1
|
Qambari H, Hein M, Balaratnasingam C, Yu P, Yu DY. Enabling visualization of GFAP-positive retinal glial cells, neurons and microvasculature in three-dimensions. Exp Eye Res 2025; 257:110410. [PMID: 40306397 DOI: 10.1016/j.exer.2025.110410] [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: 07/31/2024] [Revised: 04/22/2025] [Accepted: 04/27/2025] [Indexed: 05/02/2025]
Abstract
Glial cells are one of the most numerous cell types in the vertebrate retina and they serve to support neurovascular function. The principal glial cell in the retina is the Müller cell, accounting for approximately 90 % of all retinal glial cells. Müller cells are phenotypically elongated in shape and were first described as 'radial fibers' by Heinrich Müller in 1851. Their structure spans the entire thickness of the retina, through all retinal layers from the internal to external limiting membrane. This unique three-dimensional spatial arrangement enables Müller cells' direct contact with almost all cell types in the retina to perform its function. Despite this, the current study of Müller cells has largely been limited to thin sections or in culture, which provide limited detail about its spatial arrangement and interconnection with other cell types. The novel technique described here enables the three-dimensional visualization of GFAP-positive Müller cell processes in rodent retina and is based on the isolated arterially perfused rat eye preparation. Our micro perfusion technique utilizes the microvasculature as the delivery channel to quickly and effectively preserve all retinal elements. Intravascular labelling enables visualization of the intact three-dimensional retinal microvasculature within its normal neuronal and glial confines. Additional immersion immunolabeling and subsequent clearing with RapiClear® enables the three-dimensional visualization of different retinal elements and their physical interaction. Volume rendering of confocal image stacks acquired from these specimens can facilitate the study of such interactions in normal and disease models to further our understanding. This technique may be replicated in human donor retinae for future investigations to provide insight into Müller cell form and spatial relationship with other cell types.
Collapse
Affiliation(s)
- Hassanain Qambari
- Centre for Ophthalmology and Visual Science, The University of Western Australia, Perth, Western Australia, Australia; Lions Eye Institute, Perth, Western Australia, Australia
| | - Martin Hein
- Centre for Ophthalmology and Visual Science, The University of Western Australia, Perth, Western Australia, Australia; Lions Eye Institute, Perth, Western Australia, Australia
| | - Chandrakumar Balaratnasingam
- Centre for Ophthalmology and Visual Science, The University of Western Australia, Perth, Western Australia, Australia; Lions Eye Institute, Perth, Western Australia, Australia
| | - Paula Yu
- Centre for Ophthalmology and Visual Science, The University of Western Australia, Perth, Western Australia, Australia; Lions Eye Institute, Perth, Western Australia, Australia
| | - Dao-Yi Yu
- Centre for Ophthalmology and Visual Science, The University of Western Australia, Perth, Western Australia, Australia; Lions Eye Institute, Perth, Western Australia, Australia.
| |
Collapse
|
2
|
Jobling AI, Greferath U, Dixon MA, Quiriconi P, Eyar B, van Koeverden AK, Mills SA, Vessey KA, Bui BV, Fletcher EL. Microglial regulation of the retinal vasculature in health and during the pathology associated with diabetes. Prog Retin Eye Res 2025; 106:101349. [PMID: 40020909 DOI: 10.1016/j.preteyeres.2025.101349] [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: 11/20/2024] [Revised: 02/25/2025] [Accepted: 02/25/2025] [Indexed: 03/03/2025]
Abstract
The high metabolic demand of retinal neurons requires a precisely regulated vascular system that can deliver rapid changes in blood flow in response to neural need. In the retina, this is achieved via the action of a coordinated group of cells that form the neurovascular unit. While cells such as pericytes, Müller cells, and astrocytes have long been linked to neurovascular coupling, more recently the resident microglial population have also been implicated. In the healthy retina, microglia make extensive contact with blood vessels, as well as neuronal synapses, and are important in vascular patterning during development. Work in the brain and retina has recently indicated that microglia can directly regulate the local vasculature. In the retina, the fractalkine-Cx3cr1 signalling axis has been shown to induce local capillary constriction within the superficial vascular plexus via a mechanism involving components of the renin-angiotensin system. Furthermore, aberrant microglial induced vasoconstriction may be at the centre of early vascular reactivity changes observed in those with diabetes. This review summarizes the recent emerging evidence that microglia play multiple roles in retinal homeostasis especially in regulating the vasculature. We highlight what is known about the role of microglia under normal circumstances, and then build on this to discuss how microglia contribute to early vascular compromise during diabetes. Further understanding of the mechanisms of microglial-vascular regulation may allow alternate treatment strategies to be devised to reduce vascular pathology in diseases such as diabetic retinopathy.
Collapse
Affiliation(s)
- Andrew I Jobling
- Department of Anatomy and Physiology, The University of Melbourne, Victoria, Australia
| | - Ursula Greferath
- Department of Anatomy and Physiology, The University of Melbourne, Victoria, Australia
| | - Michael A Dixon
- Department of Anatomy and Physiology, The University of Melbourne, Victoria, Australia
| | - Pialuisa Quiriconi
- Department of Anatomy and Physiology, The University of Melbourne, Victoria, Australia
| | - Belinda Eyar
- Department of Anatomy and Physiology, The University of Melbourne, Victoria, Australia
| | - Anna K van Koeverden
- Department of Optometry and Vision Sciences, The University of Melbourne, Victoria, Australia
| | - Samuel A Mills
- Department of Anatomy and Physiology, The University of Melbourne, Victoria, Australia
| | - Kirstan A Vessey
- Department of Anatomy and Physiology, The University of Melbourne, Victoria, Australia
| | - Bang V Bui
- Department of Optometry and Vision Sciences, The University of Melbourne, Victoria, Australia
| | - Erica L Fletcher
- Department of Anatomy and Physiology, The University of Melbourne, Victoria, Australia.
| |
Collapse
|
3
|
Tekgol Uzuner G, Ozer A, Uzuner N. Combination among VEP, OCT and TCD data in patients with relapsing-remitting multiple sclerosis during attack-free period. Int J Neurosci 2025; 135:257-263. [PMID: 38088352 DOI: 10.1080/00207454.2023.2295228] [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: 04/07/2022] [Revised: 12/07/2023] [Accepted: 12/11/2023] [Indexed: 12/21/2023]
Abstract
AIMS Multiple sclerosis (MS) is characterized by inflammation, demyelination, glial cell pathology and axonal injury in the central nervous system, and also progressive axonal loss of the optic nerve in cases with optic neuritis (ON). The previous transcranial Doppler (TCD) investigations suggest cortical hyperreactivity in patients with relapsing-remitting Multiple sclerosis (RRMS). We investigated the effect of ON involvement on visual reactivity. MATERIAL AND METHODS One hundred thirty patients with RRMS during attack-free period and 12 healthy subjects were enrolled to the study. Visual evoked potential (VEP), optic coherence tomography (OCT) and TCD examinations of all subjects were performed. RESULTS Cerebrovascular reactivity measured with breath holding (BH) test was found to be normal. VEP amplitude, visual reactivity and peripapillary retinal nerve fibre layer (pRNFL) measurements were found to be low in patients with ON involvement, whereas VEP latencies were long. Visual reactivity was negatively correlated with VEP amplitude and RFNL measurements, and positively correlated with VEP latency. CONCLUSIONS The present study supports that cerebrovascular reactivity is preserved in patients with RRMS except for attacks, and neurovascular reactivity is increased in patients without ON involvement.
Collapse
Affiliation(s)
| | - Ahmet Ozer
- Department of Ophthalmology, Eskisehir Osmangazi University, Eskisehir, Turkey
| | - Nevzat Uzuner
- Department of Neurology, Eskisehir Osmangazi University, Eskisehir, Turkey
| |
Collapse
|
4
|
Dmitriev AV, Linsenmeier RA. pH in the vertebrate retina and its naturally occurring and pathological changes. Prog Retin Eye Res 2025; 104:101321. [PMID: 39608565 PMCID: PMC11711014 DOI: 10.1016/j.preteyeres.2024.101321] [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: 08/23/2024] [Revised: 11/23/2024] [Accepted: 11/25/2024] [Indexed: 11/30/2024]
Abstract
This review summarizes the existing information on the concentration of H+ (pH) in vertebrate retinae and its changes due to various reasons. Special features of H+ homeostasis that make it different from other ions will be discussed, particularly metabolic production of H+ and buffering. The transretinal distribution of extracellular H+ concentration ([H+]o) and its changes under illumination and other conditions will be described in detail, since [H+]o is more intensively investigated than intracellular pH. In vertebrate retinae, the highest [H+]o occurs in the inner part of the outer nuclear layer, and decreases toward the RPE, reaching the blood level on the apical side of the RPE. [H+]o falls toward the vitreous as well, but less, so that the inner retina is acidic to the vitreous. Light leads to complex changes with both electrogenic and metabolic origins, culminating in alkalinization. There is a rhythm of [H+]o with H+ being higher during circadian night. Extracellular pH can potentially be used as a signal in intercellular volume transmission, but evidence is against pH as a normal controller of fluid transport across the RPE or as a horizontal cell feedback signal. Pathological and experimentally created conditions (systemic metabolic acidosis, hypoxia and ischemia, vascular occlusion, excess glucose and diabetes, genetic disorders, and blockade of carbonic anhydrase) disturb H+ homeostasis, mostly producing retinal acidosis, with consequences for retinal blood flow, metabolism and function.
Collapse
Affiliation(s)
- Andrey V Dmitriev
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, USA.
| | - Robert A Linsenmeier
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, USA; Department of Neurobiology, Northwestern University, Evanston, IL, USA; Department of Ophthalmology, Northwestern University, Chicago, IL, USA.
| |
Collapse
|
5
|
Lin WL, Dickson DW. Capillary basal lamina in human brain and spinal cord has fibrillar collagen type I and type III: Ignorance may not be bliss. FREE NEUROPATHOLOGY 2025; 6:6. [PMID: 40012952 PMCID: PMC11862662 DOI: 10.17879/freeneuropathology-2025-6159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Accepted: 01/25/2025] [Indexed: 02/28/2025]
Abstract
The capillary basal lamina (BL) located between the endothelial cell, pericyte and perivascular astrocyte plays important roles in normal and diseased central nervous system (CNS). Using immunohistochemistry (IHC), electron microscopy (EM) and post-embedding immunogold EM (IEM), we studied capillary BL in biopsy and autopsy tissues of human CNS from cases with and without significant brain pathology and aged from 4 days to 49 years. In all cases, IHC showed, in the BL of microvessels, immunoreactivity for collagen types I, III, IV, VI and fibronectin. EM revealed fusion of the BL of capillary endothelial cells or pericyte with perivascular astrocyte BL, which was focally split, resulting in expanded spaces bordered by BL and containing striated fibrils. There was no significant thickening of fused or split BL. IEM showed localization of collagen I and III to banded fibrils, and of collagen IV to split and fused BL. These characteristic ultrastructural findings in human capillary BL were not found in normal or transgenic mice. Our observations of fibrillar collagen in young individuals complement previous observations of similar findings in older individuals. This raises the possibility that fibrillar collagen in human vascular BL plays a significant role in CNS capillary physiology and pathophysiology. The species-specific differences in capillary morphology between humans and mice might have relevance to poor correlations between benefits of immunotherapy and drug treatment in mice compared with human.
Collapse
Affiliation(s)
- Wen-Lang Lin
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida, USA
| | | |
Collapse
|
6
|
Galgani G, Bray G, Martelli A, Calderone V, Citi V. In Vitro Models of Diabetes: Focus on Diabetic Retinopathy. Cells 2024; 13:1864. [PMID: 39594613 PMCID: PMC11592768 DOI: 10.3390/cells13221864] [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: 10/14/2024] [Revised: 11/05/2024] [Accepted: 11/08/2024] [Indexed: 11/28/2024] Open
Abstract
Diabetic retinopathy is a major eye complication in patients with diabetes mellitus, and it is the leading cause of blindness and visual impairment in the world. Chronic hyperglycemia induces endothelial damage with consequent vascular lesions, resulting in global vasculitis, which affects the small vessels of the retina. These vascular lesions cause ischemic conditions in certain areas of the retina, with a consequent increase in the release of pro-angiogenic mediators. In addition to pharmacological interventions for controlling the blood glycaemic level, the main strategies for treating diabetic retinopathy are the intravitreal injections of drugs, surgical treatments, and vitrectomies. The complexity of diabetic retinopathy is due to its close interactions with different cell types (endothelial cells, astrocytes, and Müller cells). The evaluation of the efficacy of novel pharmacological strategies is mainly performed through in vivo models. However, the use of different animal species leads to heterogenic results and ethical concerns. For these reasons, the development of new and reliable in vitro models, such as cell co-cultures and eye organoids, represents an urgent need in this area of research. This review features an overview of the in vitro models used to date and highlights the advances in technology used to study this pathology.
Collapse
Affiliation(s)
- Giulia Galgani
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy; (G.G.); (A.M.); (V.C.); (V.C.)
| | - Giorgia Bray
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy; (G.G.); (A.M.); (V.C.); (V.C.)
| | - Alma Martelli
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy; (G.G.); (A.M.); (V.C.); (V.C.)
- Interuniversity Centre for the Promotion of the 3R Principles in Teaching and Research, Italy
| | - Vincenzo Calderone
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy; (G.G.); (A.M.); (V.C.); (V.C.)
- Interuniversity Centre for the Promotion of the 3R Principles in Teaching and Research, Italy
| | - Valentina Citi
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy; (G.G.); (A.M.); (V.C.); (V.C.)
- Interuniversity Centre for the Promotion of the 3R Principles in Teaching and Research, Italy
| |
Collapse
|
7
|
Sinclair SH, Schwartz S. Diabetic retinopathy: New concepts of screening, monitoring, and interventions. Surv Ophthalmol 2024; 69:882-892. [PMID: 38964559 DOI: 10.1016/j.survophthal.2024.07.001] [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: 03/14/2024] [Revised: 06/30/2024] [Accepted: 07/01/2024] [Indexed: 07/06/2024]
Abstract
The science of diabetes care has progressed to provide a better understanding of the oxidative and inflammatory lesions and pathophysiology of the neurovascular unit within the retina (and brain) that occur early in diabetes, even prediabetes. Screening for retinal structural abnormalities, has traditionally been performed by fundus examination or color fundus photography; however, these imaging techniques detect the disease only when there are sufficient lesions, predominantly hemorrhagic, that are recognized to occur late in the disease process after significant neuronal apoptosis and atrophy, as well as microvascular occlusion with alterations in vision. Thus, interventions have been primarily oriented toward the later-detected stages, and clinical trials, while demonstrating a slowing of the disease progression, demonstrate minimal visual improvement and modest reduction in the continued loss over prolonged periods. Similarly, vision measurement utilizing charts detects only problems of visual function late, as the process begins most often parafoveally with increasing number and progressive expansion, including into the fovea. While visual acuity has long been used to define endpoints of visual function for such trials, current methods reviewed herein are found to be imprecise. We review improved methods of testing visual function and newer imaging techniques with the recommendation that these must be utilized to discover and evaluate the injury earlier in the disease process, even in the prediabetic state. This would allow earlier therapy with ocular as well as systemic pharmacologic treatments that lower the and neuro-inflammatory processes within eye and brain. This also may include newer, micropulsed laser therapy that, if applied during the earlier cascade, should result in improved and often normalized retinal function without the adverse treatment effects of standard photocoagulation therapy.
Collapse
Affiliation(s)
| | - Stan Schwartz
- University of Pennsylvania Affiliate, Main Line Health System, USA
| |
Collapse
|
8
|
Zhu Y, Zhang Y, He S, Yi S, Feng H, Xia X, Fang X, Gong X, Zhao P. Integrating single-nucleus RNA sequencing and spatial transcriptomics to elucidate a specialized subpopulation of astrocytes, microglia and vascular cells in brains of mouse model of lipopolysaccharide-induced sepsis-associated encephalopathy. J Neuroinflammation 2024; 21:169. [PMID: 38961424 PMCID: PMC11223438 DOI: 10.1186/s12974-024-03161-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Accepted: 06/26/2024] [Indexed: 07/05/2024] Open
Abstract
BACKGROUND Understanding the mechanism behind sepsis-associated encephalopathy (SAE) remains a formidable task. This study endeavors to shed light on the complex cellular and molecular alterations that occur in the brains of a mouse model with SAE, ultimately unraveling the underlying mechanisms of this condition. METHODS We established a murine model using intraperitoneal injection of lipopolysaccharide (LPS) in wild type and Anxa1-/- mice and collected brain tissues for analysis at 0-hour, 12-hour, 24-hour, and 72-hour post-injection. Utilizing advanced techniques such as single-nucleus RNA sequencing (snRNA-seq) and Stereo-seq, we conducted a comprehensive characterization of the cellular responses and molecular patterns within the brain. RESULTS Our study uncovered notable temporal differences in the response to LPS challenge between Anxa1-/- (annexin A1 knockout) and wild type mice, specifically at the 12-hour and 24-hour time points following injection. We observed a significant increase in the proportion of Astro-2 and Micro-2 cells in these mice. These cells exhibited a colocalization pattern with the vascular subtype Vas-1, forming a distinct region known as V1A2M2, where Astro-2 and Micro-2 cells surrounded Vas-1. Moreover, through further analysis, we discovered significant upregulation of ligands and receptors such as Timp1-Cd63, Timp1-Itgb1, Timp1-Lrp1, as well as Ccl2-Ackr1 and Cxcl2-Ackr1 within this region. In addition, we observed a notable increase in the expression of Cd14-Itgb1, Cd14-Tlr2, and Cd14-C3ar1 in regions enriched with Micro-2 cells. Additionally, Cxcl10-Sdc4 showed broad upregulation in brain regions containing both Micro-2 and Astro-2 cells. Notably, upon LPS challenge, there was an observed increase in Anxa1 expression in the mouse brain. Furthermore, our study revealed a noteworthy increase in mortality rates following Anxa1 knockdown. However, we did not observe substantial differences in the types, numbers, or distribution of other brain cells between Anxa1-/- and wildtype mice over time. Nevertheless, when comparing the 24-hour post LPS injection time point, we observed a significant decrease in the proportion and distribution of Micro-2 and Astro-2 cells in the vicinity of blood vessels in Anxa1-/- mice. Additionally, we noted reduced expression levels of several ligand-receptor pairs including Cd14-Tlr2, Cd14-C3ar1, Cd14-Itgb1, Cxcl10-Sdc4, Ccl2-Ackr1, and Cxcl2-Ackr1. CONCLUSIONS By combining snRNA-seq and Stereo-seq techniques, our study successfully identified a distinctive cellular colocalization, referred to as a special pathological niche, comprising Astro-2, Micro-2, and Vas-1 cells. Furthermore, we observed an upregulation of ligand-receptor pairs within this niche. These findings suggest a potential association between this cellular arrangement and the underlying mechanisms contributing to SAE or the increased mortality observed in Anxa1 knockdown mice.
Collapse
Grants
- 2021A1515012429 Natural Science Foundation of Guangdong Province, China
- 211102114530659 Shaoguan Municipal Science and Technology Program, China
- 20221807 Shaoguan Engineering Research Center for Research and Development of Molecular and Cellular Technology in Rapid Diagnosis of Infectious Diseases and Cancer Program, China
- KEYANSHEN (2023) 01 Research Fund for Joint Laboratory for Digital and Precise Detection of Clinical Pathogens, Yuebei People's Hospital Affiliated to Shantou University Medical College, China
- RS202001 Research Project for Outstanding Scholar of Yuebei People's Hospital, Shantou University Medical College, China
- Research Fund for Joint Laboratory for Digital and Precise Detection of Clinical Pathogens, Yuebei People’s Hospital Affiliated to Shantou University Medical College, China
- Research Project for Outstanding Scholar of Yuebei People’s Hospital, Shantou University Medical College, China
Collapse
Affiliation(s)
- Yanyan Zhu
- Department of Laboratory Medicine, Yuebei People's Hospital, Shantou University Medical College, No 133, Huimin Road South, Wujiang District, Shaoguan, 512025, China
- Laboratory for Diagnosis of Clinical Microbiology and Infection, Yuebei People's Hospital, Shantou University Medical College, Shaoguan, 512025, China
- Research Center for Interdisciplinary & High-quality Innovative Development in Laboratory Medicine, Shaoguan, 512025, China
- Shaoguan Municipal Quality Control Center for Laboratory Medicine, Yuebei People's Hospital, Shantou University Medical College, Shaoguan, 512025, China
- Shaoguan Municipal Quality Control Center for Surveillance of Bacterial Resistance, Shaoguan, 512025, China
- Shaoguan Engineering Research Center for Research and Development of Molecular and Cellular Technology in Rapid Diagnosis of Infectious Diseases and Cancer, Shaoguan, 512025, China
| | - Yin Zhang
- BGI-Shenzhen, Shenzhen, 518083, China
| | - Sheng He
- Department of Laboratory Medicine, Yuebei People's Hospital, Shantou University Medical College, No 133, Huimin Road South, Wujiang District, Shaoguan, 512025, China
- Laboratory for Diagnosis of Clinical Microbiology and Infection, Yuebei People's Hospital, Shantou University Medical College, Shaoguan, 512025, China
- Research Center for Interdisciplinary & High-quality Innovative Development in Laboratory Medicine, Shaoguan, 512025, China
- Shaoguan Municipal Quality Control Center for Laboratory Medicine, Yuebei People's Hospital, Shantou University Medical College, Shaoguan, 512025, China
- Shaoguan Municipal Quality Control Center for Surveillance of Bacterial Resistance, Shaoguan, 512025, China
- Shaoguan Engineering Research Center for Research and Development of Molecular and Cellular Technology in Rapid Diagnosis of Infectious Diseases and Cancer, Shaoguan, 512025, China
| | - Sanjun Yi
- Department of Laboratory Medicine, Yuebei People's Hospital, Shantou University Medical College, No 133, Huimin Road South, Wujiang District, Shaoguan, 512025, China
- Laboratory for Diagnosis of Clinical Microbiology and Infection, Yuebei People's Hospital, Shantou University Medical College, Shaoguan, 512025, China
- Research Center for Interdisciplinary & High-quality Innovative Development in Laboratory Medicine, Shaoguan, 512025, China
- Shaoguan Municipal Quality Control Center for Laboratory Medicine, Yuebei People's Hospital, Shantou University Medical College, Shaoguan, 512025, China
- Shaoguan Municipal Quality Control Center for Surveillance of Bacterial Resistance, Shaoguan, 512025, China
- Shaoguan Engineering Research Center for Research and Development of Molecular and Cellular Technology in Rapid Diagnosis of Infectious Diseases and Cancer, Shaoguan, 512025, China
| | - Hao Feng
- Jiaxing University Master Degree Cultivation Base, Zhejiang Chinese Medical University, Jiaxing, 314001, China
| | - Xianzhu Xia
- Department of Laboratory Medicine, Yuebei People's Hospital, Shantou University Medical College, No 133, Huimin Road South, Wujiang District, Shaoguan, 512025, China
- Laboratory for Diagnosis of Clinical Microbiology and Infection, Yuebei People's Hospital, Shantou University Medical College, Shaoguan, 512025, China
- Research Center for Interdisciplinary & High-quality Innovative Development in Laboratory Medicine, Shaoguan, 512025, China
| | | | - Xiaoqian Gong
- Yuebei People's Hospital, Shantou University Medical College, Shaoguan, 512025, China.
| | - Pingsen Zhao
- Department of Laboratory Medicine, Yuebei People's Hospital, Shantou University Medical College, No 133, Huimin Road South, Wujiang District, Shaoguan, 512025, China.
- Laboratory for Diagnosis of Clinical Microbiology and Infection, Yuebei People's Hospital, Shantou University Medical College, Shaoguan, 512025, China.
- Research Center for Interdisciplinary & High-quality Innovative Development in Laboratory Medicine, Shaoguan, 512025, China.
- Shaoguan Municipal Quality Control Center for Laboratory Medicine, Yuebei People's Hospital, Shantou University Medical College, Shaoguan, 512025, China.
- Shaoguan Municipal Quality Control Center for Surveillance of Bacterial Resistance, Shaoguan, 512025, China.
- Shaoguan Engineering Research Center for Research and Development of Molecular and Cellular Technology in Rapid Diagnosis of Infectious Diseases and Cancer, Shaoguan, 512025, China.
| |
Collapse
|
9
|
Zeine F, Jafari N, Baron D, Bowirrat A, Pinhasov A, Norling B, Martinez KC, Nami M, Manavi N, Sunder K, Rabin DM, Bagchi D, Khalsa J, Gold MS, Sipple D, Barzegar M, Bodhanapati J, Khader W, Carney P, Dennen CA, Gupta A, Elman I, Badgaiyan RD, Modestino EJ, Thanos PK, Hanna C, McLaughlin T, Cadet JL, Soni D, Braverman ER, Barh D, Giordano J, Edwards D, Ashford JW, Gondre-Lewis MC, Gilley E, Murphy KT, Lewandrowski KU, Sharafshah A, Makale M, Fuehrlein B, Blum K. Solving the Global Opioid Crisis: Incorporating Genetic Addiction Risk Assessment with Personalized Dopaminergic Homeostatic Therapy and Awareness Integration Therapy. JOURNAL OF ADDICTION PSYCHIATRY 2024; 8:50-95. [PMID: 39635461 PMCID: PMC11615735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/07/2024]
Abstract
Objectives The opioid crisis in the last few decades has mounted to a global level, impacting all areas of socioeconomic, demographic, geographic, and cultural boundaries. Traditional treatments have not been deemed to show the degree of efficacy necessary to address the crisis. The authors of this review paper have set forth an unprecedented and in-depth look into multi-factorial determinants that have contributed to the opioid crisis becoming global and multi-faceted. Methods For this narrative review/opinion article, we searched PsychINFO, PubMed, Google Scholar, and Web of Science databases to identify relevant articles on topics including the "opioid crisis," "opioid mechanisms," "genetics and epigenetics," "neuropharmacology," and "clinical aspects of opioid treatment and prevention." Since this was not a systematic review the articles selected could represent unitential bias. Results Despite some success achieved through Opioid Substitution Therapy (OST) in harm reduction, the annual mortality toll in the US alone surpasses 106,699 individuals, a figure expected to climb to 165,000 by 2025. Data from the Substance Abuse and Mental Health Services Administration's (SAMHSA) National Survey on Drug Abuse and Health (NSDUH) reveals that approximately 21.4% of individuals in the US engaged in illicit drug use in 2020, with 40.3 million individuals aged 12 or older experiencing a Substance Use Disorder (SUD). Provisional figures from the Centers for Disease Control and Prevention (CDC) indicate a troubling 15% increase in overdose deaths in 2021, rising from 93,655 in 2020 to 107,622, with opioids accounting for roughly 80,816 of these deaths. Conclusions We advocate reevaluating the "standard of care" and shifting towards inducing dopamine homeostasis by manipulating key neurotransmitter systems within the brain's reward cascade. We propose a paradigm shift towards a novel "standard of care" that begins with incorporating Genetic Addiction Risk Severity (GARS) testing to assess pre-addiction risk and vulnerability to opioid-induced addiction; emphasis should be placed on inducing dopamine homeostasis through safe and non-addictive alternatives like KB220, and comprehensive treatment approaches that address psychological, spiritual, and societal aspects of addiction through Awareness Integration Therapy (AIT).
Collapse
Affiliation(s)
- Foojan Zeine
- Awareness Integration Institute, San Clemente, USA
- Department of Health Science, California State University, Long Beach, USA
| | - Nicole Jafari
- Department of Applied Clinical Psychology, The Chicago School of Professional Psychology, Los Angeles, USA
- Division of Personalized Medicine, Cross-Cultural Research and Educational Institute, San Clemente, USA
| | - David Baron
- Center for Exercise and Sport Mental Health, Western University Health Sciences, Pomona, USA
| | - Abdalla Bowirrat
- Department of Molecular Biology, Adelson School of Medicine, Ariel University, Ariel, Israel
| | - Albert Pinhasov
- Department of Molecular Biology, Adelson School of Medicine, Ariel University, Ariel, Israel
| | - Brian Norling
- MEMS Precision Technology, Inc., Santa Barbara, USA
- Acies Biomedical, Inc. Santa Barbara, USA
| | - Kathleen Carter Martinez
- Division of General Education-Berkeley College, Paramus Campus, New Jersey, USA
- Chey-Wind Center for Trauma and Healing, Peru, USA
| | - Mohammad Nami
- Brain, Cognition, and Behavior Unit, Brain Hub Academy, Dubai, UAE
| | - Nima Manavi
- College of Osteopathic Medicine, Western University of Health Sciences, Pomona, USA
| | - Keerthy Sunder
- Department of Psychiatry, University of California, UC Riverside School of Medicine, Riverside, USA
- Division of Neuromodulation Research, Karma Doctors and Karma TMS, Palm Springs, USA
| | | | - Debasis Bagchi
- Division of Nutrigenomics, Victory Nutrition International, LLC, Bonita Springs, USA
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, Texas Southern University, Houston, USA
| | - Jag Khalsa
- Department of Medicine, University of Maryland School of Medicine, Baltimore, USA
| | - Mark S. Gold
- Department of Psychiatry, Washington University, School of Medicine, St. Louis, USA
| | - Daniel Sipple
- Minnesota Institute for Pain Management, Minnesota, USA
| | - Mojtaba Barzegar
- Hamad Medical Corporation, National Center for Cancer Care and Research (NCCCR), Doha, Qatar
| | - Jothsna Bodhanapati
- Division of Neuromodulation Research, Karma Doctors and Karma TMS, Palm Springs, USA
| | - Waseem Khader
- Karma Doctors, Palm Springs, USA
- Global Medical Detox Center, Menifee, CA, USA
| | - Paul Carney
- Division of Pediatric Neurology, University of Missouri, School of Medicine, Columbia, USA
| | - Catherine A. Dennen
- Department of Family Medicine, Jefferson Health Northeast, Philadelphia, USA
| | | | - Igor Elman
- Department of Molecular Biology, Adelson School of Medicine, Ariel University, Ariel, Israel
- Department of Psychiatry, Harvard School of Medicine, Cambridge, USA
| | - Rajendra D. Badgaiyan
- Department of Psychiatry, Case Western University School of Medicine, The Metro Health System, Cleveland, USA
- Department of Psychiatry, Mt. Sinai University, Ichan School of Medicine, New York, USA
| | | | - Panayotis K. Thanos
- Department of Molecular Biology, Adelson School of Medicine, Ariel University, Ariel, Israel
- Behavioral Neuropharmacology and Neuroimaging Laboratory on Addictions, Research Institute on Addictions, University at Buffalo, Buffalo, USA
| | - Colin Hanna
- Behavioral Neuropharmacology and Neuroimaging Laboratory on Addictions, Research Institute on Addictions, University at Buffalo, Buffalo, USA
| | - Thomas McLaughlin
- Division of Primary Care Research, Reward Deficiency Syndrome Clinics of America, Inc. Austin, USA
| | - Jean Lud Cadet
- Molecular Neuropsychiatry Research Branch, NIH National Institute on Drug Abuse, Baltimore, USA
| | - Diwanshu Soni
- College of Osteopathic Medicine, Western University of Health Sciences, Pomona, USA
| | - Eric R. Braverman
- Division of Clinical Neurological Research, The Kenneth Blum Neurogenetic and Behavioral Institute, LLC., Austin, USA
| | - Debmalya Barh
- Centre for Genomics and Applied Gene Technology, Institute of Integrative Omics and Applied Biotechnology, Nonakuri, Purba Medinipur, West Bengal, India
| | | | | | - J. Wesson Ashford
- Department of Psychiatry and Behavioral Sciences, Stanford University, Palo Alto, USA
| | | | | | - Kevin T. Murphy
- Department of Radiation Oncology, University of California, San Diego, La Jolla, USA
| | - Kai-Uwe Lewandrowski
- Division of Personalized Pain Therapy Research, Center for Advanced Spine Care of Southern Arizona, Tucson, USA
- Department of Orthopaedics, Fundación Universitaria Sanitas, Bogotá, D.C., Colombia
- Department of Orthopedics, Hospital Universitário Gaffrée Guinle Universidade Federal do Estado do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Alireza Sharafshah
- Cellular and Molecular Research Center, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Milan Makale
- Department of Radiation Oncology, University of California, San Diego, La Jolla, USA
| | - Brian Fuehrlein
- Department of Psychiatry, School of Medicine, Yale University, New Haven, USA
| | - Kenneth Blum
- Center for Exercise and Sport Mental Health, Western University Health Sciences, Pomona, USA
- Department of Molecular Biology, Adelson School of Medicine, Ariel University, Ariel, Israel
- Division of Primary Care Research, Reward Deficiency Syndrome Clinics of America, Inc. Austin, USA
- Division of Clinical Neurological Research, The Kenneth Blum Neurogenetic and Behavioral Institute, LLC., Austin, USA
- Centre for Genomics and Applied Gene Technology, Institute of Integrative Omics and Applied Biotechnology, Nonakuri, Purba Medinipur, West Bengal, India
- JC’s Recovery and Counseling Center, Hollywood, USA
- Department of Psychiatry, University of Vermont, Burlington, USA
- Department of Psychiatry, Wright University Boonshoft School of Medicine, Dayton, USA
- Institute of Psychology, ELTE Eötvös Loránd University, Budapest, Hungary
- Center for Advanced Spine Care of Southern Arizona, Tucson, USA
| |
Collapse
|
10
|
Hein M, Qambari H, An D, Balaratnasingam C. Current understanding of subclinical diabetic retinopathy informed by histology and high-resolution in vivo imaging. Clin Exp Ophthalmol 2024; 52:464-484. [PMID: 38363022 DOI: 10.1111/ceo.14363] [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: 10/30/2023] [Revised: 01/21/2024] [Accepted: 01/26/2024] [Indexed: 02/17/2024]
Abstract
The escalating incidence of diabetes mellitus has amplified the global impact of diabetic retinopathy. There are known structural and functional changes in the diabetic retina that precede the fundus photography abnormalities which currently are used to diagnose clinical diabetic retinopathy. Understanding these subclinical alterations is important for effective disease management. Histology and high-resolution clinical imaging reveal that the entire neurovascular unit, comprised of retinal vasculature, neurons and glial cells, is affected in subclinical disease. Early functional manifestations are seen in the form of blood flow and electroretinography disturbances. Structurally, there are alterations in the cellular components of vasculature, glia and the neuronal network. On clinical imaging, changes to vessel density and thickness of neuronal layers are observed. How these subclinical disturbances interact and ultimately manifest as clinical disease remains elusive. However, this knowledge reveals potential early therapeutic targets and the need for imaging modalities that can detect subclinical changes in a clinical setting.
Collapse
Affiliation(s)
- Martin Hein
- Physiology and Pharmacology Group, Lions Eye Institute, Perth, Western Australia, Australia
- Centre for Ophthalmology and Visual Science, University of Western Australia, Perth, Western Australia, Australia
| | - Hassanain Qambari
- Physiology and Pharmacology Group, Lions Eye Institute, Perth, Western Australia, Australia
- Centre for Ophthalmology and Visual Science, University of Western Australia, Perth, Western Australia, Australia
| | - Dong An
- Physiology and Pharmacology Group, Lions Eye Institute, Perth, Western Australia, Australia
- Centre for Ophthalmology and Visual Science, University of Western Australia, Perth, Western Australia, Australia
| | - Chandrakumar Balaratnasingam
- Physiology and Pharmacology Group, Lions Eye Institute, Perth, Western Australia, Australia
- Centre for Ophthalmology and Visual Science, University of Western Australia, Perth, Western Australia, Australia
- Department of Ophthalmology, Sir Charles Gairdner Hospital, Perth, Western Australia, Australia
| |
Collapse
|
11
|
Linsenmeier RA, Dmitriev AV. Increased Retinal Metabolism Induced by Flicker in the Isolated Mouse Retina. eNeuro 2024; 11:ENEURO.0509-23.2024. [PMID: 38641415 PMCID: PMC11089847 DOI: 10.1523/eneuro.0509-23.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 04/10/2024] [Accepted: 04/12/2024] [Indexed: 04/21/2024] Open
Abstract
Both the retina and brain exhibit neurovascular coupling, increased blood flow during increased neural activity. In the retina increased blood flow can be evoked by flickering light, but the magnitude of the metabolic change that underlies this is not known. Local changes in oxygen consumption (QO2) are difficult to measure in vivo when both supply and demand are changing. Here we isolated the C57BL/6J mouse retina and supplied it with oxygen from both sides of the tissue. Microelectrode recordings of PO2 were made in darkness and during 20 s of high scotopic flickering light at 1 Hz. Flicker led to a PO2 increase in the outer retina and a decrease in the inner retina, indicating that outer retinal QO2 (QOR) decreased and inner retinal QO2 (QIR) increased. A four-layer oxygen diffusion model was fitted to PO2 values obtained in darkness and at the end of flicker to determine the values of QOR and QIR. QOR in flicker was 76 ± 14% (mean and SD, n = 10) of QOR in darkness. The increase in QIR was smaller, 6.4 ± 5.0%. These metabolic changes are likely smaller than the maximum changes, because with no regeneration of pigment in the isolated retina, we limited the illumination. Further modeling indicated that at high illumination, QIR could increase by up to 45%, which is comparable to the magnitude of flow changes. This suggests that the blood flow increase is at least roughly matched to the increased metabolic demands of activity in the retina.
Collapse
Affiliation(s)
- Robert A Linsenmeier
- Departments of Biomedical Engineering, Northwestern University, Evanston, Illinois 60208
- Neurobiology, Northwestern University, Evanston, Illinois 60208
- Department of Ophthalmology, Northwestern University, Chicago, Illinois 60611
| | - Andrey V Dmitriev
- Departments of Biomedical Engineering, Northwestern University, Evanston, Illinois 60208
| |
Collapse
|
12
|
Du C, Park K, Hua Y, Liu Y, Volkow ND, Pan Y. Astrocytes modulate cerebral blood flow and neuronal response to cocaine in prefrontal cortex. Mol Psychiatry 2024; 29:820-834. [PMID: 38238549 PMCID: PMC11784220 DOI: 10.1038/s41380-023-02373-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 11/30/2023] [Accepted: 12/07/2023] [Indexed: 01/30/2024]
Abstract
Cocaine affects both cerebral blood vessels and neuronal activity in brain. Cocaine can also disrupt astrocytes, which modulate neurovascular coupling-a process that regulates cerebral hemodynamics in response to neuronal activation. However, separating neuronal and astrocytic effects from cocaine's direct vasoactive effects has been challenging, partially due to limitations of neuroimaging techniques able to differentiate vascular from neuronal and glial effects at high temporal and spatial resolutions. Here, we used a newly-developed multi-channel fluorescence and optical coherence Doppler microscope (fl-ODM) that allows for simultaneous measurements of neuronal and astrocytic activities (reflected by the intracellular calcium changes in neurons Ca2+N and astrocytes Ca2+A, respectively) alongside their vascular interactions in vivo to address this challenge. Using green and red genetically-encoded Ca2+ indicators differentially expressed in astrocytes and neurons, fl-ODM enabled concomitant imaging of large-scale astrocytic and neuronal Ca2+ fluorescence and 3D cerebral blood flow velocity (CBFv) in vascular networks in the mouse cortex. We assessed cocaine's effects in the prefrontal cortex (PFC) and found that the CBFv changes triggered by cocaine were temporally correlated with astrocytic Ca2+A activity. Chemogenetic inhibition of astrocytes during the baseline state resulted in blood vessel dilation and CBFv increases but did not affect neuronal activity, suggesting modulation of spontaneous blood vessel's vascular tone by astrocytes. Chemogenetic inhibition of astrocytes during a cocaine challenge prevented its vasoconstricting effects alongside the CBFv decreases, but it also attenuated the neuronal Ca2+N increases triggered by cocaine. These results document a role of astrocytes both in regulating vascular tone and consequently blood flow, at baseline and for modulating the vasoconstricting and neuronal activation responses to cocaine in the PFC. Strategies to inhibit astrocytic activity could offer promise for ameliorating vascular and neuronal toxicity from cocaine misuse.
Collapse
Affiliation(s)
- Congwu Du
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY, 11794, USA
| | - Kichon Park
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY, 11794, USA
| | - Yueming Hua
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY, 11794, USA
| | - Yanzuo Liu
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY, 11794, USA
| | - Nora D Volkow
- National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, 20857, USA
| | - Yingtian Pan
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY, 11794, USA.
| |
Collapse
|
13
|
Nippert AR, Chiang PP, Newman EA. Whisker-evoked neurovascular coupling is preserved during hypoglycemia in mouse cortical arterioles and capillaries. J Cereb Blood Flow Metab 2024; 44:155-168. [PMID: 37728791 PMCID: PMC10993878 DOI: 10.1177/0271678x231201241] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 07/12/2023] [Accepted: 08/15/2023] [Indexed: 09/21/2023]
Abstract
Hypoglycemia is a serious complication of insulin treatment of diabetes that can lead to coma and death. Neurovascular coupling, which mediates increased local blood flow in response to neuronal activity, increases glucose availability to active neurons. This mechanism could be essential for neuronal health during hypoglycemia, when total glucose supplies are low. Previous studies suggest, however, that neurovascular coupling (a transient blood flow increase in response to an increase in neuronal activity) may be reduced during hypoglycemia. Such a reduction in blood flow increase would exacerbate the effects of hypoglycemia, depriving active neurons of glucose. We have reexamined the effects of hypoglycemia on neurovascular coupling by simultaneously monitoring neuronal and vascular responses to whisker stimulation in the awake mouse somatosensory cortex. We find that neurovascular coupling at both penetrating arterioles and at 2nd order capillaries did not change significantly during insulin-induced hypoglycemia compared to euglycemia. In addition, we show that the basal diameter of both arterioles and capillaries increases during hypoglycemia (10.3 and 9.7% increases, respectively). Our results demonstrate that both neurovascular coupling and basal increases in vessel diameter are active mechanisms which help to maintain an adequate supply of glucose to the brain during hypoglycemia.
Collapse
Affiliation(s)
| | | | - Eric A Newman
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota, USA
| |
Collapse
|
14
|
McDonald H, Gardner-Russell J, Alarcon-Martinez L. Orchestrating Blood Flow in the Retina: Interpericyte Tunnelling Nanotube Communication. Results Probl Cell Differ 2024; 73:229-247. [PMID: 39242382 DOI: 10.1007/978-3-031-62036-2_11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/09/2024]
Abstract
The retina transforms light into electrical signals, which are sent to the brain via the optic nerve to form our visual perception. This complex signal processing is performed by the retinal neuron and requires a significant amount of energy. Since neurons are unable to store energy, they must obtain glucose and oxygen from the bloodstream to produce energy to match metabolic needs. This process is called neurovascular coupling (NVC), and it is based on a precise mechanism that is not totally understood. The discovery of fine tubular processes termed tunnelling nanotubes (TNTs) set a new type of cell-to-cell communication. TNTs are extensions of the cellular membrane that allow the transfer of material between connected cells. Recently, they have been reported in the brain and retina of living mice, where they connect pericytes, which are vascular mural cells that regulate vessel diameter. Accordingly, these TNTs were termed interpericyte tunnelling nanotubes (IPTNTs), which showed a vital role in blood delivery and NVC. In this chapter, we review the involvement of TNTs in NVC and discuss their implications in retinal neurodegeneration.
Collapse
Affiliation(s)
- Hannah McDonald
- Centre for Eye Research Australia, Melbourne, VIC, Australia
- Department of Ophthalmology, University of Melbourne, Melbourne, VIC, Australia
- The Royal Victorian Eye and Ear Hospital, Melbourne, VIC, Australia
| | - Jesse Gardner-Russell
- Centre for Eye Research Australia, Melbourne, VIC, Australia
- Department of Ophthalmology, University of Melbourne, Melbourne, VIC, Australia
- The Royal Victorian Eye and Ear Hospital, Melbourne, VIC, Australia
| | - Luis Alarcon-Martinez
- Centre for Eye Research Australia, Melbourne, VIC, Australia.
- Department of Ophthalmology, University of Melbourne, Melbourne, VIC, Australia.
- The Royal Victorian Eye and Ear Hospital, Melbourne, VIC, Australia.
| |
Collapse
|
15
|
Rupareliya VP, Singh AA, Butt AM, A H, Kumar H. The "molecular soldiers" of the CNS: Astrocytes, a comprehensive review on their roles and molecular signatures. Eur J Pharmacol 2023; 959:176048. [PMID: 37758010 DOI: 10.1016/j.ejphar.2023.176048] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 08/24/2023] [Accepted: 09/13/2023] [Indexed: 09/29/2023]
Abstract
For a long time, neurons held the position of central players in the nervous system. Since there are far more astrocytes than neurons in the brain, it makes us wonder if these cells just take up space and support the neurons or if they are actively participating in central nervous system (CNS) homeostasis. Now, astrocytes' contribution to CNS physiology is appreciated as they are known to regulate ion and neurotransmitter levels, synapse formation and elimination, blood-brain barrier integrity, immune function, cerebral blood flow, and many more. In many neurological and psychiatric disorders, astrocyte functions are altered. Advancements in microscopic and transcriptomic tools revealed populations of astrocytes with varied morphology, electrophysiological properties, and transcriptomic profiles. Neuron-circuit-specific functions and neuron-specific interactions of astroglial subpopulations are found, which suggests that diversity is essential in carrying out diverse region-specific CNS functions. Investigations on heterogeneous astrocyte populations are revealing new astrocyte functions and their role in pathological conditions, opening a new therapeutic avenue for targeting neurological conditions. The true extent of astrocytic heterogeneity and its functional implications are yet to be fully explored. This review summarizes essential astrocytic functions and their relevance in pathological conditions and discusses astrocytic diversity in relation to morphology, function, and gene expression throughout the CNS.
Collapse
Affiliation(s)
- Vimal P Rupareliya
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Gandhinagar, Gujarat 382355, India
| | - Aditya A Singh
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Gandhinagar, Gujarat 382355, India
| | - Ayub Mohammed Butt
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Gandhinagar, Gujarat 382355, India
| | - Hariharan A
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Gandhinagar, Gujarat 382355, India
| | - Hemant Kumar
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Gandhinagar, Gujarat 382355, India.
| |
Collapse
|
16
|
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: 22] [Impact Index Per Article: 11.0] [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
|
17
|
Hein M, Mehnert A, Freund KB, Yu DY, Balaratnasingam C. Variability in Capillary Perfusion Is Increased in Regions of Retinal Ischemia Due to Branch Retinal Vein Occlusion. Invest Ophthalmol Vis Sci 2023; 64:30. [PMID: 37856113 PMCID: PMC10615145 DOI: 10.1167/iovs.64.13.30] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Accepted: 09/23/2023] [Indexed: 10/20/2023] Open
Abstract
Purpose To investigate alterations in macular perfusion variability due to branch retinal vein occlusion (BRVO) using a novel approach based on optical coherence tomography angiography (OCTA) coefficient of variation (CoV) analysis. Methods Thirteen eyes of 13 patients with macular ischemia due to BRVO were studied. Multiple consecutive en face OCTA images were acquired. Bias field correction, spatial alignment, and normalization of intensities across the images were performed followed by pixelwise computation of standard deviation divided by the mean to generate a CoV map. Region of interest-based CoV values, derived from this map, for arterioles, venules, and the microvasculature were compared between regions with macular ischemia and control areas of the same eye. Control areas were regions of the same macula that were not affected by the BRVO and had normal retinal vascular structure as seen on multimodal imaging and normal retinal vascular density measurements as quantified using OCTA. Results CoV increased by a mean value of 17.6% within the microvasculature of ischemic regions compared to the control microvasculature (P < 0.0001). CoV measurements of microvasculature were consistently greater in the ischemic area of all 13 eyes compared to control. There were no differences in CoV measurements between ischemic and control areas for arterioles (P = 0.13) and venules (P = 1.0). Conclusions Greater variability in microvasculature perfusion occurs at sites of macular ischemia due to BRVO. We report a novel way for quantifying macular perfusion variability using OCTA. This technique may have applicability for studying the pathophysiology of other retinal vascular diseases.
Collapse
Affiliation(s)
- Martin Hein
- Lions Eye Institute, Nedlands, Western Australia, Australia
- Centre for Ophthalmology and Visual Science, University of Western Australia, Perth, Australia
| | - Andrew Mehnert
- Lions Eye Institute, Nedlands, Western Australia, Australia
- Centre for Ophthalmology and Visual Science, University of Western Australia, Perth, Australia
| | - K. Bailey Freund
- Vitreous Retina Macula Consultants of New York, New York, New York, United States
- Department of Ophthalmology, New York University Grossman School of Medicine, New York, New York, United States
| | - Dao-Yi Yu
- Lions Eye Institute, Nedlands, Western Australia, Australia
- Centre for Ophthalmology and Visual Science, University of Western Australia, Perth, Australia
| | - Chandrakumar Balaratnasingam
- Lions Eye Institute, Nedlands, Western Australia, Australia
- Centre for Ophthalmology and Visual Science, University of Western Australia, Perth, Australia
- Department of Ophthalmology, Sir Charles Gairdner Hospital, Western Australia, Perth, Australia
| |
Collapse
|
18
|
Wu X, Li JR, Fu Y, Chen DY, Nie H, Tang ZP. From static to dynamic: live observation of the support system after ischemic stroke by two photon-excited fluorescence laser-scanning microscopy. Neural Regen Res 2023; 18:2093-2107. [PMID: 37056116 PMCID: PMC10328295 DOI: 10.4103/1673-5374.369099] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 12/21/2022] [Accepted: 01/13/2023] [Indexed: 02/17/2023] Open
Abstract
Ischemic stroke is one of the most common causes of mortality and disability worldwide. However, treatment efficacy and the progress of research remain unsatisfactory. As the critical support system and essential components in neurovascular units, glial cells and blood vessels (including the blood-brain barrier) together maintain an optimal microenvironment for neuronal function. They provide nutrients, regulate neuronal excitability, and prevent harmful substances from entering brain tissue. The highly dynamic networks of this support system play an essential role in ischemic stroke through processes including brain homeostasis, supporting neuronal function, and reacting to injuries. However, most studies have focused on postmortem animals, which inevitably lack critical information about the dynamic changes that occur after ischemic stroke. Therefore, a high-precision technique for research in living animals is urgently needed. Two-photon fluorescence laser-scanning microscopy is a powerful imaging technique that can facilitate live imaging at high spatiotemporal resolutions. Two-photon fluorescence laser-scanning microscopy can provide images of the whole-cortex vascular 3D structure, information on multicellular component interactions, and provide images of structure and function in the cranial window. This technique shifts the existing research paradigm from static to dynamic, from flat to stereoscopic, and from single-cell function to multicellular intercommunication, thus providing direct and reliable evidence to identify the pathophysiological mechanisms following ischemic stroke in an intact brain. In this review, we discuss exciting findings from research on the support system after ischemic stroke using two-photon fluorescence laser-scanning microscopy, highlighting the importance of dynamic observations of cellular behavior and interactions in the networks of the brain's support systems. We show the excellent application prospects and advantages of two-photon fluorescence laser-scanning microscopy and predict future research developments and directions in the study of ischemic stroke.
Collapse
Affiliation(s)
- Xuan Wu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Jia-Rui Li
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Yu Fu
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Dan-Yang Chen
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Hao Nie
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Zhou-Ping Tang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| |
Collapse
|
19
|
Sanicola HW, Stewart CE, Luther P, Yabut K, Guthikonda B, Jordan JD, Alexander JS. Pathophysiology, Management, and Therapeutics in Subarachnoid Hemorrhage and Delayed Cerebral Ischemia: An Overview. PATHOPHYSIOLOGY 2023; 30:420-442. [PMID: 37755398 PMCID: PMC10536590 DOI: 10.3390/pathophysiology30030032] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 08/21/2023] [Accepted: 09/07/2023] [Indexed: 09/28/2023] Open
Abstract
Subarachnoid hemorrhage (SAH) is a type of hemorrhagic stroke resulting from the rupture of an arterial vessel within the brain. Unlike other stroke types, SAH affects both young adults (mid-40s) and the geriatric population. Patients with SAH often experience significant neurological deficits, leading to a substantial societal burden in terms of lost potential years of life. This review provides a comprehensive overview of SAH, examining its development across different stages (early, intermediate, and late) and highlighting the pathophysiological and pathohistological processes specific to each phase. The clinical management of SAH is also explored, focusing on tailored treatments and interventions to address the unique pathological changes that occur during each stage. Additionally, the paper reviews current treatment modalities and pharmacological interventions based on the evolving guidelines provided by the American Heart Association (AHA). Recent advances in our understanding of SAH will facilitate clinicians' improved management of SAH to reduce the incidence of delayed cerebral ischemia in patients.
Collapse
Affiliation(s)
- Henry W. Sanicola
- Department of Neurology, Louisiana State University Health Sciences Center in Shreveport, Shreveport, LA 71103, USA;
| | - Caleb E. Stewart
- Department of Neurosurgery, Louisiana State University Health Sciences Center in Shreveport, Shreveport, LA 71103, USA;
| | - Patrick Luther
- School of Medicine, Louisiana State University Health Sciences Center in Shreveport, Shreveport, LA 71103, USA; (P.L.); (K.Y.)
| | - Kevin Yabut
- School of Medicine, Louisiana State University Health Sciences Center in Shreveport, Shreveport, LA 71103, USA; (P.L.); (K.Y.)
| | - Bharat Guthikonda
- Department of Neurosurgery, Louisiana State University Health Sciences Center in Shreveport, Shreveport, LA 71103, USA;
| | - J. Dedrick Jordan
- Department of Neurology, Louisiana State University Health Sciences Center in Shreveport, Shreveport, LA 71103, USA;
| | - J. Steven Alexander
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center in Shreveport, Shreveport, LA 71103, USA
| |
Collapse
|
20
|
Abstract
Astrocyte endfeet enwrap the entire vascular tree within the central nervous system, where they perform important functions in regulating the blood-brain barrier (BBB), cerebral blood flow, nutrient uptake, and waste clearance. Accordingly, astrocyte endfeet contain specialized organelles and proteins, including local protein translation machinery and highly organized scaffold proteins, which anchor channels, transporters, receptors, and enzymes critical for astrocyte-vascular interactions. Many neurological diseases are characterized by the loss of polarization of specific endfoot proteins, vascular dysregulation, BBB disruption, altered waste clearance, or, in extreme cases, loss of endfoot coverage. A role for astrocyte endfeet has been demonstrated or postulated in many of these conditions. This review provides an overview of the development, composition, function, and pathological changes of astrocyte endfeet and highlights the gaps in our knowledge that future research should address.
Collapse
Affiliation(s)
- Blanca Díaz-Castro
- UK Dementia Research Institute and Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, Scotland, UK;
| | - Stefanie Robel
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama, USA;
| | - Anusha Mishra
- Department of Neurology Jungers Center for Neurosciences Research and Knight Cardiovascular Institute, Oregon Health & Science University, Portland, Oregon, USA;
| |
Collapse
|
21
|
Lia A, Di Spiezio A, Speggiorin M, Zonta M. Two decades of astrocytes in neurovascular coupling. FRONTIERS IN NETWORK PHYSIOLOGY 2023; 3:1162757. [PMID: 37078069 PMCID: PMC10106690 DOI: 10.3389/fnetp.2023.1162757] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 03/23/2023] [Indexed: 04/05/2023]
Abstract
The brain is a highly energy demanding organ, which accounts in humans for the 20% of total energy consumption at resting state although comprising only 2% of the body mass. The necessary delivery of nutrients to brain parenchyma is ensured by the cerebral circulatory system, through the exchange of glucose and oxygen (O2) at the capillary level. Notably, a tight spatial and temporal correlation exists between local increases in neuronal activity and the subsequent changes in regional cerebral blood flow. The recognized concept of neurovascular coupling (NVC), also named functional hyperemia, expresses this close relationship and stands at the basis of the modern functional brain imaging techniques. Different cellular and molecular mechanisms have been proposed to mediate this tight coupling. In this context, astrocytes are ideally positioned to act as relay elements that sense neuronal activity through their perisynaptic processes and release vasodilator agents at their endfeet in contact with brain parenchymal vessels. Two decades after the astrocyte involvement in neurovascular coupling has been proposed, we here review the experimental evidence that contributed to unraveling the molecular and cellular mechanisms underlying cerebral blood flow regulation. While traveling through the different controversies that moved the research in this field, we keep a peculiar focus on those exploring the role of astrocytes in neurovascular coupling and conclude with two sections related to methodological aspects in neurovascular research and to some pathological conditions resulting in altered neurovascular coupling.
Collapse
Affiliation(s)
- Annamaria Lia
- Neuroscience Institute, National Research Council (CNR), Padua, Italy
| | - Alessandro Di Spiezio
- Neuroscience Institute, National Research Council (CNR), Padua, Italy
- Department of Biomedical Sciences, University of Padua, Padua, Italy
| | | | - Micaela Zonta
- Neuroscience Institute, National Research Council (CNR), Padua, Italy
| |
Collapse
|
22
|
Liu K, Zhu T, Gao M, Yin X, Zheng R, Yan Y, Gao L, Ding Z, Ye J, Li P. Functional OCT angiography reveals early retinal neurovascular dysfunction in diabetes with capillary resolution. BIOMEDICAL OPTICS EXPRESS 2023; 14:1670-1684. [PMID: 37078055 PMCID: PMC10110312 DOI: 10.1364/boe.485940] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 03/18/2023] [Accepted: 03/18/2023] [Indexed: 05/03/2023]
Abstract
Altered retinal neurovascular coupling may contribute to the development and progression of diabetic retinopathy (DR) but remains highly challenging to measure due to limited resolution and field of view of the existing functional hyperemia imaging. Here, we present a novel modality of functional OCT angiography (fOCTA) that allows a 3D imaging of retinal functional hyperemia across the entire vascular tree with single-capillary resolution. In fOCTA, functional hyperemia was evoked by a flicker light stimulation, recorded by a synchronized time-lapse OCTA (i.e., 4D), and extracted precisely from each capillary segment (space) and stimulation period (time) in the OCTA time series. The high-resolution fOCTA revealed that the retinal capillaries, particularly the intermediate capillary plexus, exhibited apparent hyperemic response in normal mice, and significant functional hyperemia loss (P < 0.001) at an early stage of DR with few overt signs of retinopathy and visible restoration after aminoguanidine treatment (P < 0.05). Retinal capillary functional hyperemia has strong potential to provide sensitive biomarkers of early DR, and retinal fOCTA would provide new insights into the pathophysiology, screening and treatment of early DR.
Collapse
Affiliation(s)
- Kaiyuan Liu
- State Key Laboratory of Modern Optical Instrumentation, College of Optical Science and Engineering, Zhejiang University, Hangzhou 310027, China
| | - Tiepei Zhu
- Eye Center of the Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Mengqin Gao
- State Key Laboratory of Modern Optical Instrumentation, College of Optical Science and Engineering, Zhejiang University, Hangzhou 310027, China
| | - Xiaoting Yin
- State Key Laboratory of Modern Optical Instrumentation, College of Optical Science and Engineering, Zhejiang University, Hangzhou 310027, China
| | - Rong Zheng
- State Key Laboratory of Modern Optical Instrumentation, College of Optical Science and Engineering, Zhejiang University, Hangzhou 310027, China
| | - Yan Yan
- Eye Center of the Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Lei Gao
- Jiaxing Key Laboratory of Photonic Sensing & Intelligent Imaging, Jiaxing 314000, China
- Intelligent Optics & Photonics Research Center, Jiaxing Research Institute, Zhejiang University, Jiaxing 314000, China
| | - Zhihua Ding
- State Key Laboratory of Modern Optical Instrumentation, College of Optical Science and Engineering, Zhejiang University, Hangzhou 310027, China
| | - Juan Ye
- Eye Center of the Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Peng Li
- State Key Laboratory of Modern Optical Instrumentation, College of Optical Science and Engineering, Zhejiang University, Hangzhou 310027, China
- Jiaxing Key Laboratory of Photonic Sensing & Intelligent Imaging, Jiaxing 314000, China
- Intelligent Optics & Photonics Research Center, Jiaxing Research Institute, Zhejiang University, Jiaxing 314000, China
| |
Collapse
|
23
|
Henao‐Restrepo J, López‐Murillo C, Valderrama‐Carmona P, Orozco‐Santa N, Gomez J, Gutiérrez‐Vargas J, Moraga R, Toledo J, Littau JL, Härtel S, Arboleda‐Velásquez JF, Sepulveda‐Falla D, Lopera F, Cardona‐Gómez GP, Villegas A, Posada‐Duque R. Gliovascular alterations in sporadic and familial Alzheimer's disease: APOE3 Christchurch homozygote glioprotection. Brain Pathol 2023; 33:e13119. [PMID: 36130084 PMCID: PMC10041169 DOI: 10.1111/bpa.13119] [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: 01/21/2022] [Accepted: 08/30/2022] [Indexed: 11/28/2022] Open
Abstract
In response to brain insults, astrocytes become reactive, promoting protection and tissue repair. However, astroglial reactivity is typical of brain pathologies, including Alzheimer's disease (AD). Considering the heterogeneity of the reactive response, the role of astrocytes in the course of different forms of AD has been underestimated. Colombia has the largest human group known to have familial AD (FAD). This group carries the autosomal dominant and fully penetrant mutation E280A in PSEN1, which causes early-onset AD. Recently, our group identified an E280A carrier who did not develop FAD. The individual was homozygous for the Christchurch mutation R136S in APOE3 (APOEch). Remarkably, APOE is the main genetic risk factor for developing sporadic AD (SAD) and most of cerebral ApoE is produced by astroglia. Here, we characterized astrocyte properties related to reactivity, glutamate homeostasis, and structural integrity of the gliovascular unit (GVU), as factors that could underlie the pathogenesis or protection of AD. Specifically, through histological and 3D microscopy analyses of postmortem samples, we briefly describe the histopathology and cytoarchitecture of the frontal cortex of SAD, FAD, and APOEch, and demonstrate that, while astrodegeneration and vascular deterioration are prominent in SAD, FAD is characterized by hyperreactive-like glia, and APOEch displays the mildest astrocytic and vascular alterations despite having the highest burden of Aβ. Notably, astroglial, gliovascular, and vascular disturbances, as well as brain cell death, correlate with the specific astrocytic phenotypes identified in each condition. This study provides new insights into the potential relevance of the gliovasculature in the development and protection of AD. To our knowledge, this is the first study assessing the components of the GVU in human samples of SAD, FAD, and APOEch.
Collapse
Affiliation(s)
- Julián Henao‐Restrepo
- Instituto de Biología, Facultad de Ciencias Exactas y NaturalesUniversidad de AntioquiaMedellínColombia
- Área de Neurobiología Celular y Molecular, Grupo de Neurociencias de AntioquiaUniversidad de AntioquiaMedellínColombia
| | - Carolina López‐Murillo
- Instituto de Biología, Facultad de Ciencias Exactas y NaturalesUniversidad de AntioquiaMedellínColombia
- Área de Neurobiología Celular y Molecular, Grupo de Neurociencias de AntioquiaUniversidad de AntioquiaMedellínColombia
| | - Pablo Valderrama‐Carmona
- Instituto de Biología, Facultad de Ciencias Exactas y NaturalesUniversidad de AntioquiaMedellínColombia
- Área de Neurobiología Celular y Molecular, Grupo de Neurociencias de AntioquiaUniversidad de AntioquiaMedellínColombia
| | - Natalia Orozco‐Santa
- Instituto de Biología, Facultad de Ciencias Exactas y NaturalesUniversidad de AntioquiaMedellínColombia
- Área de Neurobiología Celular y Molecular, Grupo de Neurociencias de AntioquiaUniversidad de AntioquiaMedellínColombia
| | - Johana Gomez
- Grupo de Neurociencias de Antioquia, Facultad de MedicinaSIU, Universidad de AntioquiaMedellínColombia
| | - Johanna Gutiérrez‐Vargas
- Instituto de Biología, Facultad de Ciencias Exactas y NaturalesUniversidad de AntioquiaMedellínColombia
- Health Sciences FacultyRemington University CorporationMedellínColombia
| | - Renato Moraga
- Biomedical Neuroscience Institute BNI, Faculty of MedicineUniversity of ChileSantiagoChile
| | - Jorge Toledo
- Biomedical Neuroscience Institute BNI, Faculty of MedicineUniversity of ChileSantiagoChile
| | - Jessica Lisa Littau
- Molecular Neuropathology of Alzheimer's DiseaseInstitute of Neuropathology, University Medical Center Hamburg‐EppendorfHamburgGermany
| | - Steffen Härtel
- Biomedical Neuroscience Institute BNI, Faculty of MedicineUniversity of ChileSantiagoChile
| | - Joseph F. Arboleda‐Velásquez
- Schepens Eye Research Institute of Mass Eye and Ear, Department of OphthalmologyHarvard Medical SchoolBostonMassachusettsUSA
| | - Diego Sepulveda‐Falla
- Molecular Neuropathology of Alzheimer's DiseaseInstitute of Neuropathology, University Medical Center Hamburg‐EppendorfHamburgGermany
| | - Francisco Lopera
- Grupo de Neurociencias de Antioquia, Facultad de MedicinaSIU, Universidad de AntioquiaMedellínColombia
| | - Gloria Patricia Cardona‐Gómez
- Área de Neurobiología Celular y Molecular, Grupo de Neurociencias de AntioquiaUniversidad de AntioquiaMedellínColombia
| | - Andrés Villegas
- Grupo de Neurociencias de Antioquia, Facultad de MedicinaSIU, Universidad de AntioquiaMedellínColombia
| | - Rafael Posada‐Duque
- Instituto de Biología, Facultad de Ciencias Exactas y NaturalesUniversidad de AntioquiaMedellínColombia
- Área de Neurobiología Celular y Molecular, Grupo de Neurociencias de AntioquiaUniversidad de AntioquiaMedellínColombia
| |
Collapse
|
24
|
Ryan AK, Rich W, Reilly MA. Oxidative stress in the brain and retina after traumatic injury. Front Neurosci 2023; 17:1021152. [PMID: 36816125 PMCID: PMC9935939 DOI: 10.3389/fnins.2023.1021152] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 01/13/2023] [Indexed: 02/05/2023] Open
Abstract
The brain and the retina share many physiological similarities, which allows the retina to serve as a model of CNS disease and disorder. In instances of trauma, the eye can even indicate damage to the brain via abnormalities observed such as irregularities in pupillary reflexes in suspected traumatic brain injury (TBI) patients. Elevation of reactive oxygen species (ROS) has been observed in neurodegenerative disorders and in both traumatic optic neuropathy (TON) and in TBI. In a healthy system, ROS play a pivotal role in cellular communication, but in neurodegenerative diseases and post-trauma instances, ROS elevation can exacerbate neurodegeneration in both the brain and the retina. Increased ROS can overwhelm the inherent antioxidant systems which are regulated via mitochondrial processes. The overabundance of ROS can lead to protein, DNA, and other forms of cellular damage which ultimately result in apoptosis. Even though elevated ROS have been observed to be a major cause in the neurodegeneration observed after TON and TBI, many antioxidants therapeutic strategies fail. In order to understand why these therapeutic approaches fail further research into the direct injury cascades must be conducted. Additional therapeutic approaches such as therapeutics capable of anti-inflammatory properties and suppression of other neurodegenerative processes may be needed for the treatment of TON, TBI, and neurodegenerative diseases.
Collapse
Affiliation(s)
- Annie K. Ryan
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH, United States
| | - Wade Rich
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH, United States
| | - Matthew A. Reilly
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH, United States,Department of Ophthalmology and Visual Sciences, The Ohio State University, Columbus, OH, United States,*Correspondence: Matthew A. Reilly,
| |
Collapse
|
25
|
Lee E, Lee EA, Kong E, Chon H, Llaiqui-Condori M, Park CH, Park BY, Kang NR, Yoo JS, Lee HS, Kim HS, Park SH, Choi SW, Vestweber D, Lee JH, Kim P, Lee WS, Kim I. An agonistic anti-Tie2 antibody suppresses the normal-to-tumor vascular transition in the glioblastoma invasion zone. Exp Mol Med 2023; 55:470-484. [PMID: 36828931 PMCID: PMC9981882 DOI: 10.1038/s12276-023-00939-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 12/05/2022] [Accepted: 12/08/2022] [Indexed: 02/26/2023] Open
Abstract
Tumor progression is intimately associated with the vasculature, as tumor proliferation induces angiogenesis and tumor cells metastasize to distant organs via blood vessels. However, whether tumor invasion is associated with blood vessels remains unknown. As glioblastoma (GBM) is featured by aggressive invasion and vascular abnormalities, we characterized the onset of vascular remodeling in the diffuse tumor infiltrating zone by establishing new spontaneous GBM models with robust invasion capacity. Normal brain vessels underwent a gradual transition to severely impaired tumor vessels at the GBM periphery over several days. Increasing vasodilation from the tumor periphery to the tumor core was also found in human GBM. The levels of vascular endothelial growth factor (VEGF) and VEGF receptor 2 (VEGFR2) showed a spatial correlation with the extent of vascular abnormalities spanning the tumor-invading zone. Blockade of VEGFR2 suppressed vascular remodeling at the tumor periphery, confirming the role of VEGF-VEGFR2 signaling in the invasion-associated vascular transition. As angiopoietin-2 (ANGPT2) was expressed in only a portion of the central tumor vessels, we developed a ligand-independent tunica interna endothelial cell kinase 2 (Tie2)-activating antibody that can result in Tie2 phosphorylation in vivo. This agonistic anti-Tie2 antibody effectively normalized the vasculature in both the tumor periphery and tumor center, similar to the effects of VEGFR2 blockade. Mechanistically, this antibody-based Tie2 activation induced VE-PTP-mediated VEGFR2 dephosphorylation in vivo. Thus, our study reveals that the normal-to-tumor vascular transition is spatiotemporally associated with GBM invasion and may be controlled by Tie2 activation via a novel mechanism of action.
Collapse
Affiliation(s)
- Eunhyeong Lee
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Eun-Ah Lee
- R&D Center, PharmAbcine Inc., Daejeon, 34047, Republic of Korea
| | - Eunji Kong
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Haemin Chon
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Melissa Llaiqui-Condori
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Cheon Ho Park
- R&D Center, PharmAbcine Inc., Daejeon, 34047, Republic of Korea
| | - Beom Yong Park
- R&D Center, PharmAbcine Inc., Daejeon, 34047, Republic of Korea
| | - Nu Ri Kang
- R&D Center, PharmAbcine Inc., Daejeon, 34047, Republic of Korea
| | - Jin-San Yoo
- R&D Center, PharmAbcine Inc., Daejeon, 34047, Republic of Korea
| | - Hyun-Soo Lee
- Department of Bio and Brain Engineering, KAIST, Daejeon, 34141, Republic of Korea
| | - Hyung-Seok Kim
- Department of Forensic Medicine, Chonnam National University Medical School, Gwangju, 61463, Republic of Korea
| | - Sung-Hong Park
- Department of Bio and Brain Engineering, KAIST, Daejeon, 34141, Republic of Korea
| | - Seung-Won Choi
- Department of Neurosurgery, Samsung Medical Center, Sungkyunkwan University, Seoul, 06351, Republic of Korea
| | - Dietmar Vestweber
- Max Planck Institute for Molecular Biomedicine, D-48149, Muenster, Germany
| | - Jeong Ho Lee
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea.,BioMedical Research Center, KAIST, Daejeon, 34141, Republic of Korea.,SoVarGen, Inc., Daejeon, 34051, Republic of Korea
| | - Pilhan Kim
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea.,Graduate School of Nanoscience and Technology, Daejeon, 34141, Republic of Korea.,KI for Health Science and Technology, KAIST, Daejeon, 34141, Republic of Korea
| | - Weon Sup Lee
- R&D Center, PharmAbcine Inc., Daejeon, 34047, Republic of Korea.
| | - Injune Kim
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea. .,BioMedical Research Center, KAIST, Daejeon, 34141, Republic of Korea.
| |
Collapse
|
26
|
Gudkov SV, Burmistrov DE, Kondakova EV, Sarimov RM, Yarkov RS, Franceschi C, Vedunova MV. An emerging role of astrocytes in aging/neuroinflammation and gut-brain axis with consequences on sleep and sleep disorders. Ageing Res Rev 2023; 83:101775. [PMID: 36334910 DOI: 10.1016/j.arr.2022.101775] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 10/05/2022] [Accepted: 10/30/2022] [Indexed: 11/18/2022]
Abstract
Understanding the role of astrocytes in the central nervous system has changed dramatically over the last decade. The accumulating findings indicate that glial cells are involved not only in the maintenance of metabolic and ionic homeostasis and in the implementation of trophic functions but also in cognitive functions and information processing in the brain. Currently, there are some controversies regarding the role of astrocytes in complex processes such as aging of the nervous system and the pathogenesis of age-related neurodegenerative diseases. Many findings confirm the important functional role of astrocytes in age-related brain changes, including sleep disturbance and the development of neurodegenerative diseases and particularly Alzheimer's disease. Until recent years, neurobiological research has focused mainly on neuron-glial interactions, in which individual astrocytes locally modulate neuronal activity and communication between neurons. The review considers the role of astrocytes in the physiology of sleep and as an important "player" in the development of neurodegenerative diseases. In addition, the features of the astrocytic network reorganization during aging are discussed.
Collapse
Affiliation(s)
- Sergey V Gudkov
- Prokhorov General Physics Institute of the Russian Academy of Sciences, 38 Vavilov str., 119991 Moscow, Russia; Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, 23 Gagarin ave., 603022 Nizhny Novgorod, Russia.
| | - Dmitriy E Burmistrov
- Prokhorov General Physics Institute of the Russian Academy of Sciences, 38 Vavilov str., 119991 Moscow, Russia.
| | - Elena V Kondakova
- Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, 23 Gagarin ave., 603022 Nizhny Novgorod, Russia.
| | - Ruslan M Sarimov
- Prokhorov General Physics Institute of the Russian Academy of Sciences, 38 Vavilov str., 119991 Moscow, Russia.
| | - Roman S Yarkov
- Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, 23 Gagarin ave., 603022 Nizhny Novgorod, Russia.
| | - Claudio Franceschi
- Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, 23 Gagarin ave., 603022 Nizhny Novgorod, Russia.
| | - Maria V Vedunova
- Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, 23 Gagarin ave., 603022 Nizhny Novgorod, Russia.
| |
Collapse
|
27
|
Marchese NA, Ríos MN, Guido ME. Müller glial cell photosensitivity: a novel function bringing higher complexity to vertebrate retinal physiology. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY 2023. [DOI: 10.1016/j.jpap.2023.100162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
|
28
|
Fletcher EL, Dixon MA, Mills SA, Jobling AI. Anomalies in neurovascular coupling during early diabetes: A review. Clin Exp Ophthalmol 2023; 51:81-91. [PMID: 36349522 PMCID: PMC10947109 DOI: 10.1111/ceo.14190] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Revised: 10/26/2022] [Accepted: 11/04/2022] [Indexed: 11/11/2022]
Abstract
Diabetic retinopathy is the most feared complication for those with diabetes. Although visible vascular pathology traditionally defines the management of this condition, it is now recognised that a range of cellular changes occur in the retina from an early stage of diabetes. One of the most significant functional changes that occurs in those with diabetes is a loss of vasoregulation in response to changes in neural activity. There are several retinal cell types that are critical for mediating so-called neurovascular coupling, including Müller cells, microglia and pericytes. Although there is a great deal of evidence that suggests that Müller cells are integral to regulating the vasculature, they only modulate part of the vascular tree, highlighting the complexity of vasoregulation within the retina. Recent studies suggest that retinal immune cells, microglia, play an important role in mediating vasoconstriction. Importantly, retinal microglia contact both the vasculature and neural synapses and induce vasoconstriction in response to neurally expressed chemokines such as fractalkine. This microglial-dependent regulation occurs via the vasomediator angiotensinogen. Diabetes alters the way microglia regulate the retinal vasculature, by increasing angiotensinogen expression, causing capillary vasoconstriction and contributing to a loss of vascular reactivity to physiological signals. This article summarises recent studies showing changes in vascular regulation during diabetes, the potential mechanisms by which this occurs and the significance of these early changes to the progression of diabetic retinopathy.
Collapse
Affiliation(s)
- Erica L. Fletcher
- Department of Anatomy and PhysiologyThe University of MelbourneMelbourneVictoriaAustralia
| | - Michael A. Dixon
- Department of Anatomy and PhysiologyThe University of MelbourneMelbourneVictoriaAustralia
| | - Samuel A. Mills
- Department of Anatomy and Developmental BiologyMonash UniversityMelbourneVictoriaAustralia
| | - Andrew I. Jobling
- Department of Anatomy and PhysiologyThe University of MelbourneMelbourneVictoriaAustralia
| |
Collapse
|
29
|
Križaj D, Cordeiro S, Strauß O. Retinal TRP channels: Cell-type-specific regulators of retinal homeostasis and multimodal integration. Prog Retin Eye Res 2023; 92:101114. [PMID: 36163161 PMCID: PMC9897210 DOI: 10.1016/j.preteyeres.2022.101114] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 08/03/2022] [Accepted: 08/08/2022] [Indexed: 02/05/2023]
Abstract
Transient receptor potential (TRP) channels are a widely expressed family of 28 evolutionarily conserved cationic ion channels that operate as primary detectors of chemical and physical stimuli and secondary effectors of metabotropic and ionotropic receptors. In vertebrates, the channels are grouped into six related families: TRPC, TRPV, TRPM, TRPA, TRPML, and TRPP. As sensory transducers, TRP channels are ubiquitously expressed across the body and the CNS, mediating critical functions in mechanosensation, nociception, chemosensing, thermosensing, and phototransduction. This article surveys current knowledge about the expression and function of the TRP family in vertebrate retinas, which, while dedicated to transduction and transmission of visual information, are highly susceptible to non-visual stimuli. Every retinal cell expresses multiple TRP subunits, with recent evidence establishing their critical roles in paradigmatic aspects of vertebrate vision that include TRPM1-dependent transduction of ON bipolar signaling, TRPC6/7-mediated ganglion cell phototransduction, TRP/TRPL phototransduction in Drosophila and TRPV4-dependent osmoregulation, mechanotransduction, and regulation of inner and outer blood-retina barriers. TRP channels tune light-dependent and independent functions of retinal circuits by modulating the intracellular concentration of the 2nd messenger calcium, with emerging evidence implicating specific subunits in the pathogenesis of debilitating diseases such as glaucoma, ocular trauma, diabetic retinopathy, and ischemia. Elucidation of TRP channel involvement in retinal biology will yield rewards in terms of fundamental understanding of vertebrate vision and therapeutic targeting to treat diseases caused by channel dysfunction or over-activation.
Collapse
Affiliation(s)
- David Križaj
- Departments of Ophthalmology, Neurobiology, and Bioengineering, University of Utah, Salt Lake City, USA
| | - Soenke Cordeiro
- Institute of Physiology, Faculty of Medicine, Christian-Albrechts-University Kiel, Germany
| | - Olaf Strauß
- Experimental Ophthalmology, Department of Ophthalmology, Charité - Universitätsmedizin Berlin, a Corporate Member of Freie Universität, Humboldt-University, The Berlin Institute of Health, Berlin, Germany.
| |
Collapse
|
30
|
Wang Y, Wang Y, Yue G, Zhao Y. Energy metabolism disturbance in migraine: From a mitochondrial point of view. Front Physiol 2023; 14:1133528. [PMID: 37123270 PMCID: PMC10133718 DOI: 10.3389/fphys.2023.1133528] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Accepted: 03/20/2023] [Indexed: 05/02/2023] Open
Abstract
Migraine is a serious central nervous system disease with a high incidence rate. Its pathogenesis is very complex, which brings great difficulties for clinical treatment. Recently, many studies have revealed that mitochondrial dysfunction may play a key role in migraine, which affects the hyperosmotic of Ca2+, the excessive production of free radicals, the decrease of mitochondrial membrane potential, the imbalance of mPTP opening and closing, and the decrease of oxidative phosphorylation level, which leads to neuronal energy exhaustion and apoptosis, and finally lessens the pain threshold and migraine attack. This article mainly introduces cortical spreading depression, a pathogenesis of migraine, and then damages the related function of mitochondria, which leads to migraine. Oxidative phosphorylation and the tricarboxylic acid cycle are the main ways to provide energy for the body. 95 percent of the energy needed for cell survival is provided by the mitochondrial respiratory chain. At the same time, hypoxia can lead to cell death and migraine. The pathological opening of the mitochondrial permeability transition pore can promote the interaction between pro-apoptotic protein and mitochondrial, destroy the structure of mPTP, and further lead to cell death. The increase of mPTP permeability can promote the accumulation of reactive oxygen species, which leads to a series of changes in the expression of proteins related to energy metabolism. Both Nitric oxide and Calcitonin gene-related peptide are closely related to the attack of migraine. Recent studies have shown that changes in their contents can also affect the energy metabolism of the body, so this paper reviews the above mechanisms and discusses the mechanism of brain energy metabolism of migraine, to provide new strategies for the prevention and treatment of migraine and promote the development of individualized and accurate treatment of migraine.
Collapse
Affiliation(s)
- Yicheng Wang
- Department of Neurology, The Third Affiliated Hospital of Beijing University of Chinese Medicine, Beijing, China
| | - Yongli Wang
- Department of Neurology, Xiamen Hospital of Traditional Chinese Medicine, Xiamen, China
| | - Guangxin Yue
- Institute of Basic Theory for Chinese Medicine, Chinese Academy of Chinese Medical Sciences, Beijing, China
| | - Yonglie Zhao
- Department of Neurology, The Third Affiliated Hospital of Beijing University of Chinese Medicine, Beijing, China
- *Correspondence: Yonglie Zhao,
| |
Collapse
|
31
|
Sbardella D, Tundo GR, Mecchia A, Palumbo C, Atzori MG, Levati L, Boccaccini A, Caccuri AM, Cascio P, Lacal PM, Graziani G, Varano M, Coletta M, Parravano M. A novel and atypical NF-KB pro-inflammatory program regulated by a CamKII-proteasome axis is involved in the early activation of Muller glia by high glucose. Cell Biosci 2022; 12:108. [PMID: 35842713 PMCID: PMC9287993 DOI: 10.1186/s13578-022-00839-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 06/28/2022] [Indexed: 11/29/2022] Open
Abstract
Background Diabetic retinopathy (DR) is a microvascular complication of diabetes with a heavy impact on the quality of life of subjects and with a dramatic burden for health and economic systems on a global scale. Although the pathogenesis of DR is largely unknown, several preclinical data have pointed out to a main role of Muller glia (MG), a cell type which spans across the retina layers providing nourishment and support for Retina Ganglion Cells (RGCs), in sensing hyper-glycemia and in acquiring a pro-inflammatory polarization in response to this insult. Results By using a validated experimental model of DR in vitro, rMC1 cells challenged with high glucose, we uncovered the induction of an early (within minutes) and atypical Nuclear Factor-kB (NF-kB) signalling pathway regulated by a calcium-dependent calmodulin kinase II (CamKII)-proteasome axis. Phosphorylation of proteasome subunit Rpt6 (at Serine 120) by CamKII stimulated the accelerated turnover of IkBα (i.e., the natural inhibitor of p65-50 transcription factor), regardless of the phosphorylation at Serine 32 which labels canonical NF-kB signalling. This event allowed the p65-p50 heterodimer to migrate into the nucleus and to induce transcription of IL-8, Il-1β and MCP-1. Pharmacological inhibition of CamKII as well as proteasome inhibition stopped this pro-inflammatory program, whereas introduction of a Rpt6 phospho-dead mutant (Rpt6-S120A) stimulated a paradoxical effect on NF-kB probably through the activation of a compensatory mechanism which may involve phosphorylation of 20S α4 subunit. Conclusions This study introduces a novel pathway of MG activation by high glucose and casts some light on the biological relevance of proteasome post-translational modifications in modulating pathways regulated through targeted proteolysis. Supplementary Information The online version contains supplementary material available at 10.1186/s13578-022-00839-x. High glucose quickly induces an atypical NF-kB pro-inflammatory program. CamKII phosphorylation of Rpt6 subunit of the proteasome stimulates IkBα turnover and p65-p50 release. Inhibition of either CamkII or proteasome blocks this pathway.
Collapse
|
32
|
Tabata H. Crosstalk between Blood Vessels and Glia during the Central Nervous System Development. Life (Basel) 2022; 12:1761. [PMID: 36362915 PMCID: PMC9699316 DOI: 10.3390/life12111761] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 10/21/2022] [Accepted: 10/28/2022] [Indexed: 11/07/2023] Open
Abstract
The formation of proper blood vessel patterns in the central nervous system (CNS) is crucial to deliver oxygen and nutrient to neurons efficiently. At the same time, neurons must be isolated from the outer blood circulation by a specialized structure, the blood-brain barrier (BBB), to maintain the microenvironment of brain parenchyma for the survival of neurons and proper synaptic transmission. To develop this highly organized structure, glial cells, a major component of the brain, have been reported to play essential roles. In this review, the crosstalk between the macroglia, including astrocytes and oligodendrocytes, and endothelial cells during the development of CNS will be discussed. First, the known roles of astrocytes in neuro-vascular unit and its development, and then, the requirements of astrocytes for BBB development and maintenance are shown. Then, various genetic and cellular studies revealing the roles of astrocytes in the growth of blood vessels by providing a scaffold, including laminins and fibronectin, as well as by secreting trophic factors, including vascular endothelial growth factor (VEGF) and transforming growth factor-β (TGF-β) are introduced. Finally, the interactions between oligodendrocyte progenitors and blood vessels are overviewed. Although these studies revealed the necessity for proper communication between glia and endothelial cells for CNS development, our knowledge about the detailed cellular and molecular mechanisms for them is still limited. The questions to be clarified in the future are also discussed.
Collapse
Affiliation(s)
- Hidenori Tabata
- Department of Molecular Neurobiology, Institute for Developmental Research, Aichi Developmental Disability Center, 713-8 Kamiya, Kasugai 480-0392, Japan
| |
Collapse
|
33
|
Mori A, Seki H, Mizukoshi S, Uezono T, Sakamoto K. Role of Prostaglandins in Nitric Oxide-Induced Glial Cell-Mediated Vasodilation in Rat Retina. Biomolecules 2022; 12:biom12101403. [PMID: 36291611 PMCID: PMC9599243 DOI: 10.3390/biom12101403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 09/21/2022] [Accepted: 09/29/2022] [Indexed: 11/25/2022] Open
Abstract
We previously identified that NO derived from neuronal cells acts on glial cells and causes vasodilation in the healthy rat retina via the release of epoxyeicosatrienoic acids (EETs) and prostaglandins (PGs) by activation of the arachidonic acid cascade. However, it is not clear which PG types are involved in these responses. The aim of the present study was to identify prostanoid receptors involved in glial cell-derived vasodilation induced by NO in rat retina. Male Wistar rats were used to examine the effects of intravitreal pretreatment with indomethacin, a cyclooxygenase inhibitor; PF-04418948, a prostanoid EP2 receptor antagonist; and CAY10441, a prostanoid IP receptor antagonist, on the changes in the retinal arteriolar diameter induced by intravitreal administration of NOR3, an NO donor. Retinal arteriolar diameters were measured using ocular fundus images captured with a high-resolution digital camera in vivo. The increase in the retinal arteriolar diameter induced by intravitreal injection of NOR3 was significantly suppressed by intravitreal pretreatment with indomethacin and PF-04418948, but not by CAY10441. The dose of PF-04418948 and CAY10441 injected intravitreally in the present study significantly reduced the increase in the retinal arteriolar diameter induced by prostaglandin E2 (PGE2) and prostaglandin I2 (PGI2), respectively. These results suggest that activation of the arachidonic acid cascade and subsequent stimulation of prostanoid EP2 receptors are involved in rat retinal vasodilatory responses evoked by NO-induced glial cell stimulation. Therefore, glial cell-derived PGE2, similar to EETs, may play an important role in retinal vasodilatory mechanisms.
Collapse
|
34
|
Balaratnasingam C, An D, Hein M, Yu P, Yu DY. Studies of the retinal microcirculation using human donor eyes and high-resolution clinical imaging: Insights gained to guide future research in diabetic retinopathy. Prog Retin Eye Res 2022; 94:101134. [PMID: 37154065 DOI: 10.1016/j.preteyeres.2022.101134] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 09/18/2022] [Accepted: 10/03/2022] [Indexed: 11/06/2022]
Abstract
The microcirculation plays a key role in delivering oxygen to and removing metabolic wastes from energy-intensive retinal neurons. Microvascular changes are a hallmark feature of diabetic retinopathy (DR), a major cause of irreversible vision loss globally. Early investigators have performed landmark studies characterising the pathologic manifestations of DR. Previous works have collectively informed us of the clinical stages of DR and the retinal manifestations associated with devastating vision loss. Since these reports, major advancements in histologic techniques coupled with three-dimensional image processing has facilitated a deeper understanding of the structural characteristics in the healthy and diseased retinal circulation. Furthermore, breakthroughs in high-resolution retinal imaging have facilitated clinical translation of histologic knowledge to detect and monitor progression of microcirculatory disturbances with greater precision. Isolated perfusion techniques have been applied to human donor eyes to further our understanding of the cytoarchitectural characteristics of the normal human retinal circulation as well as provide novel insights into the pathophysiology of DR. Histology has been used to validate emerging in vivo retinal imaging techniques such as optical coherence tomography angiography. This report provides an overview of our research on the human retinal microcirculation in the context of the current ophthalmic literature. We commence by proposing a standardised histologic lexicon for characterising the human retinal microcirculation and subsequently discuss the pathophysiologic mechanisms underlying key manifestations of DR, with a focus on microaneurysms and retinal ischaemia. The advantages and limitations of current retinal imaging modalities as determined using histologic validation are also presented. We conclude with an overview of the implications of our research and provide a perspective on future directions in DR research.
Collapse
Affiliation(s)
- Chandrakumar Balaratnasingam
- Lions Eye Institute, Nedlands, Western Australia, Australia; Centre for Ophthalmology and Visual Science, University of Western Australia, Perth, Australia; Department of Ophthalmology, Sir Charles Gairdner Hospital, Western Australia, Australia.
| | - Dong An
- Lions Eye Institute, Nedlands, Western Australia, Australia; Centre for Ophthalmology and Visual Science, University of Western Australia, Perth, Australia
| | - Martin Hein
- Lions Eye Institute, Nedlands, Western Australia, Australia; Centre for Ophthalmology and Visual Science, University of Western Australia, Perth, Australia
| | - Paula Yu
- Lions Eye Institute, Nedlands, Western Australia, Australia; Centre for Ophthalmology and Visual Science, University of Western Australia, Perth, Australia
| | - Dao-Yi Yu
- Lions Eye Institute, Nedlands, Western Australia, Australia; Centre for Ophthalmology and Visual Science, University of Western Australia, Perth, Australia
| |
Collapse
|
35
|
Role of Microglia and Astrocytes in Alzheimer’s Disease: From Neuroinflammation to Ca2+ Homeostasis Dysregulation. Cells 2022; 11:cells11172728. [PMID: 36078138 PMCID: PMC9454513 DOI: 10.3390/cells11172728] [Citation(s) in RCA: 80] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 08/26/2022] [Accepted: 08/30/2022] [Indexed: 12/12/2022] Open
Abstract
Alzheimer’s disease (AD) is the most common form of dementia worldwide, with a complex, poorly understood pathogenesis. Cerebral atrophy, amyloid-β (Aβ) plaques, and neurofibrillary tangles represent the main pathological hallmarks of the AD brain. Recently, neuroinflammation has been recognized as a prominent feature of the AD brain and substantial evidence suggests that the inflammatory response modulates disease progression. Additionally, dysregulation of calcium (Ca2+) homeostasis represents another early factor involved in the AD pathogenesis, as intracellular Ca2+ concentration is essential to ensure proper cellular and neuronal functions. Although growing evidence supports the involvement of Ca2+ in the mechanisms of neurodegeneration-related inflammatory processes, scant data are available on its contribution in microglia and astrocytes functioning, both in health and throughout the AD continuum. Nevertheless, AD-related aberrant Ca2+ signalling in astrocytes and microglia is crucially involved in the mechanisms underpinning neuroinflammatory processes that, in turn, impact neuronal Ca2+ homeostasis and brain function. In this light, we attempted to provide an overview of the current understanding of the interactions between the glia cells-mediated inflammatory responses and the molecular mechanisms involved in Ca2+ homeostasis dysregulation in AD.
Collapse
|
36
|
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: 13] [Impact Index Per Article: 4.3] [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
|
37
|
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
|
38
|
Burboa PC, Puebla M, Gaete PS, Durán WN, Lillo MA. Connexin and Pannexin Large-Pore Channels in Microcirculation and Neurovascular Coupling Function. Int J Mol Sci 2022; 23:ijms23137303. [PMID: 35806312 PMCID: PMC9266979 DOI: 10.3390/ijms23137303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 06/27/2022] [Accepted: 06/28/2022] [Indexed: 01/27/2023] Open
Abstract
Microcirculation homeostasis depends on several channels permeable to ions and/or small molecules that facilitate the regulation of the vasomotor tone, hyperpermeability, the blood–brain barrier, and the neurovascular coupling function. Connexin (Cxs) and Pannexin (Panxs) large-pore channel proteins are implicated in several aspects of vascular physiology. The permeation of ions (i.e., Ca2+) and key metabolites (ATP, prostaglandins, D-serine, etc.) through Cxs (i.e., gap junction channels or hemichannels) and Panxs proteins plays a vital role in intercellular communication and maintaining vascular homeostasis. Therefore, dysregulation or genetic pathologies associated with these channels promote deleterious tissue consequences. This review provides an overview of current knowledge concerning the physiological role of these large-pore molecule channels in microcirculation (arterioles, capillaries, venules) and in the neurovascular coupling function.
Collapse
Affiliation(s)
- Pía C. Burboa
- Department of Pharmacology, Physiology and Neuroscience, New Jersey Medical School, Rutgers University, 185 South Orange Avenue, Newark, NJ 07103, USA; (P.C.B.); (W.N.D.)
- Departamento de Morfología y Función, Facultad de Salud y Ciencias Sociales, Sede Santiago Centro, Universidad de las Américas, Avenue República 71, Santiago 8370040, Chile;
| | - Mariela Puebla
- Departamento de Morfología y Función, Facultad de Salud y Ciencias Sociales, Sede Santiago Centro, Universidad de las Américas, Avenue República 71, Santiago 8370040, Chile;
| | - Pablo S. Gaete
- Department of Physiology and Membrane Biology, University of California at Davis, Davis, CA 95616, USA;
| | - Walter N. Durán
- Department of Pharmacology, Physiology and Neuroscience, New Jersey Medical School, Rutgers University, 185 South Orange Avenue, Newark, NJ 07103, USA; (P.C.B.); (W.N.D.)
- Rutgers School of Graduate Studies, 185 South Orange Avenue, Newark, NJ 07103, USA
| | - Mauricio A. Lillo
- Department of Pharmacology, Physiology and Neuroscience, New Jersey Medical School, Rutgers University, 185 South Orange Avenue, Newark, NJ 07103, USA; (P.C.B.); (W.N.D.)
- Correspondence:
| |
Collapse
|
39
|
Hanssen H, Streese L, Vilser W. Retinal vessel diameters and function in cardiovascular risk and disease. Prog Retin Eye Res 2022; 91:101095. [PMID: 35760749 DOI: 10.1016/j.preteyeres.2022.101095] [Citation(s) in RCA: 64] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 06/03/2022] [Accepted: 06/13/2022] [Indexed: 12/13/2022]
Abstract
In the last two decades evidence has gradually accumulated suggesting that the eye may be a unique window for cardiovascular risk stratification based on the assessment of subclinical damage of retinal microvascular structure and function. This can be facilitated by non-invasive analysis of static retinal vessel diameters and dynamic recording of flicker light-induced and endothelial function-related dilation of both retinal arterioles and venules. Recent new findings have made retinal microvascular biomarkers strong candidates for clinical implementation as reliable risk predictors. Beyond a review of the current evidence and state of research, the article aims to discuss the methodological benefits and pitfalls and to identify research gaps and future directions. Above all, the potential use for screening and treatment monitoring of cardiovascular disease risk are highlighted. The article provides fundamental comprehension of retinal vessel imaging by explaining anatomical and physiological essentials of the retinal microcirculation leading to a detailed description of the methodological approach. This allows for better understanding of the underlying retinal microvascular pathology associated with the prevalence and development of cardiovascular disease. A body of new evidence is presented on the clinical validity and predictive value of retinal vessel diameters and function for incidence cardiovascular disease and outcome. Findings in children indicate the potential for utility in childhood cardiovascular disease prevention, and the efficacy of exercise interventions highlight the treatment sensitivity of retinal microvascular biomarkers. Finally, coming from the availability of normative data, solutions for diagnostic challenges are discussed and conceptual steps towards clinical implementation are put into perspective.
Collapse
Affiliation(s)
- Henner Hanssen
- Department of Sport, Exercise and Health, Preventive Sports Medicine and Systems Physiology, Medical Faculty, University of Basel, Switzerland.
| | - Lukas Streese
- Department of Sport, Exercise and Health, Preventive Sports Medicine and Systems Physiology, Medical Faculty, University of Basel, Switzerland
| | - Walthard Vilser
- Institute of Biomedical Engineering and Informatics, Ilmenau University of Technology, Ilmenau, Germany; Neonatology and Pediatric Intensive Care Unit, Department of Pediatrics, Jena University Hospital, Jena, Germany
| |
Collapse
|
40
|
Fort PE, Losiewicz MK, Elghazi L, Kong D, Cras-Méneur C, Fingar DC, Kimball SR, Rajala RVS, Smith AJ, Ali RR, Abcouwer SF, Gardner TW. mTORC1 regulates high levels of protein synthesis in retinal ganglion cells of adult mice. J Biol Chem 2022; 298:101944. [PMID: 35447116 PMCID: PMC9117545 DOI: 10.1016/j.jbc.2022.101944] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 03/17/2022] [Accepted: 03/18/2022] [Indexed: 02/02/2023] Open
Abstract
Mechanistic target of rapamycin (mTOR) and mTOR complex 1 (mTORC1), linchpins of the nutrient sensing and protein synthesis pathways, are present at relatively high levels in the ganglion cell layer (GCL) and retinal ganglion cells (RGCs) of rodent and human retinas. However, the role of mTORCs in the control of protein synthesis in RGC is unknown. Here, we applied the SUrface SEnsing of Translation (SUnSET) method of nascent protein labeling to localize and quantify protein synthesis in the retinas of adult mice. We also used intravitreal injection of an adeno-associated virus 2 vector encoding Cre recombinase in the eyes of mtor- or rptor-floxed mice to conditionally knockout either both mTORCs or only mTORC1, respectively, in cells within the GCL. A novel vector encoding an inactive Cre mutant (CreΔC) served as control. We found that retinal protein synthesis was highest in the GCL, particularly in RGC. Negation of both complexes or only mTORC1 significantly reduced protein synthesis in RGC. In addition, loss of mTORC1 function caused a significant reduction in the pan-RGC marker, RNA-binding protein with multiple splicing, with little decrease of the total number of cells in the RGC layer, even at 25 weeks after adeno-associated virus-Cre injection. These findings reveal that mTORC1 signaling is necessary for maintaining the high rate of protein synthesis in RGCs of adult rodents, but it may not be essential to maintain RGC viability. These findings may also be relevant to understanding the pathophysiology of RGC disorders, including glaucoma, diabetic retinopathy, and optic neuropathies.
Collapse
Affiliation(s)
- Patrice E Fort
- Ophthalmology & Visual Sciences, University of Michigan Medical School, Ann Arbor, Michigan, USA; Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Mandy K Losiewicz
- Ophthalmology & Visual Sciences, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Lynda Elghazi
- Ophthalmology & Visual Sciences, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Dejuan Kong
- Ophthalmology & Visual Sciences, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Corentin Cras-Méneur
- Internal Medicine (MEND Division), University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Diane C Fingar
- Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Scot R Kimball
- Cellular & Molecular Physiology, Penn State College of Medicine, Hershey, Pennsylvania, USA
| | - Raju V S Rajala
- Departments of Ophthalmology and Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Alexander J Smith
- Centre for Gene Therapy and Regenerative Medicine, King's College London, England, United Kingdom
| | - Robin R Ali
- Ophthalmology & Visual Sciences, University of Michigan Medical School, Ann Arbor, Michigan, USA; Centre for Gene Therapy and Regenerative Medicine, King's College London, England, United Kingdom
| | - Steven F Abcouwer
- Ophthalmology & Visual Sciences, University of Michigan Medical School, Ann Arbor, Michigan, USA.
| | - Thomas W Gardner
- Ophthalmology & Visual Sciences, University of Michigan Medical School, Ann Arbor, Michigan, USA; Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, Michigan, USA; Internal Medicine (MEND Division), University of Michigan Medical School, Ann Arbor, Michigan, USA
| |
Collapse
|
41
|
Dmitriev AV, Dmitriev AA, Linsenmeier RA. Extracellular K+ reflects light-evoked changes in retinal energy metabolism. Exp Eye Res 2022; 221:109133. [PMID: 35636490 PMCID: PMC10392107 DOI: 10.1016/j.exer.2022.109133] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 05/19/2022] [Accepted: 05/24/2022] [Indexed: 11/25/2022]
Abstract
Retinal neurons spend most of their energy to support the transmembrane movement of ions. Light-induced electrical activity is associated with a redistribution of ions, which affects the energy demand and results in a change in metabolism. Light-induced metabolic changes are expected to be different in distal and proximal retina due to differences in the light responses of different retinal cells. Extracellular K+ concentration ([K+]o) is a reliable indicator of local electrophysiological activity, and the purpose of this work was to compare [K+]o changes evoked by steady and flickering light in distal and proximal retina. Data were obtained from isolated mouse (C57Bl/6J) retinae. Double-barreled K+-selective microelectrodes were used to simultaneously record [K+]o and local ERGs. In the distal retina, photoreceptor hyperpolarization led to suppression of ion transfer, a decrease in [K+]o by 0.3-0.5 mM, reduced energy demand, and, as previously shown in vivo, decreased metabolism. Flickering light had the same effect on [K+]o in the distal retina as steady light of equivalent illumination. The conductance and voltage changes in postreceptor neurons are cell-specific, but the overall effect of steady light in the proximal retina is excitation, which is reflected in a [K+]o increase there (by a maximum of 0.2 mM). In steady light the [K+]o increase lasts only 1-2 s, but a sustained [K+]o increase is evoked by flickering light. A squarewave low frequency (1 Hz) flicker of photopic intensity produced the largest increases in [K+]o. Judging by measurements of [K+]o, steady illumination decreases energy metabolism in the distal retina, but not in the proximal retina (except for the first few seconds). Flickering light evokes the same decrease in the distal retina, but also evokes a sustained [K+]o increase in the proximal retina, suggesting an increase of metabolic demand there, especially at 1 Hz, when neurons of both on- and off-pathways appear to contribute maximally. This proximal retinal metabolic response to flicker correlates to the increase in blood flow during flicker that constitutes neurovascular coupling.
Collapse
|
42
|
Mori A, Yano E, Nishikiori M, Fujino S, Nakahara T. N-methyl-D-aspartic acid receptor-mediated vasodilation is attenuated in the retinas of diabetic rats. Curr Eye Res 2022; 47:1193-1199. [PMID: 35485610 DOI: 10.1080/02713683.2022.2072896] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
PURPOSE Activation of N-methyl-d-aspartic acid (NMDA) receptors enhances nitric oxide (NO) production in retinal neuronal cells, and in turn, NO released from neuronal cells induces glial cell-mediated dilation of retinal arterioles in rats. The purpose of this study was to examine how neuronal cell-dependent, glial cell-mediated vasodilation is impacted in diabetic rat retinas. MATERIALS AND METHODS Diabetes was induced in 6-week-old male Wistar rats by combining streptozotocin injection and D-glucose feeding. Two weeks later, the dilator function of retinal arterioles was assessed. RESULTS Compared with non-diabetic rats, the dilator responses of retinal arterioles induced by intravitreal injection of NMDA and NOR3, an NO donor, were reduced in diabetic rats. Following the blockade of large-conductance Ca2+-activated K+ (BKCa) channels with iberiotoxin, no significant difference in the retinal vasodilator response to NOR3 was observed between non-diabetic and diabetic rats. Intravitreal injection of 14,15-epoxyeicosatrienoic acid, a vasodilatory factor released from glial cells, dilated retinal arterioles, and the response was diminished by diabetes. CONCLUSION These findings suggest that the impaired BKCa channel function in vascular cells is responsible for the diminished neuronal cell-dependent, glial cell-mediated dilation of retinal arterioles during the early stage of diabetes.
Collapse
Affiliation(s)
- Asami Mori
- Department of Molecular Pharmacology, Kitasato University School of Pharmaceutical Sciences, 5-9-1 Shirokane, Minato-ku, Tokyo 108-8641, Japan.,Present address: Asami Mori, Ph.D., Laboratory of Medical Pharmacology, Department of Clinical & Pharmaceutical Sciences, Faculty of Pharma-Sciences, Teikyo University, Itabashi-ku, Tokyo 173-8605, Japan
| | - Erika Yano
- Department of Molecular Pharmacology, Kitasato University School of Pharmaceutical Sciences, 5-9-1 Shirokane, Minato-ku, Tokyo 108-8641, Japan
| | - Masato Nishikiori
- Department of Molecular Pharmacology, Kitasato University School of Pharmaceutical Sciences, 5-9-1 Shirokane, Minato-ku, Tokyo 108-8641, Japan
| | - Saho Fujino
- Department of Molecular Pharmacology, Kitasato University School of Pharmaceutical Sciences, 5-9-1 Shirokane, Minato-ku, Tokyo 108-8641, Japan
| | - Tsutomu Nakahara
- Department of Molecular Pharmacology, Kitasato University School of Pharmaceutical Sciences, 5-9-1 Shirokane, Minato-ku, Tokyo 108-8641, Japan
| |
Collapse
|
43
|
Bohra SK, Achar RR, Chidambaram SB, Pellegrino C, Laurin J, Masoodi M, Srinivasan A. CURRENT PERSPECTIVES ON MITOCHONDRIAL DYSFUNCTION IN MIGRAINE. Eur J Neurosci 2022; 56:3738-3754. [PMID: 35478208 DOI: 10.1111/ejn.15676] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 04/11/2022] [Accepted: 04/13/2022] [Indexed: 11/29/2022]
Abstract
Mitochondria is an autonomous organelle that plays a crucial role in the metabolic aspects of a cell. Cortical Spreading Depression (CSD) and fluctuations in the cerebral blood flow have for long been mechanisms underlying migraine. It is a neurovascular disorder with a unilateral manifestation of disturbing, throbbing and pulsating head pain. Migraine affects 2.6 and 21.7% of the general population and is the major cause of partial disability in the age group 15-49. Higher mutation rates, imbalance in concentration of physiologically relevant molecules, oxidative stress biomarkers have been the main themes of discussion in determining the role of mitochondrial disability in migraine. The correlation of migraine with other disorders like hemiplegic migraine, MELAS, TTH, CVS, ischemic stroke and hypertension has helped in the assessment of the physiological and morphogenetic basis of migraine. Here, we have reviewed the different nuances of mitochondrial dysfunction and migraine. The different mtDNA polymorphisms that can affect the generation and transmission of nerve impulse has been highlighted and supported with research findings. In addition to this, the genetic basis of migraine pathogenesis as a consequence of mutations in nuclear DNA that can in turn affect the synthesis of defective mitochondrial proteins is discussed along with a brief overview of epigenetic profile. This review gives an overview of the pathophysiology of migraine and explores mitochondrial dysfunction as a potential underlying mechanism. Also, therapeutic supplements for managing migraine have been discussed at different junctures in this paper.
Collapse
Affiliation(s)
- Shraman Kumar Bohra
- Department of Life Sciences, Pooja Bhagavat Memorial Mahajana Education Center, Mysore
| | - Raghu Ram Achar
- Division of Biochemistry, Faculty of Life Sciences, JSS Academy of Higher Education & Research. Mysore
| | - Saravana Babu Chidambaram
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysore
| | - Christophe Pellegrino
- Institut National de la Santé et de la Recherche Médicale, Institute of Mediterranean Neurobiology, Aix-Marseille University, Marseille, France
| | - Jerome Laurin
- Aix-Marseille University. Sport Science Faculty. Marseille. Institut de Neurobiologie de la Méditerranée, INMED (INSERM- AMU)., France
| | - Mojgan Masoodi
- Institute of Clinical Chemistry, University hospital Bern, Bern
| | - Asha Srinivasan
- Division of Nanoscience & Technology, School of Life Sciences & Centre for Excellence in Molecular Biology and Regenerative Medicine, JSS Academy of Higher Education & Research
| |
Collapse
|
44
|
Zhu BT. Biochemical mechanism underlying the pathogenesis of diabetic retinopathy and other diabetic complications in humans: the methanol-formaldehyde-formic acid hypothesis. Acta Biochim Biophys Sin (Shanghai) 2022; 54:415-451. [PMID: 35607958 PMCID: PMC9828688 DOI: 10.3724/abbs.2022012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 11/18/2021] [Indexed: 11/25/2022] Open
Abstract
Hyperglycemia in diabetic patients is associated with abnormally-elevated cellular glucose levels. It is hypothesized that increased cellular glucose will lead to increased formation of endogenous methanol and/or formaldehyde, both of which are then metabolically converted to formic acid. These one-carbon metabolites are known to be present naturally in humans, and their levels are increased under diabetic conditions. Mechanistically, while formaldehyde is a cross-linking agent capable of causing extensive cytotoxicity, formic acid is an inhibitor of mitochondrial cytochrome oxidase, capable of inducing histotoxic hypoxia, ATP deficiency and cytotoxicity. Chronic increase in the production and accumulation of these toxic one-carbon metabolites in diabetic patients can drive the pathogenesis of ocular as well as other diabetic complications. This hypothesis is supported by a large body of experimental and clinical observations scattered in the literature. For instance, methanol is known to have organ- and species-selective toxicities, including the characteristic ocular lesions commonly seen in humans and non-human primates, but not in rodents. Similarly, some of the diabetic complications (such as ocular lesions) also have a characteristic species-selective pattern, closely resembling methanol intoxication. Moreover, while alcohol consumption or combined use of folic acid plus vitamin B is beneficial for mitigating acute methanol toxicity in humans, their use also improves the outcomes of diabetic complications. In addition, there is also a large body of evidence from biochemical and cellular studies. Together, there is considerable experimental support for the proposed hypothesis that increased metabolic formation of toxic one-carbon metabolites in diabetic patients contributes importantly to the development of various clinical complications.
Collapse
Affiliation(s)
- Bao Ting Zhu
- Shenzhen Key Laboratory of Steroid Drug Discovery and DevelopmentSchool of MedicineThe Chinese University of Hong KongShenzhen518172China
- Department of PharmacologyToxicology and TherapeuticsSchool of MedicineUniversity of Kansas Medical CenterKansas CityKS66160USA
| |
Collapse
|
45
|
Turner MP, Zhao Y, Abdelkarim D, Liu P, Spence JS, Hutchison JL, Sivakolundu DK, Thomas BP, Hubbard NA, Xu C, Taneja K, Lu H, Rypma B. Altered linear coupling between stimulus-evoked blood flow and oxygen metabolism in the aging human brain. Cereb Cortex 2022; 33:135-151. [PMID: 35388407 PMCID: PMC9758587 DOI: 10.1093/cercor/bhac057] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 01/13/2022] [Accepted: 01/14/2022] [Indexed: 11/14/2022] Open
Abstract
Neural-vascular coupling (NVC) is the process by which oxygen and nutrients are delivered to metabolically active neurons by blood vessels. Murine models of NVC disruption have revealed its critical role in healthy neural function. We hypothesized that, in humans, aging exerts detrimental effects upon the integrity of the neural-glial-vascular system that underlies NVC. To test this hypothesis, calibrated functional magnetic resonance imaging (cfMRI) was used to characterize age-related changes in cerebral blood flow (CBF) and oxygen metabolism during visual cortex stimulation. Thirty-three younger and 27 older participants underwent cfMRI scanning during both an attention-controlled visual stimulation task and a hypercapnia paradigm used to calibrate the blood-oxygen-level-dependent signal. Measurement of stimulus-evoked blood flow and oxygen metabolism permitted calculation of the NVC ratio to assess the integrity of neural-vascular communication. Consistent with our hypothesis, we observed monotonic NVC ratio increases with increasing visual stimulation frequency in younger adults but not in older adults. Age-related changes in stimulus-evoked cerebrovascular and neurometabolic signal could not fully explain this disruption; increases in stimulus-evoked neurometabolic activity elicited corresponding increases in stimulus-evoked CBF in younger but not in older adults. These results implicate age-related, demand-dependent failures of the neural-glial-vascular structures that comprise the NVC system.
Collapse
Affiliation(s)
- Monroe P Turner
- School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX 75080, USA,Center for BrainHealth, University of Texas at Dallas, Dallas, TX, 75235, USA
| | - Yuguang Zhao
- School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX 75080, USA,Center for BrainHealth, University of Texas at Dallas, Dallas, TX, 75235, USA
| | - Dema Abdelkarim
- School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX 75080, USA,Center for BrainHealth, University of Texas at Dallas, Dallas, TX, 75235, USA
| | - Peiying Liu
- Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, MD 21287, USA
| | - Jeffrey S Spence
- School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX 75080, USA,Center for BrainHealth, University of Texas at Dallas, Dallas, TX, 75235, USA
| | - Joanna L Hutchison
- School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX 75080, USA,Center for BrainHealth, University of Texas at Dallas, Dallas, TX, 75235, USA
| | - Dinesh K Sivakolundu
- School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX 75080, USA,Department of Biological Sciences, University of Texas at Dallas, Richardson, TX, 75080, USA
| | - Binu P Thomas
- Advanced Imaging Research Center, University of Texas Southwestern Medical Center, Dallas, TX 75235, USA
| | - Nicholas A Hubbard
- Department of Psychology, Center for Brain, Biology, and Behavior, University of Nebraska, Lincoln, NE 68588, USA
| | - Cuimei Xu
- Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, MD 21287, USA
| | - Kamil Taneja
- Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, MD 21287, USA
| | - Hanzhang Lu
- Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, MD 21287, USA
| | - Bart Rypma
- Corresponding author: School of Behavioral and Brain Sciences, Center for Brain Health, University of Texas at Dallas, 800 West Campbell Road, Richardson, TX 75080, USA.
| |
Collapse
|
46
|
Karadayi R, Mazzocco J, Leclere L, Buteau B, Gregoire S, Belloir C, Koudsi M, Bessard P, Bizeau JB, Dubus E, Fenech C, Briand L, Bretillon L, Bron AM, Fioramonti X, Acar N. Plasmalogens Regulate Retinal Connexin 43 Expression and Müller Glial Cells Gap Junction Intercellular Communication and Migration. Front Cell Dev Biol 2022; 10:864599. [PMID: 35433704 PMCID: PMC9009447 DOI: 10.3389/fcell.2022.864599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 03/16/2022] [Indexed: 11/13/2022] Open
Abstract
Plasmalogens are a specific glycerophospholipid subtype characterized by a vinyl-ether bound at their sn-1 moiety. Their biosynthesis is initiated in the peroxisome by dihydroxyacetone phosphate-acyltransferase (DHAPAT), which is encoded by the DAPAT gene. Previous studies have shown that plasmalogen-deficient mice exhibit major physiological dysfunctions including several eye defects, among which abnormal vascular development of the retina and a reactive activation of macroglial Müller cells. Interestingly, plasmalogen deficiency in mice is also associated with a reduced expression of brain connexin 43 (Cx43). Cx43 is the main connexin subtype of retinal glial cells and is involved in several cellular mechanisms such as calcium-based gap junction intercellular communication (GJIC) or cell migration. Thus, the aim of our work was 1) to confirm the alteration of Cx43 expression in the retina of plasmalogen-deficient DAPAT−/- mice and 2) to investigate whether plasmalogens are involved in crucial functions of Müller cells such as GJIC and cell migration. First, we found that plasmalogen deficiency was associated with a significant reduction of Cx43 expression in the retina of DAPAT−/- mice in vivo. Secondly, using a siRNA targeting DHAPAT in vitro, we found that a 50%-reduction of Müller cells content in plasmalogens was sufficient to significantly downregulate Cx43 expression, while increasing its phosphorylation. Furthermore, plasmalogen-depleted Müller cells exhibited several alterations in ATP-induced GJIC, such as calcium waves of higher amplitude that propagated slower to neighboring cells, including astrocytes. Finally, in vitro plasmalogen depletion was also associated with a significant downregulation of Müller cells migration. Taken together, these data confirm that plasmalogens are critical for the regulation of Cx43 expression and for characteristics of retinal Müller glial cells such as GJIC and cell migration.
Collapse
Affiliation(s)
- Rémi Karadayi
- Eye and Nutrition Research Group, CSGA, Université de Bourgogne Franche-Comté, Dijon, France
| | - Julie Mazzocco
- Eye and Nutrition Research Group, CSGA, Université de Bourgogne Franche-Comté, Dijon, France
| | - Laurent Leclere
- Eye and Nutrition Research Group, CSGA, Université de Bourgogne Franche-Comté, Dijon, France
| | - Bénédicte Buteau
- Eye and Nutrition Research Group, CSGA, Université de Bourgogne Franche-Comté, Dijon, France
| | - Stéphane Gregoire
- Eye and Nutrition Research Group, CSGA, Université de Bourgogne Franche-Comté, Dijon, France
| | - Christine Belloir
- Taste and Olfaction Research Group, CSGA, Université de Bourgogne Franche-Comté, Dijon, France
| | - Mounzer Koudsi
- Eye and Nutrition Research Group, CSGA, Université de Bourgogne Franche-Comté, Dijon, France
| | - Pauline Bessard
- Eye and Nutrition Research Group, CSGA, Université de Bourgogne Franche-Comté, Dijon, France
| | - Jean-Baptiste Bizeau
- Eye and Nutrition Research Group, CSGA, Université de Bourgogne Franche-Comté, Dijon, France
| | - Elisabeth Dubus
- Eye and Nutrition Research Group, CSGA, Université de Bourgogne Franche-Comté, Dijon, France
| | - Claire Fenech
- Brain Nutrient Sensing and Energy Homeostasis, CSGA, Université de Bourgogne Franche-Comté, Dijon, France
| | - Loïc Briand
- Taste and Olfaction Research Group, CSGA, Université de Bourgogne Franche-Comté, Dijon, France
| | - Lionel Bretillon
- Eye and Nutrition Research Group, CSGA, Université de Bourgogne Franche-Comté, Dijon, France
| | - Alain M. Bron
- Eye and Nutrition Research Group, CSGA, Université de Bourgogne Franche-Comté, Dijon, France
- Department of Ophthalmology, University Hospital, Dijon, France
| | | | - Niyazi Acar
- Eye and Nutrition Research Group, CSGA, Université de Bourgogne Franche-Comté, Dijon, France
- *Correspondence: Niyazi Acar,
| |
Collapse
|
47
|
Carpi-Santos R, de Melo Reis RA, Gomes FCA, Calaza KC. Contribution of Müller Cells in the Diabetic Retinopathy Development: Focus on Oxidative Stress and Inflammation. Antioxidants (Basel) 2022; 11:617. [PMID: 35453302 PMCID: PMC9027671 DOI: 10.3390/antiox11040617] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 03/01/2022] [Accepted: 03/15/2022] [Indexed: 01/27/2023] Open
Abstract
Diabetic retinopathy is a neurovascular complication of diabetes and the main cause of vision loss in adults. Glial cells have a key role in maintenance of central nervous system homeostasis. In the retina, the predominant element is the Müller cell, a specialized cell with radial morphology that spans all retinal layers and influences the function of the entire retinal circuitry. Müller cells provide metabolic support, regulation of extracellular composition, synaptic activity control, structural organization of the blood-retina barrier, antioxidant activity, and trophic support, among other roles. Therefore, impairments of Müller actions lead to retinal malfunctions. Accordingly, increasing evidence indicates that Müller cells are affected in diabetic retinopathy and may contribute to the severity of the disease. Here, we will survey recently described alterations in Müller cell functions and cellular events that contribute to diabetic retinopathy, especially related to oxidative stress and inflammation. This review sheds light on Müller cells as potential therapeutic targets of this disease.
Collapse
Affiliation(s)
- Raul Carpi-Santos
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil; (R.C.-S.); (F.C.A.G.)
| | - Ricardo A. de Melo Reis
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil;
| | - Flávia Carvalho Alcantara Gomes
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil; (R.C.-S.); (F.C.A.G.)
| | - Karin C. Calaza
- Instituto de Biologia, Departamento de Neurobiologia, Universidade Federal Fluminense, Niteroi 24210-201, RJ, Brazil
| |
Collapse
|
48
|
Del Franco AP, Chiang PP, Newman EA. Dilation of cortical capillaries is not related to astrocyte calcium signaling. Glia 2022; 70:508-521. [PMID: 34767261 PMCID: PMC8732319 DOI: 10.1002/glia.24119] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 10/12/2021] [Accepted: 10/31/2021] [Indexed: 12/30/2022]
Abstract
The brain requires an adequate supply of oxygen and nutrients to maintain proper function as neuronal activity varies. This is achieved, in part, through neurovascular coupling mechanisms that mediate local increases in blood flow through the dilation of arterioles and capillaries. The role of astrocytes in mediating this functional hyperemia response is controversial. Specifically, the function of astrocyte Ca2+ signaling is unclear. Cortical arterioles dilate in the absence of astrocyte Ca2+ signaling, but previous work suggests that Ca2+ increases are necessary for capillary dilation. This question has not been fully addressed in vivo, however, and we have reexamined the role of astrocyte Ca2+ signaling in vessel dilation in the barrel cortex of awake, behaving mice. We recorded evoked vessel dilations and astrocyte Ca2+ signaling in response to whisker stimulation. Experiments were carried out on WT and IP3R2 KO mice, a transgenic model where astrocyte Ca2+ signaling is substantially reduced. Compared to WT mice at rest, Ca2+ signaling in astrocyte endfeet contacting capillaries increased by 240% when whisker stimulation evoked running. In contrast, Ca2+ signaling was reduced to 9% of WT values in IP3R2 KO mice. In all three conditions, however, the amplitude of capillary dilation was largely unchanged. In addition, the latency to the onset of astrocyte Ca2+ signaling lagged behind dilation onset in most trials, although a subset of rapid onset Ca2+ events with latencies as short as 0.15 s occurred. In summary, we found that whisker stimulation-evoked capillary dilations occurred independent of astrocyte Ca2+ increases in the cerebral cortex.
Collapse
Affiliation(s)
- Armani P Del Franco
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota, USA
| | - Pei-Pei Chiang
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota, USA
| | - Eric A Newman
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota, USA
| |
Collapse
|
49
|
Tworig JM, Feller MB. Müller Glia in Retinal Development: From Specification to Circuit Integration. Front Neural Circuits 2022; 15:815923. [PMID: 35185477 PMCID: PMC8856507 DOI: 10.3389/fncir.2021.815923] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 12/23/2021] [Indexed: 01/21/2023] Open
Abstract
Müller glia of the retina share many features with astroglia located throughout the brain including maintenance of homeostasis, modulation of neurotransmitter spillover, and robust response to injury. Here we present the molecular factors and signaling events that govern Müller glial specification, patterning, and differentiation. Next, we discuss the various roles of Müller glia in retinal development, which include maintaining retinal organization and integrity as well as promoting neuronal survival, synaptogenesis, and phagocytosis of debris. Finally, we review the mechanisms by which Müller glia integrate into retinal circuits and actively participate in neuronal signaling during development.
Collapse
Affiliation(s)
- Joshua M. Tworig
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, United States
- *Correspondence: Joshua M. Tworig,
| | - Marla B. Feller
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, United States
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA, United States
| |
Collapse
|
50
|
Markitantova YV, Simirskii VN. The Role of the Purinergic Signaling System in the Control of Histogenesis, Homeostasis, and Pathogenesis of the Vertebrate Retina. Russ J Dev Biol 2021. [DOI: 10.1134/s1062360421060084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|