Flow Metab 2022; 42:1534-1546. [PMID: 35296178 PMCID: PMC9274859 DOI: 10.1177/0271678x221089091] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 02/07/2022] [Accepted: 02/28/2022] [Indexed: 12/13/2022]
Abstract
Hypoglycemia triggers increases in cerebral blood flow (CBF), augmenting glucose supply to the brain. We have tested whether astrocytes, which can regulate vessel tone, contribute to this CBF increase. We hypothesized that hypoglycemia-induced adenosine signaling acts to increase astrocyte Ca2+ activity, which then causes the release of prostaglandins (PGs) and epoxyeicosatrienoic acids (EETs), leading to the dilation of brain arterioles and blood flow increases. We used an awake mouse model to investigate the effects of insulin-induced hypoglycemia on arterioles and astrocytes in the somatosensory cortex. During insulin-induced hypoglycemia, penetrating arterioles dilated and astrocyte Ca2+ signaling increased when blood glucose dropped below a threshold of ∼50 mg/dL. Application of the A2A adenosine receptor antagonist ZM-241385 eliminated hypoglycemia-evoked astrocyte Ca2+ increases and reduced arteriole dilations by 44% (p < 0.05). SC-560 and miconazole, which block the production of the astrocyte vasodilators PGs and EETs respectively, reduced arteriole dilations in response to hypoglycemia by 89% (p < 0.001) and 76% (p < 0.001). Hypoglycemia-induced arteriole dilations were decreased by 65% (p < 0.001) in IP3R2 knockout mice, which have reduced astrocyte Ca2+ signaling compared to wild-type. These results support the hypothesis that astrocytes contribute to hypoglycemia-induced increases in CBF by releasing vasodilators in a Ca2+-dependent manner.
Collapse
Affiliation(s)
- Amy R Nippert
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, USA
| | - Pei-Pei Chiang
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, USA
| | | | - Eric A Newman
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
8
|
Bojovic D, Stackhouse TL, Mishra A. Assaying activity-dependent arteriole and capillary responses in brain slices. NEUROPHOTONICS 2022; 9:031913. [PMID: 35558646 PMCID: PMC9089234 DOI: 10.1117/1.nph.9.3.031913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 03/31/2022] [Indexed: 06/15/2023]
Abstract
Significance: Neurovascular coupling (NVC) is the process that increases cerebral blood flow in response to neuronal activity. NVC is orchestrated by signaling between neurons, glia, and vascular cells. Elucidating the mechanisms underlying NVC at different vascular segments and in different brain regions is imperative for understanding of brain function and mechanisms of dysfunction. Aim: Our goal is to describe a protocol for concurrently monitoring stimulation-evoked neuronal activity and resultant vascular responses in acute brain slices. Approach: We describe a step-by-step protocol that allows the study of endogenous NVC mechanisms engaged by neuronal activity in a controlled, reduced preparation. Results: This ex vivo NVC assay allows researchers to disentangle the mechanisms regulating the contractile responses of different vascular segments in response to neuronal firing independent of flow and pressure mediated effects from connected vessels. It also enables easy pharmacological manipulations in a simplified, reduced system and can be combined with Ca 2 + imaging or broader electrophysiology techniques to obtain multimodal data during NVC. Conclusions: The ex vivo NVC assay will facilitate investigations of cellular and molecular mechanisms that give rise to NVC and should serve as a valuable complement to in vivo imaging methods.
Collapse
Affiliation(s)
- Danica Bojovic
- Oregon Health & Science University, Jungers Center for Neurosciences Research, Department of Neurology, Portland, Oregon, United States
- Oregon Health & Science University, Vollum Institute, Portland, Oregon, United States
| | - Teresa L. Stackhouse
- Oregon Health & Science University, Jungers Center for Neurosciences Research, Department of Neurology, Portland, Oregon, United States
| | - Anusha Mishra
- Oregon Health & Science University, Jungers Center for Neurosciences Research, Department of Neurology, Portland, Oregon, United States
- Oregon Health & Science University, Knight Cardiovascular Institute, Portland, Oregon, United States
| |
Collapse
|
9
|
Mills WA, Woo AM, Jiang S, Martin J, Surendran D, Bergstresser M, Kimbrough IF, Eyo UB, Sofroniew MV, Sontheimer H. Astrocyte plasticity in mice ensures continued endfoot coverage of cerebral blood vessels following injury and declines with age. Nat Commun 2022; 13:1794. [PMID: 35379828 PMCID: PMC8980042 DOI: 10.1038/s41467-022-29475-2] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Accepted: 03/11/2022] [Indexed: 01/30/2023] Open
Abstract
Astrocytes extend endfeet that enwrap the vasculature, and disruptions to this association which may occur in disease coincide with breaches in blood-brain barrier (BBB) integrity. Here we investigate if focal ablation of astrocytes is sufficient to disrupt the BBB in mice. Targeted two-photon chemical apoptotic ablation of astrocytes induced a plasticity response whereby surrounding astrocytes extended processes to cover vascular vacancies. In young animals, replacement processes occur in advance of endfoot retraction, but this is delayed in aged animals. Stimulation of replacement astrocytes results in constriction of pre-capillary arterioles, suggesting that replacement astrocytes are functional. Pharmacological inhibition of pSTAT3, as well as astrocyte specific deletion of pSTAT3, reduces astrocyte replacement post-ablation, without perturbations to BBB integrity. Similar endfoot replacement occurs following astrocyte cell death due to reperfusion in a stroke model. Together, these studies uncover the ability of astrocytes to maintain cerebrovascular coverage via substitution from nearby cells.
Collapse
Affiliation(s)
- William A. Mills
- grid.27755.320000 0000 9136 933XBrain, Immunology, and Glia Center, University of Virginia School of Medicine, Charlottesville, VA USA ,grid.27755.320000 0000 9136 933XDepartment of Neuroscience, University of Virginia School of Medicine, Charlottesville, VA USA ,grid.27755.320000 0000 9136 933XRobert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA USA ,grid.438526.e0000 0001 0694 4940Graduate Program in Translational Biology, Medicine, & Health, Virginia Polytechnic Institute and State University, Blacksburg, VA USA
| | - AnnaLin M. Woo
- grid.27755.320000 0000 9136 933XBrain, Immunology, and Glia Center, University of Virginia School of Medicine, Charlottesville, VA USA ,grid.27755.320000 0000 9136 933XDepartment of Neuroscience, University of Virginia School of Medicine, Charlottesville, VA USA
| | - Shan Jiang
- grid.168010.e0000000419368956Department of Material Science and Engineering, Stanford University, Stanford, CA USA ,grid.168010.e0000000419368956Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA USA
| | - Joelle Martin
- grid.438526.e0000 0001 0694 4940Graduate Program in Translational Biology, Medicine, & Health, Virginia Polytechnic Institute and State University, Blacksburg, VA USA
| | - Dayana Surendran
- grid.27755.320000 0000 9136 933XBrain, Immunology, and Glia Center, University of Virginia School of Medicine, Charlottesville, VA USA ,grid.27755.320000 0000 9136 933XDepartment of Neuroscience, University of Virginia School of Medicine, Charlottesville, VA USA
| | - Matthew Bergstresser
- grid.438526.e0000 0001 0694 4940School of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, VA USA
| | - Ian F. Kimbrough
- grid.27755.320000 0000 9136 933XBrain, Immunology, and Glia Center, University of Virginia School of Medicine, Charlottesville, VA USA ,grid.27755.320000 0000 9136 933XDepartment of Neuroscience, University of Virginia School of Medicine, Charlottesville, VA USA
| | - Ukpong B. Eyo
- grid.27755.320000 0000 9136 933XBrain, Immunology, and Glia Center, University of Virginia School of Medicine, Charlottesville, VA USA ,grid.27755.320000 0000 9136 933XDepartment of Neuroscience, University of Virginia School of Medicine, Charlottesville, VA USA ,grid.27755.320000 0000 9136 933XRobert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA USA
| | - Michael V. Sofroniew
- grid.19006.3e0000 0000 9632 6718Department of Neurobiology, University of California, Los Angeles, CA USA
| | - Harald Sontheimer
- grid.27755.320000 0000 9136 933XBrain, Immunology, and Glia Center, University of Virginia School of Medicine, Charlottesville, VA USA ,grid.27755.320000 0000 9136 933XDepartment of Neuroscience, University of Virginia School of Medicine, Charlottesville, VA USA
| |
Collapse
|
10
|
Mills SA, Jobling AI, Dixon MA, Bui BV, Vessey KA, Phipps JA, Greferath U, Venables G, Wong VHY, Wong CHY, He Z, Hui F, Young JC, Tonc J, Ivanova E, Sagdullaev BT, Fletcher EL. Fractalkine-induced microglial vasoregulation occurs within the retina and is altered early in diabetic retinopathy. Proc Natl Acad Sci U S A 2021; 118:e2112561118. [PMID: 34903661 PMCID: PMC8713803 DOI: 10.1073/pnas.2112561118] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/14/2021] [Indexed: 01/19/2023] Open
Abstract
Local blood flow control within the central nervous system (CNS) is critical to proper function and is dependent on coordination between neurons, glia, and blood vessels. Macroglia, such as astrocytes and Müller cells, contribute to this neurovascular unit within the brain and retina, respectively. This study explored the role of microglia, the innate immune cell of the CNS, in retinal vasoregulation, and highlights changes during early diabetes. Structurally, microglia were found to contact retinal capillaries and neuronal synapses. In the brain and retinal explants, the addition of fractalkine, the sole ligand for monocyte receptor Cx3cr1, resulted in capillary constriction at regions of microglial contact. This vascular regulation was dependent on microglial Cx3cr1 involvement, since genetic and pharmacological inhibition of Cx3cr1 abolished fractalkine-induced constriction. Analysis of the microglial transcriptome identified several vasoactive genes, including angiotensinogen, a constituent of the renin-angiotensin system (RAS). Subsequent functional analysis showed that RAS blockade via candesartan abolished microglial-induced capillary constriction. Microglial regulation was explored in a rat streptozotocin (STZ) model of diabetic retinopathy. Retinal blood flow was reduced after 4 wk due to reduced capillary diameter and this was coincident with increased microglial association. Functional assessment showed loss of microglial-capillary response in STZ-treated animals and transcriptome analysis showed evidence of RAS pathway dysregulation in microglia. While candesartan treatment reversed capillary constriction in STZ-treated animals, blood flow remained decreased likely due to dilation of larger vessels. This work shows microglia actively participate in the neurovascular unit, with aberrant microglial-vascular function possibly contributing to the early vascular compromise during diabetic retinopathy.
Collapse
Affiliation(s)
- Samuel A Mills
- Department of Anatomy and Physiology, University of Melbourne, Parkville, 3010 VIC, Australia
| | - Andrew I Jobling
- Department of Anatomy and Physiology, University of Melbourne, Parkville, 3010 VIC, Australia
| | - Michael A Dixon
- Department of Anatomy and Physiology, University of Melbourne, Parkville, 3010 VIC, Australia
| | - Bang V Bui
- Department of Optometry and Vision Sciences, University of Melbourne, Parkville, 3010 VIC, Australia
| | - Kirstan A Vessey
- Department of Anatomy and Physiology, University of Melbourne, Parkville, 3010 VIC, Australia
| | - Joanna A Phipps
- Department of Anatomy and Physiology, University of Melbourne, Parkville, 3010 VIC, Australia
| | - Ursula Greferath
- Department of Anatomy and Physiology, University of Melbourne, Parkville, 3010 VIC, Australia
| | - Gene Venables
- Department of Anatomy and Physiology, University of Melbourne, Parkville, 3010 VIC, Australia
| | - Vickie H Y Wong
- Department of Optometry and Vision Sciences, University of Melbourne, Parkville, 3010 VIC, Australia
| | - Connie H Y Wong
- Department of Medicine, Centre for Inflammatory Diseases, School of Clinical Sciences, Monash University, Clayton, 3800 VIC, Australia
| | - Zheng He
- Department of Optometry and Vision Sciences, University of Melbourne, Parkville, 3010 VIC, Australia
| | - Flora Hui
- Department of Optometry and Vision Sciences, University of Melbourne, Parkville, 3010 VIC, Australia
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, East Melbourne, 3002 VIC, Australia
| | - James C Young
- Department of Anatomy and Physiology, University of Melbourne, Parkville, 3010 VIC, Australia
| | - Josh Tonc
- Department of Anatomy and Physiology, University of Melbourne, Parkville, 3010 VIC, Australia
| | - Elena Ivanova
- Burke Neurological Institute, Weill Cornell Medical College, White Plains, NY 10605
| | - Botir T Sagdullaev
- Burke Neurological Institute, Weill Cornell Medical College, White Plains, NY 10605
| | - Erica L Fletcher
- Department of Anatomy and Physiology, University of Melbourne, Parkville, 3010 VIC, Australia;
| |
Collapse
|
11
|
Hu Y, Tao W. Microenvironmental Variations After Blood-Brain Barrier Breakdown in Traumatic Brain Injury. Front Mol Neurosci 2021; 14:750810. [PMID: 34899180 PMCID: PMC8662751 DOI: 10.3389/fnmol.2021.750810] [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: 07/31/2021] [Accepted: 10/18/2021] [Indexed: 12/12/2022] Open
Abstract
Traumatic brain injury (TBI) is linked to several pathologies. The blood-brain barrier (BBB) breakdown is considered to be one of the initial changes. Further, the microenvironmental alteration following TBI-induced BBB breakdown can be multi-scaled, constant, and dramatic. The microenvironmental variations after disruption of BBB includes several pathological changes, such as cerebral blood flow (CBF) alteration, brain edema, cerebral metabolism imbalances, and accumulation of inflammatory molecules. The modulation of the microenvironment presents attractive targets for TBI recovery, such as reducing toxic substances, inhibiting inflammation, and promoting neurogenesis. Herein, we briefly review the pathological alterations of the microenvironmental changes following BBB breakdown and outline potential interventions for TBI recovery based on microenvironmental modulation.
Collapse
Affiliation(s)
- Yue Hu
- School of Chinese Medicine, School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Weiwei Tao
- School of Chinese Medicine, School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China.,Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
12
|
Li Z, McConnell HL, Stackhouse TL, Pike MM, Zhang W, Mishra A. Increased 20-HETE Signaling Suppresses Capillary Neurovascular Coupling After Ischemic Stroke in Regions Beyond the Infarct. Front Cell Neurosci 2021; 15:762843. [PMID: 34819839 PMCID: PMC8606525 DOI: 10.3389/fncel.2021.762843] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 10/06/2021] [Indexed: 12/23/2022] Open
Abstract
Neurovascular coupling, the process by which neuronal activity elicits increases in the local blood supply, is impaired in stroke patients in brain regions outside the infarct. Such impairment may contribute to neurological deterioration over time, but its mechanism is unknown. Using the middle cerebral artery occlusion (MCAO) model of stroke, we show that neuronal activity-evoked capillary dilation is reduced by ∼75% in the intact cortical tissue outside the infarct border. This decrease in capillary responsiveness was not explained by a decrease in local neuronal activity or a loss of vascular contractility. Inhibiting synthesis of the vasoconstrictive molecule 20-hydroxyeicosatetraenoic acid (20-HETE), either by inhibiting its synthetic enzyme CYP450 ω-hydroxylases or by increasing nitric oxide (NO), which is a natural inhibitor of ω-hydroxylases, rescued activity-evoked capillary dilation. The capillary dilation unmasked by inhibiting 20-HETE was dependent on PGE2 activation of endoperoxide 4 (EP4) receptors, a vasodilatory pathway previously identified in healthy animals. Cortical 20-HETE levels were increased following MCAO, in agreement with data from stroke patients. Inhibition of ω-hydroxylases normalized 20-HETE levels in vivo and increased cerebral blood flow in the peri-infarct cortex. These data identify 20-HETE-dependent vasoconstriction as a mechanism underlying capillary neurovascular coupling impairment after stroke. Our results suggest that the brain's energy supply may be significantly reduced after stroke in regions previously believed to be asymptomatic and that ω-hydroxylase inhibition may restore healthy neurovascular coupling post-stroke.
Collapse
Affiliation(s)
- Zhenzhou Li
- Knight Cardiovascular Institute, Oregon Health & Science University, Portland, OR, United States
- Department of Anesthesiology, General Hospital of Ningxia Medical University, Ningxia, China
| | - Heather L. McConnell
- Department of Neurology, Jungers Center for Neurosciences Research, Oregon Health & Science University, Portland, OR, United States
| | - Teresa L. Stackhouse
- Department of Neurology, Jungers Center for Neurosciences Research, Oregon Health & Science University, Portland, OR, United States
| | - Martin M. Pike
- Advanced Imaging Research Center, Oregon Health & Science University, Portland, OR, United States
| | - Wenri Zhang
- Department of Anesthesiology and Perioperative Medicine, Oregon Health & Science University, Portland, OR, United States
| | - Anusha Mishra
- Knight Cardiovascular Institute, Oregon Health & Science University, Portland, OR, United States
- Department of Neurology, Jungers Center for Neurosciences Research, Oregon Health & Science University, Portland, OR, United States
| |
Collapse
|
13
|
Li L, Tong XK, Hosseini Kahnouei M, Vallerand D, Hamel E, Girouard H. Impaired Hippocampal Neurovascular Coupling in a Mouse Model of Alzheimer's Disease. Front Physiol 2021; 12:715446. [PMID: 34475828 PMCID: PMC8406685 DOI: 10.3389/fphys.2021.715446] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Accepted: 07/02/2021] [Indexed: 12/21/2022] Open
Abstract
Alzheimer’s disease (AD), the most common form of dementia, is characterized by neuronal degeneration and cerebrovascular dysfunction. Increasing evidence indicates that cerebrovascular dysfunction may be a key or an aggravating pathogenic factor in AD. This emphasizes the importance to investigate the tight coupling between neuronal activity and cerebral blood flow (CBF) termed neurovascular coupling (NVC). NVC depends on all cell types of the neurovascular unit within which astrocytes are important players in the progression of AD. Hence, the objective of this study was to characterize the hippocampal NVC in a mouse model of AD. Hippocampal NVC was studied in 6-month-old amyloid-beta precursor protein (APP) transgenic mice and their corresponding wild-type littermates using in vivo laser Doppler flowmetry to measure CBF in area CA1 of the hippocampus in response to Schaffer collaterals stimulation. Ex vivo two-photon microscopy experiments were performed to determine astrocytic Ca2+ and vascular responses to electrical field stimulation (EFS) or caged Ca2+ photolysis in hippocampal slices. Neuronal synaptic transmission, astrocytic endfeet Ca2+ in correlation with reactive oxygen species (ROS), and vascular reactivity in the presence or absence of Tempol, a mimetic of superoxide dismutase, were further investigated using electrophysiological, caged Ca2+ photolysis or pharmacological approaches. Whisker stimulation evoked-CBF increases and ex vivo vascular responses to EFS were impaired in APP mice compared with their age-matched controls. APP mice were also characterized by decreased basal synaptic transmission, a shorter astrocytic Ca2+ increase, and altered vascular response to elevated perivascular K+. However, long-term potentiation, astrocytic Ca2+ amplitude in response to EFS, together with vascular responses to nitric oxide remained unchanged. Importantly, we found a significantly increased Ca2+ uncaging-induced ROS production in APP mice. Tempol prevented the vascular response impairment while normalizing astrocytic Ca2+ in APP mice. These findings suggest that NVC is altered at many levels in APP mice, at least in part through oxidative stress. This points out that therapies against AD should include an antioxidative component to protect the neurovascular unit.
Collapse
Affiliation(s)
- Lin Li
- Department of Pharmacology and Physiology, Faculty of Medicine, Université de Montréal, Montréal, QC, Canada.,Groupe de Recherche sur le Système Nerveux Central (GRSNC), Université de Montréal, Montréal, QC, Canada
| | - Xin-Kang Tong
- Laboratory of Cerebrovascular Research, Montreal Neurological Institute, McGill University, Montréal, QC, Canada
| | - Mohammadamin Hosseini Kahnouei
- Department of Pharmacology and Physiology, Faculty of Medicine, Université de Montréal, Montréal, QC, Canada.,Groupe de Recherche sur le Système Nerveux Central (GRSNC), Université de Montréal, Montréal, QC, Canada.,Centre Interdisciplinaire de Recherche sur le Cerveau et l'Apprentissage (CIRCA), Université de Montréal, Montréal, QC, Canada
| | - Diane Vallerand
- Department of Pharmacology and Physiology, Faculty of Medicine, Université de Montréal, Montréal, QC, Canada.,Centre Interdisciplinaire de Recherche sur le Cerveau et l'Apprentissage (CIRCA), Université de Montréal, Montréal, QC, Canada
| | - Edith Hamel
- Laboratory of Cerebrovascular Research, Montreal Neurological Institute, McGill University, Montréal, QC, Canada
| | - Hélène Girouard
- Department of Pharmacology and Physiology, Faculty of Medicine, Université de Montréal, Montréal, QC, Canada.,Groupe de Recherche sur le Système Nerveux Central (GRSNC), Université de Montréal, Montréal, QC, Canada.,Centre Interdisciplinaire de Recherche sur le Cerveau et l'Apprentissage (CIRCA), Université de Montréal, Montréal, QC, Canada.,Centre de Recherche de l'Institut Universitaire de Gériatrie de Montréal, Montréal, QC, Canada
| |
Collapse
|
14
|
Wenceslau CF, McCarthy CG, Earley S, England SK, Filosa JA, Goulopoulou S, Gutterman DD, Isakson BE, Kanagy NL, Martinez-Lemus LA, Sonkusare SK, Thakore P, Trask AJ, Watts SW, Webb RC. Guidelines for the measurement of vascular function and structure in isolated arteries and veins. Am J Physiol Heart Circ Physiol 2021; 321:H77-H111. [PMID: 33989082 DOI: 10.1152/ajpheart.01021.2020] [Citation(s) in RCA: 83] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The measurement of vascular function in isolated vessels has revealed important insights into the structural, functional, and biomechanical features of the normal and diseased cardiovascular system and has provided a molecular understanding of the cells that constitutes arteries and veins and their interaction. Further, this approach has allowed the discovery of vital pharmacological treatments for cardiovascular diseases. However, the expansion of the vascular physiology field has also brought new concerns over scientific rigor and reproducibility. Therefore, it is appropriate to set guidelines for the best practices of evaluating vascular function in isolated vessels. These guidelines are a comprehensive document detailing the best practices and pitfalls for the assessment of function in large and small arteries and veins. Herein, we bring together experts in the field of vascular physiology with the purpose of developing guidelines for evaluating ex vivo vascular function. By using this document, vascular physiologists will have consistency among methodological approaches, producing more reliable and reproducible results.
Collapse
Affiliation(s)
- Camilla F Wenceslau
- Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
| | - Cameron G McCarthy
- Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
| | - Scott Earley
- Department of Pharmacology, Center for Molecular and Cellular Signaling in the Cardiovascular System, Reno School of Medicine, University of Nevada, Reno, Nevada
| | - Sarah K England
- Department of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis, Missouri
| | - Jessica A Filosa
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Styliani Goulopoulou
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, Texas
| | - David D Gutterman
- Department of Medicine, Medical College of Wisconsin Cardiovascular Center, Milwaukee, Wisconsin
| | - Brant E Isakson
- Department of Molecular Physiology and Biophysics, Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, Virginia
| | - Nancy L Kanagy
- Department of Cell Biology and Physiology, University of New Mexico, Albuquerque, New Mexico
| | - Luis A Martinez-Lemus
- Department of Medical Pharmacology and Physiology, Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri
| | - Swapnil K Sonkusare
- Department of Molecular Physiology and Biophysics, Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, Virginia
| | - Pratish Thakore
- Department of Pharmacology, Center for Molecular and Cellular Signaling in the Cardiovascular System, Reno School of Medicine, University of Nevada, Reno, Nevada
| | - Aaron J Trask
- Center for Cardiovascular Research, The Heart Center, The Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, Ohio.,Department of Pediatrics, The Ohio State University College of Medicine, Columbus, Ohio
| | - Stephanie W Watts
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan
| | - R Clinton Webb
- Cardiovascular Translational Research Center, Department of Cell Biology and Anatomy, University of South Carolina, Columbia, South Carolina
| |
Collapse
|
15
|
Longitudinal stability of retinal blood flow regulation in response to flicker stimulation and systemic hyperoxia in mice assessed with laser speckle flowgraphy. Sci Rep 2020; 10:19796. [PMID: 33188259 PMCID: PMC7666208 DOI: 10.1038/s41598-020-75296-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Accepted: 10/08/2020] [Indexed: 11/08/2022] Open
Abstract
This study aimed to evaluate longitudinal changes in retinal blood flow in response to flicker stimulation and systemic hyperoxia in mice using a laser speckle flowgraphy (LSFG-Micro). The retinal blood flow in vascular area surrounding the optic nerve head was measured in 8-week-old male mice every 2 weeks until age 20-week. The coefficient of variation of retinal blood flow under resting condition was analyzed every 2 weeks to validate the consistency of the measurement. On day 1 of the experiment, retinal blood flow was assessed every 20 s for 6 min during and after 3 min flicker light (12 Hz) stimulation; on day 2, retinal blood flow was measured every minute for 20 min during and after 10 min systemic hyperoxia; and on day 3, electroretinography (ERG) was performed. Body weight, systemic blood pressure, and ocular perfusion pressure increased significantly with age, but the resting retinal blood flow and ERG parameters remained unchanged. Retinal blood flow significantly increased with flicker stimulation and decreased with systemic hyperoxia, independent of age. The LSFG-Micro provides consistent and reproducible retinal blood flow measurement in adult mice. Longitudinal assessments of retinal blood flow in response to flicker stimulation and systemic hyperoxia may be useful indexes for noninvasive monitoring of vascular function in retinas.
Collapse
|
16
|
Abstract
Blood flow in the retina increases in response to light-evoked neuronal activity, ensuring that retinal neurons receive an adequate supply of oxygen and nutrients as metabolic demands vary. This response, termed "functional hyperemia," is disrupted in diabetic retinopathy. The reduction in functional hyperemia may result in retinal hypoxia and contribute to the development of retinopathy. This review will discuss the neurovascular coupling signaling mechanisms that generate the functional hyperemia response in the retina, the changes to neurovascular coupling that occur in diabetic retinopathy, possible treatments for restoring functional hyperemia and retinal oxygen levels, and changes to functional hyperemia that occur in the diabetic brain.
Collapse
|
17
|
Haselden WD, Kedarasetti RT, Drew PJ. Spatial and temporal patterns of nitric oxide diffusion and degradation drive emergent cerebrovascular dynamics. PLoS Comput Biol 2020; 16:e1008069. [PMID: 32716940 PMCID: PMC7410342 DOI: 10.1371/journal.pcbi.1008069] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 08/06/2020] [Accepted: 06/17/2020] [Indexed: 02/07/2023] Open
Abstract
Nitric oxide (NO) is a gaseous signaling molecule that plays an important role in neurovascular coupling. NO produced by neurons diffuses into the smooth muscle surrounding cerebral arterioles, driving vasodilation. However, the rate of NO degradation in hemoglobin is orders of magnitude higher than in brain tissue, though how this might impact NO signaling dynamics is not completely understood. We used simulations to investigate how the spatial and temporal patterns of NO generation and degradation impacted dilation of a penetrating arteriole in cortex. We found that the spatial location of NO production and the size of the vessel both played an important role in determining its responsiveness to NO. The much higher rate of NO degradation and scavenging of NO in the blood relative to the tissue drove emergent vascular dynamics. Large vasodilation events could be followed by post-stimulus constrictions driven by the increased degradation of NO by the blood, and vasomotion-like 0.1-0.3 Hz oscillations could also be generated. We found that these dynamics could be enhanced by elevation of free hemoglobin in the plasma, which occurs in diseases such as malaria and sickle cell anemia, or following blood transfusions. Finally, we show that changes in blood flow during hypoxia or hyperoxia could be explained by altered NO degradation in the parenchyma. Our simulations suggest that many common vascular dynamics may be emergent phenomena generated by NO degradation by the blood or parenchyma.
Collapse
Affiliation(s)
- William Davis Haselden
- Neuroscience Graduate Program, MD/PhD Medical Scientist Training Program, Pennsylvania State University, University Park, Pennsylvania, United States of America
- Department of Engineering Science and Mechanics, Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Ravi Teja Kedarasetti
- Department of Engineering Science and Mechanics, Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Patrick J. Drew
- Neuroscience Graduate Program, MD/PhD Medical Scientist Training Program, Pennsylvania State University, University Park, Pennsylvania, United States of America
- Department of Engineering Science and Mechanics, Pennsylvania State University, University Park, Pennsylvania, United States of America
- Departments of Biomedical Engineering and Neurosurgery, Pennsylvania State University, University Park, Pennsylvania, United States of America
| |
Collapse
|
18
|
Retinal capillary oximetry with visible light optical coherence tomography. Proc Natl Acad Sci U S A 2020; 117:11658-11666. [PMID: 32398376 DOI: 10.1073/pnas.1918546117] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Assessing oxygen saturation (sO2) remains challenging but is nonetheless necessary for understanding retinal metabolism. We and others previously achieved oximetry on major retinal vessels and measured the total retinal oxygen metabolic rate in rats using visible-light optical coherence tomography. Here we extend oximetry measurements to capillaries and investigate all three retinal vascular plexuses by amplifying and extracting the spectroscopic signal from each capillary segment under the guidance of optical coherence tomography (OCT) angiography. Using this approach, we measured capillary sO2 in the retinal circulation in rats, demonstrated reproducibility of the results, validated the measurements in superficial capillaries with known perfusion pathways, and determined sO2 responses to hypoxia and hyperoxia in the different retinal capillary beds. OCT capillary oximetry has the potential to provide new insights into the retinal circulation in the normal eye as well as in retinal vascular diseases.
Collapse
|
19
|
Drew PJ. Vascular and neural basis of the BOLD signal. Curr Opin Neurobiol 2019; 58:61-69. [PMID: 31336326 DOI: 10.1016/j.conb.2019.06.004] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Accepted: 06/22/2019] [Indexed: 12/26/2022]
Abstract
Neural activity in the brain is usually coupled to increases in local cerebral blood flow, leading to the increase in oxygenation that generates the BOLD fMRI signal. Recent work has begun to elucidate the vascular and neural mechanisms underlying the BOLD signal. The dilatory response is distributed throughout the vascular network. Arteries actively dilate within a second following neural activity increases, while venous distensions are passive and have a time course that last tens of seconds. Vasodilation, and thus local blood flow, is controlled by the activity of both neurons and astrocytes via multiple different pathways. The relationship between sensory-driven neural activity and the vascular dynamics in sensory areas are well-captured with a linear convolution model. However, depending on the behavioral state or brain region, the coupling between neural activity and hemodynamic signals can be weak or even inverted.
Collapse
Affiliation(s)
- Patrick J Drew
- Departments of Engineering Science and Mechanics, Biomedical Engineering and Neurosurgery, Pennsylvania State University, University Park, PA 16802, United States.
| |
Collapse
|
20
|
McConnell HL, Li Z, Woltjer RL, Mishra A. Astrocyte dysfunction and neurovascular impairment in neurological disorders: Correlation or causation? Neurochem Int 2019; 128:70-84. [PMID: 30986503 DOI: 10.1016/j.neuint.2019.04.005] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 04/08/2019] [Accepted: 04/09/2019] [Indexed: 12/14/2022]
Abstract
The neurovascular unit, consisting of neurons, astrocytes, and vascular cells, has become the focus of much discussion in the last two decades and emerging literature now suggests an association between neurovascular dysfunction and neurological disorders. In this review, we synthesize the known and suspected contributions of astrocytes to neurovascular dysfunction in disease. Throughout the brain, astrocytes are centrally positioned to dynamically mediate interactions between neurons and the cerebral vasculature, and play key roles in blood-brain barrier maintenance and neurovascular coupling. It is increasingly apparent that the changes in astrocytes in response to a variety of insults to brain tissue -collectively referred to as "reactive astrogliosis" - are not just an epiphenomenon restricted to morphological alterations, but comprise functional changes in astrocytes that contribute to the phenotype of neurological diseases with both beneficial and detrimental effects. In the context of the neurovascular unit, astrocyte dysfunction accompanies, and may contribute to, blood-brain barrier impairment and neurovascular dysregulation, highlighting the need to determine the exact nature of the relationship between astrocyte dysfunction and neurovascular impairments. Targeting astrocytes may represent a new strategy in combinatorial therapeutics for preventing the mismatch of energy supply and demand that often accompanies neurological disorders.
Collapse
Affiliation(s)
- Heather L McConnell
- Knight Cardiovascular Institute, Oregon Health & Science University, Portland, OR, United States
| | - Zhenzhou Li
- Knight Cardiovascular Institute, Oregon Health & Science University, Portland, OR, United States; Department of Anesthesiology, General Hospital of Ningxia Medical University, Yinchuan City, China
| | - Randall L Woltjer
- Department of Neuropathology, Oregon Health & Science University, Portland, OR, United States
| | - Anusha Mishra
- Knight Cardiovascular Institute, Oregon Health & Science University, Portland, OR, United States.
| |
Collapse
|
21
|
Retinal capillary perfusion: Spatial and temporal heterogeneity. Prog Retin Eye Res 2019; 70:23-54. [PMID: 30769149 DOI: 10.1016/j.preteyeres.2019.01.001] [Citation(s) in RCA: 84] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 01/18/2019] [Accepted: 01/19/2019] [Indexed: 01/17/2023]
Abstract
The central role of the cardiovascular system is to maintain adequate capillary perfusion. The spatially and temporally heterogeneous nature of capillary perfusion has been reported in some organs. However, such heterogeneous perfusion properties have not been sufficiently explored in the retina. Arguably, spatial and temporal heterogeneity of capillary perfusion could be more predominant in the retina than that in other organs. This is because the retina is one of the highest metabolic demand neural tissues yet it has a limited blood supply due to optical requirements. In addition, the unique heterogeneous distribution of retinal neural cells within different layers and regions, and the significant heterogeneity of intraretinal oxygen distribution and consumption add to the complexity. Retinal blood flow distribution must match consumption of nutrients such as oxygen and glucose within the retina at the cellular level in order to effectively maintain cell survival and function. Sophisticated local blood flow control in the microcirculation is likely required to control the retinal capillary perfusion to supply local retinal tissue and accommodate temporal and spatial variations in metabolic supply and demand. The authors would like to update the knowledge of the retinal microvessel and capillary network and retinal oxidative metabolism from their own studies and the work of others. The coupling between blood supply and energy demands in the retina is particularly interesting. We will mostly describe information regarding the retinal microvessel network and retinal oxidative metabolism relevant to the spatial and temporal heterogeneity of capillary perfusion. We believe that there is significant and necessary spatial and temporal heterogeneity and active regulation of retinal blood flow in the retina, particularly in the macular region. Recently, retinal optical coherence tomography angiography (OCTA) has been widely used in ophthalmology, both experimentally and clinically. OCTA could be a valuable tool for examining retinal microvessel and capillary network structurally and has potential for determining retinal capillary perfusion and its control. We have demonstrated spatial and temporal heterogeneity of capillary perfusion in the retina both experimentally and clinically. We have also found close relationships between the smallest arterioles and capillaries within paired arterioles and venules and determined the distribution of smooth muscle cell contraction proteins in these vessels. Spatial and temporal heterogeneity of retinal capillary perfusion could be a useful parameter to determine retinal microvessel regulatory capability as an early assay for retinal vascular diseases. This topic will be of great interest, not only for the eye but also other organs. The retina could be the best model for such investigations. Unlike cerebral vessels, retinal vessels can be seen even at the capillary level. The purpose of this manuscript is to share our current understanding with the readers and encourage more researchers and clinicians to investigate this field. We begin by reviewing the general principles of microcirculation properties and the spatial and temporal heterogeneity of the capillary perfusion in other organs, before considering the special requirements of the retina. The local heterogeneity of oxygen supply and demand in the retina and the need to have a limited and well-regulated retinal circulation to preserve the transparency of the retina is discussed. We then consider how such a delicate balance of metabolic supply and consumption is achieved. Finally we discuss how new imaging methodologies such as optical coherence tomography angiography may be able to detect the presence of spatial and temporal heterogeneity of capillary perfusion in a clinical setting. We also provide some new information of the control role of very small arterioles in the modulation of retinal capillary perfusion which could be an interesting topic for further investigation.
Collapse
|
22
|
Blair NP, Tan MR, Felder AE, Teng PY, Wanek J, Shahidi M. Retinal tissue oxygen tension and consumption during light flicker stimulation in rat. Exp Eye Res 2018; 175:207-211. [PMID: 30121195 DOI: 10.1016/j.exer.2018.08.007] [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: 04/13/2018] [Revised: 08/09/2018] [Accepted: 08/12/2018] [Indexed: 10/28/2022]
Abstract
Light flicker stimulation has been shown to increase inner retinal oxygen metabolism and supply. The purpose of the study was to test the hypothesis that sustained light flicker stimulation of various durations alters the depth profile metrics of oxygen partial pressure in the retinal tissue (tPO2) but not the outer retinal oxygen consumption rate (QO2). In 17 rats, tPO2 depth profiles were derived by phosphorescence lifetime imaging after intravitreal injection of an oxyphor. tPO2 profile metrics, including mean inner retinal tPO2, maximum outer retinal tPO2 and minimum outer retinal tPO2 were determined. QO2 was calculated using a one-dimensional oxygen diffusion model. Data were acquired at baseline (constant light illumination) and during light flicker stimulation at 10 Hz under the same mean illumination levels, and differences between values obtained during flicker and baseline were calculated. None of the tPO2 profile metrics or QO2 differences depended on the duration of light flicker stimulation (R2 ≤ 0.03). No significant change in any of the tPO2 profile metrics was detected with light flicker compared with constant light (P ≥ 0.08). Light flicker decreased QO2 from 0.53 ± 0.29 to 0.38 ± 0.30 mL O2/(min*100 gm), a reduction of 28% (P = 0.02). The retinal compensatory responses to the physiologic challenge of light flicker stimulation were effective in maintaining the levels of oxygen at or near baseline in the inner retina. Oxygen availability to the inner retina during light flicker may also have been enhanced by the decrease in QO2.
Collapse
Affiliation(s)
- Norman P Blair
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, 1855 West Taylor Street, Chicago, IL, 60612, USA.
| | - Michael R Tan
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, 1855 West Taylor Street, Chicago, IL, 60612, USA.
| | - Anthony E Felder
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, 1855 West Taylor Street, Chicago, IL, 60612, USA.
| | - Pang-Yu Teng
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, 1855 West Taylor Street, Chicago, IL, 60612, USA.
| | - Justin Wanek
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, 1855 West Taylor Street, Chicago, IL, 60612, USA.
| | - Mahnaz Shahidi
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, 1855 West Taylor Street, Chicago, IL, 60612, USA.
| |
Collapse
|
23
|
Hall CN, Howarth C, Kurth-Nelson Z, Mishra A. Interpreting BOLD: towards a dialogue between cognitive and cellular neuroscience. Philos Trans R Soc Lond B Biol Sci 2017; 371:rstb.2015.0348. [PMID: 27574302 DOI: 10.1098/rstb.2015.0348] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/11/2016] [Indexed: 12/11/2022] Open
Abstract
Cognitive neuroscience depends on the use of blood oxygenation level-dependent (BOLD) functional magnetic resonance imaging (fMRI) to probe brain function. Although commonly used as a surrogate measure of neuronal activity, BOLD signals actually reflect changes in brain blood oxygenation. Understanding the mechanisms linking neuronal activity to vascular perfusion is, therefore, critical in interpreting BOLD. Advances in cellular neuroscience demonstrating differences in this neurovascular relationship in different brain regions, conditions or pathologies are often not accounted for when interpreting BOLD. Meanwhile, within cognitive neuroscience, the increasing use of high magnetic field strengths and the development of model-based tasks and analyses have broadened the capability of BOLD signals to inform us about the underlying neuronal activity, but these methods are less well understood by cellular neuroscientists. In 2016, a Royal Society Theo Murphy Meeting brought scientists from the two communities together to discuss these issues. Here, we consolidate the main conclusions arising from that meeting. We discuss areas of consensus about what BOLD fMRI can tell us about underlying neuronal activity, and how advanced modelling techniques have improved our ability to use and interpret BOLD. We also highlight areas of controversy in understanding BOLD and suggest research directions required to resolve these issues.This article is part of the themed issue 'Interpreting BOLD: a dialogue between cognitive and cellular neuroscience'.
Collapse
Affiliation(s)
| | - Clare Howarth
- Department of Psychology, University of Sheffield, Sheffield, UK
| | - Zebulun Kurth-Nelson
- Max Planck UCL Centre for Computational Psychiatry and Ageing Research, University College London, London, UK
| | - Anusha Mishra
- Department of Neuroscience, Physiology and Pharmacology, University College London, London, UK
| |
Collapse
|
24
|
Abstract
Müller glia, the principal macroglia of the retina, express diverse subtypes of adenosine and metabotropic purinergic (P2Y) receptors. Müller cells of several species, including man, also express ionotropic P2X7 receptors. ATP is liberated from Müller cells after activation of metabotropic glutamate receptors and during osmotic and mechanical induction of membrane stretch; adenosine is released through equilibrative nucleoside transporters. Müller cell-derived purines modulate the neuronal activity and have autocrine effects, for example, induction of glial calcium waves and regulation of the cellular volume. Glial calcium waves induced by neuron-derived ATP mediate functional hyperemia in the retina. Purinergic signaling contributes to the induction of Müller cell gliosis, for example, of cellular proliferation and downregulation of potassium channels, which are important for the homeostatic functions of Müller cells. Purinergic glial calcium waves may also promote the long-range propagation of gliosis and neuronal degeneration across the retinal tissue. The osmotic ATP release is inhibited under pathological conditions. Inhibition of the ATP release may result in osmotic Müller cell swelling and dysregulation of the water transport through the cells; both may contribute to the development of retinal edema. Suppression of the osmotic ATP release and upregulation of the ecto-apyrase (NTPDase1), which facilitate the extracellular degradation of ATP and the formation of adenosine, may protect neurons and photoreceptors from death due to overactivation of P2X receptors. Pharmacological inhibition of P2X7 receptors and stimulation of adenosine receptors may represent clinical approaches to prevent retinal cell death and dysregulated cell proliferation, and to treat retinal edema.
Collapse
Affiliation(s)
- Andreas Reichenbach
- 1 Paul Flechsig Institute of Brain Research, University of Leipzig , Leipzig, Germany
| | - Andreas Bringmann
- 2 Department of Ophthalmology and Eye Hospital, University of Leipzig , Leipzig, Germany
| |
Collapse
|
25
|
Toft-Kehler AK, Skytt DM, Kolko M. A Perspective on the Müller Cell-Neuron Metabolic Partnership in the Inner Retina. Mol Neurobiol 2017; 55:5353-5361. [PMID: 28929338 DOI: 10.1007/s12035-017-0760-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Accepted: 08/31/2017] [Indexed: 10/18/2022]
Abstract
The Müller cells represent the predominant macroglial cell in the retina. In recent decades, Müller cells have been acknowledged to be far more influential on neuronal homeostasis in the retina than previously assumed. With its unique localization, spanning the entire retina being interposed between the vessels and neurons, Müller cells are responsible for the functional and metabolic support of the surrounding neurons. As a consequence of major energy demands in the retina, high levels of glucose are consumed and processed by Müller cells. The present review provides a perspective on the symbiotic relationship between Müller cells and inner retinal neurons on a cellular level by emphasizing the essential role of energy metabolism within Müller cells in relation to retinal neuron survival.
Collapse
Affiliation(s)
- A K Toft-Kehler
- Department of Drug Design and Pharmacology, University of Copenhagen, Universitetsparken 2, 2100, Copenhagen, Denmark
| | - D M Skytt
- Department of Drug Design and Pharmacology, University of Copenhagen, Universitetsparken 2, 2100, Copenhagen, Denmark
| | - Miriam Kolko
- Department of Drug Design and Pharmacology, University of Copenhagen, Universitetsparken 2, 2100, Copenhagen, Denmark. .,Department of Ophthalmology, Zealand University Hospital, Vestermarksvej 23, 4000, Roskilde, Denmark. .,Department of Ophthalmology, Copenhagen University Hospital, Rigshospitalet, Nordre Ringvej 57, 2600, Glostrup, Denmark.
| |
Collapse
|
26
|
Effects of aeration on metabolic profiles of Mortierella alpina during the production of arachidonic acid. ACTA ACUST UNITED AC 2017; 44:1225-1235. [DOI: 10.1007/s10295-017-1950-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Accepted: 05/04/2017] [Indexed: 12/27/2022]
Abstract
Abstract
To investigate the metabolic regulation against oxygen supply, comparative metabolomics was performed to explore the metabolic responses of Mortierella alpina in the process of arachidonic acid (ARA) production. More than 110 metabolites involved in Embden–Meyerhof–Parnas pathway, pentose phosphate pathway, tricarboxylic acid cycle, inositol phosphate metabolism, fatty acid biosynthesis, and amino acid metabolism were identified by gas chromatography–mass spectrometry. Samples at different aeration rates were clearly distinguished by principal components analysis and partial least squares analysis, indicating that oxygen supply had a profound effect on the metabolism of M. alpina. Eleven major metabolites were identified as potential biomarkers to be primarily responsible for the difference of metabolism. Further study of metabolic changes with the relevant pathways demonstrated that the levels of several intermediate metabolites in relation to central carbon metabolism changed remarkably via both processes and citrate and malate was supposed to play vital roles in polyunsaturated acid (PUFA) synthesis. Increase of myo-inositol and sorbitol were probably for osmo-regulation and redox balance, while enhanced phosphoric acid and pyroglutamic acid were supposed to have function in the activation of signal transduction pathway for stress resistance. The present study provides a novel insight into the metabolic responses of M. alpina to aeration rates and the metabolic characteristics during the ARA fermentation.
Collapse
|
27
|
Kisler K, Nelson AR, Montagne A, Zlokovic BV. Cerebral blood flow regulation and neurovascular dysfunction in Alzheimer disease. Nat Rev Neurosci 2017; 18:419-434. [PMID: 28515434 PMCID: PMC5759779 DOI: 10.1038/nrn.2017.48] [Citation(s) in RCA: 780] [Impact Index Per Article: 111.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Cerebral blood flow (CBF) regulation is essential for normal brain function. The mammalian brain has evolved a unique mechanism for CBF control known as neurovascular coupling. This mechanism ensures a rapid increase in the rate of CBF and oxygen delivery to activated brain structures. The neurovascular unit is composed of astrocytes, mural vascular smooth muscle cells and pericytes, and endothelia, and regulates neurovascular coupling. This Review article examines the cellular and molecular mechanisms within the neurovascular unit that contribute to CBF control, and neurovascular dysfunction in neurodegenerative disorders such as Alzheimer disease.
Collapse
Affiliation(s)
- Kassandra Kisler
- Zilkha Neurogenetic Institute, 1501 San Pablo Street, Los Angeles, California 90089, USA
| | - Amy R Nelson
- Zilkha Neurogenetic Institute, 1501 San Pablo Street, Los Angeles, California 90089, USA
| | - Axel Montagne
- Zilkha Neurogenetic Institute, 1501 San Pablo Street, Los Angeles, California 90089, USA
| | - Berislav V Zlokovic
- Zilkha Neurogenetic Institute, 1501 San Pablo Street, Los Angeles, California 90089, USA
| |
Collapse
|
28
|
Nippert AR, Biesecker KR, Newman EA. Mechanisms Mediating Functional Hyperemia in the Brain. Neuroscientist 2017; 24:73-83. [PMID: 28403673 DOI: 10.1177/1073858417703033] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Neuronal activity within the brain evokes local increases in blood flow, a response termed functional hyperemia. This response ensures that active neurons receive sufficient oxygen and nutrients to maintain tissue function and health. In this review, we discuss the functions of functional hyperemia, the types of vessels that generate the response, and the signaling mechanisms that mediate neurovascular coupling, the communication between neurons and blood vessels. Neurovascular coupling signaling is mediated primarily by the vasoactive metabolites of arachidonic acid (AA), by nitric oxide, and by K+. While much is known about these pathways, many contentious issues remain. We highlight two controversies, the role of glial cell Ca2+ signaling in mediating neurovascular coupling and the importance of capillaries in generating functional hyperemia. We propose signaling pathways that resolve these controversies. In this scheme, capillary dilations are generated by Ca2+ increases in astrocyte endfeet, leading to production of AA metabolites. In contrast, arteriole dilations are generated by Ca2+ increases in neurons, resulting in production of nitric oxide and AA metabolites. Arachidonic acid from neurons also diffuses into astrocyte endfeet where it is converted into additional vasoactive metabolites. While this scheme resolves several discrepancies in the field, many unresolved challenges remain and are discussed in the final section of the review.
Collapse
Affiliation(s)
- Amy R Nippert
- 1 Department of Neuroscience, University of Minnesota-Twin Cities, Minneapolis, MN, USA
| | - Kyle R Biesecker
- 1 Department of Neuroscience, University of Minnesota-Twin Cities, Minneapolis, MN, USA
| | - Eric A Newman
- 1 Department of Neuroscience, University of Minnesota-Twin Cities, Minneapolis, MN, USA
| |
Collapse
|
29
|
Linsenmeier RA, Zhang HF. Retinal oxygen: from animals to humans. Prog Retin Eye Res 2017; 58:115-151. [PMID: 28109737 DOI: 10.1016/j.preteyeres.2017.01.003] [Citation(s) in RCA: 159] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Revised: 01/13/2017] [Accepted: 01/17/2017] [Indexed: 10/20/2022]
Abstract
This article discusses retinal oxygenation and retinal metabolism by focusing on measurements made with two of the principal methods used to study O2 in the retina: measurements of PO2 with oxygen-sensitive microelectrodes in vivo in animals with a retinal circulation similar to that of humans, and oximetry, which can be used non-invasively in both animals and humans to measure O2 concentration in retinal vessels. Microelectrodes uniquely have high spatial resolution, allowing the mapping of PO2 in detail, and when combined with mathematical models of diffusion and consumption, they provide information about retinal metabolism. Mathematical models, grounded in experiments, can also be used to simulate situations that are not amenable to experimental study. New methods of oximetry, particularly photoacoustic ophthalmoscopy and visible light optical coherence tomography, provide depth-resolved methods that can separate signals from blood vessels and surrounding tissues, and can be combined with blood flow measures to determine metabolic rate. We discuss the effects on retinal oxygenation of illumination, hypoxia and hyperoxia, and describe retinal oxygenation in diabetes, retinal detachment, arterial occlusion, and macular degeneration. We explain how the metabolic measurements obtained from microelectrodes and imaging are different, and how they need to be brought together in the future. Finally, we argue for revisiting the clinical use of hyperoxia in ophthalmology, particularly in retinal arterial occlusions and retinal detachment, based on animal research and diffusion theory.
Collapse
Affiliation(s)
- Robert A Linsenmeier
- Biomedical Engineering Department, Northwestern University, 2145 Sheridan Road, Evanston 60208-3107, IL, USA; Neurobiology Department, Northwestern University, 2205 Tech Drive, Evanston 60208-3520, IL, USA; Ophthalmology Department, Northwestern University, 645 N. Michigan Ave, Suite 440, Chicago 60611, IL, USA.
| | - Hao F Zhang
- Biomedical Engineering Department, Northwestern University, 2145 Sheridan Road, Evanston 60208-3107, IL, USA; Ophthalmology Department, Northwestern University, 645 N. Michigan Ave, Suite 440, Chicago 60611, IL, USA.
| |
Collapse
|
30
|
Mishra A, Reynolds JP, Chen Y, Gourine AV, Rusakov DA, Attwell D. Astrocytes mediate neurovascular signaling to capillary pericytes but not to arterioles. Nat Neurosci 2016; 19:1619-1627. [PMID: 27775719 PMCID: PMC5131849 DOI: 10.1038/nn.4428] [Citation(s) in RCA: 372] [Impact Index Per Article: 46.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Accepted: 09/20/2016] [Indexed: 12/11/2022]
Abstract
Active neurons increase their energy supply by dilating nearby arterioles and capillaries. This neurovascular coupling underlies blood oxygen level-dependent functional imaging signals, but its mechanism is controversial. Canonically, neurons release glutamate to activate metabotropic glutamate receptor 5 (mGluR5) on astrocytes, evoking Ca2+ release from internal stores, activating phospholipase A2 and generating vasodilatory arachidonic acid derivatives. However, adult astrocytes lack mGluR5, and knockout of the inositol 1,4,5-trisphosphate receptors that release Ca2+ from stores does not affect neurovascular coupling. We now show that buffering astrocyte Ca2+ inhibits neuronally evoked capillary dilation, that astrocyte [Ca2+]i is raised not by release from stores but by entry through ATP-gated channels, and that Ca2+ generates arachidonic acid via phospholipase D2 and diacylglycerol lipase rather than phospholipase A2. In contrast, dilation of arterioles depends on NMDA receptor activation and Ca2+-dependent NO generation by interneurons. These results reveal that different signaling cascades regulate cerebral blood flow at the capillary and arteriole levels.
Collapse
Affiliation(s)
- Anusha Mishra
- Department of Neuroscience, Physiology &Pharmacology, University College London, London, UK
| | | | - Yang Chen
- Department of Neuroscience, Physiology &Pharmacology, University College London, London, UK
| | - Alexander V Gourine
- Department of Neuroscience, Physiology &Pharmacology, University College London, London, UK
| | | | - David Attwell
- Department of Neuroscience, Physiology &Pharmacology, University College London, London, UK
| |
Collapse
|
31
|
Mishra A. Binaural blood flow control by astrocytes: listening to synapses and the vasculature. J Physiol 2016; 595:1885-1902. [PMID: 27619153 DOI: 10.1113/jp270979] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Accepted: 07/15/2016] [Indexed: 12/28/2022] Open
Abstract
Astrocytes are the most common glial cells in the brain with fine processes and endfeet that intimately contact both neuronal synapses and the cerebral vasculature. They play an important role in mediating neurovascular coupling (NVC) via several astrocytic Ca2+ -dependent signalling pathways such as K+ release through BK channels, and the production and release of arachidonic acid metabolites. They are also involved in maintaining the resting tone of the cerebral vessels by releasing ATP and COX-1 derivatives. Evidence also supports a role for astrocytes in maintaining blood pressure-dependent change in cerebrovascular tone, and perhaps also in blood vessel-to-neuron signalling as posited by the 'hemo-neural hypothesis'. Thus, astrocytes are emerging as new stars in preserving the intricate balance between the high energy demand of active neurons and the supply of oxygen and nutrients from the blood by maintaining both resting blood flow and activity-evoked changes therein. Following neuropathology, astrocytes become reactive and many of their key signalling mechanisms are altered, including those involved in NVC. Furthermore, as they can respond to changes in vascular pressure, cardiovascular diseases might exert previously unknown effects on the central nervous system by altering astrocyte function. This review discusses the role of astrocytes in neurovascular signalling in both physiology and pathology, and the impact of these findings on understanding BOLD-fMRI signals.
Collapse
Affiliation(s)
- Anusha Mishra
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, London, WC1E 6BT, UK
| |
Collapse
|
32
|
Filosa JA, Morrison HW, Iddings JA, Du W, Kim KJ. Beyond neurovascular coupling, role of astrocytes in the regulation of vascular tone. Neuroscience 2016; 323:96-109. [PMID: 25843438 PMCID: PMC4592693 DOI: 10.1016/j.neuroscience.2015.03.064] [Citation(s) in RCA: 138] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2014] [Revised: 03/24/2015] [Accepted: 03/27/2015] [Indexed: 12/22/2022]
Abstract
The brain possesses two intricate mechanisms that fulfill its continuous metabolic needs: cerebral autoregulation, which ensures constant cerebral blood flow over a wide range of arterial pressures and functional hyperemia, which ensures rapid delivery of oxygen and glucose to active neurons. Over the past decade, a number of important studies have identified astrocytes as key intermediaries in neurovascular coupling (NVC), the mechanism by which active neurons signal blood vessels to change their diameter. Activity-dependent increases in astrocytic Ca(2+) activity are thought to contribute to the release of vasoactive substances that facilitate arteriole vasodilation. A number of vasoactive signals have been identified and their role on vessel caliber assessed both in vitro and in vivo. In this review, we discuss mechanisms implicating astrocytes in NVC-mediated vascular responses, limitations encountered as a result of the challenges in maintaining all the constituents of the neurovascular unit intact and deliberate current controversial findings disputing a main role for astrocytes in NVC. Finally, we briefly discuss the potential role of pericytes and microglia in NVC-mediated processes.
Collapse
Affiliation(s)
- J A Filosa
- Georgia Regents University, 1120 15th Street, Augusta, GA 30912, United States.
| | - H W Morrison
- University of Arizona, 1305 N. Martin Avenue, P.O. Box 210203, Tucson, AZ 85721, United States
| | - J A Iddings
- Georgia Regents University, 1120 15th Street, Augusta, GA 30912, United States
| | - W Du
- Georgia Regents University, 1120 15th Street, Augusta, GA 30912, United States
| | - K J Kim
- Georgia Regents University, 1120 15th Street, Augusta, GA 30912, United States
| |
Collapse
|
33
|
Prada D, Harris A, Guidoboni G, Siesky B, Huang AM, Arciero J. Autoregulation and neurovascular coupling in the optic nerve head. Surv Ophthalmol 2016; 61:164-86. [DOI: 10.1016/j.survophthal.2015.10.004] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2015] [Revised: 10/02/2015] [Accepted: 10/02/2015] [Indexed: 12/23/2022]
|
34
|
Newman EA. Glial cell regulation of neuronal activity and blood flow in the retina by release of gliotransmitters. Philos Trans R Soc Lond B Biol Sci 2016; 370:rstb.2014.0195. [PMID: 26009774 DOI: 10.1098/rstb.2014.0195] [Citation(s) in RCA: 141] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Astrocytes in the brain release transmitters that actively modulate neuronal excitability and synaptic efficacy. Astrocytes also release vasoactive agents that contribute to neurovascular coupling. As reviewed in this article, Müller cells, the principal retinal glial cells, modulate neuronal activity and blood flow in the retina. Stimulated Müller cells release ATP which, following its conversion to adenosine by ectoenzymes, hyperpolarizes retinal ganglion cells by activation of A1 adenosine receptors. This results in the opening of G protein-coupled inwardly rectifying potassium (GIRK) channels and small conductance Ca(2+)-activated K(+) (SK) channels. Tonic release of ATP also contributes to the generation of tone in the retinal vasculature by activation of P2X receptors on vascular smooth muscle cells. Vascular tone is lost when glial cells are poisoned with the gliotoxin fluorocitrate. The glial release of vasoactive metabolites of arachidonic acid, including prostaglandin E2 (PGE2) and epoxyeicosatrienoic acids (EETs), contributes to neurovascular coupling in the retina. Neurovascular coupling is reduced when neuronal stimulation of glial cells is interrupted and when the synthesis of arachidonic acid metabolites is blocked. Neurovascular coupling is compromised in diabetic retinopathy owing to the loss of glial-mediated vasodilation. This loss can be reversed by inhibiting inducible nitric oxide synthase. It is likely that future research will reveal additional important functions of the release of transmitters from glial cells.
Collapse
Affiliation(s)
- Eric A Newman
- Department of Neuroscience, University of Minnesota, 6-145 Jackson Hall, 321 Church Street SE, Minneapolis, MN 55455, USA
| |
Collapse
|
35
|
Rosenegger DG, Gordon GR. A slow or modulatory role of astrocytes in neurovascular coupling. Microcirculation 2015; 22:197-203. [PMID: 25556627 DOI: 10.1111/micc.12184] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Accepted: 12/16/2014] [Indexed: 11/30/2022]
Abstract
Astrocytes are thought to play an important role in NVC, a process that allows the brain to locally control blood flow in response to changes in activity. However, there is an ongoing debate as to when, and under what conditions astrocyte activity is required. In the following review we set forth the hypotheses that astrocytes: (i) act to modulate but not initiate functional hyperemia and (ii) help set the basal tone state of the brain microvasculature by the tonic release of vaso-active messengers. Through these actions astrocytes could help match metabolic demand with supply over a spectrum of activity timescales.
Collapse
Affiliation(s)
- David George Rosenegger
- Hotchkiss Brain Institute, Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | | |
Collapse
|
36
|
Palkovits S, Lasta M, Told R, Schmidl D, Werkmeister R, Cherecheanu AP, Garhöfer G, Schmetterer L. Relation of retinal blood flow and retinal oxygen extraction during stimulation with diffuse luminance flicker. Sci Rep 2015; 5:18291. [PMID: 26672758 PMCID: PMC4682144 DOI: 10.1038/srep18291] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Accepted: 09/02/2015] [Indexed: 02/06/2023] Open
Abstract
Cerebral and retinal blood flow are dependent on local neuronal activity. Several studies quantified the increase in cerebral blood flow and oxygen consumption during activity. In the present study we investigated the relation between changes in retinal blood flow and oxygen extraction during stimulation with diffuse luminance flicker and the influence of breathing gas mixtures with different fractions of O2 (FiO2; 100% 15% and 12%). Twenty-four healthy subjects were included. Retinal blood flow was studied by combining measurement of vessel diameters using the Dynamic Vessel Analyser with measurements of blood velocity using laser Doppler velocimetry. Oxygen saturation was measured using spectroscopic reflectometry and oxygen extraction was calculated. Flicker stimulation increased retinal blood flow (57.7 ± 17.8%) and oxygen extraction (34.6 ± 24.1%; p < 0.001 each). During 100% oxygen breathing the response of retinal blood flow and oxygen extraction was increased (p < 0.01 each). By contrast, breathing gas mixtures with 12% and 15% FiO2 did not alter flicker–induced retinal haemodynamic changes. The present study indicates that at a comparable increase in blood flow the increase in oxygen extraction in the retina is larger than in the brain. During systemic hyperoxia the blood flow and oxygen extraction responses to neural stimulation are augmented. The underlying mechanism is unknown.
Collapse
Affiliation(s)
- Stefan Palkovits
- Department of Clinical Pharmacology Medical University of Vienna Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Michael Lasta
- Department of Clinical Pharmacology Medical University of Vienna Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Reinhard Told
- Department of Clinical Pharmacology Medical University of Vienna Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Doreen Schmidl
- Department of Clinical Pharmacology Medical University of Vienna Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - René Werkmeister
- Department of Clinical Pharmacology Medical University of Vienna Waehringer Guertel 18-20, 1090 Vienna, Austria.,Center for Medical Physics and Biomedical Engineering Medical University of Vienna Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Alina Popa Cherecheanu
- Department of Ophthalmology, Emergency University Hospital 169, Splaiul Independentei St., District 5, Bucharest, Romania
| | - Gerhard Garhöfer
- Department of Clinical Pharmacology Medical University of Vienna Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Leopold Schmetterer
- Department of Clinical Pharmacology Medical University of Vienna Waehringer Guertel 18-20, 1090 Vienna, Austria.,Center for Medical Physics and Biomedical Engineering Medical University of Vienna Waehringer Guertel 18-20, 1090 Vienna, Austria
| |
Collapse
|
37
|
Grosche A, Hauser A, Lepper MF, Mayo R, von Toerne C, Merl-Pham J, Hauck SM. The Proteome of Native Adult Müller Glial Cells From Murine Retina. Mol Cell Proteomics 2015; 15:462-80. [PMID: 26324419 PMCID: PMC4739667 DOI: 10.1074/mcp.m115.052183] [Citation(s) in RCA: 116] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Indexed: 12/26/2022] Open
Abstract
To date, the proteomic profiling of Müller cells, the dominant macroglia of the retina, has been hampered because of the absence of suitable enrichment methods. We established a novel protocol to isolate native, intact Müller cells from adult murine retinae at excellent purity which retain in situ morphology and are well suited for proteomic analyses. Two different strategies of sample preparation - an in StageTips (iST) and a subcellular fractionation approach including cell surface protein profiling were used for quantitative liquid chromatography-mass spectrometry (LC-MSMS) comparing Müller cell-enriched to depleted neuronal fractions. Pathway enrichment analyses on both data sets enabled us to identify Müller cell-specific functions which included focal adhesion kinase signaling, signal transduction mediated by calcium as second messenger, transmembrane neurotransmitter transport and antioxidant activity. Pathways associated with RNA processing, cellular respiration and phototransduction were enriched in the neuronal subpopulation. Proteomic results were validated for selected Müller cell genes by quantitative real time PCR, confirming the high expression levels of numerous members of the angiogenic and anti-inflammatory annexins and antioxidant enzymes (e.g. paraoxonase 2, peroxiredoxin 1, 4 and 6). Finally, the significant enrichment of antioxidant proteins in Müller cells was confirmed by measurements on vital retinal cells using the oxidative stress indicator CM-H2DCFDA. In contrast to photoreceptors or bipolar cells, Müller cells were most efficiently protected against H2O2-induced reactive oxygen species formation, which is in line with the protein repertoire identified in the proteomic profiling. Our novel approach to isolate intact glial cells from adult retina in combination with proteomic profiling enabled the identification of novel Müller glia specific proteins, which were validated as markers and for their functional impact in glial physiology. This provides the basis to allow the discovery of novel glial specializations and will enable us to elucidate the role of Müller cells in retinal pathologies — a topic still controversially discussed.
Collapse
Affiliation(s)
- Antje Grosche
- From the ‡Insitute of Human Genetics, University of Regensburg, D-93053 Regensburg, Germany;
| | - Alexandra Hauser
- From the ‡Insitute of Human Genetics, University of Regensburg, D-93053 Regensburg, Germany
| | - Marlen Franziska Lepper
- §Research Unit Protein Science, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), D-85764 Neuherberg, Germany
| | - Rebecca Mayo
- From the ‡Insitute of Human Genetics, University of Regensburg, D-93053 Regensburg, Germany
| | - Christine von Toerne
- §Research Unit Protein Science, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), D-85764 Neuherberg, Germany
| | - Juliane Merl-Pham
- §Research Unit Protein Science, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), D-85764 Neuherberg, Germany
| | - Stefanie M Hauck
- §Research Unit Protein Science, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), D-85764 Neuherberg, Germany
| |
Collapse
|
38
|
The progress in understanding and treatment of diabetic retinopathy. Prog Retin Eye Res 2015; 51:156-86. [PMID: 26297071 DOI: 10.1016/j.preteyeres.2015.08.001] [Citation(s) in RCA: 633] [Impact Index Per Article: 70.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Revised: 08/12/2015] [Accepted: 08/13/2015] [Indexed: 12/15/2022]
Abstract
Diabetic retinopathy is the most frequently occurring complication of diabetes mellitus and remains a leading cause of vision loss globally. Its aetiology and pathology have been extensively studied for half a century, yet there are disappointingly few therapeutic options. Although some new treatments have been introduced for diabetic macular oedema (DMO) (e.g. intravitreal vascular endothelial growth factor inhibitors ('anti-VEGFs') and new steroids), up to 50% of patients fail to respond. Furthermore, for people with proliferative diabetic retinopathy (PDR), laser photocoagulation remains a mainstay therapy, even though it is an inherently destructive procedure. This review summarises the clinical features of diabetic retinopathy and its risk factors. It describes details of retinal pathology and how advances in our understanding of pathogenesis have led to identification of new therapeutic targets. We emphasise that although there have been significant advances, there is still a pressing need for a better understanding basic mechanisms enable development of reliable and robust means to identify patients at highest risk, and to intervene effectively before vision loss occurs.
Collapse
|
39
|
Noonan JE, Lamoureux EL, Sarossy M. Neuronal activity-dependent regulation of retinal blood flow. Clin Exp Ophthalmol 2015; 43:673-82. [PMID: 25824961 DOI: 10.1111/ceo.12530] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Accepted: 03/23/2015] [Indexed: 02/01/2023]
Abstract
Blood flow in the retina is intrinsically regulated to meet the metabolic demands of its constituent cells. Flickering light or stationary contrast reversals induce an increase in blood flow within seconds of the stimulus onset. This phenomenon is thought to compensate for an increase in ganglion cell activity and energy consumption. Ganglion cell activity is in turn dependent on signals from photoreceptors, bipolar cells, horizontal cells and amacrine cells. The physiological properties of these neurons determine how each type is affected by a particular light characteristic. Neuronal activity then triggers the release of signalling molecules that dilate local blood vessels and increase blood flow. Nitric oxide has been implicated as an important mediator, but metabolites of arachidonic acid may also be involved. Detailed elucidation of these mechanisms, together with advances in imaging technology, may facilitate the use of neurovascular tests to improve the detection of retinal damage in pathological conditions.
Collapse
Affiliation(s)
- Jonathan E Noonan
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, University of Melbourne, Melbourne, Victoria, Australia
| | - Ecosse L Lamoureux
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, University of Melbourne, Melbourne, Victoria, Australia.,Singapore Eye Research Institute, Department of Ophthalmology, National University of Singapore, Singapore.,Duke-NUS Graduate Medical School, National University of Singapore, Singapore
| | - Marc Sarossy
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
40
|
Reichenbach A, Bringmann A. Purinergic signaling in retinal degeneration and regeneration. Neuropharmacology 2015; 104:194-211. [PMID: 25998275 DOI: 10.1016/j.neuropharm.2015.05.005] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Revised: 05/07/2015] [Accepted: 05/07/2015] [Indexed: 02/01/2023]
Abstract
Purinergic signaling is centrally involved in mediating the degeneration of the injured and diseased retina, the induction of retinal gliosis, and the protection of the retinal tissue from degeneration. Dysregulated calcium signaling triggered by overactivation of P2X7 receptors is a crucial step in the induction of neuronal and microvascular cell death under pathogenic conditions like ischemia-hypoxia, elevated intraocular pressure, and diabetes, respectively. Overactivation of P2X7 plays also a pathogenic role in inherited and age-related photoreceptor cell death and in the age-related dysfunction and degeneration of the retinal pigment epithelium. Gliosis of micro- and macroglial cells, which is induced and/or modulated by purinergic signaling and associated with an impaired homeostatic support to neurons, and the ATP-mediated propagation of retinal gliosis from a focal injury into the surrounding noninjured tissue are involved in inducing secondary cell death in the retina. On the other hand, alterations in the glial metabolism of extracellular nucleotides, resulting in a decreased level of ATP and an increased level of adenosine, may be neuroprotective in the diseased retina. Purinergic signals stimulate the proliferation of retinal glial cells which contributes to glial scarring which has protective effects on retinal degeneration and adverse effects on retinal regeneration. Pharmacological modulation of purinergic receptors, e.g., inhibition of P2X and activation of adenosine receptors, may have clinical importance for the prevention of photoreceptor, neuronal, and microvascular cell death in diabetic retinopathy, retinitis pigmentosa, age-related macular degeneration, and glaucoma, respectively, for the clearance of retinal edema, and the inhibition of dysregulated cell proliferation in proliferative retinopathies. This article is part of a Special Issue entitled 'Purines in Neurodegeneration and Neuroregeneration'.
Collapse
Affiliation(s)
- Andreas Reichenbach
- Paul Flechsig Institute of Brain Research, University of Leipzig, Leipzig, Germany.
| | - Andreas Bringmann
- Department of Ophthalmology and Eye Hospital, University of Leipzig, Leipzig, Germany
| |
Collapse
|
41
|
Tran CHT, Gordon GR. Astrocyte and Microvascular Imaging in Awake Animals Using Two-Photon Microscopy. Microcirculation 2015; 22:219-27. [DOI: 10.1111/micc.12188] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2014] [Accepted: 01/06/2015] [Indexed: 12/22/2022]
Affiliation(s)
- Cam Ha T. Tran
- Hotchkiss Brain Institute; Department of Physiology and Pharmacology; Cumming School of Medicine; University of Calgary; Calgary Alberta Canada
| | - Grant R. Gordon
- Hotchkiss Brain Institute; Department of Physiology and Pharmacology; Cumming School of Medicine; University of Calgary; Calgary Alberta Canada
| |
Collapse
|
42
|
MacVicar BA, Newman EA. Astrocyte regulation of blood flow in the brain. Cold Spring Harb Perspect Biol 2015; 7:cshperspect.a020388. [PMID: 25818565 DOI: 10.1101/cshperspect.a020388] [Citation(s) in RCA: 227] [Impact Index Per Article: 25.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Neuronal activity results in increased blood flow in the brain, a response named functional hyperemia. Astrocytes play an important role in mediating this response. Neurotransmitters released from active neurons evoke Ca(2+) increases in astrocytes, leading to the release of vasoactive metabolites of arachidonic acid from astrocyte endfeet onto blood vessels. Synthesis of prostaglandin E2 (PGE2) and epoxyeicosatrienoic acids (EETs) dilate blood vessels, whereas 20-hydroxyeicosatetraenoic acid (20-HETE) constricts vessels. The release of K(+) from astrocyte endfeet may also contribute to vasodilation. Oxygen modulates astrocyte regulation of blood flow. Under normoxic conditions, astrocytic Ca(2+) signaling results in vasodilation, whereas under hyperoxic conditions, vasoconstriction is favored. Astrocytes also contribute to the generation of vascular tone. Tonic release of both 20-HETE and ATP from astrocytes constricts vascular smooth muscle cells, generating vessel tone. Under pathological conditions, including Alzheimer's disease and diabetic retinopathy, disruption of normal astrocyte physiology can compromise the regulation of blood flow.
Collapse
Affiliation(s)
- Brian A MacVicar
- Djavad Mowafaghian Centre for Brain Health, Department of Psychiatry, University of British Columbia, Vancouver, British Columbia V6T 2B5, Canada
| | - Eric A Newman
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota 55455
| |
Collapse
|
43
|
|
44
|
Effect of increased oxygen tension on flicker-induced vasodilatation in the human retina. J Cereb Blood Flow Metab 2014; 34:1914-8. [PMID: 25248833 PMCID: PMC4269745 DOI: 10.1038/jcbfm.2014.161] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2014] [Revised: 08/04/2014] [Accepted: 08/12/2014] [Indexed: 12/04/2022]
Abstract
In the retina, blood flow and neural activity are tightly coupled. Stimulation of the retina with flickering light is accompanied by an increase in blood flow. The current study seeks to investigate whether an increase in oxygen tension modulates flicker (FL)-induced vasodilatation in the human retina. A total of 52 healthy volunteers were included. Via a breathing mask, 100% oxygen (O(2)) was administered in one, a mixture of 8% carbon dioxide and 92% oxygen (C/O) in a second cohort. Retinal vessel diameters were measured with a Vessel Analyzer and FL responses were assessed before and during the breathing periods. At baseline, FL stimulation increased retinal vessel diameters by +3.7±2.3% in arteries and by +5.1±3.7% in veins. Breathing of C/O led to a decrease in arterial (-9.0±6.9%) and venous (-11.3±5.9%) vessel calibers. Flicker response was increased to 5.7±2.5% in arteries and to 8.6±4.1% in veins. Breathing of pure O2 induced a vasoconstriction of vessel diameters by -14.0±5.3% in arteries and -18.4±7.0% in veins and increased FL responses in arteries (+6.2±2.8%) and veins (+7.2±3.1%). Systemic hyperoxia increases FL-induced retinal vasodilatation in the retina. The mechanism by which oxygen modulates the hyperemic response to FL stimulation remains to be elucidated.
Collapse
|
45
|
Astrocytic Gq-GPCR-linked IP3R-dependent Ca2+ signaling does not mediate neurovascular coupling in mouse visual cortex in vivo. J Neurosci 2014; 34:13139-50. [PMID: 25253859 DOI: 10.1523/jneurosci.2591-14.2014] [Citation(s) in RCA: 149] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Local blood flow is modulated in response to changing patterns of neuronal activity (Roy and Sherrington, 1890), a process termed neurovascular coupling. It has been proposed that the central cellular pathway driving this process is astrocytic Gq-GPCR-linked IP3R-dependent Ca(2+) signaling, though in vivo tests of this hypothesis are largely lacking. We examined the impact of astrocytic Gq-GPCR and IP3R-dependent Ca(2+) signaling on cortical blood flow in awake, lightly sedated, responsive mice using multiphoton laser-scanning microscopy and novel genetic tools that enable the selective manipulation of astrocytic signaling pathways in vivo. Selective stimulation of astrocytic Gq-GPCR cascades and downstream Ca(2+) signaling with the hM3Dq DREADD (designer receptors exclusively activated by designer drugs) designer receptor system was insufficient to modulate basal cortical blood flow. We found no evidence of observable astrocyte endfeet Ca(2+) elevations following physiological visual stimulation despite robust dilations of adjacent arterioles using cyto-GCaMP3 and Lck-GCaMP6s, the most sensitive Ca(2+) indicator available. Astrocytic Ca(2+) elevations could be evoked when inducing the startle response with unexpected air puffs. However, startle-induced astrocytic Ca(2+) signals did not precede corresponding startle-induced hemodynamic changes. Further, neurovascular coupling was intact in lightly sedated, responsive mice genetically lacking astrocytic IP3R-dependent Ca(2+) signaling (IP3R2 KO). These data demonstrate that astrocytic Gq-GPCR-linked IP3R-dependent Ca(2+) signaling does not mediate neurovascular coupling in visual cortex of awake, lightly sedated, responsive mice.
Collapse
|
46
|
Age-related visual signal changes induced by hypoxemic hypoxia: a study on aircraft pilots of different ages. J Clin Neurophysiol 2014; 31:469-73. [PMID: 25271687 DOI: 10.1097/wnp.0000000000000079] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
PURPOSE Exposure to high altitude leads to a series of alterations of higher nervous functions because of hypobaric hypoxia. Sensory systems, mainly the visual one, seem to be particularly involved. This study aimed to assess the effects of hypoxemic hypoxia on the transmission of the visual stimulus simulating a condition of breathing at an altitude of 18,000 feet (5,486 m) through the administration of an air mixture with 10% O2. METHODS The subjects involved in the study were 98 pilots of military aircraft (male, acclimated, healthy, 20/20 Uncorrected Visual Acuity (UCVA)/Best Corrected Visual Acuity (BCVA), and aged between 26 and 49 years) divided into 2 groups according to age (A: 26-36 years; B: 37-49 years). The visual evoked potentials were initially recorded at sea level (760 mm Hg) and subsequently at a simulated altitude of 18,000 feet (5,486 m) through the administration of an air mixture with 10% O2 that induced a blood saturation of 80% O2 after 15 minutes. The analysis was carried out using two different kinds of stimulus (15' and 60' of arc). The latency and the amplitude of N-75 (N1) and P-100 (P1) waves have been evaluated. Results obtained from visual evoked potentials were analyzed with Student t-test. RESULTS In the first group (pilots aged 26-36 years), an increase in both latency and amplitude of P-100 wave was observed and in the second group (pilots aged 37-49 years), an increase was found in latency and a significant reduction in amplitude. CONCLUSIONS The study suggests the existence of a mechanism or a particular anatomic and physiologic condition (probably the neurovascular coupling) that connects the local neuronal activity and the resulting changes in cerebral perfusion. This complex series of events binds together different structures and cell types, and it seems that younger people have a better resistance against the hypoxic insult to the central nervous system because of more efficient compensatory mechanisms.
Collapse
|
47
|
Abstract
Astrocytes have been found to play important roles in physiology being fundamental for ionic homeostasis and glutamate clearance from the synaptic cleft by their plasma membrane glutamate transporters. Astrocytes are electrically non-excitable, but they exhibit Ca(2+) signaling, which now has been demonstrated to serve as an indirect mediator of neuron-glia bidirectional interactions through gliotransmission via tripartite synapses and to modulate synaptic function and plasticity. Spontaneous astrocytic Ca(2+) signaling was observed in vivo. Intercellular Ca(2+) waves in astrocytes can be evoked by a variety of stimulations. Astrocytes are critically involved in many pathological conditions including ischemic stroke. For example, it is well known that astrocytes become reactive and form glial scar after stroke. In animal models of some brain disorders, astrocytes have been shown to exhibit enhanced Ca(2+) excitability featured as regenerative intercellular Ca(2+) waves. This chapter briefly summarizes astrocytic Ca(2+) signaling pathways under normal conditions and in experimental in vitro and in vivo ischemic models. It discusses the possible mechanisms and therapeutic implication underlying the enhanced astrocytic Ca(2+) excitability in stroke.
Collapse
Affiliation(s)
- Shinghua Ding
- Dalton Cardiovascular Research Center, Department of Bioengineering, University of Missouri-Columbia, 134 Research Park Drive, Columbia, MO, 65211, USA,
| |
Collapse
|
48
|
Howarth C. The contribution of astrocytes to the regulation of cerebral blood flow. Front Neurosci 2014; 8:103. [PMID: 24847203 PMCID: PMC4023041 DOI: 10.3389/fnins.2014.00103] [Citation(s) in RCA: 128] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2014] [Accepted: 04/18/2014] [Indexed: 12/31/2022] Open
Abstract
In order to maintain normal brain function, it is critical that cerebral blood flow (CBF) is matched to neuronal metabolic needs. Accordingly, blood flow is increased to areas where neurons are more active (a response termed functional hyperemia). The tight relationships between neuronal activation, glial cell activity, cerebral energy metabolism, and the cerebral vasculature, known as neurometabolic and neurovascular coupling, underpin functional MRI (fMRI) signals but are incompletely understood. As functional imaging techniques, particularly BOLD fMRI, become more widely used, their utility hinges on our ability to accurately and reliably interpret the findings. A growing body of data demonstrates that astrocytes can serve as a "bridge," relaying information on the level of neural activity to blood vessels in order to coordinate oxygen and glucose delivery with the energy demands of the tissue. It is widely assumed that calcium-dependent release of vasoactive substances by astrocytes results in arteriole dilation and the increased blood flow which accompanies neuronal activity. However, the signaling molecules responsible for this communication between astrocytes and blood vessels are yet to be definitively confirmed. Indeed, there is controversy over whether activity-induced changes in astrocyte calcium are widespread and fast enough to elicit such functional hyperemia responses. In this review, I will summarize the evidence which has convincingly demonstrated that astrocytes are able to modify the diameter of cerebral arterioles. I will discuss the prevalence, presence, and timing of stimulus-induced astrocyte calcium transients and describe the evidence for and against the role of calcium-dependent formation and release of vasoactive substances by astrocytes. I will also review alternative mechanisms of astrocyte-evoked changes in arteriole diameter and consider the questions which remain to be answered in this exciting area of research.
Collapse
Affiliation(s)
- Clare Howarth
- Department of Psychology, University of Sheffield Sheffield, UK
| |
Collapse
|
49
|
Kur J, Newman EA. Purinergic control of vascular tone in the retina. J Physiol 2014; 592:491-504. [PMID: 24277867 PMCID: PMC3930435 DOI: 10.1113/jphysiol.2013.267294] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2013] [Accepted: 11/18/2013] [Indexed: 12/19/2022] Open
Abstract
Purinergic control of vascular tone in the CNS has been largely unexplored. This study examines the contribution of endogenous extracellular ATP, acting on vascular smooth muscle cells, in controlling vascular tone in the in vivo rat retina. Retinal vessels were labelled by i.v. injection of a fluorescent dye and imaged with scanning laser confocal microscopy. The diameters of primary arterioles were monitored under control conditions and following intravitreal injection of pharmacological agents. Apyrase (500 units ml(-1)), an ATP hydrolysing enzyme, dilated retinal arterioles by 40.4 ± 2.8%, while AOPCP (12.5 mm), an ecto-5'-nucleotidase inhibitor that increases extracellular ATP levels, constricted arterioles by 58.0 ± 3.8% (P < 0.001 for both), demonstrating the importance of ATP in the control of basal vascular tone. Suramin (500 μm), a broad-spectrum P2 receptor antagonist, dilated retinal arterioles by 50.9 ± 3.7% (P < 0.001). IsoPPADS (300 μm) and TNP-ATP (50 μm), more selective P2X antagonists, dilated arterioles by 41.0 ± 5.3% and 55.2 ± 6.1% respectively (P < 0.001 for both). NF023 (50 μm), a potent antagonist of P2X1 receptors, dilated retinal arterioles by 32.1 ± 2.6% (P < 0.001). A438079 (500 μm) and AZ10606120 (50 μm), P2X7 antagonists, had no effect on basal vascular tone (P = 0.99 and P = 1.00 respectively). In the ex vivo retina, the P2X1 receptor agonist α,β-methylene ATP (300 nm) evoked sustained vasoconstrictions of 18.7 ± 3.2% (P < 0.05). In vivo vitreal injection of the gliotoxin fluorocitrate (150 μm) dilated retinal vessels by 52.3 ± 1.1% (P < 0.001) and inhibited the vasodilatory response to NF023 (50 μm, 7.9 ± 2.0%; P < 0.01). These findings suggest that vascular tone in rat retinal arterioles is maintained by tonic release of ATP from the retina. ATP acts on P2X1 receptors, although contributions from other P2X and P2Y receptors cannot be ruled out. Retinal glial cells are a possible source of the vasoconstricting ATP.
Collapse
Affiliation(s)
- Joanna Kur
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA.
| | | |
Collapse
|
50
|
Mishra A, O'Farrell FM, Reynell C, Hamilton NB, Hall CN, Attwell D. Imaging pericytes and capillary diameter in brain slices and isolated retinae. Nat Protoc 2014; 9:323-36. [PMID: 24434801 DOI: 10.1038/nprot.2014.019] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
The cerebral circulation is highly specialized, both structurally and functionally, and it provides a fine-tuned supply of oxygen and nutrients to active regions of the brain. Our understanding of blood flow regulation by cerebral arterioles has evolved rapidly. Recent work has opened new avenues in microvascular research; for example, it has been demonstrated that contractile pericytes found on capillary walls induce capillary diameter changes in response to neurotransmitters, suggesting that pericytes could have a role in neurovascular coupling. This concept is at odds with traditional models of brain blood flow regulation, which assume that only arterioles control cerebral blood flow. The investigation of mechanisms underlying neurovascular coupling at the capillary level requires a range of approaches, which involve unique technical challenges. Here we provide detailed protocols for the successful physiological and immunohistochemical study of pericytes and capillaries in brain slices and isolated retinae, allowing investigators to probe the role of capillaries in neurovascular coupling. This protocol can be completed within 6-8 h; however, immunohistochemical experiments may take 3-6 d.
Collapse
Affiliation(s)
- Anusha Mishra
- 1] Department of Neuroscience, Physiology and Pharmacology, University College London, London, UK. [2]
| | - Fergus M O'Farrell
- 1] Department of Neuroscience, Physiology and Pharmacology, University College London, London, UK. [2]
| | - Clare Reynell
- Department of Neuroscience, Physiology and Pharmacology, University College London, London, UK
| | - Nicola B Hamilton
- Department of Neuroscience, Physiology and Pharmacology, University College London, London, UK
| | - Catherine N Hall
- Department of Neuroscience, Physiology and Pharmacology, University College London, London, UK
| | - David Attwell
- Department of Neuroscience, Physiology and Pharmacology, University College London, London, UK
| |
Collapse
|