51
|
Velloso FJ, Shankar S, Parpura V, Rakic P, Levison SW. Neural Stem Cells in Adult Mammals are not Astrocytes. ASN Neuro 2022; 14:17590914221134739. [PMID: 36330653 PMCID: PMC9638700 DOI: 10.1177/17590914221134739] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 09/27/2022] [Accepted: 10/01/2022] [Indexed: 11/06/2022] Open
Abstract
At the turn of the 21st century studies of the cells that resided in the adult mammalian subventricular zone (SVZ) characterized the neural stem cells (NSCs) as a subtype of astrocyte. Over the ensuing years, numerous studies have further characterized the properties of these NSCs and compared them to parenchymal astrocytes. Here we have evaluated the evidence collected to date to establish whether classifying the NSCs as astrocytes is appropriate and useful. We also performed a meta-analysis with 4 previously published datasets that used cell sorting and unbiased single-cell RNAseq to highlight the distinct gene expression profiles of adult murine NSCs and niche astrocytes. On the basis of our understanding of the properties and functions of astrocytes versus the properties and functions of NSCs, and from our comparative transcriptomic analyses we conclude that classifying the adult mammalian NSC as an astrocyte is potentially misleading. From our vantage point, it is more appropriate to refer to the cells in the adult mammalian SVZ that retain the capacity to produce new neurons and macroglia as NSCs without attaching the term "astrocyte-like."
Collapse
Affiliation(s)
- Fernando Janczur Velloso
- Department of Pharmacology, Physiology & Neuroscience, New
Jersey Medical School, Rutgers University, Newark, NJ, USA
| | - Sandhya Shankar
- Department of Pharmacology, Physiology & Neuroscience, New
Jersey Medical School, Rutgers University, Newark, NJ, USA
| | - Vladimir Parpura
- Department of Neurobiology, The University of Alabama at Birmingham,
Birmingham, AL, USA
| | - Pasko Rakic
- Department of Neuroscience, Yale School of Medicine, New Haven, CT,
USA
- Kavli Institute for Neuroscience, Yale School of Medicine, New
Haven, CT, USA
| | - Steven W. Levison
- Department of Pharmacology, Physiology & Neuroscience, New
Jersey Medical School, Rutgers University, Newark, NJ, USA
| |
Collapse
|
52
|
Papp A, Horváth A, Virág M, Tóth Z, Borbély C, Gombos F, Szűcs A, Kamondi A. Sleep alterations are related to cognitive symptoms in Parkinson's disease: A 24-hour ambulatory polygraphic EEG study. Int J Psychophysiol 2022; 173:93-103. [DOI: 10.1016/j.ijpsycho.2022.01.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 01/17/2022] [Accepted: 01/19/2022] [Indexed: 11/16/2022]
|
53
|
Bilateral hyperplasia of choroid plexus with severe CSF production: a case report and review of the glymphatic system. Childs Nerv Syst 2021; 37:3521-3529. [PMID: 34410450 DOI: 10.1007/s00381-021-05325-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 08/04/2021] [Indexed: 10/20/2022]
Abstract
BACKGROUND An important feature of hydrocephalus is the alteration of the cerebral spinal fluid (CSF) homeostasis. New insights in the understanding of production, secretion, and absorption of CSF, along with the discovery of the glymphatic system (GS), can be useful for a better understanding and treatment of hydrocephalus in disorders with CSF overproduction. CASE DESCRIPTION A 1-year-old patient was diagnosed with communicating hydrocephalus; ventricle peritoneal shunt (VPS) is installed and ascites developed. VPS is exposed, yielding volumes of 1000-1200ml/day CSF per day. MRI is performed showing generalized choroidal plexus hyperplasia. Bilateral endoscopic coagulation of thechoroid plexus was performed in 2 stages (CPC) however the high rate of CSF production persisted, needing a bilateral plexectomy through septostomy, which finally decreased the CSF outflow. DISCUSSION New knowledge about the CSF physiology will help to propose better treatment depending on the cause of the hydrocephalus. The GS is becoming an additional reason to better study and develop new therapies focused of the modulation of alternative CSF reabsorption. CONCLUSION Despite the current knowledge about hydrocephalus, we remain without a complete understanding of the pathophysiology of this condition. GS could be more important than conventional concept of reabsorption of CSF in the arachnoid villi, therefore GS could be a new key point, which will guide future investigations.
Collapse
|
54
|
Cao J, Yao D, Li R, Guo X, Hao J, Xie M, Li J, Pan D, Luo X, Yu Z, Wang M, Wang W. Digoxin Ameliorates Glymphatic Transport and Cognitive Impairment in a Mouse Model of Chronic Cerebral Hypoperfusion. Neurosci Bull 2021; 38:181-199. [PMID: 34704235 PMCID: PMC8821764 DOI: 10.1007/s12264-021-00772-y] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 07/04/2021] [Indexed: 02/03/2023] Open
Abstract
The glymphatic system plays a pivotal role in maintaining cerebral homeostasis. Chronic cerebral hypoperfusion, arising from small vessel disease or carotid stenosis, results in cerebrometabolic disturbances ultimately manifesting in white matter injury and cognitive dysfunction. However, whether the glymphatic system serves as a potential therapeutic target for white matter injury and cognitive decline during hypoperfusion remains unknown. Here, we established a mouse model of chronic cerebral hypoperfusion via bilateral common carotid artery stenosis. We found that the hypoperfusion model was associated with significant white matter injury and initial cognitive impairment in conjunction with impaired glymphatic system function. The glymphatic dysfunction was associated with altered cerebral perfusion and loss of aquaporin 4 polarization. Treatment of digoxin rescued changes in glymphatic transport, white matter structure, and cognitive function. Suppression of glymphatic functions by treatment with the AQP4 inhibitor TGN-020 abolished this protective effect of digoxin from hypoperfusion injury. Our research yields new insight into the relationship between hemodynamics, glymphatic transport, white matter injury, and cognitive changes after chronic cerebral hypoperfusion.
Collapse
Affiliation(s)
- Jie Cao
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 China
| | - Di Yao
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 China
| | - Rong Li
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 China ,Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 China
| | - Xuequn Guo
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 China ,Department of Respiratory Medicine, Quanzhou First Hospital Affiliated to Fujian Medical University, Quanzhou, 362000 China
| | - Jiahuan Hao
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 China
| | - Minjie Xie
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 China
| | - Jia Li
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 China
| | - Dengji Pan
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 China
| | - Xiang Luo
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 China
| | - Zhiyuan Yu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 China
| | - Minghuan Wang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 China
| | - Wei Wang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 China ,Key Laboratory of Neurological Diseases of the Chinese Ministry of Education, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 China
| |
Collapse
|
55
|
Nguyen H, Zerimech S, Baltan S. Astrocyte Mitochondria in White-Matter Injury. Neurochem Res 2021; 46:2696-2714. [PMID: 33527218 PMCID: PMC8935665 DOI: 10.1007/s11064-021-03239-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 01/05/2021] [Accepted: 01/06/2021] [Indexed: 12/11/2022]
Abstract
This review summarizes the diverse structure and function of astrocytes to describe the bioenergetic versatility required of astrocytes that are situated at different locations. The intercellular domain of astrocyte mitochondria defines their roles in supporting and regulating astrocyte-neuron coupling and survival against ischemia. The heterogeneity of astrocyte mitochondria, and how subpopulations of astrocyte mitochondria adapt to interact with other glia and regulate axon function, require further investigation. It has become clear that mitochondrial permeability transition pores play a key role in a wide variety of human diseases, whose common pathology may be based on mitochondrial dysfunction triggered by Ca2+ and potentiated by oxidative stress. Reactive oxygen species cause axonal degeneration and a reduction in axonal transport, leading to axonal dystrophies and neurodegeneration including Alzheimer's disease, amyotrophic lateral sclerosis, Parkinson's disease, and Huntington's disease. Developing new tools to allow better investigation of mitochondrial structure and function in astrocytes, and techniques to specifically target astrocyte mitochondria, can help to unravel the role of mitochondrial health and dysfunction in a more inclusive context outside of neuronal cells. Overall, this review will assess the value of astrocyte mitochondria as a therapeutic target to mitigate acute and chronic injury in the CNS.
Collapse
Affiliation(s)
- Hung Nguyen
- Anesthesiology and Peri-Operative Medicine (APOM), Oregon Health and Science University, Portland, OR, 97239, USA
| | - Sarah Zerimech
- Anesthesiology and Peri-Operative Medicine (APOM), Oregon Health and Science University, Portland, OR, 97239, USA
| | - Selva Baltan
- Anesthesiology and Peri-Operative Medicine (APOM), Oregon Health and Science University, Portland, OR, 97239, USA.
| |
Collapse
|
56
|
Cerebrovascular alterations in NAFLD: Is it increasing our risk of Alzheimer's disease? Anal Biochem 2021; 636:114387. [PMID: 34537182 DOI: 10.1016/j.ab.2021.114387] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 07/27/2021] [Accepted: 09/15/2021] [Indexed: 02/07/2023]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a multisystem disease, which has been classified as an emerging epidemic not only confined to liver-related morbidity and mortality. It is also becoming apparent that NAFLD is associated with moderate cerebral dysfunction and cognitive decline. A possible link between NAFLD and Alzheimer's disease (AD) has only recently been proposed due to the multiple shared genes and pathological mechanisms contributing to the development of these conditions. Although AD is a progressive neurodegenerative disease, the exact pathophysiological mechanism remains ambiguous and similarly to NAFLD, currently available pharmacological therapies have mostly failed in clinical trials. In addition to the usual suspects (inflammation, oxidative stress, blood-brain barrier alterations and ageing) that could contribute to the NAFLD-induced development and progression of AD, changes in the vasculature, cerebral perfusion and waste clearance could be the missing link between these two diseases. Here, we review the most recent literature linking NAFLD and AD, focusing on cerebrovascular alterations and the brain's clearance system as risk factors involved in the development and progression of AD, with the aim of promoting further research using neuroimaging techniques and new mechanism-based therapeutic interventions.
Collapse
|
57
|
Abstract
UNLABELLED Exercise is associated with higher cognitive function and is a promising intervention to reduce the risk of dementia. With advancing age, there are changes in the vasculature that have important clinical implications for brain health and cognition. Primary aging and vascular risk factors are associated with increases in arterial stiffness and pulse pressure, and reductions in peripheral vascular function. OBJECTIVE The purpose is to discuss the epidemiological, observational, and mechanistic evidence regarding the link between age-related changes in vascular health and brain health. METHODS We performed a literature review and integrated with our published data. RESULTS Epidemiological evidence suggests a link between age-related increases in arterial stiffness and lower cognitive function, which may be mediated by cerebral vascular function, including cerebral vasoreactivity and cerebral pulsatility. Age-associated impairments in central arterial stiffness and peripheral vascular function have been attenuated or reversed through lifestyle behaviors such as exercise. Greater volumes of habitual exercise and higher cardiorespiratory fitness are associated with beneficial effects on both peripheral vascular health and cognition. Yet, the extent to which exercise directly influences cerebral vascular function and brain health, as well as the associated mechanisms remains unclear. CONCLUSION Although there is evidence that exercise positively impacts cerebral vascular function, more research is necessary in humans to optimize experimental protocols and address methodological limitations and physiological considerations. Understanding the impact of exercise on cerebral vascular function is important for understanding the association between exercise and brain health and may inform future intervention studies that seek to improve cognition.
Collapse
|
58
|
Meyerhoff J, Chakraborty N, Hammamieh R. Glymphatics: A Transformative Development in Medical Neuroscience Relevant to Injuries in Military Central Nervous System. Mil Med 2021; 187:e1086-e1090. [PMID: 34453167 DOI: 10.1093/milmed/usab344] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 07/07/2021] [Accepted: 08/06/2021] [Indexed: 11/13/2022] Open
Abstract
INTRODUCTION The glia-operated glymphatic system, analogous to but separate from the lymphatics in the periphery, is unique to brain and retina, where it is very closely aligned with the arteriolar system. This intimate relationship leads to a "blood vessel like" distribution pattern of glymphatic vessels in the brain. The spatial relationship of glymphatics, including their essential component aquaporin-4 with vascular pericytes of brain arterioles is critical to functionality and is termed "polarization". MATERIALS AND METHODS We review the available literature on the factors affecting the resting state of glymphatics under normal conditions, including the important role of sleep in supporting normal glymphatic function (including waste removal) as well as the critical role of "polarization" under normal conditions. We then examine the effects of traumatic brain injury (TBI) or seizures on the glymphatic system and its state of "polarization". RESULTS Injury, such as TBI, can disrupt polarization resulting in "depolarization" leading to brain edema. CONCLUSION Damage to the glymphatic system might explain the brain edema so often seen following TBI or other insult. Moreover, similar damage should be expected in response to seizures, which can often be associated with chemical exposures as well as with TBI. Military operations, whether night operations or continuous operations, quite often impose limitations on sleep. As glymphatic function is sleep-dependent, sleep deprivation alone could compromise glymphatic function, as well, and might in addition, explain some of the well-known performance deficits associated with sleep deprivation. Possible effects of submarine and diving operations, chemical agents (including seizures), as well as high altitude exposure and other threats should be considered. In addition to the brain, the retina is also served and protected by the glymphatic system. Accordingly, the effect of military-related risks (e.g., exposure to laser or other threats) to retinal glymphatic function should also be considered. An intact glymphatic system is absolutely essential to support normal central nervous system functionality, including cognition. This effects a broad range of military threats on brain and retinal glymphatics should be explored. Possible preventive and therapeutic measures should be proposed and evaluated, as well.
Collapse
Affiliation(s)
- James Meyerhoff
- Geneva Foundation, Medical Readiness Systems Biology, CMPN, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA.,Medical Readiness Systems Biology, CMPN, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
| | - Nabarun Chakraborty
- Medical Readiness Systems Biology, CMPN, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
| | - Rasha Hammamieh
- Medical Readiness Systems Biology, CMPN, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
| |
Collapse
|
59
|
Tan ZX, Dong F, Wu LY, Feng YS, Zhang F. The Beneficial Role of Exercise on Treating Alzheimer's Disease by Inhibiting β-Amyloid Peptide. Mol Neurobiol 2021; 58:5890-5906. [PMID: 34415486 DOI: 10.1007/s12035-021-02514-7] [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: 02/10/2021] [Accepted: 08/03/2021] [Indexed: 02/07/2023]
Abstract
Alzheimer's disease (AD) is associated with a very large burden on global healthcare systems. Thus, it is imperative to find effective treatments of the disease. One feature of AD is the accumulation of neurotoxic β-amyloid peptide (Aβ). Aβ induces multiple pathological processes that are deleterious to nerve cells. Despite the development of medications that target the reduction of Aβ to treat AD, none has proven to be effective to date. Non-pharmacological interventions, such as physical exercise, are also being studied. The benefits of exercise on AD are widely recognized. Experimental and clinical studies have been performed to verify the role that exercise plays in reducing Aβ deposition to alleviate AD. This paper reviewed the various mechanisms involved in the exercise-induced reduction of Aβ, including the regulation of amyloid precursor protein cleaved proteases, the glymphatic system, brain-blood transport proteins, degrading enzymes and autophagy, which is beneficial to promote exercise therapy as a means of prevention and treatment of AD and indicates that exercise may provide new therapeutic targets for the treatment of AD.
Collapse
Affiliation(s)
- Zi-Xuan Tan
- Department of Rehabilitation Medicine, The Third Hospital of Hebei Medical University, No. 139 Ziqiang Road, Shijiazhuang, Hebei, 050051, People's Republic of China
| | - Fang Dong
- Department of Clinical Laboratory Medicine, The Third Hospital of Hebei Medical University, Shijiazhuang, 050051, People's Republic of China
| | - Lin-Yu Wu
- Department of Rehabilitation Medicine, The Third Hospital of Hebei Medical University, No. 139 Ziqiang Road, Shijiazhuang, Hebei, 050051, People's Republic of China
| | - Ya-Shuo Feng
- Department of Rehabilitation Medicine, The Third Hospital of Hebei Medical University, No. 139 Ziqiang Road, Shijiazhuang, Hebei, 050051, People's Republic of China
| | - Feng Zhang
- Department of Rehabilitation Medicine, The Third Hospital of Hebei Medical University, No. 139 Ziqiang Road, Shijiazhuang, Hebei, 050051, People's Republic of China. .,Hebei Key Laboratory of Critical Disease Mechanism and Intervention, Shijiazhuang, 050051, People's Republic of China.
| |
Collapse
|
60
|
Mostovenko E, Saunders S, Muldoon PP, Bishop L, Campen MJ, Erdely A, Ottens AK. Carbon Nanotube Exposure Triggers a Cerebral Peptidomic Response: Barrier Compromise, Neuroinflammation, and a Hyperexcited State. Toxicol Sci 2021; 182:107-119. [PMID: 33892499 DOI: 10.1093/toxsci/kfab042] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
The unique physicochemical properties of carbon nanomaterials and their ever-growing utilization generate a serious concern for occupational risk. Pulmonary exposure to these nanoparticles induces local and systemic inflammation, cardiovascular dysfunction, and even cognitive deficits. Although multiple routes of extrapulmonary toxicity have been proposed, the mechanism for and manner of neurologic effects remain minimally understood. Here, we examine the cerebral spinal fluid (CSF)-derived peptidomic fraction as a reflection of neuropathological alterations induced by pulmonary carbon nanomaterial exposure. Male C57BL/6 mice were exposed to 10 or 40 µg of multiwalled carbon nanotubes (MWCNT) by oropharyngeal aspiration. Serum and CSFs were collected 4 h post exposure. An enriched peptide fraction of both biofluids was analyzed using ion mobility-enabled data-independent mass spectrometry for label-free quantification. MWCNT exposure induced a prominent peptidomic response in the blood and CSF; however, correlation between fluids was limited. Instead, we determined that a MWCNT-induced peptidomic shift occurred specific to the CSF with 292 significant responses found that were not in serum. Identified MWCNT-responsive peptides depicted a mechanism involving aberrant fibrinolysis (fibrinopeptide A), blood-brain barrier permeation (homeobox protein A4), neuroinflammation (transmembrane protein 131L) with reactivity by astrocytes and microglia, and a pro-degradative (signal transducing adapter molecule, phosphoglycerate kinase), antiplastic (AF4/FMR2 family member 1, vacuolar protein sorting-associated protein 18) state with the excitation-inhibition balance shifted to a hyperexcited (microtubule-associated protein 1B) phenotype. Overall, the significant pathologic changes observed were consistent with early neurodegenerative disease and were diagnostically reflected in the CSF peptidome.
Collapse
Affiliation(s)
- Ekaterina Mostovenko
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, Virginia 23298, USA
| | - Samantha Saunders
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, Virginia 23298, USA
| | - Pretal P Muldoon
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, Virginia 23298, USA
| | - Lindsey Bishop
- Pathology and Physiology Research Branch, National Institute for Occupational Safety and Health, Morgantown, West Virginia 26505, USA
| | - Matthew J Campen
- Department of Pharmaceutical Sciences, University of New Mexico, Albuquerque, New Mexico 87131, USA
| | - Aaron Erdely
- Pathology and Physiology Research Branch, National Institute for Occupational Safety and Health, Morgantown, West Virginia 26505, USA
| | - Andrew K Ottens
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, Virginia 23298, USA
| |
Collapse
|
61
|
Cumulative Damage: Cell Death in Posthemorrhagic Hydrocephalus of Prematurity. Cells 2021; 10:cells10081911. [PMID: 34440681 PMCID: PMC8393895 DOI: 10.3390/cells10081911] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 07/23/2021] [Accepted: 07/25/2021] [Indexed: 12/19/2022] Open
Abstract
Globally, approximately 11% of all infants are born preterm, prior to 37 weeks’ gestation. In these high-risk neonates, encephalopathy of prematurity (EoP) is a major cause of both morbidity and mortality, especially for neonates who are born very preterm (<32 weeks gestation). EoP encompasses numerous types of preterm birth-related brain abnormalities and injuries, and can culminate in a diverse array of neurodevelopmental impairments. Of note, posthemorrhagic hydrocephalus of prematurity (PHHP) can be conceptualized as a severe manifestation of EoP. PHHP impacts the immature neonatal brain at a crucial timepoint during neurodevelopment, and can result in permanent, detrimental consequences to not only cerebrospinal fluid (CSF) dynamics, but also to white and gray matter development. In this review, the relevant literature related to the diverse mechanisms of cell death in the setting of PHHP will be thoroughly discussed. Loss of the epithelial cells of the choroid plexus, ependymal cells and their motile cilia, and cellular structures within the glymphatic system are of particular interest. Greater insights into the injuries, initiating targets, and downstream signaling pathways involved in excess cell death shed light on promising areas for therapeutic intervention. This will bolster current efforts to prevent, mitigate, and reverse the consequential brain remodeling that occurs as a result of hydrocephalus and other components of EoP.
Collapse
|
62
|
Zhou W, Yang J, Wang Q, Wang Y, Yan Y, Wu S, Chen S, Wei W. Systemic Stressors and Retinal Microvascular Alterations in People Without Diabetes: The Kailuan Eye Study. Invest Ophthalmol Vis Sci 2021; 62:20. [PMID: 33595612 PMCID: PMC7900855 DOI: 10.1167/iovs.62.2.20] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Purpose The purpose of this study was to determine systemic stressors, including fasting plasma glucose (FPG), and other major atherosclerotic cardiovascular disease (ASCVD) risk factors of the retinal microvasculature in people without diabetes. Methods The Kailuan Eye Study enrolled applicants from the community-based longitudinal Kailuan Study. Applicants underwent optical coherence tomographic angiography (OCTA) and systemic examinations. Both the macula and optic disc were screened, whereas superficial capillary plexus (SCP), deep capillary plexus (DCP), foveal vessel density in the 300 µm ring (FD-300), and radial peripapillary capillaries (RPCs) density were measured in the study. Results This study included 353 eligible applicants (mean age = 49.86 ± 11.41 years; 47% men; FPG =5.32 ± 1.19 mmol/L). Lower DCP density was associated with elder age (P = 0.001), male gender (P < 0.001), and higher FPG (P = 0.008). Male gender (P < 0.001), axial length (P < 0.001), and FPG (P = 0.029) were inversely associated with RPC density. Meanwhile, a higher FPG concentration was significantly correlated with lower DCP density (P = 0.006) and higher intraocular pressure (P = 0.006), after adjusting mean arterial blood pressure (P = 0.001) and sex (P = 0.042). Conclusions DCP density showed a significantly negative correlation with FPG concentration in people without diabetes. These data suggest hyperglycemia could cause early retinal capillary alterations in patients without clinical signs of retinopathy and indicate the potential clinical applications of routine OCTA may be beneficial to screen for subclinical microvasculature and monitor patients with high risks of ASCVD.
Collapse
Affiliation(s)
- Wenjia Zhou
- Beijing Tongren Eye Center, Beijing Tongren Hospital, Beijing Ophthalmology and Visual Science Key Lab, Beijing Key Laboratory of Intraocular Tumor Diagnosis and Treatment, Capital Medical University, Beijing, China
| | - Jingyan Yang
- Beijing Tongren Eye Center, Beijing Tongren Hospital, Beijing Ophthalmology and Visual Science Key Lab, Beijing Key Laboratory of Intraocular Tumor Diagnosis and Treatment, Capital Medical University, Beijing, China
| | - Qian Wang
- Beijing Tongren Eye Center, Beijing Tongren Hospital, Beijing Ophthalmology and Visual Science Key Lab, Beijing Key Laboratory of Intraocular Tumor Diagnosis and Treatment, Capital Medical University, Beijing, China
| | - Yaxing Wang
- Beijing Institute of Ophthalmology, Beijing Key Laboratory of Ophthalmology and Visual Sciences, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University; Beijing, China
| | - Yanni Yan
- Beijing Tongren Eye Center, Beijing Tongren Hospital, Beijing Ophthalmology and Visual Science Key Lab, Beijing Key Laboratory of Intraocular Tumor Diagnosis and Treatment, Capital Medical University, Beijing, China
| | - Shouling Wu
- Department of Cardiology, Kailuan General Hospital, Tangshan, China
| | - Shuohua Chen
- Health Care Center, Kailuan Group, Tangshan, China
| | - Wenbin Wei
- Beijing Tongren Eye Center, Beijing Tongren Hospital, Beijing Ophthalmology and Visual Science Key Lab, Beijing Key Laboratory of Intraocular Tumor Diagnosis and Treatment, Capital Medical University, Beijing, China
| |
Collapse
|
63
|
Hayden MR, Banks WA. Deficient Leptin Cellular Signaling Plays a Key Role in Brain Ultrastructural Remodeling in Obesity and Type 2 Diabetes Mellitus. Int J Mol Sci 2021; 22:5427. [PMID: 34063911 PMCID: PMC8196569 DOI: 10.3390/ijms22115427] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 05/13/2021] [Accepted: 05/15/2021] [Indexed: 12/11/2022] Open
Abstract
The triad of obesity, metabolic syndrome (MetS), Type 2 diabetes mellitus (T2DM) and advancing age are currently global societal problems that are expected to grow over the coming decades. This triad is associated with multiple end-organ complications of diabetic vasculopathy (maco-microvessel disease), neuropathy, retinopathy, nephropathy, cardiomyopathy, cognopathy encephalopathy and/or late-onset Alzheimer's disease. Further, obesity, MetS, T2DM and their complications are associated with economical and individual family burdens. This review with original data focuses on the white adipose tissue-derived adipokine/hormone leptin and how its deficient signaling is associated with brain remodeling in hyperphagic, obese, or hyperglycemic female mice. Specifically, the ultrastructural remodeling of the capillary neurovascular unit, brain endothelial cells (BECs) and their endothelial glycocalyx (ecGCx), the blood-brain barrier (BBB), the ventricular ependymal cells, choroid plexus, blood-cerebrospinal fluid barrier (BCSFB), and tanycytes are examined in female mice with impaired leptin signaling from either dysfunction of the leptin receptor (DIO and db/db models) or the novel leptin deficiency (BTBR ob/ob model).
Collapse
Affiliation(s)
- Melvin R. Hayden
- Departments of Internal Medicine, Endocrinology Diabetes and Metabolism, Diabetes and Cardiovascular Disease Center, University of Missouri-Columbia School of Medicine, One Hospital Drive, Columbia, MO 65212, USA;
| | - William A. Banks
- Geriatrics Research, Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, 1660 S. Columbian Way, 810C/Bldg 1, Seattle, WA 98108, USA
- Department of Medicine, Division of Gerontology and Geriatric Medicine, University of Washington, Seattle, WA 98108, USA
| |
Collapse
|
64
|
Nanjundaiah S, Chidambaram H, Chandrashekar M, Chinnathambi S. Role of Microglia in Regulating Cholesterol and Tau Pathology in Alzheimer's Disease. Cell Mol Neurobiol 2021; 41:651-668. [PMID: 32468440 DOI: 10.1007/s10571-020-00883-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Accepted: 05/19/2020] [Indexed: 01/21/2023]
Abstract
Cholesterol, a principal constituent of the cell membrane, plays a crucial role in the brain by regulating the synaptic transmission, neuronal signaling, as well as neurodegenerative diseases. Defects in the cholesterol trafficking are associated with enhanced generation of hyperphosphorylated Tau and Amyloid-β protein. Tau, a major microtubule-associated protein in the brain, is the key regulator of the mature neuron. Abnormally hyperphosphorylated Tau hampers the major functions related to microtubule assembly by promoting neurofibrillary tangles of paired helical filaments, twisted ribbons, and straight filaments. The observed pathological changes due to impaired cholesterol and Tau protein accumulation cause Alzheimer's disease. Thus, in order to regulate the pathogenesis of Alzheimer's disease, regulation of cholesterol metabolism, as well as Tau phosphorylation, is essential. The current review provides an overview of (1) cholesterol synthesis in the brain, neurons, astrocytes, and microglia; (2) the mechanism involved in modulating cholesterol concentration between the astrocytes and brain; (3) major mechanisms involved in the hyperphosphorylation of Tau and amyloid-β protein; and (4) microglial involvement in its regulation. Thus, the answering key questions will provide an in-depth information on microglia involvement in managing the pathogenesis of cholesterol-modulated hyperphosphorylated Tau protein.
Collapse
Affiliation(s)
- Shwetha Nanjundaiah
- Neurobiology Group, Division of Biochemical Sciences, CSIR-National Chemical Laboratory (CSIR-NCL), Dr. Homi Bhabha Road, Pune, 411008, India
| | - Hariharakrishnan Chidambaram
- Neurobiology Group, Division of Biochemical Sciences, CSIR-National Chemical Laboratory (CSIR-NCL), Dr. Homi Bhabha Road, Pune, 411008, India
- Academy of Scientific and Innovative Research (AcSIR), New Delhi, 110025, India
| | - Madhura Chandrashekar
- School of Biomedical Engineering and Sciences, MIT University, Loni Kalbhor, Pune, 412201, India
| | - Subashchandrabose Chinnathambi
- Neurobiology Group, Division of Biochemical Sciences, CSIR-National Chemical Laboratory (CSIR-NCL), Dr. Homi Bhabha Road, Pune, 411008, India.
- Academy of Scientific and Innovative Research (AcSIR), New Delhi, 110025, India.
| |
Collapse
|
65
|
Ong HS, Lim CS, Png ALC, Kong JW, Peh ALH. Chronobiology and the case for sleep health interventions in the community. Singapore Med J 2021; 62:220-224. [PMID: 34409473 PMCID: PMC8801860 DOI: 10.11622/smedj.2021058] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Our sleep-wake cycle is determined by the interaction between our homeostatic sleep drive and circadian rhythm. Each of us has a personalised biological rhythm or chronotype that determines the optimal time to fall asleep and wake up. Chronic sleep deprivation has been linked to the development of several physical and mental health disorders, as well as accidents and occupational errors. Around the world, growing recognition of the importance of sleep has led to the adoption of practices that promote sleep health. Given that Singaporeans were consistently found to be one of the most sleep-deprived populations in the world, we believe that there is an urgent need to pursue the introduction of community-based sleep health interventions here. This includes sleep education and promotion of sleep hygiene, adopting practices to reduce social jetlag and improve sleep health, and enhancing screening and treatment of sleep disorders.
Collapse
Affiliation(s)
- Hatta Santoso Ong
- Department of Psychological Medicine, Changi General Hospital, Singapore
- Department of Sleep Medicine, Surgery and Science, Changi General Hospital, Singapore
| | - Chau Sian Lim
- Department of Psychological Medicine, Changi General Hospital, Singapore
- Department of Sleep Medicine, Surgery and Science, Changi General Hospital, Singapore
| | - Ai-Li Constance Png
- Department of Sleep Medicine, Surgery and Science, Changi General Hospital, Singapore
- Department of Clinical Psychology, Changi General Hospital, Singapore
| | - Jing Wen Kong
- Hougang Polyclinic, National Healthcare Group Polyclinics, Singapore
| | | |
Collapse
|
66
|
Haacke EM, Ge Y, Sethi SK, Buch S, Zamboni P. An Overview of Venous Abnormalities Related to the Development of Lesions in Multiple Sclerosis. Front Neurol 2021; 12:561458. [PMID: 33981281 PMCID: PMC8107266 DOI: 10.3389/fneur.2021.561458] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 03/26/2021] [Indexed: 12/21/2022] Open
Abstract
The etiology of multiple sclerosis (MS) is currently understood to be autoimmune. However, there is a long history and growing evidence for disrupted vasculature and flow within the disease pathology. A broad review of the literature related to vascular effects in MS revealed a suggestive role for abnormal flow in the medullary vein system. Evidence for venous involvement in multiple sclerosis dates back to the early pathological work by Charcot and Bourneville, in the mid-nineteenth century. Pioneering work by Adams in the 1980s demonstrated vasculitis within the walls of veins and venules proximal to active MS lesions. And more recently, magnetic resonance imaging (MRI) has been used to show manifestations of the central vein as a precursor to the development of new MS lesions, and high-resolution MRI using Ferumoxytol has been used to reveal the microvasculature that has previously only been demonstrated in cadaver brains. Both approaches may shed new light into the structural changes occurring in MS lesions. The material covered in this review shows that multiple pathophysiological events may occur sequentially, in parallel, or in a vicious circle which include: endothelial damage, venous collagenosis and fibrin deposition, loss of vessel compliance, venous hypertension, perfusion reduction followed by ischemia, medullary vein dilation and local vascular remodeling. We come to the conclusion that a potential source of MS lesions is due to locally disrupted flow which in turn leads to remodeling of the medullary veins followed by endothelial damage with the subsequent escape of glial cells, cytokines, etc. These ultimately lead to the cascade of inflammatory and demyelinating events which ensue in the course of the disease.
Collapse
Affiliation(s)
- E. Mark Haacke
- Department of Radiology, Wayne State University, Detroit, MI, United States
| | - Yulin Ge
- Department of Radiology, Center for Biomedical Imaging, NYU Grossman School of Medicine, New York, NY, United States
| | - Sean K. Sethi
- Department of Radiology, Wayne State University, Detroit, MI, United States
| | - Sagar Buch
- Department of Radiology, Wayne State University, Detroit, MI, United States
| | - Paolo Zamboni
- Vascular Diseases Center, University of Ferrara, Ferrara, Italy
| |
Collapse
|
67
|
Chen J, Wang L, Xu H, Wang Y, Liang Q. The lymphatic drainage system of the CNS plays a role in lymphatic drainage, immunity, and neuroinflammation in stroke. J Leukoc Biol 2021; 110:283-291. [PMID: 33884651 DOI: 10.1002/jlb.5mr0321-632r] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Revised: 03/19/2021] [Accepted: 03/26/2021] [Indexed: 12/31/2022] Open
Abstract
The lymphatic drainage system of the central nervous system (CNS) plays an important role in maintaining interstitial fluid balance and regulating immune responses and immune surveillance. The impaired lymphatic drainage system of the CNS might be involved in the onset and progression of various neurodegenerative diseases, neuroinflammation, and cerebrovascular diseases. A significant immune response and brain edema are observed after stroke, resulting from disrupted homeostasis in the brain. Thus, understanding the lymphatic drainage system of the CNS in stroke may lead to the development of new approaches for therapeutic interventions in the future. Here, we review recent evidence implicating the lymphatic drainage system of the CNS in stroke.
Collapse
Affiliation(s)
- Jinman Chen
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Institute of Spine, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Key Laboratory of theory and therapy of muscles and bones, Ministry of Education (Shanghai University of Traditional Chinese Medicine), Shanghai, China
| | - Linmei Wang
- Department of Anatomy, School of Basic Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Hao Xu
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Institute of Spine, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Key Laboratory of theory and therapy of muscles and bones, Ministry of Education (Shanghai University of Traditional Chinese Medicine), Shanghai, China
| | - Yongjun Wang
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Institute of Spine, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Key Laboratory of theory and therapy of muscles and bones, Ministry of Education (Shanghai University of Traditional Chinese Medicine), Shanghai, China
| | - Qianqian Liang
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Institute of Spine, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Key Laboratory of theory and therapy of muscles and bones, Ministry of Education (Shanghai University of Traditional Chinese Medicine), Shanghai, China
| |
Collapse
|
68
|
Natário KHP, Aguiar GBD, Vieira MADCES. The glymphatic system and its relation with neurological diseases. Rev Assoc Med Bras (1992) 2021; 67:620-624. [DOI: 10.1590/1806-9282.20200925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Accepted: 01/03/2021] [Indexed: 11/21/2022] Open
|
69
|
Transient changes in white matter microstructure during general anesthesia. PLoS One 2021; 16:e0247678. [PMID: 33770816 PMCID: PMC7997710 DOI: 10.1371/journal.pone.0247678] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Accepted: 02/10/2021] [Indexed: 01/01/2023] Open
Abstract
Cognitive dysfunction after surgery under general anesthesia is a well-recognized clinical phenomenon in the elderly. Physiological effects of various anesthetic agents have been studied at length. Very little is known about potential effects of anesthesia on brain structure. In this study we used Diffusion Tensor Imaging to compare the white matter microstructure of healthy control subjects under sevoflurane anesthesia with their awake state. Fractional Anisotropy, a white mater integrity index, transiently decreases throughout the brain during sevoflurane anesthesia and then returns back to baseline. Other DTI metrics such as mean diffusivity, axial diffusivity and radial diffusivity were increased under sevoflurane anesthesia. Although DTI metrics are age dependent, the transient changes due to sevoflurane were independent of age and sex. Volumetric analysis shows various white matter volumes decreased whereas some gray matter volumes increased during sevoflurane anesthesia. These results suggest that sevoflurane anesthesia has a significant, but transient, effect on white matter microstructure. In spite of the transient effects of sevoflurane anesthesia there were no measurable effects on brain white matter as determined by the DTI metrics at 2 days and 7 days following anesthesia. The role of white matter in the loss of consciousness under anesthesia will need to be studied and MRI studies with subjects under anesthesia will need to take these results into account.
Collapse
|
70
|
Ji C, Yu X, Xu W, Lenahan C, Tu S, Shao A. The role of glymphatic system in the cerebral edema formation after ischemic stroke. Exp Neurol 2021; 340:113685. [PMID: 33676917 DOI: 10.1016/j.expneurol.2021.113685] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 02/25/2021] [Accepted: 03/01/2021] [Indexed: 12/20/2022]
Abstract
Cerebral edema following ischemic stroke is predictive of the severity of the eventual stroke related damage, however the effective treatment is limited. The glymphatic system is a recently identified waste clearance pathway in the brain, found in the paravascular space and mainly composed of astrocytes and their aquaporin-4 (AQP4) water channels. In this review, we primarily focus on the role of the glymphatic system in the formation of cerebral edema after ischemic stroke. There is still no definite conclusion whether the influx of cerebrospinal fluid (CSF) in the glymphatic system is increased or not after ischemic stroke. However, the reduced interstitial fluid (ISF) clearance after ischemic stroke is definite. Additionally, AQP4 as the most important part of glymphatic system plays a complex bimodal in cerebral edema after ischemic stroke. Most of the research has found that AQP4 deletion in animals reduces cerebral edema after acute ischemic stroke compared with wild type animal models. The mislocalization of astrocytic AQP4 was also presented after ischemic stroke. As the cerebral edema after ischemic stroke is difficult to treat, we discuss several potential treatment targets related to glymphatic system. More studies are needed to explore the role of glymphatic system in the formation of cerebral edema after ischemic stroke and develop probable treatment strategies.
Collapse
Affiliation(s)
- Caihong Ji
- Department of Neurology, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Xing Yu
- Department of Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Weilin Xu
- Department of Neurosurgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Cameron Lenahan
- Center for Neuroscience Research, Loma Linda University School of Medicine, Loma Linda, CA, USA; Burrell College of Osteopathic Medicine, Las Cruces, NM, USA
| | - Sheng Tu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China.
| | - Anwen Shao
- Department of Neurosurgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
71
|
Shahsavani N, Kataria H, Karimi-Abdolrezaee S. Mechanisms and repair strategies for white matter degeneration in CNS injury and diseases. Biochim Biophys Acta Mol Basis Dis 2021; 1867:166117. [PMID: 33667627 DOI: 10.1016/j.bbadis.2021.166117] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 02/22/2021] [Accepted: 02/23/2021] [Indexed: 12/14/2022]
Abstract
White matter degeneration is an important pathophysiological event of the central nervous system that is collectively characterized by demyelination, oligodendrocyte loss, axonal degeneration and parenchymal changes that can result in sensory, motor, autonomic and cognitive impairments. White matter degeneration can occur due to a variety of causes including trauma, neurotoxic exposure, insufficient blood flow, neuroinflammation, and developmental and inherited neuropathies. Regardless of the etiology, the degeneration processes share similar pathologic features. In recent years, a plethora of cellular and molecular mechanisms have been identified for axon and oligodendrocyte degeneration including oxidative damage, calcium overload, neuroinflammatory events, activation of proteases, depletion of adenosine triphosphate and energy supply. Extensive efforts have been also made to develop neuroprotective and neuroregenerative approaches for white matter repair. However, less progress has been achieved in this area mainly due to the complexity and multifactorial nature of the degeneration processes. Here, we will provide a timely review on the current understanding of the cellular and molecular mechanisms of white matter degeneration and will also discuss recent pharmacological and cellular therapeutic approaches for white matter protection as well as axonal regeneration, oligodendrogenesis and remyelination.
Collapse
Affiliation(s)
- Narjes Shahsavani
- Department of Physiology and Pathophysiology, Regenerative Medicine Program, Spinal Cord Research Centre, Children's Hospital Research Institute of Manitoba, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Hardeep Kataria
- Department of Physiology and Pathophysiology, Regenerative Medicine Program, Spinal Cord Research Centre, Children's Hospital Research Institute of Manitoba, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Soheila Karimi-Abdolrezaee
- Department of Physiology and Pathophysiology, Regenerative Medicine Program, Spinal Cord Research Centre, Children's Hospital Research Institute of Manitoba, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada.
| |
Collapse
|
72
|
Yang Z, Zhang X, Li C, Chi S, Xie A. Molecular Mechanisms Underlying Reciprocal Interactions Between Sleep Disorders and Parkinson's Disease. Front Neurosci 2021; 14:592989. [PMID: 33642969 PMCID: PMC7902929 DOI: 10.3389/fnins.2020.592989] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Accepted: 11/27/2020] [Indexed: 01/11/2023] Open
Abstract
Sleep-wake disruptions are among the most prevalent and burdensome non-motor symptoms of Parkinson's disease (PD). Clinical studies have demonstrated that these disturbances can precede the onset of typical motor symptoms by years, indicating that they may play a primary function in the pathogenesis of PD. Animal studies suggest that sleep facilitates the removal of metabolic wastes through the glymphatic system via convective flow from the periarterial space to the perivenous space, upregulates antioxidative defenses, and promotes the maintenance of neuronal protein homeostasis. Therefore, disruptions to the sleep-wake cycle have been associated with inefficient metabolic clearance and increased oxidative stress in the central nervous system (CNS). This leads to excessive accumulation of alpha-synuclein and the induction of neuronal loss, both of which have been proposed to be contributing factors to the pathogenesis and progression of PD. Additionally, recent studies have suggested that PD-related pathophysiological alterations during the prodromal phase disrupt sleep and circadian rhythms. Taken together, these findings indicate potential mechanistic interactions between sleep-wake disorders and PD progression as proposed in this review. Further research into the hypothetical mechanisms underlying these interactions would be valuable, as positive findings may provide promising insights into novel therapeutic interventions for PD.
Collapse
Affiliation(s)
- Zhengjie Yang
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xiaona Zhang
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Chengqian Li
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Song Chi
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Anmu Xie
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
73
|
Manouchehrian O, Ramos M, Bachiller S, Lundgaard I, Deierborg T. Acute systemic LPS-exposure impairs perivascular CSF distribution in mice. J Neuroinflammation 2021; 18:34. [PMID: 33514389 PMCID: PMC7844902 DOI: 10.1186/s12974-021-02082-6] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 01/13/2021] [Indexed: 12/22/2022] Open
Abstract
Background The exchange of cerebrospinal (CSF) and interstitial fluid is believed to be vital for waste clearance in the brain. The sleep-dependent glymphatic system, which is comprised of perivascular flow of CSF and is largely dependent on arterial pulsatility and astrocytic aquaporin-4 (AQP4) expression, facilitates much of this brain clearance. During the last decade, several observations have indicated that impaired glymphatic function goes hand in hand with neurodegenerative diseases. Since pathologies of the brain carry inflammatory components, we wanted to know how acute inflammation, e.g., with lipopolysaccharide (LPS) injections, would affect the glymphatic system. In this study, we aim to measure the effect of LPS on perivascular CSF distribution as a measure of glymphatic function. Methods Three hours after injection of LPS (1 mg/kg i.p.), C57bl/6 mice were (1) imaged for two CSF tracers, injected into cisterna magna, (2) transcardially perfused with buffer, or (3) used for physiological readouts. Tracer flow was imaged using a low magnification microscope on fixed brains, as well as using vibratome-cut slices for measuring tracer penetration in the brain. Cytokines, glial, and BBB-permeability markers were measured with ELISAs, Western blots, and immunohistochemistry. Cerebral blood flow was approximated using laser Doppler flowmetry, respiration and heart rate with a surgical monitor, and AQP4-polarization was quantified using confocal microscopy of immunolabeled brain sections. Results LPS-injections significantly lowered perivascular CSF tracer flow and penetration into the parenchyma. No differences in AQP4 polarization, cytokines, astroglial and BBB markers, cerebral blood flow, or respiration were detected in LPS-injected mice, although LPS did elevate cortical Iba1+ area and heart rate. Conclusions This study reports another physiological response after acute exposure to the bacterial endotoxin LPS, namely the statistically significant decrease in perivascular distribution of CSF. These observations may benefit our understanding of the role of systemic inflammation in brain clearance.
Collapse
Affiliation(s)
- Oscar Manouchehrian
- Experimental Neuroinflammation Laboratory, Department of Experimental Medical Science, Lund University, SE-221 84, Lund, Sweden.
| | - Marta Ramos
- Department of Experimental Medical Science, Lund University, SE-221 84, Lund, Sweden.,Wallenberg Centre for Molecular Medicine, Lund University, SE-223 62, Lund, Sweden
| | - Sara Bachiller
- Experimental Neuroinflammation Laboratory, Department of Experimental Medical Science, Lund University, SE-221 84, Lund, Sweden
| | - Iben Lundgaard
- Department of Experimental Medical Science, Lund University, SE-221 84, Lund, Sweden.,Wallenberg Centre for Molecular Medicine, Lund University, SE-223 62, Lund, Sweden
| | - Tomas Deierborg
- Experimental Neuroinflammation Laboratory, Department of Experimental Medical Science, Lund University, SE-221 84, Lund, Sweden
| |
Collapse
|
74
|
Sun Y, Sun X. Exploring the interstitial system in the brain: the last mile of drug delivery. Rev Neurosci 2021; 32:363-377. [PMID: 33550781 DOI: 10.1515/revneuro-2020-0057] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Accepted: 12/08/2020] [Indexed: 11/15/2022]
Abstract
Brain interstitial system (ISS) is a nanoscale network of continuously connected tubes and sheets surrounding each neural cell in the central nervous system. ISS usually accounts for ∼20% of the brain volume, far more than the cerebral blood vessels, which account for 3%. The neuronal function, signaling pathways, and drug delivery are all closely related to the microenvironment provided by ISS. The objective of this paper is to give the readers a clear outline of detection, anatomy, function, and applications of ISS. This review describes the techniques propelling the exploration for ISS in chronological order, physiological function and pathological dysfunction of ISS, and strategies for drug delivery based on ISS. Biophysical features are the focus of ISS research, in which the diffusion characteristics have dominated. The various techniques that explore ISS take advantage of this feature. ISS provides an essential microenvironment for the health of cells and brain homeostasis, which plays an important functional role in brain health and disease. Direct intracranial administration allows the diffusion of drugs directly through ISS to successfully bypass the blood-brain barrier that prevents most drugs from reaching the brain. With the deepening of understanding of the brain ISS, the new research model that takes into account brain cells, cerebral vessels, and ISS will provide a new perspective and direction for understanding, utilizing, and protecting the brain.
Collapse
Affiliation(s)
- Yi Sun
- National Key Research Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing 100191, P. R. China.,Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences, Peking University, Beijing 100191, P. R. China
| | - Xinping Sun
- Clinical Laboratory, Peking University International Hospital, Beijing 102206, P. R. China
| |
Collapse
|
75
|
Akiyama Y, Yokoyama R, Takashima H, Kawata Y, Arihara M, Chiba R, Kimura Y, Mikami T, Mikuni N. Accumulation of Macromolecules in Idiopathic Normal Pressure Hydrocephalus. Neurol Med Chir (Tokyo) 2021; 61:211-218. [PMID: 33504733 PMCID: PMC7966205 DOI: 10.2176/nmc.oa.2020-0274] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The clearance system in the brain is not completely understood. The aim of this study was to prove the presence of the “glymphatic system” in the human brain using magnetic resonance spectroscopy (MRS). Spectral data of the brain white matter were obtained from healthy volunteers and patients with hydrocephalic dementia and used to measure intracerebral metabolites, including macromolecules (MMs) and lipids. Data were transferred from the MRS scanners to a workstation, and metabolites were quantified with the spectrogram-based eddy current method and water scaling. MM levels were significantly higher in patients with a slow gait and executive dysfunction due to normal pressure hydrocephalus (NPH) than in asymptomatic volunteers (p <0.01). In contrast, the N-acetyl aspartate (NAA) level was significantly lower in patients with executive dysfunction than in asymptomatic volunteers (p <0.01). There were no statistically significant differences in metabolites, including alanine, aspartate, creatine, γ-amino butyric acid, D-glucose, glutamine, glutamate, glycerophosphorylcholine, phosphorylcholine, lactate, myoinositol, N-acetyl-aspartyl-glutamate, scyllo-inositol, taurine, creatine methylene, and guanine, in the centrum semiovale between patients with NPH and asymptomatic volunteers. We quantitatively evaluated cerebral metabolites, particularly in the centrum semiovale, with MRS. In the brain of patients with a slow gait and executive dysfunction due to NPH, MRS revealed significantly higher MM levels and lower NAA levels compared to healthy volunteers. Therefore, it may be concluded that the patients have a dysfunctional glymphatic system in the brain.
Collapse
Affiliation(s)
- Yukinori Akiyama
- Department of Neurosurgery, Sapporo Medical University, Sapporo, Hokkaido, Japan
| | - Rintaro Yokoyama
- Department of Neurosurgery, Sapporo Medical University, Sapporo, Hokkaido, Japan
| | - Hiroyuki Takashima
- Division of Radiology and Nuclear Medicine, Sapporo Medical University, Sapporo, Hokkaido, Japan
| | - Yuka Kawata
- Department of Neurology, Sapporo Medical University, Sapporo, Hokkaido, Japan
| | - Masayasu Arihara
- Department of Neurosurgery, Sapporo Medical University, Sapporo, Hokkaido, Japan
| | - Ryohei Chiba
- Department of Neurosurgery, Sapporo Medical University, Sapporo, Hokkaido, Japan
| | - Yusuke Kimura
- Department of Neurosurgery, Sapporo Medical University, Sapporo, Hokkaido, Japan
| | - Takeshi Mikami
- Department of Neurosurgery, Sapporo Medical University, Sapporo, Hokkaido, Japan
| | - Nobuhiro Mikuni
- Department of Neurosurgery, Sapporo Medical University, Sapporo, Hokkaido, Japan
| |
Collapse
|
76
|
Mezey É, Szalayova I, Hogden CT, Brady A, Dósa Á, Sótonyi P, Palkovits M. An immunohistochemical study of lymphatic elements in the human brain. Proc Natl Acad Sci U S A 2021; 118:e2002574118. [PMID: 33446503 PMCID: PMC7826383 DOI: 10.1073/pnas.2002574118] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Almost 150 papers about brain lymphatics have been published in the last 150 years. Recently, the information in these papers has been synthesized into a picture of central nervous system (CNS) "glymphatics," but the fine structure of lymphatic elements in the human brain based on imaging specific markers of lymphatic endothelium has not been described. We used LYVE1 and PDPN antibodies to visualize lymphatic marker-positive cells (LMPCs) in postmortem human brain samples, meninges, cavernous sinus (cavum trigeminale), and cranial nerves and bolstered our findings with a VEGFR3 antibody. LMPCs were present in the perivascular space, the walls of small and large arteries and veins, the media of large vessels along smooth muscle cell membranes, and the vascular adventitia. Lymphatic marker staining was detected in the pia mater, in the arachnoid, in venous sinuses, and among the layers of the dura mater. There were many LMPCs in the perineurium and endoneurium of cranial nerves. Soluble waste may move from the brain parenchyma via perivascular and paravascular routes to the closest subarachnoid space and then travel along the dura mater and/or cranial nerves. Particulate waste products travel along the laminae of the dura mater toward the jugular fossa, lamina cribrosa, and perineurium of the cranial nerves to enter the cervical lymphatics. CD3-positive T cells appear to be in close proximity to LMPCs in perivascular/perineural spaces throughout the brain. Both immunostaining and qPCR confirmed the presence of adhesion molecules in the CNS known to be involved in T cell migration.
Collapse
Affiliation(s)
- Éva Mezey
- Adult Stem Cell Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892;
| | - Ildikó Szalayova
- Adult Stem Cell Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892
| | - Christopher T Hogden
- Adult Stem Cell Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892
| | - Alexandra Brady
- Adult Stem Cell Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892
| | - Ágnes Dósa
- Department of Forensic Sciences, Semmelweis University, H-1091 Budapest, Hungary
| | - Péter Sótonyi
- Department of Forensic Sciences, Semmelweis University, H-1091 Budapest, Hungary
| | - Miklós Palkovits
- Human Brain Tissue Bank, Semmelweis University, H-1094 Budapest, Hungary
| |
Collapse
|
77
|
Tajbakhsh A, Read M, Barreto GE, Ávila-Rodriguez M, Gheibi-Hayat SM, Sahebkar A. Apoptotic neurons and amyloid-beta clearance by phagocytosis in Alzheimer's disease: Pathological mechanisms and therapeutic outlooks. Eur J Pharmacol 2021; 895:173873. [PMID: 33460611 DOI: 10.1016/j.ejphar.2021.173873] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 01/06/2021] [Accepted: 01/11/2021] [Indexed: 12/13/2022]
Abstract
Neuronal survival and axonal renewal following central nervous system damage and in neurodegenerative illnesses, such as Alzheimer's disease (AD), can be enhanced by fast clearance of neuronal apoptotic debris, as well as the removal of amyloid beta (Aβ) by phagocytic cells through the process of efferocytosis. This process quickly inhibits the release of proinflammatory and antigenic autoimmune constituents, enhancing the formation of a microenvironment vital for neuronal survival and axonal regeneration. Therefore, the detrimental features associated with microglial phagocytosis uncoupling, such as the accumulation of apoptotic cells, inflammation and phagoptosis, could exacerbate the pathology in brain disease. Some mechanisms of efferocytosis could be targeted by several promising agents, such as curcumin, URMC-099 and Y-P30, which have emerged as potential treatments for AD. This review aims to investigate and update the current research regarding the signaling molecules and pathways involved in efferocytosis and how these could be targeted as a potential therapy in AD.
Collapse
Affiliation(s)
- Amir Tajbakhsh
- Department of Modern Sciences & Technologies, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Morgayn Read
- Department of Pharmacology, School of Medical Sciences, University of Otago, Dunedin, New Zealand
| | - George E Barreto
- Department of Biological Sciences, University of Limerick, Limerick, Ireland; Health Research Institute, University of Limerick, Limerick, Ireland
| | | | - Seyed Mohammad Gheibi-Hayat
- Department of Medical Biotechnology, Faculty of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Polish Mother's Memorial Hospital Research Institute (PMMHRI), Lodz, Poland.
| |
Collapse
|
78
|
Li B, Xia M, Zorec R, Parpura V, Verkhratsky A. Astrocytes in heavy metal neurotoxicity and neurodegeneration. Brain Res 2021; 1752:147234. [PMID: 33412145 DOI: 10.1016/j.brainres.2020.147234] [Citation(s) in RCA: 64] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Revised: 11/15/2020] [Accepted: 12/07/2020] [Indexed: 12/14/2022]
Abstract
With the industrial development and progressive increase in environmental pollution, the mankind overexposure to heavy metals emerges as a pressing public health issue. Excessive intake of heavy metals, such as arsenic (As), manganese (Mn), mercury (Hg), aluminium (Al), lead (Pb), nickel (Ni), bismuth (Bi), cadmium (Cd), copper (Cu), zinc (Zn), and iron (Fe), is neurotoxic and it promotes neurodegeneration. Astrocytes are primary homeostatic cells in the central nervous system. They protect neurons against all types of insults, in particular by accumulating heavy metals. However, this makes astrocytes the main target for heavy metals neurotoxicity. Intake of heavy metals affects astroglial homeostatic and neuroprotective cascades including glutamate/GABA-glutamine shuttle, antioxidative machinery and energy metabolism. Deficits in these astroglial pathways facilitate or even instigate neurodegeneration. In this review, we provide a concise outlook on heavy metal-induced astrogliopathies and their association with major neurodegenerative disorders. In particular, we focus on astroglial mechanisms of iron-induced neurotoxicity. Iron deposits in the brain are detected in main neurodegenerative diseases, such as Alzheimer's disease, Parkinson's disease and amyotrophic lateral sclerosis. Accumulation of iron in the brain is associated with motor and cognitive impairments and iron-induced histopathological manifestations may be considered as the potential diagnostic biomarker of neurodegenerative diseases. Effective management of heavy metal neurotoxicity can be regarded as a potential strategy to prevent or retard neurodegenerative pathologies.
Collapse
Affiliation(s)
- Baoman Li
- Practical Teaching Centre, School of Forensic Medicine, China Medical University, Shenyang, People's Republic of China.
| | - Maosheng Xia
- Department of Orthopaedics, The First Hospital, China Medical University, Shenyang, People's Republic of China
| | - Robert Zorec
- Celica BIOMEDICAL, Tehnološki Park 24, 1000 Ljubljana, Slovenia; Laboratory of Neuroendocrinology-Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Zaloška 4, 1000 Ljubljana, Slovenia
| | - Vladimir Parpura
- Department of Neurobiology, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Alexei Verkhratsky
- Practical Teaching Centre, School of Forensic Medicine, China Medical University, Shenyang, People's Republic of China; Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK; Achucarro Center for Neuroscience, IKERBASQUE, Basque Foundation for Science, 48011 Bilbao, Spain.
| |
Collapse
|
79
|
DeKosky ST, Kochanek PM, Valadka AB, Clark RS, Chou SHY, Au AK, Horvat C, Jha RM, Mannix R, Wisniewski SR, Wintermark M, Rowell SE, Welch RD, Lewis L, House S, Tanzi RE, Smith DR, Vittor AY, Denslow ND, Davis MD, Glushakova OY, Hayes RL. Blood Biomarkers for Detection of Brain Injury in COVID-19 Patients. J Neurotrauma 2021; 38:1-43. [PMID: 33115334 PMCID: PMC7757533 DOI: 10.1089/neu.2020.7332] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) virus attacks multiple organs of coronavirus disease 2019 (COVID-19) patients, including the brain. There are worldwide descriptions of neurological deficits in COVID-19 patients. Central nervous system (CNS) symptoms can be present early in the course of the disease. As many as 55% of hospitalized COVID-19 patients have been reported to have neurological disturbances three months after infection by SARS-CoV-2. The mutability of the SARS-COV-2 virus and its potential to directly affect the CNS highlight the urgency of developing technology to diagnose, manage, and treat brain injury in COVID-19 patients. The pathobiology of CNS infection by SARS-CoV-2 and the associated neurological sequelae of this infection remain poorly understood. In this review, we outline the rationale for the use of blood biomarkers (BBs) for diagnosis of brain injury in COVID-19 patients, the research needed to incorporate their use into clinical practice, and the improvements in patient management and outcomes that can result. BBs of brain injury could potentially provide tools for detection of brain injury in COVID-19 patients. Elevations of BBs have been reported in cerebrospinal fluid (CSF) and blood of COVID-19 patients. BB proteins have been analyzed in CSF to detect CNS involvement in patients with infectious diseases, including human immunodeficiency virus and tuberculous meningitis. BBs are approved by the U.S. Food and Drug Administration for diagnosis of mild versus moderate traumatic brain injury and have identified brain injury after stroke, cardiac arrest, hypoxia, and epilepsy. BBs, integrated with other diagnostic tools, could enhance understanding of viral mechanisms of brain injury, predict severity of neurological deficits, guide triage of patients and assignment to appropriate medical pathways, and assess efficacy of therapeutic interventions in COVID-19 patients.
Collapse
Affiliation(s)
- Steven T. DeKosky
- McKnight Brain Institute, University of Florida College of Medicine, Gainesville, Florida, USA
| | - Patrick M. Kochanek
- Department of Critical Care Medicine, Department of Anesthesiology, Pediatrics, Bioengineering, and Clinical and Translational Science, Safar Center for Resuscitation Research, University of Pittsburgh School of Medicine, UPMC Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Alex B. Valadka
- Department of Neurosurgery, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Robert S.B. Clark
- Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Sherry H.-Y. Chou
- Department of Critical Care Medicine, Neurology, and Neurosurgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Alicia K. Au
- University of Pittsburgh, UPMC Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Christopher Horvat
- Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Division of Pediatric Critical Care, UPMC Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Ruchira M. Jha
- Departments of Critical Care Medicine, Neurology, Neurological Surgery, Clinical and Translational Science Institute, Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Rebekah Mannix
- Department of Pediatrics and Emergency Medicine, Harvard Medical School, Department of Medicine, Division of Emergency Medicine, Boston Children's Hospital, Boston, Massachusetts, USA
| | | | - Max Wintermark
- Department of Neuroradiology, Stanford University, Stanford, California, USA
| | - Susan E. Rowell
- Duke University School of Medicine, Durham, North Carolina, USA
| | - Robert D. Welch
- Department of Emergency Medicine, Wayne State University School of Medicine, Detroit Receiving Hospital/University Health Center, Detroit, Michigan, USA
| | - Lawrence Lewis
- Department of Emergency Medicine, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Stacey House
- Department of Emergency Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Rudolph E. Tanzi
- Genetics and Aging Research Unit, Massachusetts General Hospital, McCance Center for Brain Health, Massachusetts General Hospital, MassGeneral Institute for Neurodegenerative Diseases, Massachusetts General Hospital, Department of Neurology (Research), Massachusetts General Hospital, Department of Neurology, Harvard Medical School, Charlestown, Massachusetts, USA
| | - Darci R. Smith
- Immunodiagnostics Department, Naval Medical Research Center, Biological Defense Research Directorate, Fort Detrick, Maryland, USA
| | - Amy Y. Vittor
- Division of Infectious Disease and Global Medicine, University of Florida, Emerging Pathogens Institute, Gainesville, Florida, USA
| | - Nancy D. Denslow
- Departments of Physiological Sciences and Biochemistry and Molecular Biology, University of Florida, Center for Environmental and Human Toxicology, Gainesville, Florida
| | - Michael D. Davis
- Department of Pediatrics, Wells Center for Pediatric Research/Pulmonology, Allergy, and Sleep Medicine, Riley Hospital for Children at Indiana University, Indianapolis, Indiana, USA
| | | | | |
Collapse
|
80
|
Volumetric distribution of perivascular space in relation to mild cognitive impairment. Neurobiol Aging 2020; 99:28-43. [PMID: 33422892 DOI: 10.1016/j.neurobiolaging.2020.12.010] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 11/25/2020] [Accepted: 12/08/2020] [Indexed: 12/19/2022]
Abstract
Vascular contributions to early cognitive decline are increasingly recognized, prompting further investigation into the nature of related changes in perivascular spaces (PVS). Using magnetic resonance imaging, we show that, compared to a cognitively normal sample, individuals with early cognitive dysfunction have altered PVS presence and distribution, irrespective of Amyloid-β. Surprisingly, we noted lower PVS presence in the anterosuperior medial temporal lobe (asMTL) (1.29 times lower PVS volume fraction in cognitively impaired individuals, p < 0.0001), which was associated with entorhinal neurofibrillary tau tangle deposition (beta (standard error) = -0.98 (0.4); p = 0.014), one of the hallmarks of early Alzheimer's disease pathology. We also observed higher PVS volume fraction in centrum semi-ovale of the white matter, but only in female participants (1.47 times higher PVS volume fraction in cognitively impaired individuals, p = 0.0011). We also observed PVS changes in participants with history of hypertension (higher in the white matter and lower in the asMTL). Our results suggest that anatomically specific alteration of the PVS is an early neuroimaging feature of cognitive impairment in aging adults, which is differentially manifested in female.
Collapse
|
81
|
Novel PET Biomarkers to Disentangle Molecular Pathways across Age-Related Neurodegenerative Diseases. Cells 2020; 9:cells9122581. [PMID: 33276490 PMCID: PMC7761606 DOI: 10.3390/cells9122581] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 11/25/2020] [Accepted: 11/28/2020] [Indexed: 12/11/2022] Open
Abstract
There is a need to disentangle the etiological puzzle of age-related neurodegenerative diseases, whose clinical phenotypes arise from known, and as yet unknown, pathways that can act distinctly or in concert. Enhanced sub-phenotyping and the identification of in vivo biomarker-driven signature profiles could improve the stratification of patients into clinical trials and, potentially, help to drive the treatment landscape towards the precision medicine paradigm. The rapidly growing field of neuroimaging offers valuable tools to investigate disease pathophysiology and molecular pathways in humans, with the potential to capture the whole disease course starting from preclinical stages. Positron emission tomography (PET) combines the advantages of a versatile imaging technique with the ability to quantify, to nanomolar sensitivity, molecular targets in vivo. This review will discuss current research and available imaging biomarkers evaluating dysregulation of the main molecular pathways across age-related neurodegenerative diseases. The molecular pathways focused on in this review involve mitochondrial dysfunction and energy dysregulation; neuroinflammation; protein misfolding; aggregation and the concepts of pathobiology, synaptic dysfunction, neurotransmitter dysregulation and dysfunction of the glymphatic system. The use of PET imaging to dissect these molecular pathways and the potential to aid sub-phenotyping will be discussed, with a focus on novel PET biomarkers.
Collapse
|
82
|
Lu H, Zhan Y, Ai L, Chen H, Chen J. AQP4-siRNA alleviates traumatic brain edema by altering post-traumatic AQP4 polarity reversal in TBI rats. J Clin Neurosci 2020; 81:113-119. [PMID: 33222898 DOI: 10.1016/j.jocn.2020.09.015] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 07/13/2020] [Accepted: 09/06/2020] [Indexed: 11/18/2022]
Abstract
The spatial and temporal distribution of aquaporin-4 (AQP4) expression in rat brain following brain trauma and AQP4-siRNA treatment, as well as corresponding pathological changes, were studied to explore the mechanism underlying the effect of AQP4-siRNA treatment on traumatic brain injury (TBI). The rats in the sham operation group had normal structure, with AQP4 located in the perivascular end-foot membranes and astrocytic membranes in a polarized pattern. The accelerated polarity reversal was observed in the TBI group in 1-12 h after TBI. During this period, AQP4 abundance on the astrocytic membrane is gradually increased, while AQP4 abundance on the perivascular end-foot membrane declined rapidly. Twelve hours after TBI, AQP4 expression was depolarized, showing a shift from the perivascular end-foot membrane to the astrocytic membrane. Pathological observation showed that vasogenic edema occurred immediately after TBI, at which time the extracellular space was expanded, leading to severe intracellular edema. AQP4-siRNA reduced the polarity reversal index at the early stage of TBI recovery and reduced edema, demonstrating the potential benefit of reduced AQP4 expression during recovery from TBI.
Collapse
Affiliation(s)
- Hong Lu
- Department of Radiology, Chongqing The Seventh Peoplés Hospital, Chongqing, China
| | - Yuefu Zhan
- Department of Radiology, Hainan Women and Children's Medical Center, No. 15, Long Kun Nan road, Haikou, Hainan 572500, China.
| | - Li Ai
- Department of Radiology, Chongqing The Seventh Peoplés Hospital, Chongqing, China
| | - Haixia Chen
- Department of Radiology, Chongqing The Seventh Peoplés Hospital, Chongqing, China
| | - Jianqiang Chen
- Department of Radiology, Xiangya School of Medicine Affiliated Haikou Hospital, Central South University, Haikou, Hainan 570208, China.
| |
Collapse
|
83
|
Gordleeva S, Kanakov O, Ivanchenko M, Zaikin A, Franceschi C. Brain aging and garbage cleaning : Modelling the role of sleep, glymphatic system, and microglia senescence in the propagation of inflammaging. Semin Immunopathol 2020; 42:647-665. [PMID: 33034735 DOI: 10.1007/s00281-020-00816-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Accepted: 07/30/2020] [Indexed: 01/01/2023]
Abstract
Brain aging is a complex process involving many functions of our body and described by the interplay of a sleep pattern and changes in the metabolic waste concentration regulated by the microglial function and the glymphatic system. We review the existing modelling approaches to this topic and derive a novel mathematical model to describe the crosstalk between these components within the conceptual framework of inflammaging. Analysis of the model gives insight into the dynamics of garbage concentration and linked microglial senescence process resulting from a normal or disrupted sleep pattern, hence, explaining an underlying mechanism behind healthy or unhealthy brain aging. The model incorporates accumulation and elimination of garbage, induction of glial activation by garbage, and glial senescence by over-activation, as well as the production of pro-inflammatory molecules by their senescence-associated secretory phenotype (SASP). Assuming that insufficient sleep leads to the increase of garbage concentration and promotes senescence, the model predicts that if the accumulation of senescent glia overcomes an inflammaging threshold, further progression of senescence becomes unstoppable even if a normal sleep pattern is restored. Inverting this process by "rejuvenating the brain" is only possible via a reset of concentration of senescent glia below this threshold. Our model approach enables analysis of space-time dynamics of senescence, and in this way, we show that heterogeneous patterns of inflammation will accelerate the propagation of senescence profile through a network, confirming a negative effect of heterogeneity.
Collapse
Affiliation(s)
- Susanna Gordleeva
- Laboratory of Systems Medicine of Healthy Aging, Lobachevsky Univeristy, Nizhny Novgorod, Russia.
- Neuroscience and Cognitive Technology Laboratory, Center for Technologies in Robotics and Mechatronics Components, Innopolis University, Innopolis, Russia.
| | - Oleg Kanakov
- Laboratory of Systems Medicine of Healthy Aging, Lobachevsky Univeristy, Nizhny Novgorod, Russia
| | - Mikhail Ivanchenko
- Laboratory of Systems Medicine of Healthy Aging, Lobachevsky Univeristy, Nizhny Novgorod, Russia
| | - Alexey Zaikin
- Laboratory of Systems Medicine of Healthy Aging, Lobachevsky Univeristy, Nizhny Novgorod, Russia
- Institute for Women's Health and Department of Mathematics, University College London, London, UK
- Centre for Analysis of Complex Systems, Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - Claudio Franceschi
- Laboratory of Systems Medicine of Healthy Aging, Lobachevsky Univeristy, Nizhny Novgorod, Russia
- Department of Experimental Pathology, University of Bologna, Bologna, Italy
| |
Collapse
|
84
|
Fang Y, Shi H, Ren R, Huang L, Okada T, Lenahan C, Gamdzyk M, Travis ZD, Lu Q, Tang L, Huang Y, Zhou K, Tang J, Zhang J, Zhang JH. Pituitary Adenylate Cyclase-Activating Polypeptide Attenuates Brain Edema by Protecting Blood-Brain Barrier and Glymphatic System After Subarachnoid Hemorrhage in Rats. Neurotherapeutics 2020; 17:1954-1972. [PMID: 32918234 PMCID: PMC7851266 DOI: 10.1007/s13311-020-00925-3] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/28/2020] [Indexed: 12/14/2022] Open
Abstract
Brain edema is a vital contributor to early brain injury after subarachnoid hemorrhage (SAH), which is responsible for prolonged hospitalization and poor outcomes. Pharmacological therapeutic targets on edema formation have been the focus of research for decades. Pituitary adenylate cyclase-activating polypeptide (PACAP) has been shown to participate in neural development and brain injury. Here, we used PACAP knockout CRISPR to demonstrate that endogenous PACAP plays an endogenous neuroprotective role against brain edema formation after SAH in rats. The exogenous PACAP treatment provided both short- and long-term neurological benefits by preserving the function of the blood-brain barrier and glymphatic system after SAH. Pretreatment of inhibitors of PACAP receptors showed that the PACAP-involved anti-edema effect and neuroprotection after SAH was facilitated by the selective PACAP receptor (PAC1). Further administration of adenylyl cyclase (AC) inhibitor and sulfonylurea receptor 1 (SUR1) CRISPR activator suggested that the AC-cyclic adenosine monophosphate (cAMP)-protein kinase A (PKA) axis participated in PACAP signaling after SAH, which inhibited the expression of edema-related proteins, SUR1 and aquaporin-4 (AQP4), through SUR1 phosphorylation. Thus, PACAP may serve as a potential clinical treatment to alleviate brain edema in patients with SAH.
Collapse
Affiliation(s)
- Yuanjian Fang
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, 88 Jiefang Road, Zhejiang, 310009, Hangzhou, China
| | - Hui Shi
- Department of Neurosurgery, Yongchuan Hospital, Chongqing Medical University, Chongqing, China
| | - Reng Ren
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, 88 Jiefang Road, Zhejiang, 310009, Hangzhou, China
| | - Lei Huang
- Department of Neurosurgery, Loma Linda University, Loma Linda, California, USA
- Department of Physiology and Pharmacology, Loma Linda University, 11041 Campus St, Risley Hall, Room 219, Loma Linda, California, 92354, USA
| | - Takeshi Okada
- Department of Neurosurgery, Loma Linda University, Loma Linda, California, USA
- Department of Physiology and Pharmacology, Loma Linda University, 11041 Campus St, Risley Hall, Room 219, Loma Linda, California, 92354, USA
| | - Cameron Lenahan
- Department of Neurosurgery, Loma Linda University, Loma Linda, California, USA
- Burrell College of Osteopathic Medicine, Las Cruces, New Mexico, USA
| | - Marcin Gamdzyk
- Department of Neurosurgery, Loma Linda University, Loma Linda, California, USA
| | - Zachary D Travis
- Department of Neurosurgery, Loma Linda University, Loma Linda, California, USA
| | - Qin Lu
- Department of Neurosurgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Lihui Tang
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, 88 Jiefang Road, Zhejiang, 310009, Hangzhou, China
| | - Yi Huang
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, 88 Jiefang Road, Zhejiang, 310009, Hangzhou, China
| | - Keren Zhou
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, 88 Jiefang Road, Zhejiang, 310009, Hangzhou, China
| | - Jiping Tang
- Department of Neurosurgery, Loma Linda University, Loma Linda, California, USA
- Department of Physiology and Pharmacology, Loma Linda University, 11041 Campus St, Risley Hall, Room 219, Loma Linda, California, 92354, USA
- Department of Anesthesiology, Loma Linda University, Loma Linda, California, USA
| | - Jianmin Zhang
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, 88 Jiefang Road, Zhejiang, 310009, Hangzhou, China.
| | - John H Zhang
- Department of Neurosurgery, Loma Linda University, Loma Linda, California, USA.
- Department of Physiology and Pharmacology, Loma Linda University, 11041 Campus St, Risley Hall, Room 219, Loma Linda, California, 92354, USA.
- Department of Anesthesiology, Loma Linda University, Loma Linda, California, USA.
| |
Collapse
|
85
|
Benveniste H, Elkin R, Heerdt PM, Koundal S, Xue Y, Lee H, Wardlaw J, Tannenbaum A. The glymphatic system and its role in cerebral homeostasis. J Appl Physiol (1985) 2020; 129:1330-1340. [PMID: 33002383 DOI: 10.1152/japplphysiol.00852.2019] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The brain's high bioenergetic state is paralleled by high metabolic waste production. Authentic lymphatic vasculature is lacking in brain parenchyma. Cerebrospinal fluid (CSF) flow has long been thought to facilitate central nervous system detoxification in place of lymphatics, but the exact processes involved in toxic waste clearance from the brain remain incompletely understood. Over the past 8 yr, novel data in animals and humans have begun to shed new light on these processes in the form of the "glymphatic system," a brain-wide perivascular transit passageway dedicated to CSF transport and interstitial fluid exchange that facilitates metabolic waste drainage from the brain. Here we will discuss glymphatic system anatomy and methods to visualize and quantify glymphatic system (GS) transport in the brain and also discuss physiological drivers of its function in normal brain and in neurodegeneration.
Collapse
Affiliation(s)
- Helene Benveniste
- Department of Anesthesiology, Yale School of Medicine, New Haven, Connecticut
| | - Rena Elkin
- Departments of Computer Science and Applied Mathematics & Statistics, Stony Brook University, Stony Brook, New York
| | - Paul M Heerdt
- Department of Anesthesiology, Yale School of Medicine, New Haven, Connecticut
| | - Sunil Koundal
- Department of Anesthesiology, Yale School of Medicine, New Haven, Connecticut
| | - Yuechuan Xue
- Department of Anesthesiology, Yale School of Medicine, New Haven, Connecticut
| | - Hedok Lee
- Department of Anesthesiology, Yale School of Medicine, New Haven, Connecticut
| | - Joanna Wardlaw
- Brain Research Imaging Centre, Centre for Clinical Brain Sciences, Dementia Research Institute at the University of Edinburgh, Edinburgh, United Kingdom
| | - Allen Tannenbaum
- Departments of Computer Science and Applied Mathematics & Statistics, Stony Brook University, Stony Brook, New York
| |
Collapse
|
86
|
Back DB, Choi BR, Han JS, Kwon KJ, Choi DH, Shin CY, Lee J, Kim HY. Characterization of Tauopathy in a Rat Model of Post-Stroke Dementia Combining Acute Infarct and Chronic Cerebral Hypoperfusion. Int J Mol Sci 2020; 21:ijms21186929. [PMID: 32967251 PMCID: PMC7555397 DOI: 10.3390/ijms21186929] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 09/14/2020] [Accepted: 09/18/2020] [Indexed: 12/13/2022] Open
Abstract
Post-stroke dementia (PSD) is a major neurodegenerative consequence of stroke. Tauopathy has been reported in diverse neurodegenerative diseases. We investigated the cognitive impairment and pathomechanism associated with tauopathy in a rat model of PSD by modeling acute ischemic stroke and underlying chronic cerebral hypoperfusion (CCH). We performed middle cerebral artery occlusion (MCAO) surgery in rats to mimic acute ischemic stroke, followed by bilateral common carotid artery occlusion (BCCAo) surgery to mimic CCH. We performed behavioral tests and focused on the characterization of tauopathy through histology. Parenchymal infiltration of cerebrospinal fluid (CSF) tracers after intracisternal injection was examined to evaluate glymphatic function. In an animal model of PSD, cognitive impairment was aggravated when BCCAo was combined with MCAO. Tauopathy, manifested by tau hyperphosphorylation, was prominent in the peri-infarct area when CCH was combined. Synergistic accentuation of tauopathy was evident in the white matter. Microtubules in the neuronal axon and myelin sheath showed partial colocalization with the hyperphosphorylated tau, whereas oligodendrocytes showed near-complete colocalization. Parenchymal infiltration of CSF tracers was attenuated in the PSD model. Our experimental results suggest a hypothesis that CCH may aggravate cognitive impairment and tau hyperphosphorylation in a rat model of PSD by interfering with tau clearance through the glymphatic system. Therapeutic strategies to improve the clearance of brain metabolic wastes, including tau, may be a promising approach to prevent PSD after stroke.
Collapse
Affiliation(s)
- Dong Bin Back
- Department of Neurology, Research Institute of Medical Science, Konkuk University School of Medicine, Seoul 05029, Korea; (D.B.B.); (B.-R.C.); (K.J.K.)
| | - Bo-Ryoung Choi
- Department of Neurology, Research Institute of Medical Science, Konkuk University School of Medicine, Seoul 05029, Korea; (D.B.B.); (B.-R.C.); (K.J.K.)
| | - Jung-Soo Han
- Department of Biological Sciences, Konkuk University, Seoul 05029, Korea;
| | - Kyoung Ja Kwon
- Department of Neurology, Research Institute of Medical Science, Konkuk University School of Medicine, Seoul 05029, Korea; (D.B.B.); (B.-R.C.); (K.J.K.)
- Department of Medicine, Konkuk University School of Medicine, Seoul 05029, Korea;
| | - Dong-Hee Choi
- Department of Medicine, Konkuk University School of Medicine, Seoul 05029, Korea;
| | - Chan Young Shin
- Department of Pharmacology, Konkuk University School of Medicine, Seoul 05029, Korea;
| | - Jongmin Lee
- Department of Rehabilitation Medicine, Konkuk University School of Medicine, Seoul 05029, Korea;
| | - Hahn Young Kim
- Department of Neurology, Research Institute of Medical Science, Konkuk University School of Medicine, Seoul 05029, Korea; (D.B.B.); (B.-R.C.); (K.J.K.)
- Correspondence: or ; Tel.: +82-2-2030-7563
| |
Collapse
|
87
|
Javierre-Petit C, Schneider JA, Kapasi A, Makkinejad N, Tamhane AA, Leurgans SE, Mehta RI, Barnes LL, Bennett DA, Arfanakis K. Neuropathologic and Cognitive Correlates of Enlarged Perivascular Spaces in a Community-Based Cohort of Older Adults. Stroke 2020; 51:2825-2833. [PMID: 32757750 PMCID: PMC7484322 DOI: 10.1161/strokeaha.120.029388] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
BACKGROUND AND PURPOSE Enlarged perivascular spaces (EPVS) have been associated with aging, increased stroke risk, decreased cognitive function, and vascular dementia. However, the relationship of EPVS with age-related neuropathologies is not well understood. Therefore, the purpose of this study was to assess the neuropathologic correlates of EPVS in a large community-based cohort of older adults. The cognitive correlates of EPVS over and beyond those of other pathologies were also assessed. METHODS This study included 654 older deceased and autopsied participants of 3 longitudinal community-based studies of aging that had available data on cognition, ex vivo brain magnetic resonance imaging, and detailed neuropathologic examination. EPVS seen on ex vivo magnetic resonance imaging were histologically validated. Experienced observers rated EPVS burden in ex vivo magnetic resonance imaging using a semiquantitative 4-level scale. Elastic-net regularized ordinal logistic regression was used to investigate associations of EPVS burden with age-related neuropathologies. Mixed-effects models of cognition controlling for neuropathologies, demographics, and clinical factors, were used to determine whether EPVS burden has additional contributions to cognitive decline. RESULTS EPVS burden in the whole group was associated with gross infarcts (odds ratio=1.67, P=0.0017) and diabetes mellitus (odds ratio=1.73, P=0.004). When considering only nondemented participants (with mild or no cognitive impairment), EPVS burden was associated with gross infarcts (odds ratio=1.74, P=0.016) and microscopic infarcts (odds ratio=1.79, P=0.013). EPVS burden was associated with faster decline in visuospatial abilities (estimate=-0.009, P=0.028), in the whole group, as well as lower levels of semantic memory (estimate=-0.13, P=0.048) and visuospatial abilities (estimate=-0.11, P=0.016) at the time of death. CONCLUSIONS EPVS and infarcts may share similar neurobiological pathways regardless of dementia status. EPVS burden is linked to diabetes mellitus independently of neuropathologies, extending recent findings in animal studies implicating diabetes mellitus in impairment of the glymphatic system. Finally, EPVS burden may reflect additional brain tissue injury that may contribute to cognitive decline, not captured with traditional neuropathologic measures.
Collapse
Affiliation(s)
- Carles Javierre-Petit
- Department of Biomedical Engineering, Illinois Institute of Technology, Chicago (C.J.P., N.M., K.A.)
| | - Julie A Schneider
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL (J.A.S., A.K., A.A.T., S.E.L., R.I.M., L.L.B., D.A.B., K.A.)
| | - Alifiya Kapasi
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL (J.A.S., A.K., A.A.T., S.E.L., R.I.M., L.L.B., D.A.B., K.A.)
| | - Nazanin Makkinejad
- Department of Biomedical Engineering, Illinois Institute of Technology, Chicago (C.J.P., N.M., K.A.)
| | - Ashish A Tamhane
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL (J.A.S., A.K., A.A.T., S.E.L., R.I.M., L.L.B., D.A.B., K.A.)
| | - Sue E Leurgans
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL (J.A.S., A.K., A.A.T., S.E.L., R.I.M., L.L.B., D.A.B., K.A.)
| | - Rupal I Mehta
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL (J.A.S., A.K., A.A.T., S.E.L., R.I.M., L.L.B., D.A.B., K.A.)
| | - Lisa L Barnes
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL (J.A.S., A.K., A.A.T., S.E.L., R.I.M., L.L.B., D.A.B., K.A.)
| | - David A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL (J.A.S., A.K., A.A.T., S.E.L., R.I.M., L.L.B., D.A.B., K.A.)
| | - Konstantinos Arfanakis
- Department of Biomedical Engineering, Illinois Institute of Technology, Chicago (C.J.P., N.M., K.A.).,Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL (J.A.S., A.K., A.A.T., S.E.L., R.I.M., L.L.B., D.A.B., K.A.)
| |
Collapse
|
88
|
Johnson SE, McKnight CD, Lants SK, Juttukonda MR, Fusco M, Chitale R, Donahue PC, Claassen DO, Donahue MJ. Choroid plexus perfusion and intracranial cerebrospinal fluid changes after angiogenesis. J Cereb Blood Flow Metab 2020; 40:1658-1671. [PMID: 31500523 PMCID: PMC7370367 DOI: 10.1177/0271678x19872563] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Recent studies have provided evidence that cortical brain ischemia may influence choroid plexus function, and such communication may be mediated by either traditional CSF circulation pathways and/or a possible glymphatic pathway. Here we investigated the hypothesis that improvements in arterial health following neoangiogenesis alter (i) intracranial CSF volume and (ii) choroid plexus perfusion in humans. CSF and tissue volume measurements were obtained from T1-weighted MRI, and cortical and choroid plexus perfusion were obtained from perfusion-weighted arterial spin labeling MRI, in patients with non-atherosclerotic intracranial stenosis (e.g. Moyamoya). Measurements were repeated after indirect surgical revascularization, which elicits cortical neoangiogenesis near the revascularization site (n = 23; age = 41.8 ± 13.4 years), or in a cohort of participants at two time points without interval surgeries (n = 10; age = 41.7 ± 10.7 years). Regression analyses were used to evaluate dependence of perfusion and volume on state (time 1 vs. 2). Post-surgery, neither CSF nor tissue volumes changed significantly. In surgical patients, cortical perfusion increased and choroid plexus perfusion decreased after surgery; in participants without surgeries, cortical perfusion reduced and choroid plexus perfusion increased between time points. Findings are discussed in the context of a homeostatic mechanism, whereby arterial health, paravascular flow, and/or ischemia can affect choroid plexus perfusion.
Collapse
Affiliation(s)
- Skylar E Johnson
- Department of Radiology and Radiological Sciences, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Colin D McKnight
- Department of Radiology and Radiological Sciences, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Sarah K Lants
- Department of Radiology and Radiological Sciences, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Meher R Juttukonda
- Department of Radiology and Radiological Sciences, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Matthew Fusco
- Department of Neurosurgery, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Rohan Chitale
- Department of Neurosurgery, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Paula C Donahue
- Department of Physical Medicine and Rehabilitation, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Daniel O Claassen
- Department of Neurology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Manus J Donahue
- Department of Radiology and Radiological Sciences, Vanderbilt University School of Medicine, Nashville, TN, USA
- Department of Neurology, Vanderbilt University School of Medicine, Nashville, TN, USA
- Department of Psychiatry, Vanderbilt University School of Medicine, Nashville, TN, USA
- Manus J Donahue, Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN 37232, USA.
| |
Collapse
|
89
|
Frič R, Eide PK. Chiari type 1-a malformation or a syndrome? A critical review. Acta Neurochir (Wien) 2020; 162:1513-1525. [PMID: 31656982 DOI: 10.1007/s00701-019-04100-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Accepted: 10/08/2019] [Indexed: 12/14/2022]
Abstract
OBJECTIVE As the understanding of pathophysiology behind Chiari malformation still is limited, the treatment of Chiari malformation type 1 remains rather empirical. This may result in suboptimal treatment strategy and outcome in many cases. In this review, we critically address whether the condition known today as Chiari malformation type I should rather be denoted Chiari syndrome. METHODS The current knowledge of Chiari malformation type 1 is summarized from the historical, etymological, genetic, clinical, and in particular pathophysiological perspectives. RESULTS There are several lines of evidence that Chiari malformation type 1 represents a condition significantly different from types 2 to 4. Unlike the other types, the type 1 should rather be considered a syndrome, thus supporting the reasons to reappraise the traditional classification of Chiari malformations. CONCLUSION We propose that Chiari malformation type 1 should rather be denoted Chiari syndrome, while the notation malformation is maintained for types 2-4.
Collapse
Affiliation(s)
- Radek Frič
- Department of Neurosurgery, Oslo University Hospital - Rikshospitalet, Oslo, Norway.
| | - Per Kristian Eide
- Department of Neurosurgery, Oslo University Hospital - Rikshospitalet, Oslo, Norway
- Faculty of Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
90
|
Kelliny S, Lam HY, Parikh A, Wang YJ, Bobrovskaya L, Upton R, Zhou XF. Preclinical Study of the Pharmacokinetics of p75ECD-Fc, a Novel Human Recombinant Protein for Treatment of Alzheimer’s Disease, in Sprague Dawley Rats. Curr Drug Metab 2020; 21:235-244. [DOI: 10.2174/1389200221666200502015203] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 02/11/2020] [Accepted: 03/26/2020] [Indexed: 01/08/2023]
Abstract
Background:
p75ECD-Fc is a recombinant human protein that has recently been developed as a novel
therapy for Alzheimer’s disease. Current studies showed that it is able to alleviate Alzheimer’s disease pathologies in
animal models of dementia. Thus, knowledge about the pharmacokinetic behavior and tissue distribution of this
novel protein is crucial in order to better understand its pharmacodynamics and more importantly for its clinical
development.
Methods:
The aim of this study is to characterize the pharmacokinetics of p75ECD-Fc after single intravenous and
subcutaneous injection of 3mg/kg in Sprague Dawley rats. We calculated the bioavailability of the SC route and
studied the distribution of that protein in different tissues, cerebrospinal fluid and urine using ELISA and immunofluorescence
techniques. In-vitro stability of the drug was also assessed. Data obtained were analyzed with
Non-compartmental pharmacokinetic method using R.
Results:
Results showed that the bioavailability of SC route was 66.15%. Half-life time was 7.5 ± 1.7 and 6.2 ± 2.4
days for IV and SC injection, respectively. Tissue distribution of p75ECD-Fc was modest with the ability to penetrate
the blood brain barrier. It showed high in vitro stability in human plasma.
Conclusion:
These acceptable pharmacokinetic properties of p75ECD-Fc present it as a potential candidate for clinical
development for the treatment of Alzheimer’s disease.
Collapse
Affiliation(s)
- Sally Kelliny
- School of Pharmacy and Medical Sciences, Division of Health Sciences, University of South Australia, Adelaide, Australia
| | - Ho Yin Lam
- School of Pharmacy and Medical Sciences, Division of Health Sciences, University of South Australia, Adelaide, Australia
| | - Ankit Parikh
- School of Pharmacy and Medical Sciences, Division of Health Sciences, University of South Australia, Adelaide, Australia
| | - Yan-Jiang Wang
- Department of Neurology, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Larisa Bobrovskaya
- School of Pharmacy and Medical Sciences, Division of Health Sciences, University of South Australia, Adelaide, Australia
| | - Richard Upton
- Australian Centre for Pharmacometrics, University of South Australia, Adelaide, Australia
| | - Xin-Fu Zhou
- School of Pharmacy and Medical Sciences, Division of Health Sciences, University of South Australia, Adelaide, Australia
| |
Collapse
|
91
|
Iron Aggravates the Depressive Phenotype of Stressed Mice by Compromising the Glymphatic System. Neurosci Bull 2020; 36:1542-1546. [PMID: 32578069 DOI: 10.1007/s12264-020-00539-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Accepted: 04/02/2020] [Indexed: 12/11/2022] Open
|
92
|
Kananen J, Helakari H, Korhonen V, Huotari N, Järvelä M, Raitamaa L, Raatikainen V, Rajna Z, Tuovinen T, Nedergaard M, Jacobs J, LeVan P, Ansakorpi H, Kiviniemi V. Respiratory-related brain pulsations are increased in epilepsy-a two-centre functional MRI study. Brain Commun 2020; 2:fcaa076. [PMID: 32954328 PMCID: PMC7472909 DOI: 10.1093/braincomms/fcaa076] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 04/29/2020] [Accepted: 05/05/2020] [Indexed: 01/03/2023] Open
Abstract
Resting-state functional MRI has shown potential for detecting changes in cerebral blood oxygen level-dependent signal in patients with epilepsy, even in the absence of epileptiform activity. Furthermore, it has been suggested that coefficient of variation mapping of fast functional MRI signal may provide a powerful tool for the identification of intrinsic brain pulsations in neurological diseases such as dementia, stroke and epilepsy. In this study, we used fast functional MRI sequence (magnetic resonance encephalography) to acquire ten whole-brain images per second. We used the functional MRI data to compare physiological brain pulsations between healthy controls (n = 102) and patients with epilepsy (n = 33) and furthermore to drug-naive seizure patients (n = 9). Analyses were performed by calculating coefficient of variation and spectral power in full band and filtered sub-bands. Brain pulsations in the respiratory-related frequency sub-band (0.11-0.51 Hz) were significantly (P < 0.05) increased in patients with epilepsy, with an increase in both signal variance and power. At the individual level, over 80% of medicated and drug-naive seizure patients exhibited areas of abnormal brain signal power that correlated well with the known clinical diagnosis, while none of the controls showed signs of abnormality with the same threshold. The differences were most apparent in the basal brain structures, respiratory centres of brain stem, midbrain and temporal lobes. Notably, full-band, very low frequency (0.01-0.1 Hz) and cardiovascular (0.8-1.76 Hz) brain pulses showed no differences between groups. This study extends and confirms our previous results of abnormal fast functional MRI signal variance in epilepsy patients. Only respiratory-related brain pulsations were clearly increased with no changes in either physiological cardiorespiratory rates or head motion between the subjects. The regional alterations in brain pulsations suggest that mechanisms driving the cerebrospinal fluid homeostasis may be altered in epilepsy. Magnetic resonance encephalography has both increased sensitivity and high specificity for detecting the increased brain pulsations, particularly in times when other tools for locating epileptogenic areas remain inconclusive.
Collapse
Affiliation(s)
- Janne Kananen
- Oulu Functional NeuroImaging (OFNI), Department of Diagnostic Radiology, Oulu University Hospital, Oulu 90029, Finland
- Medical Imaging, Physics and Technology (MIPT), Faculty of Medicine, University of Oulu, Oulu 90220, Finland
- Medical Research Center (MRC), Oulu 90220, Finland
| | - Heta Helakari
- Oulu Functional NeuroImaging (OFNI), Department of Diagnostic Radiology, Oulu University Hospital, Oulu 90029, Finland
- Medical Imaging, Physics and Technology (MIPT), Faculty of Medicine, University of Oulu, Oulu 90220, Finland
- Medical Research Center (MRC), Oulu 90220, Finland
| | - Vesa Korhonen
- Oulu Functional NeuroImaging (OFNI), Department of Diagnostic Radiology, Oulu University Hospital, Oulu 90029, Finland
- Medical Imaging, Physics and Technology (MIPT), Faculty of Medicine, University of Oulu, Oulu 90220, Finland
- Medical Research Center (MRC), Oulu 90220, Finland
| | - Niko Huotari
- Oulu Functional NeuroImaging (OFNI), Department of Diagnostic Radiology, Oulu University Hospital, Oulu 90029, Finland
- Medical Imaging, Physics and Technology (MIPT), Faculty of Medicine, University of Oulu, Oulu 90220, Finland
- Medical Research Center (MRC), Oulu 90220, Finland
| | - Matti Järvelä
- Oulu Functional NeuroImaging (OFNI), Department of Diagnostic Radiology, Oulu University Hospital, Oulu 90029, Finland
- Medical Imaging, Physics and Technology (MIPT), Faculty of Medicine, University of Oulu, Oulu 90220, Finland
- Medical Research Center (MRC), Oulu 90220, Finland
| | - Lauri Raitamaa
- Oulu Functional NeuroImaging (OFNI), Department of Diagnostic Radiology, Oulu University Hospital, Oulu 90029, Finland
- Medical Imaging, Physics and Technology (MIPT), Faculty of Medicine, University of Oulu, Oulu 90220, Finland
- Medical Research Center (MRC), Oulu 90220, Finland
| | - Ville Raatikainen
- Oulu Functional NeuroImaging (OFNI), Department of Diagnostic Radiology, Oulu University Hospital, Oulu 90029, Finland
- Medical Imaging, Physics and Technology (MIPT), Faculty of Medicine, University of Oulu, Oulu 90220, Finland
- Medical Research Center (MRC), Oulu 90220, Finland
| | - Zalan Rajna
- Oulu Functional NeuroImaging (OFNI), Department of Diagnostic Radiology, Oulu University Hospital, Oulu 90029, Finland
- Center for Machine Vision and Signal Analysis (CMVS), University of Oulu, Oulu 90014, Finland
| | - Timo Tuovinen
- Oulu Functional NeuroImaging (OFNI), Department of Diagnostic Radiology, Oulu University Hospital, Oulu 90029, Finland
- Medical Imaging, Physics and Technology (MIPT), Faculty of Medicine, University of Oulu, Oulu 90220, Finland
- Medical Research Center (MRC), Oulu 90220, Finland
| | - Maiken Nedergaard
- Center for Translational Neuromedicine, Department of Neurosurgery, University of Rochester Medical Center, Rochester, NY 14642, USA
- Center for Translational Neuromedicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark
| | - Julia Jacobs
- Department of Pediatric Neurology and Muscular Disease, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Freiburg 79110, Germany
- Department of Paediatrics, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
- Department of Neuroscience, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
- Hotchkiss Brain Institute and Alberta Children’s Hospital Research Institute, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Pierre LeVan
- Department of Paediatrics, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
- Department of Neuroscience, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
- Hotchkiss Brain Institute and Alberta Children’s Hospital Research Institute, University of Calgary, Calgary, AB T2N 4N1, Canada
- Department of Radiology, Medical Physics, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Freiburg 79110, Germany
- Department of Radiology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Hanna Ansakorpi
- Medical Research Center (MRC), Oulu 90220, Finland
- Research Unit of Neuroscience, Neurology, University of Oulu, Oulu 90220, Finland
- Department of Neurology, Oulu University Hospital, Oulu 90029, Finland
| | - Vesa Kiviniemi
- Oulu Functional NeuroImaging (OFNI), Department of Diagnostic Radiology, Oulu University Hospital, Oulu 90029, Finland
- Medical Imaging, Physics and Technology (MIPT), Faculty of Medicine, University of Oulu, Oulu 90220, Finland
- Medical Research Center (MRC), Oulu 90220, Finland
| |
Collapse
|
93
|
Bouvy WH, van Veluw SJ, Kuijf HJ, Zwanenburg JJ, Kappelle JL, Luijten PR, Koek HL, Geerlings MI, Biessels GJ. Microbleeds colocalize with enlarged juxtacortical perivascular spaces in amnestic mild cognitive impairment and early Alzheimer's disease: A 7 Tesla MRI study. J Cereb Blood Flow Metab 2020; 40:739-746. [PMID: 30890076 PMCID: PMC7074594 DOI: 10.1177/0271678x19838087] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
MRI-visible perivascular spaces (PVS) in the semioval centre are associated with cerebral amyloid angiopathy (CAA), but it is unknown if PVS co-localize with MRI markers of CAA. To examine this, we assessed the topographical association between cortical cerebral microbleeds (CMBs) - as an indirect marker of CAA - and dilatation of juxtacortical perivascular spaces (jPVS) in 46 patients with amnestic mild cognitive impairment (aMCI) or early Alzheimer's disease (eAD). The degree of dilatation of jPVS <1 cm around each cortical CMBs was compared with a similar reference site (no CMB) in the contralateral hemisphere, using a 4-point scale. Also, jPVS dilatation was compared between patients with and without cortical CMBs. Eleven patients (24%) had cortical CMBs [total=35, median=1, range=1-14] of whom five had >1 cortical CMBs. The degree of jPVS dilatation was higher around CMBs than at the reference sites [Wilcoxon signed rank test, Z = 2.2, p = 0.03]. Patients with >1 cortical CMBs had a higher degree of jPVS dilation [median=2.2, IQR = 1.8-2.3] than patients without cortical CMBs [median=1.4, IQR = 1.0-1.8], p = 0.02. We found a topographical association between a high degree of jPVS dilatation and cortical CMBs, supporting a common underlying pathophysiology - most likely CAA.
Collapse
Affiliation(s)
- Willem H Bouvy
- Brain Center Rudolf Magnus, Department of Neurology, University Medical Center Utrecht, the Netherlands
| | - Susanne J van Veluw
- J. Philip Kistler Stroke Research Center, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Hugo J Kuijf
- Image Sciences Institute, University Medical Center Utrecht, the Netherlands
| | - Jaco Jm Zwanenburg
- Department of Radiology, University Medical Center Utrecht, the Netherlands
| | - Jaap L Kappelle
- Brain Center Rudolf Magnus, Department of Neurology, University Medical Center Utrecht, the Netherlands
| | - Peter R Luijten
- Department of Radiology, University Medical Center Utrecht, the Netherlands
| | - Huiberdina L Koek
- Department of Geriatrics, University Medical Center Utrecht, the Netherlands
| | - Mirjam I Geerlings
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, the Netherlands
| | - Geert J Biessels
- Brain Center Rudolf Magnus, Department of Neurology, University Medical Center Utrecht, the Netherlands
| | | |
Collapse
|
94
|
Piotrowska A, Winter K, Carare RO, Bechmann I. Vital Functions Contribute to the Spread of Extracellular Fluids in the Brain: Comparison Between Life and Death. Front Aging Neurosci 2020; 12:15. [PMID: 32116648 PMCID: PMC7027336 DOI: 10.3389/fnagi.2020.00015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Accepted: 01/16/2020] [Indexed: 12/25/2022] Open
Abstract
Vascular pulsations, contractions of vascular smooth muscle cells and breathing have been reported to foster movement and clearance of interstitial and cerebrospinal fluids from the brain. The aim of this study was to estimate the contribution of these vital functions. We compared the spread of an injected hydrophilic tracer (Fluoro-Emerald, a 10 kDa fluorescein-coupled dextran amine) in the brains of live anesthetized and sacrificed rats at 30 and 90 min after injection. To determine the overall pattern of distribution of tracers, we created 3D-reconstructions of the horizontal transections of the whole brain. Immunofluorescence staining with laminin and collagen IV was performed to determine the pattern of distribution of tracer in relation to the cerebrovascular basement membranes. We found that diffusion was widely restricted to the periventricular region in sacrificed rats with no spread to the contralateral hemisphere, while the bulk flow occurred along the vasculature and reached the surface and the contralateral hemisphere as soon as 30 min after injection in live anesthetized animals. The tracer appeared to be localized along the vascular basement membranes and along fiber tracts as reported previously. Thus, our data indicate that vital functions are essential for the remote movement of extracellular fluids within the cerebral parenchyma.
Collapse
Affiliation(s)
| | - Karsten Winter
- Institute of Anatomy, Leipzig University, Leipzig, Germany
| | - Roxana O Carare
- Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Ingo Bechmann
- Institute of Anatomy, Leipzig University, Leipzig, Germany
| |
Collapse
|
95
|
Liu JJ, Raskin JS, McFarlane R, Samatham R, Cetas JS. Subarachnoid Hemorrhage Pattern Predicts Acute Cerebral Blood Flow Response in the Rat. ACTA NEUROCHIRURGICA. SUPPLEMENT 2020; 127:83-89. [PMID: 31407068 DOI: 10.1007/978-3-030-04615-6_14] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
There is considerable variability in the presentation of patients with acute subarachnoid hemorrhage (aSAH). Evidence suggests that a thick, diffuse clot better predicts the development of delayed cerebral ischemia and poor outcomes. In a rodent model of acute SAH, we directly measured the effects of the volume of blood injected versus the pattern of distribution of hemorrhage in the subarachnoid space on markers of early brain injury, namely, cerebral blood flow (CBF), cerebrospinal fluid (CSF) concentrations of P450 eicosanoids and catecholamines, and cortical spreading depolarizations (CSDs). There is a significant decrease in CBF, an increase in CSF biomarkers, and a trend toward increasing frequency and severity of CSDs when grouped by severity of hemorrhage but not by volume of blood injected. In severe hemorrhage grade animals, there was a progressive decrease in CBF after successive CSD events. These results suggest that the pattern of SAH (thick diffuse clots) correlates with the "clinical" severity of SAH.
Collapse
Affiliation(s)
- Jesse J Liu
- Department of Neurological Surgery, Oregon Health & Science University, Portland, OR, USA
| | - Jeffrey S Raskin
- Department of Neurological Surgery, Oregon Health & Science University, Portland, OR, USA
| | | | - Ravi Samatham
- Department of Neurological Surgery, Oregon Health & Science University, Portland, OR, USA
| | - Justin S Cetas
- Department of Neurological Surgery, Oregon Health & Science University, Portland, OR, USA.
- Portland VA Medical Center, Portland, OR, USA.
| |
Collapse
|
96
|
Dong Y, Stewart T, Bai L, Li X, Xu T, Iliff J, Shi M, Zheng D, Yuan L, Wei T, Yang X, Zhang J. Coniferaldehyde attenuates Alzheimer's pathology via activation of Nrf2 and its targets. Am J Cancer Res 2020; 10:179-200. [PMID: 31903114 PMCID: PMC6929631 DOI: 10.7150/thno.36722] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 09/02/2019] [Indexed: 01/22/2023] Open
Abstract
Background: Alzheimer's disease (AD) currently lacks a cure. Because substantial neuronal damage usually occurs before AD is advanced enough for diagnosis, the best hope for disease-modifying AD therapies likely relies on early intervention or even prevention, and targeting multiple pathways implicated in early AD pathogenesis rather than focusing exclusively on excessive production of β-amyloid (Aβ) species. Methods: Coniferaldehyde (CFA), a food flavoring and agonist of NF-E2-related factor 2 (Nrf2), was selected by multimodal in vitro screening, followed by investigation of several downstream effects potentially involved. Furthermore, in the APP/PS1 AD mouse model, the therapeutic effects of CFA (0.2 mmol kg-1d-1) were tested beginning at 3 months of age. Behavioral phenotypes related to learning and memory capacity, brain pathology and biochemistry, including Aβ transport, were assessed at different time intervals. Results: CFA promoted neuron viability and showed potent neuroprotective effects, especially on mitochondrial structure and functions. In addition, CFA greatly enhanced the brain clearance of Aβ in both free and extracellular vesicle (EV)-contained Aβ forms. In the APP/PS1 mouse model, CFA effectively abolished brain Aβ deposits and reduced the level of toxic soluble Aβ peptides, thus eliminating AD-like pathological changes in the hippocampus and cerebral cortex and preserving learning and memory capacity of the mice. Conclusion: The experimental evidence overall indicated that Nrf2 activation may contribute to the potent anti-AD effects of CFA. With an excellent safety profile, further clinical investigation of coniferaldehyde might bring hope for AD prevention/therapy.
Collapse
|
97
|
Klebe D, McBride D, Krafft PR, Flores JJ, Tang J, Zhang JH. Posthemorrhagic hydrocephalus development after germinal matrix hemorrhage: Established mechanisms and proposed pathways. J Neurosci Res 2020; 98:105-120. [PMID: 30793349 PMCID: PMC6703985 DOI: 10.1002/jnr.24394] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Revised: 12/05/2018] [Accepted: 01/14/2019] [Indexed: 01/17/2023]
Abstract
In addition to being the leading cause of morbidity and mortality in premature infants, germinal matrix hemorrhage (GMH) is also the leading cause of acquired infantile hydrocephalus. The pathophysiology of posthemorrhagic hydrocephalus (PHH) development after GMH is complex and vaguely understood, although evidence suggests fibrosis and gliosis in the periventricular and subarachnoid spaces disrupts normal cerebrospinal fluid (CSF) dynamics. Theories explaining general hydrocephalus etiology have substantially evolved from the original bulk flow theory developed by Dr. Dandy over a century ago. Current clinical and experimental evidence supports a new hydrodynamic theory for hydrocephalus development involving redistribution of vascular pulsations and disruption of Starling forces in the brain microcirculation. In this review, we discuss CSF flow dynamics, history and development of theoretical hydrocephalus pathophysiology, and GMH epidemiology and etiology as it relates to PHH development. We highlight known mechanisms and propose new avenues that will further elucidate GMH pathophysiology, specifically related to hydrocephalus.
Collapse
Affiliation(s)
- Damon Klebe
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, CA 92350
| | - Devin McBride
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, CA 92350
| | - Paul R Krafft
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, CA 92350
- Department of Neurosurgery, Loma Linda University School of Medicine, Loma Linda, CA 92350
| | - Jerry J Flores
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, CA 92350
| | - Jiping Tang
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, CA 92350
| | - John H Zhang
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, CA 92350
- Department of Anesthesiology and Neurosurgery, Loma Linda University School of Medicine, Loma Linda, CA 92350
| |
Collapse
|
98
|
Carroll JE, Small BJ, Tometich D, Zhai W, Zhou X, Luta G, Ahles TA, Saykin AJ, Nudelman KNH, Clapp JD, Jim H, Jacobsen PB, Hurria A, Graham D, McDonald BC, Denduluri N, Extermann M, Isaacs C, Dilawari AA, Root J, Stern R, Mandelblatt JS. Sleep disturbance and neurocognitive outcomes in older patients with breast cancer: Interaction with genotype. Cancer 2019; 125:4516-4524. [PMID: 31553501 PMCID: PMC6891125 DOI: 10.1002/cncr.32489] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Revised: 06/03/2019] [Accepted: 07/09/2019] [Indexed: 11/12/2022]
Abstract
BACKGROUND Sleep disturbance and genetic profile are risks for cognitive decline in noncancer populations, yet their role in cancer-related cognitive problems remains understudied. This study examined whether sleep disturbance was associated with worse neurocognitive outcomes in breast cancer survivors and whether sleep effects on cognition varied by genotype. METHODS Newly diagnosed female patients (n = 319) who were 60 years old or older and had stage 0 to III breast cancer were recruited from August 2010 to December 2015. Assessments were performed before systemic therapy and 12 and 24 months later. Neuropsychological testing measured attention, processing speed, executive function, learning, and memory; self-perceived cognitive functioning was also assessed. Sleep disturbance was defined by self-report of routine poor or restless sleep. Genotyping included APOE, BDNF, and COMT polymorphisms. Random effects fluctuation models tested associations of between-person and within-person differences in sleep, genotype, and sleep-genotype interactions and cognition and controlled for age, reading level, race, site, and treatment. RESULTS One-third of the patients reported sleep disturbances at each time point. There was a sleep-APOE ε4 interaction (P = .001) in which patients with the APOE ε4 allele and sleep disturbances had significantly lower learning and memory scores than those who were APOE ε4-negative and without sleep disturbances. There was also a sleep disturbance-COMT genotype interaction (P = .02) in which COMT Val carriers with sleep disturbances had lower perceived cognition than noncarriers. CONCLUSIONS Sleep disturbance was common and was associated with worse cognitive performance in older breast cancer survivors, especially those with a genetic risk for cognitive decline. Survivorship care should include sleep assessments and interventions to address sleep problems.
Collapse
Affiliation(s)
- Judith E. Carroll
- Jane and Terry Semel Institute for Neuroscience and Human Behavior, Jonsson Comprehensive Cancer Center, UCLA Cousins Center for PNI, Psychiatry & Biobehavioral Sciences, Los Angeles, CA
| | | | | | - Wanting Zhai
- Lombardi Comprehensive Cancer Center, Georgetown University
| | - Xiangtao Zhou
- Lombardi Comprehensive Cancer Center, Georgetown University
| | - George Luta
- Lombardi Comprehensive Cancer Center, Georgetown University
| | - Tim A. Ahles
- Memorial Sloan Kettering Cancer Center, New York, NY
| | - Andrew J. Saykin
- Indiana University School of Medicine, Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, IN
| | - Kelly N. H. Nudelman
- Indiana University School of Medicine, Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, IN
| | | | | | - Paul B. Jacobsen
- Healthcare Delivery Research Program, National Cancer Institute, Bethesda, MD
| | | | | | - Brenna C. McDonald
- Indiana University School of Medicine, Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, IN
| | | | | | | | | | - James Root
- Memorial Sloan Kettering Cancer Center, New York, NY
| | - Robert Stern
- Boston University School of Medicine, Boston University
| | | | | |
Collapse
|
99
|
Katsouri L, Birch AM, Renziehausen AWJ, Zach C, Aman Y, Steeds H, Bonsu A, Palmer EOC, Mirzaei N, Ries M, Sastre M. Ablation of reactive astrocytes exacerbates disease pathology in a model of Alzheimer's disease. Glia 2019; 68:1017-1030. [PMID: 31799735 PMCID: PMC7383629 DOI: 10.1002/glia.23759] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 11/20/2019] [Accepted: 11/21/2019] [Indexed: 02/06/2023]
Abstract
The role of astrocytes in the progression of Alzheimer's disease (AD) remains poorly understood. We assessed the consequences of ablating astrocytic proliferation in 9 months old double transgenic APP23/GFAP-TK mice. Treatment of these mice with the antiviral agent ganciclovir conditionally ablates proliferating reactive astrocytes. The loss of proliferating astrocytes resulted in significantly increased levels of monomeric amyloid-β (Aβ) in brain homogenates, associated with reduced enzymatic degradation and clearance mechanisms. In addition, our data revealed exacerbated memory deficits in mice lacking proliferating astrocytes concomitant with decreased levels of synaptic markers and higher expression of pro-inflammatory cytokines. Our data suggest that loss of reactive astrocytes in AD aggravates amyloid pathology and memory loss, possibly via disruption of amyloid clearance and enhanced neuroinflammation.
Collapse
Affiliation(s)
- Loukia Katsouri
- Department of Brain Sciences, Imperial College London, Hammersmith Hospital, London, UK
| | - Amy M Birch
- Department of Brain Sciences, Imperial College London, Hammersmith Hospital, London, UK
| | | | - Carolin Zach
- Department of Brain Sciences, Imperial College London, Hammersmith Hospital, London, UK
| | - Yahyah Aman
- Department of Brain Sciences, Imperial College London, Hammersmith Hospital, London, UK
| | - Hannah Steeds
- Department of Brain Sciences, Imperial College London, Hammersmith Hospital, London, UK
| | - Angela Bonsu
- Department of Brain Sciences, Imperial College London, Hammersmith Hospital, London, UK
| | - Emily O C Palmer
- Department of Brain Sciences, Imperial College London, Hammersmith Hospital, London, UK
| | - Nazanin Mirzaei
- Department of Brain Sciences, Imperial College London, Hammersmith Hospital, London, UK
| | - Miriam Ries
- Department of Brain Sciences, Imperial College London, Hammersmith Hospital, London, UK
| | - Magdalena Sastre
- Department of Brain Sciences, Imperial College London, Hammersmith Hospital, London, UK
| |
Collapse
|
100
|
Abstract
Leukodystrophies are genetically determined disorders affecting the white matter of the central nervous system. The combination of MRI pattern recognition and next-generation sequencing for the definition of novel disease entities has recently demonstrated that many leukodystrophies are due to the primary involvement and/or mutations in genes selectively expressed by cell types other than the oligodendrocytes, the myelin-forming cells in the brain. This has led to a new definition of leukodystrophies as genetic white matter disorders resulting from the involvement of any white matter structural component. As a result, the research has shifted its main focus from oligodendrocytes to other types of neuroglia. Astrocytes are the housekeeping cells of the nervous system, responsible for maintaining homeostasis and normal brain physiology and to orchestrate repair upon injury. Several lines of evidence show that astrocytic interactions with the other white matter cellular constituents play a primary pathophysiologic role in many leukodystrophies. These are thus now classified as astrocytopathies. This chapter addresses how the crosstalk between astrocytes, other glial cells, axons and non-neural cells are essential for the integrity and maintenance of the white matter in health. It also addresses the current knowledge of the cellular pathomechanisms of astrocytic leukodystrophies, and specifically Alexander disease, vanishing white matter, megalencephalic leukoencephalopathy with subcortical cysts and Aicardi-Goutière Syndrome.
Collapse
Affiliation(s)
- M S Jorge
- Department of Pathology, Free University Medical Centre, Amsterdam, The Netherlands
| | - Marianna Bugiani
- Department of Pathology, Free University Medical Centre, Amsterdam, The Netherlands.
| |
Collapse
|