1
|
Aleksandrowicz M. Role of oxidative/nitrosative stress in dysfunction of rat's intracerebral parenchymal arterioles in low sodium environment in the presence of vasopressin. Pflugers Arch 2025:10.1007/s00424-025-03097-1. [PMID: 40434496 DOI: 10.1007/s00424-025-03097-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 04/29/2025] [Accepted: 05/18/2025] [Indexed: 05/29/2025]
Abstract
Hyponatremia is the most common electrolyte disturbance in hospitalized patients. Symptoms of hyponatremia include attention deficits and cognitive impairments. The cause of such abnormalities may be disturbances in the regulation of microcirculation. Previous studies have shown that increased vasopressin (AVP) concentration to 15 pg/ml in the presence of decreased Na+ concentration to 121 mM, which mimics AVP-associated hyponatremia in vivo leads to dysfunction, i.e., constriction and impaired endothelial regulation of small intracerebral blood vessels-parenchymal arterioles (PA). One of the possible causes of this dysfunction may be excessive production of superoxide anion (O2•-). The superoxide anion binds nitric oxide (NO) in a reaction that produces aggressive nitrogen-free radical, peroxynitrite (ONOO-), which simultaneously reduces the bioavailability of NO. The present studies were performed in the organ chamber on isolated, perfused, and pressurized rats' PA in low sodium environment in the presence of AVP. These studies aimed to investigate the mechanism leading to PA dysfunction, i.e., constriction and disturbed endothelial regulation. L-NAME (N(ω)-nitro-L-arginine methyl ester) did not elicit constriction of PA, indicating reduced involvement of NO in maintaining basal tone of PA. Vasopressin V1a receptor antagonist (SR 49059), endothelin ETA/ETB receptors antagonist (PD 142,893), peroxynitrite decomposition catalyst (FeTMPyP) and ROS scavengers: superoxide dismutase (SOD) and catalase (CAT) improved studied responses. Dihydroethidium (DHE) staining confirmed the increased superoxide anion formation in low sodium environment in the presence of AVP. Thromboxane A2/prostaglandin H2 receptor blocker (SQ 29,548), an inhibitor of the production of 20-HETE (HET0016), and L-arginine, a precursor of NO, did not improve dysfunctions of PA. Thus, in studied conditions, endothelial dysfunction occurs due to oxidative/nitrosative stress. These findings provide novel insight into the detrimental effects of decreased Na+ concentration in the presence of increased AVP concentration that mimic hyponatremia, on the regulation of cerebral microcirculation.
Collapse
Affiliation(s)
- Marta Aleksandrowicz
- Laboratory of Preclinical Research and Environmental Agents, Mossakowski Medical Research Institute, Polish Academy of Sciences, A. Pawińskiego Str. 5, 02-106, Warsaw, Poland.
| |
Collapse
|
2
|
Gomez JR, Bhende BU, Mathur R, Gonzalez LF, Shah VA. Individualized autoregulation-guided arterial blood pressure management in neurocritical care. Neurotherapeutics 2025; 22:e00526. [PMID: 39828496 PMCID: PMC11840358 DOI: 10.1016/j.neurot.2025.e00526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 01/07/2025] [Accepted: 01/08/2025] [Indexed: 01/22/2025] Open
Abstract
Cerebral autoregulation (CA) is the physiological process by which cerebral blood flow is maintained during fluctuations in arterial blood pressure (ABP). There are various validated methods to measure CA, either invasively, with intracranial pressure or brain tissue oxygenation monitors, or noninvasively, with transcranial Doppler ultrasound or near-infrared spectroscopy. Utilizing these monitors, researchers have been able to discern CA patterns in several pathological states, such as but not limited to acute ischemic stroke, spontaneous intracranial hemorrhage, aneurysmal subarachnoid hemorrhage, sepsis, and post-cardiac arrest, and they have found CA to be altered in these patients. CA disturbances predispose patients suffering from these ailments to worse outcomes. Much focus has been placed on CA monitoring in these populations, with an emphasis on arterial blood pressure optimization. Many guidelines recommend universal static ABP targets; however, in patients with altered CA, these targets may make them susceptible to hypoperfusion and further neurological injury. Based on this observation, there has been much investigation on individualized ABP goals and their effect on clinical outcomes. The scope of this review includes (1) a summary of the physiology of CA in healthy adults; (2) a review of the evidence on CA monitoring in healthy individuals; (3) a summary of CA changes and its effect on outcomes in various diseased states including acute ischemic stroke, spontaneous intracranial hemorrhage, aneurysmal subarachnoid hemorrhage, sepsis and meningitis, post-cardiac arrest, hypoxic-ischemic encephalopathy, surgery, and moyamoya disease; and (4) a review of the current evidence on individualized ABP changes in various patient populations.
Collapse
Affiliation(s)
- Jonathan R Gomez
- Division of Neurosciences Critical Care, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Neurology, Johns Hopkins University School of Medicine, USA; Department of Anesthesiology & Critical Care Medicine, Johns Hopkins University School of Medicine, USA
| | - Bhagyashri U Bhende
- Division of Neurosciences Critical Care, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Neurology, Johns Hopkins University School of Medicine, USA; Department of Anesthesiology & Critical Care Medicine, Johns Hopkins University School of Medicine, USA
| | - Rohan Mathur
- Division of Neurosciences Critical Care, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Neurology, Johns Hopkins University School of Medicine, USA; Department of Anesthesiology & Critical Care Medicine, Johns Hopkins University School of Medicine, USA
| | - L Fernando Gonzalez
- Department of Neurosurgery, Johns Hopkins University School of Medicine, USA; Division of Vascular and Endovascular Neurosurgery, Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Vishank A Shah
- Division of Neurosciences Critical Care, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Neurology, Johns Hopkins University School of Medicine, USA; Department of Anesthesiology & Critical Care Medicine, Johns Hopkins University School of Medicine, USA.
| |
Collapse
|
3
|
Carr JMJR, Hoiland RL, Fernandes IA, Schrage WG, Ainslie PN. Recent insights into mechanisms of hypoxia-induced vasodilatation in the human brain. J Physiol 2024; 602:5601-5618. [PMID: 37655827 DOI: 10.1113/jp284608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 08/07/2023] [Indexed: 09/02/2023] Open
Abstract
The cerebral vasculature manages oxygen delivery by adjusting arterial blood in-flow in the face of reductions in oxygen availability. Hypoxic cerebral vasodilatation, and the associated hypoxic cerebral blood flow reactivity, involve many vascular, erythrocytic and cerebral tissue mechanisms that mediate elevations in cerebral blood flow via micro- and macrovascular dilatation. This contemporary review focuses on in vivo human work - with reference to seminal preclinical work where necessary - on hypoxic cerebrovascular reactivity, particularly where recent advancements have been made. We provide updates with the following information: in humans, hypoxic cerebral vasodilatation is partially mediated via a - likely non-obligatory - combination of: (1) nitric oxide synthases, (2) deoxygenation-coupled S-nitrosothiols, (3) potassium channel-related vascular smooth muscle hyperpolarization, and (4) prostaglandin mechanisms with some contribution from an interrelationship with reactive oxygen species. And finally, we discuss the fact that, due to the engagement of deoxyhaemoglobin-related mechanisms, reductions in O2 content via haemoglobin per se seem to account for ∼50% of that seen with hypoxic cerebral vasodilatation during hypoxaemia. We further highlight the issue that methodological impediments challenge the complete elucidation of hypoxic cerebral reactivity mechanisms in vivo in healthy humans. Future research is needed to confirm recent advancements and to reconcile human and animal findings. Further investigations are also required to extend these findings to address questions of sex-, heredity-, age-, and disease-related differences. The final step is to then ultimately translate understanding of these mechanisms into actionable, targetable pathways for the prevention and treatment of cerebral vascular dysfunction and cerebral hypoxic brain injury.
Collapse
Affiliation(s)
- Jay M J R Carr
- Centre for Heart, Lung and Vascular Health, University of British Columbia Okanagan, Kelowna, British Columbia, Canada
| | - Ryan L Hoiland
- Department of Anesthesiology, Pharmacology and Therapeutics, Vancouver General Hospital, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, British Columbia, Canada
- International Collaboration on Repair Discoveries, University of British Columbia, Vancouver, British Columbia, Canada
- Collaborative Entity for Researching Brain Ischemia (CEREBRI), University of British Columbia, Vancouver, British Columbia, Canada
| | - Igor A Fernandes
- Department of Health and Kinesiology, Purdue University, Indiana, USA
| | - William G Schrage
- Department of Kinesiology, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Philip N Ainslie
- Centre for Heart, Lung and Vascular Health, University of British Columbia Okanagan, Kelowna, British Columbia, Canada
| |
Collapse
|
4
|
Collée M, Rajkumar R, Farrher E, Hagen J, Ramkiran S, Schnellbächer GJ, Khudeish N, Shah NJ, Veselinović T, Neuner I. Predicting performance in attention by measuring key metabolites in the PCC with 7T MRS. Sci Rep 2024; 14:17099. [PMID: 39048626 PMCID: PMC11269673 DOI: 10.1038/s41598-024-67866-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 07/16/2024] [Indexed: 07/27/2024] Open
Abstract
The posterior cingulate cortex (PCC) is a key hub of the default mode network and is known to play an important role in attention. Using ultra-high field 7 Tesla magnetic resonance spectroscopy (MRS) to quantify neurometabolite concentrations, this exploratory study investigated the effect of the concentrations of myo-inositol (Myo-Ins), glutamate (Glu), glutamine (Gln), aspartate or aspartic acid (Asp) and gamma-amino-butyric acid (GABA) in the PCC on attention in forty-six healthy participants. Each participant underwent an MRS scan and cognitive testing, consisting of a trail-making test (TMT A/B) and a test of attentional performance. After a multiple regression analysis and bootstrapping for correction, the findings show that Myo-Ins and Asp significantly influence (p < 0.05) attentional tasks. On one hand, Myo-Ins shows it can improve the completion times of both TMT A and TMT B. On the other hand, an increase in aspartate leads to more mistakes in Go/No-go tasks and shows a trend towards enhancing reaction time in Go/No-go tasks and stability of alertness without signal. No significant (p > 0.05) influence of Glu, Gln and GABA was observed.
Collapse
Affiliation(s)
- M Collée
- Department of Psychiatry, Psychotherapy and Psychosomatics, RWTH Aachen University, Aachen, Germany
- Institute of Neuroscience and Medicine 4, INM-4, Forschungszentrum Jülich GmbH, 52425, Jülich, Germany
| | - R Rajkumar
- Department of Psychiatry, Psychotherapy and Psychosomatics, RWTH Aachen University, Aachen, Germany
- Institute of Neuroscience and Medicine 4, INM-4, Forschungszentrum Jülich GmbH, 52425, Jülich, Germany
- JARA - BRAIN - Translational Medicine, Aachen, Germany
| | - E Farrher
- Institute of Neuroscience and Medicine 4, INM-4, Forschungszentrum Jülich GmbH, 52425, Jülich, Germany
| | - J Hagen
- Department of Psychiatry, Psychotherapy and Psychosomatics, RWTH Aachen University, Aachen, Germany
- Institute of Neuroscience and Medicine 4, INM-4, Forschungszentrum Jülich GmbH, 52425, Jülich, Germany
| | - S Ramkiran
- Department of Psychiatry, Psychotherapy and Psychosomatics, RWTH Aachen University, Aachen, Germany
- Institute of Neuroscience and Medicine 4, INM-4, Forschungszentrum Jülich GmbH, 52425, Jülich, Germany
| | - G J Schnellbächer
- Department of Psychiatry, Psychotherapy and Psychosomatics, RWTH Aachen University, Aachen, Germany
- Institute of Neuroscience and Medicine 4, INM-4, Forschungszentrum Jülich GmbH, 52425, Jülich, Germany
| | - N Khudeish
- Department of Psychiatry, Psychotherapy and Psychosomatics, RWTH Aachen University, Aachen, Germany
- Institute of Neuroscience and Medicine 4, INM-4, Forschungszentrum Jülich GmbH, 52425, Jülich, Germany
| | - N J Shah
- Department of Psychiatry, Psychotherapy and Psychosomatics, RWTH Aachen University, Aachen, Germany
- Institute of Neuroscience and Medicine 4, INM-4, Forschungszentrum Jülich GmbH, 52425, Jülich, Germany
- JARA - BRAIN - Translational Medicine, Aachen, Germany
- Institute of Neuroscience and Medicine 11, INM-11, Forschungszentrum Jülich, Jülich, Germany
- Department of Neurology, RWTH Aachen University, Aachen, Germany
| | - T Veselinović
- Department of Psychiatry, Psychotherapy and Psychosomatics, RWTH Aachen University, Aachen, Germany
- Institute of Neuroscience and Medicine 4, INM-4, Forschungszentrum Jülich GmbH, 52425, Jülich, Germany
- JARA - BRAIN - Translational Medicine, Aachen, Germany
| | - I Neuner
- Department of Psychiatry, Psychotherapy and Psychosomatics, RWTH Aachen University, Aachen, Germany.
- Institute of Neuroscience and Medicine 4, INM-4, Forschungszentrum Jülich GmbH, 52425, Jülich, Germany.
- JARA - BRAIN - Translational Medicine, Aachen, Germany.
| |
Collapse
|
5
|
Hristovska I, Binette AP, Kumar A, Gaiteri C, Karlsson L, Strandberg O, Janelidze S, van Westen D, Stomrud E, Palmqvist S, Ossenkoppele R, Mattsson-Carlgren N, Vogel JW, Hansson O. Identification of distinct and shared biomarker panels in different manifestations of cerebral small vessel disease through proteomic profiling. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.06.10.24308599. [PMID: 38947084 PMCID: PMC11213103 DOI: 10.1101/2024.06.10.24308599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
The pathophysiology underlying various manifestations of cerebral small vessel disease (cSVD) remains obscure. Using cerebrospinal fluid proximity extension assays and co-expression network analysis of 2,943 proteins, we found common and distinct proteomic signatures between white matter lesions (WML), microbleeds and infarcts measured in 856 living patients, and validated WML-associated proteins in three additional datasets. Proteins indicative of extracellular matrix dysregulation and vascular remodeling, including ELN, POSTN, CCN2 and MMP12 were elevated across all cSVD manifestations, with MMP12 emerging as an early cSVD indicator. cSVD-associated proteins formed a co-abundance network linked to metabolism and enriched in endothelial and arterial smooth muscle cells, showing elevated levels at early disease manifestations. Later disease stages involved changes in microglial proteins, associated with longitudinal WML progression, and changes in neuronal proteins mediating WML-associated cognitive decline. These findings provide an atlas of novel cSVD biomarkers and a promising roadmap for the next generation of cSVD therapeutics.
Collapse
Affiliation(s)
- Ines Hristovska
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
| | - Alexa Pichet Binette
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
| | - Atul Kumar
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
| | - Chris Gaiteri
- Department of Psychiatry, SUNY Upstate Medical University, Syracuse, NY, USA
- Rush University Alzheimer's Disease Center, Rush University, Chicago IL, USA
| | - Linda Karlsson
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
| | - Olof Strandberg
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
| | - Shorena Janelidze
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
| | - Danielle van Westen
- Diagnostic Radiology, Department of Clinical Sciences Lund, Lund University
- Imaging and Function, Skåne University Hospital, Lund, Sweden
| | - Erik Stomrud
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Sebastian Palmqvist
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Rik Ossenkoppele
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands
| | - Niklas Mattsson-Carlgren
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
- Department of Neurology, Skåne University Hospital, Lund, Sweden
- Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden
| | - Jacob W Vogel
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
- Department of Clinical Sciences, Malmö, SciLifeLab, Lund University, Lund, Sweden
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| |
Collapse
|
6
|
Lénárt N, Cserép C, Császár E, Pósfai B, Dénes Á. Microglia-neuron-vascular interactions in ischemia. Glia 2024; 72:833-856. [PMID: 37964690 DOI: 10.1002/glia.24487] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 10/24/2023] [Accepted: 10/24/2023] [Indexed: 11/16/2023]
Abstract
Cerebral ischemia is a devastating condition that results in impaired blood flow in the brain leading to acute brain injury. As the most common form of stroke, occlusion of cerebral arteries leads to a characteristic sequence of pathophysiological changes in the brain tissue. The mechanisms involved, and comorbidities that determine outcome after an ischemic event appear to be highly heterogeneous. On their own, the processes leading to neuronal injury in the absence of sufficient blood supply to meet the metabolic demand of the cells are complex and manifest at different temporal and spatial scales. While the contribution of non-neuronal cells to stroke pathophysiology is increasingly recognized, recent data show that microglia, the main immune cells of the central nervous system parenchyma, play previously unrecognized roles in basic physiological processes beyond their inflammatory functions, which markedly change during ischemic conditions. In this review, we aim to discuss some of the known microglia-neuron-vascular interactions assumed to contribute to the acute and delayed pathologies after cerebral ischemia. Because the mechanisms of neuronal injury have been extensively discussed in several excellent previous reviews, here we focus on some recently explored pathways that may directly or indirectly shape neuronal injury through microglia-related actions. These discoveries suggest that modulating gliovascular processes in different forms of stroke and other neurological disorders might have presently unexplored therapeutic potential in combination with neuroprotective and flow restoration strategies.
Collapse
Affiliation(s)
- Nikolett Lénárt
- Momentum Laboratory of Neuroimmunology, Institute of Experimental Medicine, Budapest, Hungary
| | - Csaba Cserép
- Momentum Laboratory of Neuroimmunology, Institute of Experimental Medicine, Budapest, Hungary
| | - Eszter Császár
- Momentum Laboratory of Neuroimmunology, Institute of Experimental Medicine, Budapest, Hungary
| | - Balázs Pósfai
- Momentum Laboratory of Neuroimmunology, Institute of Experimental Medicine, Budapest, Hungary
| | - Ádám Dénes
- Momentum Laboratory of Neuroimmunology, Institute of Experimental Medicine, Budapest, Hungary
| |
Collapse
|
7
|
Villa RF, Ferrari F, Gorini A. Effects of Chronic Hypertension on the Energy Metabolism of Cerebral Cortex Mitochondria in Normotensive and in Spontaneously Hypertensive Rats During Aging. Neuromolecular Med 2024; 26:2. [PMID: 38393429 DOI: 10.1007/s12017-023-08772-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Accepted: 12/02/2023] [Indexed: 02/25/2024]
Abstract
In this study the subcellular modifications undergone by cerebral cortex mitochondrial metabolism in chronic hypertension during aging were evaluated. The catalytic properties of regulatory energy-linked enzymes of Tricarboxylic Acid Cycle (TCA), Electron Transport Chain (ETC) and glutamate metabolism were assayed on non-synaptic mitochondria (FM, located in post-synaptic compartment) and on intra-synaptic mitochondria of pre-synaptic compartment, furtherly divided in "light" (LM) and "heavy" (HM) mitochondria, purified form cerebral cortex of normotensive Wistar Kyoto Rats (WKY) versus Spontaneously Hypertensive Rats (SHR) at 6, 12 and 18 months. During physiological aging, the metabolic machinery was differently expressed in pre- and post-synaptic compartments: LM and above all HM were more affected by aging, displaying lower ETC activities. In SHR at 6 months, FM and LM showed an uncoupling between TCA and ETC, likely as initial adaptive response to hypertension. During pathological aging, HM were particularly affected at 12 months in SHR, as if the adaptive modifications in FM and LM at 6 months granted a mitochondrial functional balance, while at 18 months all the neuronal mitochondria displayed decreased metabolic fluxes versus WKY. This study describes the effects of chronic hypertension on cerebral mitochondrial energy metabolism during aging through functional proteomics of enzymes at subcellular levels, i.e. in neuronal soma and synapses. In addition, this represents the starting point to envisage an experimental physiopathological model which could be useful also for pharmacological studies, to assess drug actions during the development of age-related pathologies that could coexist and/or are provoked by chronic hypertension.
Collapse
Affiliation(s)
- Roberto Federico Villa
- Department of Biology and Biotechnology, Laboratory of Pharmacology and Molecular Medicine of Central Nervous System, University of Pavia, Via Ferrata, 9, 27100, Pavia, Italy.
| | - Federica Ferrari
- Department of Biology and Biotechnology, Laboratory of Pharmacology and Molecular Medicine of Central Nervous System, University of Pavia, Via Ferrata, 9, 27100, Pavia, Italy
- School of Neurology, Department of Brain and Behavioral Sciences, University of Pavia, Via Bassi. 21, 27100, Pavia, Italy
| | - Antonella Gorini
- Department of Biology and Biotechnology, Laboratory of Pharmacology and Molecular Medicine of Central Nervous System, University of Pavia, Via Ferrata, 9, 27100, Pavia, Italy
| |
Collapse
|
8
|
Yuan WQ, Huang WP, Jiang YC, Xu H, Duan CS, Chen NH, Liu YJ, Fu XM. The function of astrocytes and their role in neurological diseases. Eur J Neurosci 2023; 58:3932-3961. [PMID: 37831013 DOI: 10.1111/ejn.16160] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 09/11/2023] [Accepted: 09/13/2023] [Indexed: 10/14/2023]
Abstract
Astrocytes have countless links with neurons. Previously, astrocytes were only considered a scaffold of neurons; in fact, astrocytes perform a variety of functions, including providing support for neuronal structures and energy metabolism, offering isolation and protection and influencing the formation, function and elimination of synapses. Because of these functions, astrocytes play an critical role in central nervous system (CNS) diseases. The regulation of the secretiory factors, receptors, channels and pathways of astrocytes can effectively inhibit the occurrence and development of CNS diseases, such as neuromyelitis optica (NMO), multiple sclerosis, Alzheimer's disease (AD), Parkinson's disease (PD) and Huntington's disease. The expression of aquaporin 4 in AS is directly related to NMO and indirectly involved in the clearance of Aβ and tau proteins in AD. Connexin 43 has a bidirectional effect on glutamate diffusion at different stages of stroke. Interestingly, astrocytes reduce the occurrence of PD through multiple effects such as secretion of related factors, mitochondrial autophagy and aquaporin 4. Therefore, this review is focused on the structure and function of astrocytes and the correlation between astrocytes and CNS diseases and drug treatment to explore the new functions of astrocytes with the astrocytes as the target. This, in turn, would provide a reference for the development of new drugs to protect neurons and promote the recovery of nerve function.
Collapse
Affiliation(s)
- Wen-Qin Yuan
- College of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, China
| | - Wei-Peng Huang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica and Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- College of Pharmacy, Minzu University of China, Beijing, China
| | - Yang-Chao Jiang
- College of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, China
| | - Hao Xu
- College of Economics and Management, Jiangxi University of Chinese Medicine, Nanchang, China
| | - Chong-Shen Duan
- College of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, China
| | - Nai-Hong Chen
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica and Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ying-Jiao Liu
- College of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, China
| | - Xiao-Mei Fu
- College of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, China
| |
Collapse
|
9
|
Huang L, Cheng F, Zhang X, Zielonka J, Nystoriak MA, Xiang W, Raygor K, Wang S, Lakshmanan A, Jiang W, Yuan S, Hou KS, Zhang J, Wang X, Syed AU, Juric M, Takahashi T, Navedo MF, Wang RA. Nitric oxide synthase and reduced arterial tone contribute to arteriovenous malformation. SCIENCE ADVANCES 2023; 9:eade7280. [PMID: 37235659 PMCID: PMC10219588 DOI: 10.1126/sciadv.ade7280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Accepted: 04/20/2023] [Indexed: 05/28/2023]
Abstract
Mechanisms underlying arteriovenous malformations (AVMs) are poorly understood. Using mice with endothelial cell (EC) expression of constitutively active Notch4 (Notch4*EC), we show decreased arteriolar tone in vivo during brain AVM initiation. Reduced vascular tone is a primary effect of Notch4*EC, as isolated pial arteries from asymptomatic mice exhibited reduced pressure-induced arterial tone ex vivo. The nitric oxide (NO) synthase (NOS) inhibitor NG-nitro-l-arginine (L-NNA) corrected vascular tone defects in both assays. L-NNA treatment or endothelial NOS (eNOS) gene deletion, either globally or specifically in ECs, attenuated AVM initiation, assessed by decreased AVM diameter and delayed time to moribund. Administering nitroxide antioxidant 4-hydroxy-2,2,6,6-tetramethylpiperidine-1-oxyl also attenuated AVM initiation. Increased NOS-dependent production of hydrogen peroxide, but not NO, superoxide, or peroxynitrite was detected in isolated Notch4*EC brain vessels during AVM initiation. Our data suggest that eNOS is involved in Notch4*EC-mediated AVM formation by up-regulating hydrogen peroxide and reducing vascular tone, thereby permitting AVM initiation and progression.
Collapse
Affiliation(s)
- Lawrence Huang
- Laboratory for Accelerated Vascular Research, Department of Surgery, University of California San Francisco, San Francisco, CA 94143, USA
| | - Feng Cheng
- Laboratory for Accelerated Vascular Research, Department of Surgery, University of California San Francisco, San Francisco, CA 94143, USA
| | - Xuetao Zhang
- Laboratory for Accelerated Vascular Research, Department of Surgery, University of California San Francisco, San Francisco, CA 94143, USA
| | - Jacek Zielonka
- Free Radical Research Laboratory, Department of Biophysics, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Matthew A. Nystoriak
- Department of Pharmacology, University of California, Davis, Davis, CA 95616, USA
| | - Weiwei Xiang
- Laboratory for Accelerated Vascular Research, Department of Surgery, University of California San Francisco, San Francisco, CA 94143, USA
| | - Kunal Raygor
- Laboratory for Accelerated Vascular Research, Department of Surgery, University of California San Francisco, San Francisco, CA 94143, USA
| | - Shaoxun Wang
- Laboratory for Accelerated Vascular Research, Department of Surgery, University of California San Francisco, San Francisco, CA 94143, USA
| | - Aditya Lakshmanan
- Laboratory for Accelerated Vascular Research, Department of Surgery, University of California San Francisco, San Francisco, CA 94143, USA
| | - Weiya Jiang
- Laboratory for Accelerated Vascular Research, Department of Surgery, University of California San Francisco, San Francisco, CA 94143, USA
| | - Sai Yuan
- Laboratory for Accelerated Vascular Research, Department of Surgery, University of California San Francisco, San Francisco, CA 94143, USA
| | - Kevin S. Hou
- Laboratory for Accelerated Vascular Research, Department of Surgery, University of California San Francisco, San Francisco, CA 94143, USA
| | - Jiayi Zhang
- Laboratory for Accelerated Vascular Research, Department of Surgery, University of California San Francisco, San Francisco, CA 94143, USA
| | - Xitao Wang
- Laboratory for Accelerated Vascular Research, Department of Surgery, University of California San Francisco, San Francisco, CA 94143, USA
| | - Arsalan U. Syed
- Department of Pharmacology, University of California, Davis, Davis, CA 95616, USA
| | - Matea Juric
- Free Radical Research Laboratory, Department of Biophysics, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Takamune Takahashi
- Division of Nephrology and Hypertension, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Manuel F. Navedo
- Department of Pharmacology, University of California, Davis, Davis, CA 95616, USA
| | - Rong A. Wang
- Laboratory for Accelerated Vascular Research, Department of Surgery, University of California San Francisco, San Francisco, CA 94143, USA
| |
Collapse
|
10
|
Johnson AC. Hippocampal Vascular Supply and Its Role in Vascular Cognitive Impairment. Stroke 2023; 54:673-685. [PMID: 36848422 PMCID: PMC9991081 DOI: 10.1161/strokeaha.122.038263] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 12/22/2022] [Indexed: 03/01/2023]
Abstract
The incidence of age-related dementia is increasing as the world population ages and due to lack of effective treatments for dementia. Vascular contributions to cognitive impairment and dementia are increasing as the prevalence of pathologies associated with cerebrovascular disease rise, including chronic hypertension, diabetes, and ischemic stroke. The hippocampus is a bilateral deep brain structure that is central to learning, memory, and cognitive function and highly susceptible to hypoxic/ischemic injury. Compared with cortical brain regions such as the somatosensory cortex, less is known about the function of the hippocampal vasculature that is critical in maintaining neurocognitive health. This review focuses on the hippocampal vascular supply, presenting what is known about hippocampal hemodynamics and blood-brain barrier function during health and disease, and discusses evidence that supports its contribution to vascular cognitive impairment and dementia. Understanding vascular-mediated hippocampal injury that contributes to memory dysfunction during healthy aging and cerebrovascular disease is essential to develop effective treatments to slow cognitive decline. The hippocampus and its vasculature may represent one such therapeutic target to mitigate the dementia epidemic.
Collapse
Affiliation(s)
- Abbie C Johnson
- Department of Neurological Sciences, University of Vermont Larner College of Medicine, Burlington
| |
Collapse
|
11
|
Kokoti L, Al-Karagholi MAM, Waldorff Nielsen CA, Ashina M. Glibenclamide Posttreatment Does Not Inhibit Levcromakalim Induced Headache in Healthy Participants: A Randomized Clinical Trial. Neurotherapeutics 2023; 20:389-398. [PMID: 36763326 PMCID: PMC10121935 DOI: 10.1007/s13311-023-01350-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/29/2023] [Indexed: 02/11/2023] Open
Abstract
Intravenous infusion of ATP-sensitive potassium channel (KATP) opener levcromakalim causes headache in humans and implicates KATP channels in headache pathophysiology. Whether KATP channel blocker glibenclamide inhibits levcromakalim-induced headache has not yet been elucidated. We aimed to investigate the effect of posttreatment with glibenclamide on levcromakalim-induced headache in healthy participants. In a double blind, randomized, three-arm, placebo-controlled, crossover study, 20 healthy participants were randomized to receive 20 mL of levcromakalim (0.05 mg/min (50 mg/mL)) or 20 mL placebo (isotonic saline) intravenously over 20 min followed by oral administration of 10 mg glibenclamide or placebo. Fifteen participants completed all three study days. The primary endpoint was the difference in incidence of headache (0-12 h) between glibenclamide and placebo. More participants developed headache on levcromakalim-placebo day (15/15, 100%) (P = 0.013) and levcromakalim-glibenclamide day (13/15, 86%) compared to placebo-placebo day (7/15, 46%) (P = 0.041). We found no difference in headache incidence between levcromakalim-placebo day and levcromakalim-glibenclamide day (P = 0.479). The AUC0-12 h for headache intensity was significantly larger in levcromakalim-placebo day and levcromakalim-glibenclamide day compared to placebo-placebo day (106.3 ± 215.8) (P < 0.01). There was no difference in the AUC0-12 h for headache intensity between the levcromakalim-placebo day (494 ± 336.6) and the levcromakalim-glibenclamide day (417 ± 371.6) (P = 0.836). We conclude that non-specific KATP channel inhibitor glibenclamide did not attenuate levcromakalim-induced headache in healthy volunteers. Future studies should clarify the involvement of the distinct isoforms of sulfonylurea receptor subunits of KATP channels in the pathogenesis of headache and migraine.
Collapse
Affiliation(s)
- Lili Kokoti
- Danish Headache Center, Department of Neurology, Rigshospitalet – Glostrup, Faculty of Health and Medical Sciences, University of Copenhagen, Valdemar Hansen Vej 5, 2600 Glostrup, Denmark
| | - Mohammad Al-Mahdi Al-Karagholi
- Danish Headache Center, Department of Neurology, Rigshospitalet – Glostrup, Faculty of Health and Medical Sciences, University of Copenhagen, Valdemar Hansen Vej 5, 2600 Glostrup, Denmark
| | - Cherie Amalie Waldorff Nielsen
- Danish Headache Center, Department of Neurology, Rigshospitalet – Glostrup, Faculty of Health and Medical Sciences, University of Copenhagen, Valdemar Hansen Vej 5, 2600 Glostrup, Denmark
| | - Messoud Ashina
- Danish Headache Center, Department of Neurology, Rigshospitalet – Glostrup, Faculty of Health and Medical Sciences, University of Copenhagen, Valdemar Hansen Vej 5, 2600 Glostrup, Denmark
- Danish Headache Knowledge Center, Rigshospitalet – Glostrup, Glostrup, Denmark
| |
Collapse
|
12
|
Blackwell JA, Silva JF, Louis EM, Savu A, Largent-Milnes TM, Brooks HL, Pires PW. Cerebral arteriolar and neurovascular dysfunction after chemically induced menopause in mice. Am J Physiol Heart Circ Physiol 2022; 323:H845-H860. [PMID: 36149767 PMCID: PMC9602916 DOI: 10.1152/ajpheart.00276.2022] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 09/20/2022] [Accepted: 09/20/2022] [Indexed: 12/14/2022]
Abstract
Cognitive decline is linked to decreased cerebral blood flow, particularly in women after menopause. Impaired cerebrovascular function precedes the onset of dementia, possibly because of reduced functional dilation in parenchymal arterioles. These vessels are bottlenecks of the cerebral microcirculation, and dysfunction can limit functional hyperemia in the brain. Large-conductance Ca2+-activated K+ channels (BKCa) are the final effectors of several pathways responsible for functional hyperemia, and their expression is modulated by estrogen. However, it remains unknown whether BKCa function is altered in cerebral parenchymal arterioles after menopause. Using a chemically induced model of menopause, the 4-vinylcyclohexene diepoxide (VCD) model, which depletes follicles while maintaining intact ovaries, we hypothesized that menopause would be associated with reduced functional vasodilatory responses in cerebral parenchymal arterioles of wild-type mice via reduced BKCa function. Using pressure myography of isolated parenchymal arterioles, we observed that menopause (Meno) induced a significant increase in spontaneous myogenic tone. Endothelial function, assessed as nitric oxide production and dilation after cholinergic stimulation or endothelium-dependent hyperpolarization pathways, was unaffected by Meno. BKCa function was significantly impaired in Meno compared with control, without changes in voltage-gated K+ channel activity. Cerebral functional hyperemia, measured by laser-speckle contrast imaging during whisker stimulation, was significantly blunted in Meno mice, without detectable changes in basal perfusion. However, behavioral testing identified no change in cognition. These findings suggest that menopause induces cerebral microvascular and neurovascular deficits.NEW & NOTEWORTHY Cerebral parenchymal arterioles from menopause mice showed increased myogenic tone. We identified an impairment in smooth muscle cell BKCa channel activity, without a reduction in endothelium-dependent dilation or nitric oxide production. Microvascular dysfunction was associated with a reduction in neurovascular responses after somatosensory stimulation. Despite the neurovascular impairment, cognitive abilities were maintained in menopausal mice.
Collapse
Affiliation(s)
- Jade A Blackwell
- Department of Physiology, University of Arizona, Tucson, Arizona
| | - Josiane F Silva
- Department of Physiology, University of Arizona, Tucson, Arizona
| | - Emma M Louis
- Department of Physiology, University of Arizona, Tucson, Arizona
| | - Andrea Savu
- Department of Physiology, University of Arizona, Tucson, Arizona
| | - Tally M Largent-Milnes
- Department of Pharmacology, University of Arizona, Tucson, Arizona
- Bio5 Institute, University of Arizona, Tucson, Arizona
| | - Heddwen L Brooks
- Department of Physiology, University of Arizona, Tucson, Arizona
- Bio5 Institute, University of Arizona, Tucson, Arizona
- Sarver Heart Center, University of Arizona, Tucson, Arizona
| | - Paulo W Pires
- Department of Physiology, University of Arizona, Tucson, Arizona
- Bio5 Institute, University of Arizona, Tucson, Arizona
- Sarver Heart Center, University of Arizona, Tucson, Arizona
| |
Collapse
|
13
|
Brain Bioenergetics in Chronic Hypertension: Risk Factor for Acute Ischemic Stroke. Biochem Pharmacol 2022; 205:115260. [PMID: 36179931 DOI: 10.1016/j.bcp.2022.115260] [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: 07/30/2022] [Revised: 09/13/2022] [Accepted: 09/14/2022] [Indexed: 11/23/2022]
Abstract
Chronic hypertension is one of the key modifiable risk factors for acute ischemic stroke, also contributing to determine greater neurological deficits and worse functional outcome when an acute cerebrovascular event would occur. A tight relationship exists between cerebrovascular autoregulation, neuronal activity and brain bioenergetics. In chronic hypertension, progressive adaptations of these processes occur as an attempt to cope with the demanding necessity of brain functions, creating a new steady-state homeostatic condition. However, these adaptive modifications are insufficient to grant an adequate response to possible pathological perturbations of the established fragile hemodynamic and metabolic homeostasis. In this narrative review, we will discuss the main mechanisms by which alterations in brain bioenergetics and mitochondrial function in chronic hypertension could lead to increased risk of acute ischemic stroke, stressing the interconnections between hemodynamic factors (i.e. cerebral autoregulation and neurovascular coupling) and metabolic processes. Both experimental and clinical pieces of evidence will be discussed. Moreover, the potential role of mitochondrial dysfunction in determining, or at least sustaining, the pathogenesis and progression of chronic neurogenic hypertension will be considered. In the perspective of novel therapeutic strategies aiming at improving brain bioenergetics, we propose some determinant factors to consider in future studies focused on the cause-effect relationships between chronic hypertension and brain bioenergetic abnormalities (and vice versa), so to help translational research in this so-far unfilled gap.
Collapse
|
14
|
DuBose LE, Weng TB, Pierce GL, Wharff C, Reist L, Hamilton C, O'Deen A, Dubishar K, Lane-Cordova A, Voss MW. Association between cardiorespiratory fitness and cerebrovascular reactivity to a breath-hold stimulus in older adults: influence of aerobic exercise training. J Appl Physiol (1985) 2022; 132:1468-1479. [PMID: 35482329 PMCID: PMC9208436 DOI: 10.1152/japplphysiol.00599.2021] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 04/22/2022] [Accepted: 04/22/2022] [Indexed: 11/22/2022] Open
Abstract
Cerebrovascular reactivity (CVR) to a physiological stimulus is a commonly used surrogate of cerebrovascular health. Cross-sectional studies using blood oxygen level dependent (BOLD) neuroimaging demonstrated lower BOLD-CVR to hypercapnia among adults with high compared with lower cardiorespiratory fitness (CRF) in contrast to transcranial Doppler studies. However, whether BOLD-CVR changes following chronic aerobic exercise in older, cognitively intact adults is unclear. This study evaluated relations between BOLD-CVR with CRF (V̇o2peak) using a cross-sectional and interventional study design. We hypothesized that 1) greater CRF would be associated with lower BOLD-CVR in older adults (n = 114; 65 ± 6.5 yr) with a wide range of CRF and 2) BOLD-CVR would be attenuated after exercise training in a subset (n = 33) randomized to 3-mo of moderate- or light-intensity cycling. CVR was quantified as the change in the BOLD signal in response to acute hypercapnia using a blocked breath-hold design from a region-of-interest analysis for cortical networks. In the cross-sectional analysis, there was a quadratic relation between V̇o2peak (P = 0.03), but not linear (P = 0.87) and cortical BOLD-CVR. BOLD-CVR increased until a V̇o2peak ∼28 mL/kg/min after which BOLD-CVR declined. The nonlinear trend was consistent across all networks (P = 0.04-0.07). In the intervention, both the active and light-intensity exercise groups improved CRF similarly (6% vs. 10.8%, P = 0.28). The percent change in CRF was positively associated with change in BOLD-CVR in the default mode network only. These data suggest that BOLD-CVR is nonlinearly associated with CRF and that in lower-fit adults default mode network may be most sensitive to CRF-related increases in BOLD-CVR.NEW & NOTEWORTHY Earlier studies evaluating associations between cardiorespiratory fitness (CRF) and cerebrovascular reactivity (CVR) have demonstrated conflicting findings dependent on imaging modality or subject characteristics in individuals across a narrow range of CRF. This study demonstrates that CRF is nonlinearly associated with CVR measured by blood oxygen level dependent (BOLD) fMRI in a large sample of middle-aged and older adults across a wide range of CRF, suggesting that conflicting prior findings are related to the range of CRFs studied.
Collapse
Affiliation(s)
- Lyndsey E DuBose
- Department of Health and Human Physiology, University of Iowa, Iowa City, Iowa
| | - Timothy B Weng
- Department Psychological and Brain Sciences, University of Iowa, Iowa City, Iowa
| | - Gary L Pierce
- Department of Health and Human Physiology, University of Iowa, Iowa City, Iowa
- Department Internal Medicine, University of Iowa, Iowa City, Iowa
| | - Conner Wharff
- Department Psychological and Brain Sciences, University of Iowa, Iowa City, Iowa
| | - Lauren Reist
- Department Psychological and Brain Sciences, University of Iowa, Iowa City, Iowa
| | - Chase Hamilton
- Department Psychological and Brain Sciences, University of Iowa, Iowa City, Iowa
| | - Abby O'Deen
- Department Psychological and Brain Sciences, University of Iowa, Iowa City, Iowa
| | - Kaitlyn Dubishar
- Department of Health and Human Physiology, University of Iowa, Iowa City, Iowa
| | - Abbi Lane-Cordova
- Department of Health and Human Physiology, University of Iowa, Iowa City, Iowa
| | - Michelle W Voss
- Department Psychological and Brain Sciences, University of Iowa, Iowa City, Iowa
- Iowa Neuroscience Institute, University of Iowa, Iowa City, Iowa
| |
Collapse
|
15
|
Ando K, Tong L, Peng D, Vázquez-Liébanas E, Chiyoda H, He L, Liu J, Kawakami K, Mochizuki N, Fukuhara S, Grutzendler J, Betsholtz C. KCNJ8/ABCC9-containing K-ATP channel modulates brain vascular smooth muscle development and neurovascular coupling. Dev Cell 2022; 57:1383-1399.e7. [PMID: 35588738 DOI: 10.1016/j.devcel.2022.04.019] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 12/22/2021] [Accepted: 04/26/2022] [Indexed: 12/30/2022]
Abstract
Loss- or gain-of-function mutations in ATP-sensitive potassium channel (K-ATP)-encoding genes, KCNJ8 and ABCC9, cause human central nervous system disorders with unknown pathogenesis. Here, using mice, zebrafish, and cell culture models, we investigated cellular and molecular causes of brain dysfunctions derived from altered K-ATP channel function. We show that genetic/chemical inhibition or activation of KCNJ8/ABCC9-containing K-ATP channel function leads to brain-selective suppression or promotion of arterial/arteriolar vascular smooth muscle cell (VSMC) differentiation, respectively. We further show that brain VSMCs develop from KCNJ8/ABCC9-containing K-ATP channel-expressing mural cell progenitor and that K-ATP channel cell autonomously regulates VSMC differentiation through modulation of intracellular Ca2+ oscillation via voltage-dependent calcium channels. Consistent with defective VSMC development, Kcnj8 knockout mice showed deficiency in vasoconstrictive capacity and neuronal-evoked vasodilation leading to local hyperemia. Our results demonstrate a role for KCNJ8/ABCC9-containing K-ATP channels in the differentiation of brain VSMC, which in turn is necessary for fine-tuning of cerebral blood flow.
Collapse
Affiliation(s)
- Koji Ando
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Dag Hammarskjölds väg 20, 751 85 Uppsala, Sweden; Department of Molecular Pathophysiology, Institute of Advanced Medical Science, Nippon Medical School, 1-1-5 Sendagi Bunkyo-ku, Tokyo 113-8602, Japan; Department of Regenerative Medicine and Tissue Engineering, National Cerebral and Cardiovascular Center Research Institute, 6-1 Kishibe-Shinmachi, Suita, Osaka 564-8565, Japan.
| | - Lei Tong
- Department of Neurology, Yale School of Medicine, New Haven, CT, USA
| | - Di Peng
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Dag Hammarskjölds väg 20, 751 85 Uppsala, Sweden
| | - Elisa Vázquez-Liébanas
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Dag Hammarskjölds väg 20, 751 85 Uppsala, Sweden
| | - Hirohisa Chiyoda
- Department of Molecular Pathophysiology, Institute of Advanced Medical Science, Nippon Medical School, 1-1-5 Sendagi Bunkyo-ku, Tokyo 113-8602, Japan; Department of Regenerative Medicine and Tissue Engineering, National Cerebral and Cardiovascular Center Research Institute, 6-1 Kishibe-Shinmachi, Suita, Osaka 564-8565, Japan
| | - Liqun He
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Dag Hammarskjölds väg 20, 751 85 Uppsala, Sweden
| | - Jianping Liu
- Department of Medicine Huddinge (MedH), Karolinska Institute, Campus Flemingsburg, Neo, Blickagången 16, 141 57 Huddinge, Sweden
| | - Koichi Kawakami
- Laboratory of Molecular and Developmental Biology, National Institute of Genetics, 1111 Yata, Mishima, Shizuoka 411-8540, Japan; Department of Genetics, SOKENDAI (The Graduate University for Advanced Studies), 1111 Yata, Mishima, Shizuoka 411-8540, Japan
| | - Naoki Mochizuki
- Department of Cell Biology, National Cerebral and Cardiovascular Center Research Institute, 6-1 Kishibe-Shinmachi, Suita, Osaka 564-8565, Japan
| | - Shigetomo Fukuhara
- Department of Molecular Pathophysiology, Institute of Advanced Medical Science, Nippon Medical School, 1-1-5 Sendagi Bunkyo-ku, Tokyo 113-8602, Japan
| | - Jaime Grutzendler
- Department of Neurology, Yale School of Medicine, New Haven, CT, USA
| | - Christer Betsholtz
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Dag Hammarskjölds väg 20, 751 85 Uppsala, Sweden; Department of Medicine Huddinge (MedH), Karolinska Institute, Campus Flemingsburg, Neo, Blickagången 16, 141 57 Huddinge, Sweden.
| |
Collapse
|
16
|
Cerebral Blood Flow in Healthy Subjects with Different Hypnotizability Scores. Brain Sci 2022; 12:brainsci12050558. [PMID: 35624945 PMCID: PMC9138886 DOI: 10.3390/brainsci12050558] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 04/11/2022] [Accepted: 04/26/2022] [Indexed: 12/19/2022] Open
Abstract
Hypnotizability is a cognitive trait associated with differences in the brachial artery flow-mediated dilatation of individuals with high hypnotizability (highs) and low hypnotizability scores (lows). The study investigated possible hypnotizability-related cerebrovascular differences. Among 24 healthy volunteers, the Stanford Hypnotic Susceptibility Scale Form A identified 13 medium-to-lows (med-lows), 11 medium-to-highs (med-highs), and 1 medium hypnotizable. Hypnotizability did not influence the significant changes produced by the trail making task (TMT), mental arithmetic task (MAT), hyperventilation (HVT), and rebreathing (RBT) on heart rate (HR), arterial blood pressure (ABP), and partial pressure of end-tidal CO2 (PETCO2), but moderated the correlations between the changes occurring during tasks with respect to basal conditions (Δ) in ABP and PETCO2 with middle cerebral artery flow velocity (MCAv). In HVT, med-lows exhibited a significant correlation between ΔMCAv and ΔPETCO2, and med-highs showed a significant correlation between ΔABP and ΔMCAv. Cerebrovascular reactivity (CVR) and conductance (ΔCVCi) were significantly correlated with ΔMCAv only in med-lows during HVT and RBT. For the first time, cerebrovascular reactivity related to hypnotizability was investigated, evidencing different correlations among hemodynamic variables in med-highs and med-lows.
Collapse
|
17
|
Al-Kawaz M, Cho SM, Gottesman RF, Suarez JI, Rivera-Lara L. Impact of Cerebral Autoregulation Monitoring in Cerebrovascular Disease: A Systematic Review. Neurocrit Care 2022; 36:1053-1070. [PMID: 35378665 DOI: 10.1007/s12028-022-01484-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Accepted: 03/01/2022] [Indexed: 12/16/2022]
Abstract
Cerebral autoregulation (CA) prevents brain injury by maintaining a relatively constant cerebral blood flow despite fluctuations in cerebral perfusion pressure. This process is disrupted consequent to various neurologic pathologic processes, which may result in worsening neurologic outcomes. Herein, we aim to highlight evidence describing CA changes and the impact of CA monitoring in patients with cerebrovascular disease, including ischemic stroke, intracerebral hemorrhage (ICH), and aneurysmal subarachnoid hemorrhage (aSAH). The study was preformed according to the Preferred Reporting Items for Systematic Reviews and Meta-Analysis guidelines. English language publications were identified through a systematic literature conducted in Ovid Medline, PubMed, and Embase databases. The search spanned the dates of each database's inception through January 2021. We selected case-control studies, cohort observational studies, and randomized clinical trials for adult patients (≥ 18 years) who were monitored with continuous metrics using transcranial Doppler, near-infrared spectroscopy, and intracranial pressure monitors. Of 2799 records screened, 48 studies met the inclusion criteria. There were 23 studies on ischemic stroke, 18 studies on aSAH, 5 studies on ICH, and 2 studies on systemic hypertension. CA impairment was reported after ischemic stroke but generally improved after tissue plasminogen activator administration and successful mechanical thrombectomy. Persistent impairment in CA was associated with hemorrhagic transformation, malignant cerebral edema, and need for hemicraniectomy. Studies that investigated large ICHs described bilateral CA impairment up to 12 days from the ictus, especially in the presence of small vessel disease. In aSAH, impairment of CA was associated with angiographic vasospasm, delayed cerebral ischemia, and poor functional outcomes at 6 months. This systematic review highlights the available evidence for CA disruption during cerebrovascular diseases and its possible association with long-term neurological outcome. CA may be disrupted even before acute stroke in patients with untreated chronic hypertension. Monitoring CA may help in establishing individualized management targets in patients with cerebrovascular disease.
Collapse
Affiliation(s)
- Mais Al-Kawaz
- Department of Neurosurgery, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Neurology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Sung-Min Cho
- Department of Neurology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Anesthesiology and Critical Care Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Rebecca F Gottesman
- Stroke Branch, National Institute of Neurological Disorders and Stroke Intramural Program, National Institutes of Health, Bethesda, MD, USA
| | - Jose I Suarez
- Department of Neurology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Anesthesiology and Critical Care Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Neurosurgery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Lucia Rivera-Lara
- Division of Stroke and Neurocritical Care, Stanford University, Palo Alto, CA, USA
| |
Collapse
|
18
|
Li T, Xu T, Zhao J, Gao H, Xie W. Depletion of iNOS-positive inflammatory cells decelerates neuronal degeneration and alleviates cerebral ischemic damage by suppressing the inflammatory response. Free Radic Biol Med 2022; 181:209-220. [PMID: 35150825 DOI: 10.1016/j.freeradbiomed.2022.02.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 01/25/2022] [Accepted: 02/08/2022] [Indexed: 01/05/2023]
Abstract
Ischemic stroke leads to neuronal damage and severe inflammation that activate iNOS expression in different cell types, especially inflammatory cells in the brain. It is shown that NO released from iNOS contributes to the pathological development of cerebral ischemia. However, the role of these iNOS-expressing inflammatory cells in ischemic stroke has not been fully elucidated. Our purpose is to test if ischemia-induced iNOS+ inflammatory cells may exaggerate cerebral inflammation to exacerbate neuronal deficit. We studied the dynamics of iNOS+ cells after stroke and found an early and sustained iNOS expression at lesion site. Since iNOS is highly expressed in inflammatory cells after injury, we depleted the iNOS + inflammatory cells via the selective scavenger GdCl3, and investigated its effect on stroke outcome, neuronal and vascular deficit, and inflammatory response. After GdCl3 treatment, half of iNOS+ inflammatory cells were depleted, including mainly activated microglia/macrophages and some astrocytes. Selective depletion of iNOS+ inflammatory cells resulted in a pronounced reduction in brain damage, resulting in improvement of motor ability. Histologic studies and in vivo two-photon imaging data revealed a slowdown of neuronal degeneration after the depletion of iNOS+ inflammatory cells. In contrast to iNOS inhibition alone, depletion of iNOS+ inflammatory cells profoundly altered the immune microenvironment profile, in addition to reducing NO production. qRT-PCR analysis showed that depletion of iNOS+ inflammatory cells significantly restrained the production of pro-inflammatory cytokines, which moderated the immune microenvironment at the lesion site. Taken together, our data demonstrate that depleting iNOS+ inflammatory cells prevents neuronal damage not only by inhibiting NO, but also importantly by suppressing the inflammatory response, which is beneficial to ischemic injury. These results provide evidence that iNOS+ inflammatory cells, as a vital source of pro-inflammatory cytokines, contribute to the development of ischemic damage and could be a potential therapeutic target for the treatment of ischemia.
Collapse
Affiliation(s)
- Ting Li
- Gansu Key Laboratory of Biomonitoring and Bioremediation for Environmental Pollution, School of Life Sciences, Lanzhou University, Lanzhou, 73000, China.
| | - Ting Xu
- Gansu Key Laboratory of Biomonitoring and Bioremediation for Environmental Pollution, School of Life Sciences, Lanzhou University, Lanzhou, 73000, China
| | - Jin Zhao
- Gansu Key Laboratory of Biomonitoring and Bioremediation for Environmental Pollution, School of Life Sciences, Lanzhou University, Lanzhou, 73000, China
| | - Hao Gao
- Gansu Key Laboratory of Biomonitoring and Bioremediation for Environmental Pollution, School of Life Sciences, Lanzhou University, Lanzhou, 73000, China
| | - Wenguang Xie
- Gansu Key Laboratory of Biomonitoring and Bioremediation for Environmental Pollution, School of Life Sciences, Lanzhou University, Lanzhou, 73000, China
| |
Collapse
|
19
|
Fukuda S, Nabetani M, Goldman RD, Shinomoto T, Kobata K, Yutaka N, Sano H. Experience of cases with inhaled nitric oxide and therapeutic hypothermia. Pediatr Int 2022; 64:e14901. [PMID: 34170585 DOI: 10.1111/ped.14901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 05/22/2021] [Accepted: 06/22/2021] [Indexed: 12/01/2022]
Abstract
BACKGROUND Neonates with hypoxic-ischemic encephalopathy (HIE) on therapeutic hypothermia (TH) therapy may show persistent pulmonary hypertension of the newborn (PPHN). In Japan, the reported mortality rate is lower than in the US, possibly due to treatment differences of newborns with moderate to severe HIE and PPHN. This study aimed to determine the feasibility and long-term outcomes of inhaled nitric oxide (iNO) and TH therapy in newborns with moderate to severe HIE and PPHN. METHODS This was a retrospective review of neonates with moderate to severe HIE that were treated with TH from 2008 to 2017 at a large medical center in Japan. We documented their long-term neurological prognosis, measuring their developmental and Gross Motor Function Classification System level at 18 months old. RESULTS A total of 37 neonates with moderate to severe HIE underwent TH therapy and six of them were started with iNO therapy for PPHN. iNO with TH was safely administered to all six newborns with moderate to severe HIE with PPHN. In two neonates TH was discontinued because of intraventricular hemorrhage (IVH) and severe hypotension. Neurological outcomes were similar in newborns who were treated with iNO and TH and those who were treated with TH alone. CONCLUSION These initial findings suggest that monitoring hematological and cardiovascular status is important with iNO for severe asphyxia in infants with PPHN. Safer and more feasible protocols are needed for when iNO and TH therapy are administered together.
Collapse
Affiliation(s)
- Sayaka Fukuda
- Department of Pediatrics, Yodogawa Christian Hospital, Osaka City, Osaka, Japan
| | - Makoto Nabetani
- Department of Pediatrics, Yodogawa Christian Hospital, Osaka City, Osaka, Japan
| | - Ran D Goldman
- Division of Pediatric Clinical Pharmacology, Department of Pediatrics, BC Children's Hospital, BC Children's Research Institute, University of British Columbia, Vancouver, BC, Canada
| | - Tadashi Shinomoto
- Department of Pediatrics, Takatsuki General Hospital, Takatsuki City, Osaka, Japan
| | - Keisuke Kobata
- Department of Pediatrics, Yodogawa Christian Hospital, Osaka City, Osaka, Japan
| | - Nanae Yutaka
- Department of Pediatrics, Yodogawa Christian Hospital, Osaka City, Osaka, Japan
| | - Hiroyuki Sano
- Department of Pediatrics, Yodogawa Christian Hospital, Osaka City, Osaka, Japan
| |
Collapse
|
20
|
Parmar S, Tadavarty R, Sastry BR. G-protein coupled receptors and synaptic plasticity in sleep deprivation. World J Psychiatry 2021; 11:954-980. [PMID: 34888167 PMCID: PMC8613756 DOI: 10.5498/wjp.v11.i11.954] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 06/05/2021] [Accepted: 09/19/2021] [Indexed: 02/06/2023] Open
Abstract
Insufficient sleep has been correlated to many physiological and psychoneurological disorders. Over the years, our understanding of the state of sleep has transcended from an inactive period of rest to a more active state involving important cellular and molecular processes. In addition, during sleep, electrophysiological changes also occur in pathways in specific regions of the mammalian central nervous system (CNS). Activity mediated synaptic plasticity in the CNS can lead to long-term and sometimes permanent strengthening and/or weakening synaptic strength affecting neuronal network behaviour. Memory consolidation and learning that take place during sleep cycles, can be affected by changes in synaptic plasticity during sleep disturbances. G-protein coupled receptors (GPCRs), with their versatile structural and functional attributes, can regulate synaptic plasticity in CNS and hence, may be potentially affected in sleep deprived conditions. In this review, we aim to discuss important functional changes that can take place in the CNS during sleep and sleep deprivation and how changes in GPCRs can lead to potential problems with therapeutics with pharmacological interventions.
Collapse
Affiliation(s)
- Shweta Parmar
- Department of Anesthesiology, Pharmacology and Therapeutics, The University of British Columbia, Vancouver V6T 1Z3, British Columbia, Canada
| | - Ramakrishna Tadavarty
- Department of Anesthesiology, Pharmacology and Therapeutics, The University of British Columbia, Vancouver V6T 1Z3, British Columbia, Canada
| | - Bhagavatula R Sastry
- Department of Anesthesiology, Pharmacology and Therapeutics, The University of British Columbia, Vancouver V6T 1Z3, British Columbia, Canada
| |
Collapse
|
21
|
Tamayo A, Siepmann T. Regulation of Blood Flow in the Cerebral Posterior Circulation by Parasympathetic Nerve Fibers: Physiological Background and Possible Clinical Implications in Patients With Vertebrobasilar Stroke. Front Neurol 2021; 12:660373. [PMID: 34777191 PMCID: PMC8585859 DOI: 10.3389/fneur.2021.660373] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 09/23/2021] [Indexed: 01/14/2023] Open
Abstract
Posterior circulation involves the vertebrobasilar arteries, which supply oxygen and glucose to vital human brainstem structures and other areas. This complex circulatory- perfusion system is not homogenous throughout the day; rather, its hemodynamic changes rely on physiological demands, ensuring brainstem perfusion. This dynamic autoregulatory pattern maintains cerebral perfusion during blood pressure changes. Accumulative evidence suggests that activity within the autonomic nervous system is involved in the regulation of cerebral blood flow. Neither the sympathetic nor parasympathetic nervous systems work independently. Functional studies have shown a tight and complicated cross talk between these systems. In pathological processes where sympathetic stimulation is present, systemic vasoconstriction is followed, representing the most important CNS parasympathetic trigger that will promote local vasodilation. Stroke is a clear example of this process. The posterior circulation is affected in 30% of strokes, causing high morbidity and mortality outcomes. Currently, the management of ischemic stroke is focused on thrombolytic treatment and endovascular thrombectomy within an overall tight 4.5 to 6 h ischemic time window. Therefore, the autonomic nervous system could represent a potential therapeutic target to modulate reperfusion after cerebral ischemia through vasodilation, which could potentially decrease infarct size and increase the thrombolytic therapeutic ischemic window. In addition, shifting the autonomic nervous system balance toward its parasympathetic branch has shown to enhance neurogenesis and decrease local inflammation. Regretfully, the vast majority of animal models and human research on neuromodulation during brain ischemia have been focused on anterior circulation with disappointing results. In addition, the source of parasympathetic inputs in the vertebrobasilar system in humans is poorly understood, substantiating a gap and controversy in this area. Here, we reviewed current available literature regarding the parasympathetic vascular function and challenges of its stimulation in the vertebrobasilar system.
Collapse
Affiliation(s)
- Arturo Tamayo
- The Max Rady Faculty of Health Sciences, Department of Medicine, Section of Neurology, WRHA, Winnipeg and Brandon Regional Health Centre, University of Manitoba, Winnipeg, MB, Canada
- Department of Health Care Sciences, Center for Clinical Research and Management Education, Dresden International University, Dresden, Germany
| | - Timo Siepmann
- Department of Health Care Sciences, Center for Clinical Research and Management Education, Dresden International University, Dresden, Germany
- Department of Neurology, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| |
Collapse
|
22
|
Claassen JAHR, Thijssen DHJ, Panerai RB, Faraci FM. Regulation of cerebral blood flow in humans: physiology and clinical implications of autoregulation. Physiol Rev 2021; 101:1487-1559. [PMID: 33769101 PMCID: PMC8576366 DOI: 10.1152/physrev.00022.2020] [Citation(s) in RCA: 457] [Impact Index Per Article: 114.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Brain function critically depends on a close matching between metabolic demands, appropriate delivery of oxygen and nutrients, and removal of cellular waste. This matching requires continuous regulation of cerebral blood flow (CBF), which can be categorized into four broad topics: 1) autoregulation, which describes the response of the cerebrovasculature to changes in perfusion pressure; 2) vascular reactivity to vasoactive stimuli [including carbon dioxide (CO2)]; 3) neurovascular coupling (NVC), i.e., the CBF response to local changes in neural activity (often standardized cognitive stimuli in humans); and 4) endothelium-dependent responses. This review focuses primarily on autoregulation and its clinical implications. To place autoregulation in a more precise context, and to better understand integrated approaches in the cerebral circulation, we also briefly address reactivity to CO2 and NVC. In addition to our focus on effects of perfusion pressure (or blood pressure), we describe the impact of select stimuli on regulation of CBF (i.e., arterial blood gases, cerebral metabolism, neural mechanisms, and specific vascular cells), the interrelationships between these stimuli, and implications for regulation of CBF at the level of large arteries and the microcirculation. We review clinical implications of autoregulation in aging, hypertension, stroke, mild cognitive impairment, anesthesia, and dementias. Finally, we discuss autoregulation in the context of common daily physiological challenges, including changes in posture (e.g., orthostatic hypotension, syncope) and physical activity.
Collapse
Affiliation(s)
- Jurgen A H R Claassen
- Department of Geriatrics, Radboud University Medical Center, Donders Institute for Brain, Cognition, and Behaviour, Nijmegen, The Netherlands
| | - Dick H J Thijssen
- Department of Physiology, Radboud Institute for Health Sciences, Nijmegen, The Netherlands
- Research Institute for Sport and Exercise Sciences, Liverpool John Moores University, Liverpool, United Kingdom
| | - Ronney B Panerai
- Department of Cardiovascular Sciences, University of Leicester, Leicester, United Kingdom
- >National Institute for Health Research Leicester Biomedical Research Centre, University of Leicester, Leicester, United Kingdom
| | - Frank M Faraci
- Departments of Internal Medicine, Neuroscience, and Pharmacology, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| |
Collapse
|
23
|
Evans LE, Taylor JL, Smith CJ, Pritchard HAT, Greenstein AS, Allan SM. Cardiovascular co-morbidities, inflammation and cerebral small vessel disease. Cardiovasc Res 2021; 117:2575-2588. [PMID: 34499123 DOI: 10.1093/cvr/cvab284] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Indexed: 12/15/2022] Open
Abstract
Cerebral small vessel disease (cSVD) is the most common cause of vascular cognitive impairment and affects all levels of the brain's vasculature. Features include diverse structural and functional changes affecting small arteries and capillaries that lead to a decline in cerebral perfusion. Due to an aging population, incidence of cerebral small vessel disease (cSVD) is continually rising. Despite its prevalence and its ability to cause multiple debilitating illnesses, such as stroke and dementia, there are currently no therapeutic strategies for the treatment of cSVD. In the healthy brain, interactions between neuronal, vascular and inflammatory cells are required for normal functioning. When these interactions are disturbed, chronic pathological inflammation can ensue. The interplay between cSVD and inflammation has attracted much recent interest and this review discusses chronic cardiovascular diseases, particularly hypertension, and explores how the associated inflammation may impact on the structure and function of the small arteries of the brain in cSVD. Molecular approaches in animal studies are linked to clinical outcomes in patients and novel hypotheses regarding inflammation and cSVD are proposed that will hopefully stimulate further discussion and study in this important area.
Collapse
Affiliation(s)
- Lowri E Evans
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom.,Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance & University of Manchester, Manchester, UK
| | - Jade L Taylor
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom.,Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance & University of Manchester, Manchester, UK
| | - Craig J Smith
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom.,Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance & University of Manchester, Manchester, UK.,Manchester Centre for Clinical Neurosciences, Manchester Academic Health Science Centre, Salford Royal Hospital, Manchester Academic Health Sciences Centre (MAHSC)
| | - Harry A T Pritchard
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom.,Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance & University of Manchester, Manchester, UK
| | - Adam S Greenstein
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom.,Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance & University of Manchester, Manchester, UK
| | - Stuart M Allan
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance & University of Manchester, Manchester, UK.,Division of Neuroscience and Experimental Psychology, The University of Manchester, Manchester, UK
| |
Collapse
|
24
|
Kumar G, Mukherjee S, Kumar S, Patnaik R. Rapid Determination of Nitrate in Brain Regions and Cerebrospinal Fluid of Transient Bilateral Common Carotid Artery Occlusion Rat Model by HPLC–UV. PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES, INDIA SECTION A: PHYSICAL SCIENCES 2021; 91:361-368. [DOI: 10.1007/s40010-020-00666-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Revised: 01/29/2020] [Accepted: 02/03/2020] [Indexed: 10/25/2022]
|
25
|
Haraikawa M, Kudo H, Shibuya T, Kogure Y, Takase M, Inage H, Yokota T, Htun H, Tagaya E, Fan R, Houshito H, Taniguchi G, Sakamoto K, Nagahara A. Efficacy of CO₂ Infusion for Preoperative Computed Tomographic Angiography with Computed Tomographic Colonography. Med Sci Monit 2021; 27:e931055. [PMID: 33993185 PMCID: PMC8135977 DOI: 10.12659/msm.931055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND Computed tomographic colonography (CTC) is useful for patients for whom colonoscopy may be difficult to perform and is widely employed to examine the vasculature prior to colorectal cancer surgery. Computed tomographic angiography (CTA) was shown to be beneficial intraoperatively to manipulate blood vessels and prevent vascular injury. Three-dimensional (3D)-CTA combined with CTC (3D-CTA with CTC) is useful for preoperative evaluations of the anatomy of mesenteric vessels, colon, and lymph nodes. We observed that when the intestine was dilated with carbon dioxide (CO₂), the arteriovenous delineation was often more pronounced than without CO₂. To clarify the effects of gas injection with and without CO₂ on hemodynamics and vascular passage, we compared the effect of contrast for blood vessels. MATERIAL AND METHODS Thirty patients with resectable colorectal cancer who underwent a preoperative CT examination at our institution from January to October 2019 were study participants. Of these, 15 underwent 3D-CTA and 15 had 3D-CTA with CTC. Three board-certified radiologists independently and blindly evaluated 18 blood vessels. CT values for each blood vessel were measured on each image. RESULTS CT values for 3D-CTA with CTC were significantly higher with CO₂ than without CO₂. The quality of 3D-CTA with CTC images for visualization of blood vessels was also significantly greater than that of 3D-CTA, especially those of arterial and intramesenteric venous systems. CONCLUSIONS Based on the higher image quality and CT values obtained by 3D-CTA with CTC for vessels, compared with by 3D-CTA imaging, 3D-CTA with CTC imaging might be useful in evaluating colorectal cancers.
Collapse
Affiliation(s)
- Mayuko Haraikawa
- Department of Gastroenterology, Juntendo University School of Medicine, Tokyo, Japan
| | - Hikaru Kudo
- Department of Radiology, Juntendo University School of Medicine, Tokyo, Japan
| | - Tomoyoshi Shibuya
- Department of Gastroenterology, Juntendo University School of Medicine, Tokyo, Japan
| | - Yosuke Kogure
- Department of Radiology, Juntendo University School of Medicine, Tokyo, Japan
| | - Makoto Takase
- Department of Radiology, Juntendo University School of Medicine, Tokyo, Japan
| | - Hidekazu Inage
- Department of Radiology, Juntendo University School of Medicine, Tokyo, Japan
| | - Takuya Yokota
- Department of Radiology, Juntendo University School of Medicine, Tokyo, Japan
| | - Hanni Htun
- Department of Radiology, Juntendo University School of Medicine, Tokyo, Japan
| | - Eita Tagaya
- Department of Radiology, Juntendo University School of Medicine, Tokyo, Japan
| | - Ruiheng Fan
- Department of Radiology, Juntendo University School of Medicine, Tokyo, Japan
| | - Haruyoshi Houshito
- Department of Radiology, Juntendo University School of Medicine, Tokyo, Japan
| | - Gentaro Taniguchi
- Department of Gastroenterology, Juntendo University School of Medicine, Tokyo, Japan
| | - Kazuhiro Sakamoto
- Department of Coloproctological Surgery, Juntendo University School of Medicine, Tokyo, Japan
| | - Akihito Nagahara
- Department of Gastroenterology, Juntendo University School of Medicine, Tokyo, Japan
| |
Collapse
|
26
|
Chen JJ, Gauthier CJ. The Role of Cerebrovascular-Reactivity Mapping in Functional MRI: Calibrated fMRI and Resting-State fMRI. Front Physiol 2021; 12:657362. [PMID: 33841190 PMCID: PMC8027080 DOI: 10.3389/fphys.2021.657362] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Accepted: 03/02/2021] [Indexed: 12/14/2022] Open
Abstract
Task and resting-state functional MRI (fMRI) is primarily based on the same blood-oxygenation level-dependent (BOLD) phenomenon that MRI-based cerebrovascular reactivity (CVR) mapping has most commonly relied upon. This technique is finding an ever-increasing role in neuroscience and clinical research as well as treatment planning. The estimation of CVR has unique applications in and associations with fMRI. In particular, CVR estimation is part of a family of techniques called calibrated BOLD fMRI, the purpose of which is to allow the mapping of cerebral oxidative metabolism (CMRO2) using a combination of BOLD and cerebral-blood flow (CBF) measurements. Moreover, CVR has recently been shown to be a major source of vascular bias in computing resting-state functional connectivity, in much the same way that it is used to neutralize the vascular contribution in calibrated fMRI. Furthermore, due to the obvious challenges in estimating CVR using gas challenges, a rapidly growing field of study is the estimation of CVR without any form of challenge, including the use of resting-state fMRI for that purpose. This review addresses all of these aspects in which CVR interacts with fMRI and the role of CVR in calibrated fMRI, provides an overview of the physiological biases and assumptions underlying hypercapnia-based CVR and calibrated fMRI, and provides a view into the future of non-invasive CVR measurement.
Collapse
Affiliation(s)
- J Jean Chen
- Baycrest Centre for Geriatric Care, Rotman Research Institute, Toronto, ON, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Claudine J Gauthier
- Department of Physics, Concordia University, Montreal, QC, Canada.,Montreal Heart Institute, Montreal, QC, Canada
| |
Collapse
|
27
|
Leon RL, Ortigoza EB, Ali N, Angelis D, Wolovits JS, Chalak LF. Cerebral Blood Flow Monitoring in High-Risk Fetal and Neonatal Populations. Front Pediatr 2021; 9:748345. [PMID: 35087771 PMCID: PMC8787287 DOI: 10.3389/fped.2021.748345] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 12/15/2021] [Indexed: 12/18/2022] Open
Abstract
Cerebrovascular pressure autoregulation promotes stable cerebral blood flow (CBF) across a range of arterial blood pressures. Cerebral autoregulation (CA) is a developmental process that reaches maturity around term gestation and can be monitored prenatally with both Doppler ultrasound and magnetic resonance imaging (MRI) techniques. Postnatally, there are key advantages and limitations to assessing CA with Doppler ultrasound, MRI, and near-infrared spectroscopy. Here we review these CBF monitoring techniques as well as their application to both fetal and neonatal populations at risk of perturbations in CBF. Specifically, we discuss CBF monitoring in fetuses with intrauterine growth restriction, anemia, congenital heart disease, neonates born preterm and those with hypoxic-ischemic encephalopathy. We conclude the review with insights into the future directions in this field with an emphasis on collaborative science and precision medicine approaches.
Collapse
Affiliation(s)
- Rachel L Leon
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Eric B Ortigoza
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Noorjahan Ali
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Dimitrios Angelis
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Joshua S Wolovits
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Lina F Chalak
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, United States.,Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, United States
| |
Collapse
|
28
|
Latest Insights into the Pathophysiology of Migraine: the ATP-Sensitive Potassium Channels. Curr Pain Headache Rep 2020; 24:77. [DOI: 10.1007/s11916-020-00911-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/15/2020] [Indexed: 12/15/2022]
|
29
|
Arginine and Arginine/ADMA Ratio Predict 90-Day Mortality in Patients with Out-of-Hospital Cardiac Arrest-Results from the Prospective, Observational COMMUNICATE Trial. J Clin Med 2020; 9:jcm9123815. [PMID: 33255752 PMCID: PMC7760544 DOI: 10.3390/jcm9123815] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 11/08/2020] [Accepted: 11/19/2020] [Indexed: 02/07/2023] Open
Abstract
(1) Background: In patients with shock, the L-arginine nitric oxide pathway is activated, causing an elevation of nitric oxide, asymmetric dimethylarginine (ADMA) and symmetric dimethylarginine (SDMA) levels. Whether these metabolites provide prognostic information in patients after out-of-hospital cardiac arrest (OHCA) remains unclear. (2) Methods: We prospectively included OHCA patients, recorded clinical parameters and measured plasma ADMA, SDMA and Arginine levels by liquid chromatography tandem mass spectrometry (LC-MS). The primary endpoint was 90-day mortality. (3) Results: Of 263 patients, 130 (49.4%) died within 90 days after OHCA. Compared to survivors, non-survivors had significantly higher levels of ADMA and lower Arginine and Arginine/ADMA ratios in univariable regression analyses. Arginine levels and Arginine/ADMA ratio were significantly associated with 90-day mortality (OR 0.51 (95%CI 0.34 to 0.76), p < 0.01 and OR 0.40 (95%CI 0.26 to 0.61), p < 0.001, respectively). These associations remained significant in several multivariable models. Arginine/ADMA ratio had the highest predictive value with an area under the curve (AUC) of 0.67 for 90-day mortality. Results for secondary outcomes were similar with significant associations with in-hospital mortality and neurological outcome. (4) Conclusion: Arginine and Arginine/ADMA ratio were independently associated with 90-day mortality and other adverse outcomes in patients after OHCA. Whether therapeutic modification of the L-arginine-nitric oxide pathway has the potential to improve outcome should be evaluated.
Collapse
|
30
|
Mitra S, Kaushik N, Moon IS, Choi EH, Kaushik NK. Utility of Reactive Species Generation in Plasma Medicine for Neuronal Development. Biomedicines 2020; 8:E348. [PMID: 32932745 PMCID: PMC7555638 DOI: 10.3390/biomedicines8090348] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 09/07/2020] [Accepted: 09/09/2020] [Indexed: 12/13/2022] Open
Abstract
Reactive oxygen species (ROS) are critical signaling molecules for neuronal physiology that stimulate growth and development and play vital roles in several pathways when in a balanced state, but they cause neurodegeneration when unbalanced. As ROS levels above a certain threshold cause the activation of the autophagy system, moderate levels of ROS can be used as treatment strategies. Currently, such treatments are used together with low-level laser or photodynamic therapies, photo-bio modulation, or infrared treatments, in different chronic diseases but not in the treatment of neurodegeneration. Recently, non-thermal plasma has been successfully used in biomedical applications and treatments, and beneficial effects such as differentiation, cell growth, and proliferation, stimulation of ROS based pathways have been observed. Besides the activation of a wide range of biological signaling pathways by generating ROS, plasma application can be an effective treatment in neuronal regeneration, as well as in neuronal diseases. In this review, we summarize the generation and role of ROS in neurons and provide critical insights into their potential benefits on neurons. We also discuss the underlying mechanisms of ROS on neuronal development. Regarding clinical applications, we focus on ROS-based neuronal growth and regeneration strategies and in the usage of non-thermal plasma in neuronal and CNS injury treatments.
Collapse
Affiliation(s)
- Sarmistha Mitra
- Plasma Bioscience Research Center/Applied Plasma Medicine Center, Department of Electrical and Biological Physics, Kwangwoon University, Seoul 01897, Korea;
- Department of Anatomy, Dongguk University College of Medicine, Gyeongju 38066, Korea;
| | - Neha Kaushik
- Department of Biotechnology, University of Suwon, Hwaseong 18323, Korea;
| | - Il Soo Moon
- Department of Anatomy, Dongguk University College of Medicine, Gyeongju 38066, Korea;
| | - Eun Ha Choi
- Plasma Bioscience Research Center/Applied Plasma Medicine Center, Department of Electrical and Biological Physics, Kwangwoon University, Seoul 01897, Korea;
| | - Nagendra Kumar Kaushik
- Plasma Bioscience Research Center/Applied Plasma Medicine Center, Department of Electrical and Biological Physics, Kwangwoon University, Seoul 01897, Korea;
| |
Collapse
|
31
|
Thaung Zaw JJ, Howe PR, Wong RH. Long-term effects of resveratrol on cognition, cerebrovascular function and cardio-metabolic markers in postmenopausal women: A 24-month randomised, double-blind, placebo-controlled, crossover study. Clin Nutr 2020; 40:820-829. [PMID: 32900519 DOI: 10.1016/j.clnu.2020.08.025] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 07/08/2020] [Accepted: 08/20/2020] [Indexed: 12/14/2022]
Abstract
Ageing and menopause contribute to endothelial dysfunction, causing impaired cerebral perfusion, which is in turn associated with accelerated cognitive decline. In a 14-week pilot study, we showed that supplementation with low-dose resveratrol, a phytoestrogen that can enhance endothelial function, improved cerebrovascular and cognitive functions in postmenopausal women. We sought to confirm these benefits in a larger, longer-term trial. A 24-month randomized, placebo-controlled crossover trial was undertaken in 125 postmenopausal women, aged 45-85 years, who took 75 mg trans-resveratrol or placebo twice-daily for 12 months and then crossover to the alternative treatment for another 12 months. We evaluated within individual differences between each treatment period in measures of cognition (primary outcome), cerebrovascular function in the middle cerebral artery (cerebral blood flow velocity: CBFV, cerebrovascular responsiveness: CVR) and cardio-metabolic markers as secondary outcomes. Subgroup analyses examined effects of resveratrol by life stages. Compared to placebo, resveratrol supplementation resulted a significant 33% improvement in overall cognitive performance (Cohen's d = 0.170, P = 0.005). Women ≥65 years of age showed a relative improvement in verbal memory with resveratrol compared to those younger than 65 years. Furthermore, resveratrol improved secondary outcomes including resting mean CBFV (d = 0.275, P = 0.001), CVR to hypercapnia (d = 0.307, P = 0.027), CVR to cognitive stimuli (d = 0.259, P = 0.032), fasting insulin (d = 0.174, P = 0.025) and insulin resistance index (d = 0.102, P = 0.034). Regular supplementation with low-dose resveratrol can enhance cognition, cerebrovascular function and insulin sensitivity in postmenopausal women. This may translate into a slowing of the accelerated cognitive decline due to ageing and menopause, especially in late-life women. Further studies are warranted to observe whether these cognitive benefits of resveratrol can reduce the risk of dementia.
Collapse
Affiliation(s)
- Jay Jay Thaung Zaw
- University of Newcastle, School of Biomedical Sciences and Pharmacy, Callaghan, NSW, 2308, Australia
| | - Peter Rc Howe
- University of Newcastle, School of Biomedical Sciences and Pharmacy, Callaghan, NSW, 2308, Australia; University of Southern Queensland, Institute for Resilient Regions, Springfield Central, QLD, 4300, Australia; University of South Australia, School of Health Sciences, Adelaide, SA, 5000, Australia
| | - Rachel Hx Wong
- University of Newcastle, School of Biomedical Sciences and Pharmacy, Callaghan, NSW, 2308, Australia; University of Southern Queensland, Institute for Resilient Regions, Springfield Central, QLD, 4300, Australia.
| |
Collapse
|
32
|
Pan P, Xu L, Zhang H, Liu Y, Lu X, Chen G, Tang H, Wu J. A Review of Hematoma Components Clearance Mechanism After Subarachnoid Hemorrhage. Front Neurosci 2020; 14:685. [PMID: 32733194 PMCID: PMC7358443 DOI: 10.3389/fnins.2020.00685] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Accepted: 06/04/2020] [Indexed: 12/18/2022] Open
Abstract
Subarachnoid hemorrhage (SAH) is a complicated clinical syndrome, which is caused by several kinds of cerebrovascular disorders, with high morbidity, disability and mortality rate. In recent years, several studies have shown that early brain injury (EBI) is an important factor leading to the poor prognosis of SAH. A major cause of EBI has been attributed that hematoma components invade into the brain parenchyma, resulting in neuronal cell death. Therefore, the clearance of hematoma components is essential in the clinical outcome of patients after SAH. Here, in the review, we provide a summary of the current known hematoma components clearance mechanisms and simultaneously propose a new hypothesis for hematoma components clearance.
Collapse
Affiliation(s)
- Pengjie Pan
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Li Xu
- Intensive Care Unit of Department of Anesthesiology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Hongrong Zhang
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yuan Liu
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Xiaocheng Lu
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Gang Chen
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Hailiang Tang
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Jiang Wu
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
33
|
Hugelshofer M, Buzzi RM, Schaer CA, Richter H, Akeret K, Anagnostakou V, Mahmoudi L, Vaccani R, Vallelian F, Deuel JW, Kronen PW, Kulcsar Z, Regli L, Baek JH, Pires IS, Palmer AF, Dennler M, Humar R, Buehler PW, Kircher PR, Keller E, Schaer DJ. Haptoglobin administration into the subarachnoid space prevents hemoglobin-induced cerebral vasospasm. J Clin Invest 2020; 129:5219-5235. [PMID: 31454333 DOI: 10.1172/jci130630] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Accepted: 08/20/2019] [Indexed: 12/13/2022] Open
Abstract
Delayed ischemic neurological deficit (DIND) is a major driver of adverse outcomes in patients with aneurysmal subarachnoid hemorrhage (aSAH), defining an unmet need for therapeutic development. Cell-free hemoglobin that is released from erythrocytes into the cerebrospinal fluid (CSF) is suggested to cause vasoconstriction and neuronal toxicity, and correlates with the occurrence of DIND. Cell-free hemoglobin in the CSF of patients with aSAH disrupted dilatory NO signaling ex vivo in cerebral arteries, which shifted vascular tone balance from dilation to constriction. We found that selective removal of hemoglobin from patient CSF with a haptoglobin-affinity column or its sequestration in a soluble hemoglobin-haptoglobin complex was sufficient to restore physiological vascular responses. In a sheep model, administration of haptoglobin into the CSF inhibited hemoglobin-induced cerebral vasospasm and preserved vascular NO signaling. We identified 2 pathways of hemoglobin delocalization from CSF into the brain parenchyma and into the NO-sensitive compartment of small cerebral arteries. Both pathways were critical for hemoglobin toxicity and were interrupted by the large hemoglobin-haptoglobin complex that inhibited spatial requirements for hemoglobin reactions with NO in tissues. Collectively, our data show that compartmentalization of hemoglobin by haptoglobin provides a novel framework for innovation aimed at reducing hemoglobin-driven neurological damage after subarachnoid bleeding.
Collapse
Affiliation(s)
- Michael Hugelshofer
- Department of Neurosurgery, Clinical Neuroscience Center, University Hospital and University of Zurich, Zurich, Switzerland
| | - Raphael M Buzzi
- Division of Internal Medicine, University Hospital of Zurich, Zurich, Switzerland
| | - Christian A Schaer
- Division of Internal Medicine, University Hospital of Zurich, Zurich, Switzerland
| | - Henning Richter
- Clinic for Diagnostic Imaging, Department of Clinical Diagnostics and Services, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
| | - Kevin Akeret
- Department of Neurosurgery, Clinical Neuroscience Center, University Hospital and University of Zurich, Zurich, Switzerland
| | - Vania Anagnostakou
- Department of Neuroradiology, Clinical Neuroscience Center, University Hospital and University of Zurich, Zurich, Switzerland
| | - Leila Mahmoudi
- Division of Internal Medicine, University Hospital of Zurich, Zurich, Switzerland.,Department of Chemistry and Applied Biosciences, ETH Zurich, Zurich, Switzerland
| | - Raphael Vaccani
- Division of Internal Medicine, University Hospital of Zurich, Zurich, Switzerland
| | - Florence Vallelian
- Division of Internal Medicine, University Hospital of Zurich, Zurich, Switzerland
| | - Jeremy W Deuel
- Division of Internal Medicine, University Hospital of Zurich, Zurich, Switzerland
| | - Peter W Kronen
- Veterinary Anaesthesia Services - International, Winterthur, Switzerland.,Center for Applied Biotechnology and Molecular Medicine (CABMM), University of Zurich, Zurich, Switzerland
| | - Zsolt Kulcsar
- Department of Neuroradiology, Clinical Neuroscience Center, University Hospital and University of Zurich, Zurich, Switzerland
| | - Luca Regli
- Department of Neurosurgery, Clinical Neuroscience Center, University Hospital and University of Zurich, Zurich, Switzerland
| | - Jin Hyen Baek
- Center of Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland, USA
| | - Ivan S Pires
- William G. Lowrie Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, Ohio, USA
| | - Andre F Palmer
- William G. Lowrie Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, Ohio, USA
| | - Matthias Dennler
- Clinic for Diagnostic Imaging, Department of Clinical Diagnostics and Services, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
| | - Rok Humar
- Division of Internal Medicine, University Hospital of Zurich, Zurich, Switzerland
| | - Paul W Buehler
- Center of Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland, USA
| | - Patrick R Kircher
- Clinic for Diagnostic Imaging, Department of Clinical Diagnostics and Services, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
| | - Emanuela Keller
- Neurointensive Care Unit, University Hospital of Zurich, Zurich, Switzerland
| | - Dominik J Schaer
- Division of Internal Medicine, University Hospital of Zurich, Zurich, Switzerland
| |
Collapse
|
34
|
Wang Q, Sun H, Qi X, Zhou M. eNOS rs2070744 polymorphism might influence predisposition to hemorrhagic cerebral vascular diseases in East Asians: A meta-analysis. Brain Behav 2020; 10:e01538. [PMID: 32220011 PMCID: PMC7218252 DOI: 10.1002/brb3.1538] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 01/05/2020] [Accepted: 01/08/2020] [Indexed: 12/18/2022] Open
Abstract
INTRODUCTION Endothelial nitric oxide synthase (eNOS) polymorphisms might influence predisposition to hemorrhagic cerebral vascular diseases, but the results of already published studies regarding relationship between eNOS polymorphisms and hemorrhagic cerebral vascular diseases were still controversial. METHODS The authors performed this meta-analysis to estimate relationship between eNOS polymorphisms and hemorrhagic cerebral vascular diseases in a larger pooled population by combing the results of already published related studies. The authors searched Pubmed, Embase, Web of Science, and CNKI for already published studies. RESULTS Eighteen already published studies were pooled analyzed in this meta-analysis. The pooled meta-analyses results showed that eNOS rs2070744 polymorphism was significantly associated with predisposition to hemorrhagic cerebral vascular diseases in East Asians (dominant comparison: OR = 0.77, p = .01; overdominant comparison: OR = 1.24, p = .04; allele comparison: OR = 0.78, p = .006) Nevertheless, the pooled meta-analyses did not reveal any positive results for eNOS rs1799983 and rs869109213 polymorphisms. CONCLUSIONS This meta-analysis suggested that eNOS rs2070744 polymorphism, but not rs1799983 and rs869109213 polymorphisms, might influence predisposition to hemorrhagic cerebral vascular diseases in East Asians.
Collapse
Affiliation(s)
- Qiuling Wang
- Department of Nursing, Changyi Peoples Hospital, Changyi, China
| | - Hongri Sun
- Department of Neurology, Changyi Peoples Hospital, Changyi, China
| | - Xiaoguang Qi
- Department of General Surgery, Changyi Peoples Hospital, Changyi, China
| | - Minfeng Zhou
- Department of General practice, Zhuji Affiliated Hospital of Shaoxing University, Zhuji, China
| |
Collapse
|
35
|
Kauser NI, Weisel M, Zhong YL, Lo MMC, Ali A. Calcium Dialkylamine Diazeniumdiolates: Synthesis, Stability, and Nitric Oxide Generation. J Org Chem 2020; 85:4807-4812. [DOI: 10.1021/acs.joc.0c00020] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Nabeelah I. Kauser
- Department of Discovery Chemistry, Merck & Co., Inc., Kenilworth, New Jersey 07033, United States
| | - Mark Weisel
- Department of Process Research and Development, Merck & Co., Inc., Kenilworth, New Jersey 07033, United States
| | - Yong-Li Zhong
- Department of Process Research and Development, Merck & Co., Inc., Kenilworth, New Jersey 07033, United States
| | - Michael Man-Chu Lo
- Department of Discovery Chemistry, Merck & Co., Inc., Kenilworth, New Jersey 07033, United States
| | - Amjad Ali
- Department of Discovery Chemistry, Merck & Co., Inc., Kenilworth, New Jersey 07033, United States
| |
Collapse
|
36
|
Robicsek SA, Bhattacharya A, Rabai F, Shukla K, Doré S. Blood-Related Toxicity after Traumatic Brain Injury: Potential Targets for Neuroprotection. Mol Neurobiol 2019; 57:159-178. [PMID: 31617072 DOI: 10.1007/s12035-019-01766-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 08/29/2019] [Indexed: 02/07/2023]
Abstract
Emergency visits, hospitalizations, and deaths due to traumatic brain injury (TBI) have increased significantly over the past few decades. While the primary early brain trauma is highly deleterious to the brain, the secondary injury post-TBI is postulated to significantly impact mortality. The presence of blood, particularly hemoglobin, and its breakdown products and key binding proteins and receptors modulating their clearance may contribute significantly to toxicity. Heme, hemin, and iron, for example, cause membrane lipid peroxidation, generate reactive oxygen species, and sensitize cells to noxious stimuli resulting in edema, cell death, and increased morbidity and mortality. A wide range of other mechanisms such as the immune system play pivotal roles in mediating secondary injury. Effective scavenging of all of these pro-oxidant and pro-inflammatory metabolites as well as controlling maladaptive immune responses is essential for limiting toxicity and secondary injury. Hemoglobin metabolism is mediated by key molecules such as haptoglobin, heme oxygenase, hemopexin, and ferritin. Genetic variability and dysfunction affecting these pathways (e.g., haptoglobin and heme oxygenase expression) have been implicated in the difference in susceptibility of individual patients to toxicity and may be target pathways for potential therapeutic interventions in TBI. Ongoing collaborative efforts are required to decipher the complexities of blood-related toxicity in TBI with an overarching goal of providing effective treatment options to all patients with TBI.
Collapse
Affiliation(s)
- Steven A Robicsek
- Department of Anesthesiology, Center for Translational Research in Neurodegenerative Disease and McKnight Brain Institute, College of Medicine, University of Florida, 1275 Center Drive, Biomed Sci J493, Gainesville, FL, 32610, USA. .,Departments of Neurosurgery, Neuroscience, College of Medicine, University of Florida, Gainesville, FL, USA.
| | - Ayon Bhattacharya
- Department of Anesthesiology, Center for Translational Research in Neurodegenerative Disease and McKnight Brain Institute, College of Medicine, University of Florida, 1275 Center Drive, Biomed Sci J493, Gainesville, FL, 32610, USA.,Department of Pharmacology, KPC Medical College, West Bengal University of Health Sciences, Kolkata, West Bengal, India
| | - Ferenc Rabai
- Department of Anesthesiology, Center for Translational Research in Neurodegenerative Disease and McKnight Brain Institute, College of Medicine, University of Florida, 1275 Center Drive, Biomed Sci J493, Gainesville, FL, 32610, USA
| | - Krunal Shukla
- Department of Anesthesiology, Center for Translational Research in Neurodegenerative Disease and McKnight Brain Institute, College of Medicine, University of Florida, 1275 Center Drive, Biomed Sci J493, Gainesville, FL, 32610, USA
| | - Sylvain Doré
- Department of Anesthesiology, Center for Translational Research in Neurodegenerative Disease and McKnight Brain Institute, College of Medicine, University of Florida, 1275 Center Drive, Biomed Sci J493, Gainesville, FL, 32610, USA. .,Departments of Neurology, Psychiatry, Pharmaceutics and Neuroscience, College of Medicine, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
37
|
Abstract
Haemoglobin is released into the CNS during the breakdown of red blood cells after intracranial bleeding. Extracellular free haemoglobin is directly neurotoxic. Haemoglobin scavenging mechanisms clear haemoglobin and reduce toxicity; these mechanisms include erythrophagocytosis, haptoglobin binding of haemoglobin, haemopexin binding of haem and haem oxygenase breakdown of haem. However, the capacity of these mechanisms is limited in the CNS, and they easily become overwhelmed. Targeting of haemoglobin toxicity and scavenging is, therefore, a rational therapeutic strategy. In this Review, we summarize the neurotoxic mechanisms of extracellular haemoglobin and the peculiarities of haemoglobin scavenging pathways in the brain. Evidence for a role of haemoglobin toxicity in neurological disorders is discussed, with a focus on subarachnoid haemorrhage and intracerebral haemorrhage, and emerging treatment strategies based on the molecular pathways involved are considered. By focusing on a fundamental biological commonality between diverse neurological conditions, we aim to encourage the application of knowledge of haemoglobin toxicity and scavenging across various conditions. We also hope that the principles highlighted will stimulate research to explore the potential of the pathways discussed. Finally, we present a consensus opinion on the research priorities that will help to bring about clinical benefits.
Collapse
|
38
|
Fan JL, O'Donnell T, Gray CL, Croft K, Noakes AK, Koch H, Tzeng YC. Dietary nitrate supplementation enhances cerebrovascular CO 2 reactivity in a sex-specific manner. J Appl Physiol (1985) 2019; 127:760-769. [PMID: 31318615 DOI: 10.1152/japplphysiol.01116.2018] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Insufficient nitric oxide (NO) bioavailability plays an important role in endothelial dysfunction, and increased NO has the potential to enhance cerebral blood flow (CBF). Dietary supplementation with sodium nitrate, a precursor of NO, could improve cerebrovascular function, but this has not been investigated. In 17 individuals, we examined the effects of a 7-day supplementation of dietary nitrate (0.1 mmol·kg-1·day -1) on cerebrovascular function using a randomized, single-blinded placebo-controlled crossover design. We hypothesized that 7-day dietary nitrate supplementation increases CBF response to CO2 (cerebrovascular CO2 reactivity) and cerebral autoregulation (CA). We assessed middle cerebral artery blood velocity (MCAv) and blood pressure (BP) at rest and during CO2 breathing. Transfer function analysis was performed on resting beat-to-beat MCAv and BP to determine CA, from which phase, gain, and coherence of the BP-MCAv data were derived. Dietary nitrate elevated plasma nitrate concentration by ~420% (P < 0.001) and lowered gain (d = 1.2, P = 0.025) and phase of the BP-MCAv signal compared with placebo treatment (d = 0.7, P = 0.043), while coherence was unaffected (P = 0.122). Dietary nitrate increased the MCAv-CO2 slope in a sex-specific manner (interaction: P = 0.016). Dietary nitrate increased the MCAv-CO2 slope in men (d = 1.0, P = 0.014 vs. placebo), but had no effect in women (P = 0.919). Our data demonstrate that dietary nitrate greatly increased cerebrovascular CO2 reactivity in healthy individuals, while its effect on CA remains unclear. The selective increase in the MCAv-CO2 slope observed in men indicates a clear sexual dimorphic role of NO in cerebrovascular function.NEW & NOTEWORTHY We found dietary nitrate supplementation improved the brain blood vessels' response to CO2, cerebrovascular CO2 reactivity, without affecting blood pressure in a group of healthy individuals. Meanwhile, the effect of dietary nitrate on the relationship between blood pressure and brain blood flow, cerebral autoregulation, was inconclusive. The improvement in cerebrovascular CO2 reactivity was only observed in the male participants, alluding to a sex difference in the effect of dietary nitrate on brain blood flow control. Our findings indicate that dietary nitrate could be an effective strategy to enhance cerebrovascular CO2 reactivity.
Collapse
Affiliation(s)
- Jui-Lin Fan
- Wellington Medical Technology Group, Department of Surgery and Anaesthesia, University of Otago, Wellington, New Zealand.,Centre for Translational Physiology, University of Otago, Wellington, New Zealand
| | - Terrence O'Donnell
- Wellington Medical Technology Group, Department of Surgery and Anaesthesia, University of Otago, Wellington, New Zealand.,Centre for Translational Physiology, University of Otago, Wellington, New Zealand
| | - Clint Lee Gray
- Centre for Translational Physiology, University of Otago, Wellington, New Zealand.,Paediatrics and Child Health, University of Otago, Wellington, New Zealand
| | - Kevin Croft
- School of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Western Australia, Perth, Australia
| | - Annabel Kate Noakes
- Wellington Medical Technology Group, Department of Surgery and Anaesthesia, University of Otago, Wellington, New Zealand.,Centre for Translational Physiology, University of Otago, Wellington, New Zealand
| | - Henrietta Koch
- School of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Western Australia, Perth, Australia
| | - Yu-Chieh Tzeng
- Wellington Medical Technology Group, Department of Surgery and Anaesthesia, University of Otago, Wellington, New Zealand.,Centre for Translational Physiology, University of Otago, Wellington, New Zealand
| |
Collapse
|
39
|
Aggarwal H, Kanuri BN, Dikshit M. Role of iNOS in Insulin Resistance and Endothelial Dysfunction. OXIDATIVE STRESS IN HEART DISEASES 2019:461-482. [DOI: 10.1007/978-981-13-8273-4_21] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
40
|
Cipolla MJ, Liebeskind DS, Chan SL. The importance of comorbidities in ischemic stroke: Impact of hypertension on the cerebral circulation. J Cereb Blood Flow Metab 2018; 38:2129-2149. [PMID: 30198826 PMCID: PMC6282213 DOI: 10.1177/0271678x18800589] [Citation(s) in RCA: 227] [Impact Index Per Article: 32.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Comorbidities are a hallmark of stroke that both increase the incidence of stroke and worsen outcome. Hypertension is prevalent in the stroke population and the most important modifiable risk factor for stroke. Hypertensive disorders promote stroke through increased shear stress, endothelial dysfunction, and large artery stiffness that transmits pulsatile flow to the cerebral microcirculation. Hypertension also promotes cerebral small vessel disease through several mechanisms, including hypoperfusion, diminished autoregulatory capacity and localized increase in blood-brain barrier permeability. Preeclampsia, a hypertensive disorder of pregnancy, also increases the risk of stroke 4-5-fold compared to normal pregnancy that predisposes women to early-onset cognitive impairment. In this review, we highlight how comorbidities and concomitant disorders are not only risk factors for ischemic stroke, but alter the response to acute ischemia. We focus on hypertension as a comorbidity and its effects on the cerebral circulation that alters the pathophysiology of ischemic stroke and should be considered in guiding future therapeutic strategies.
Collapse
Affiliation(s)
- Marilyn J Cipolla
- 1 Department of Neurological Sciences, University of Vermont Larner College of Medicine, Burlington, VT, USA
| | - David S Liebeskind
- 2 Neurovascular Imaging Research Core and Stroke Center, Department of Neurology, University of California at Los Angeles, Los Angeles, CA, USA
| | - Siu-Lung Chan
- 1 Department of Neurological Sciences, University of Vermont Larner College of Medicine, Burlington, VT, USA
| |
Collapse
|
41
|
Toma S, MacIntosh BJ, Swardfager W, Goldstein BI. Cerebral blood flow in bipolar disorder: A systematic review. J Affect Disord 2018; 241:505-513. [PMID: 30149339 DOI: 10.1016/j.jad.2018.08.040] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2018] [Revised: 08/01/2018] [Accepted: 08/10/2018] [Indexed: 10/28/2022]
Abstract
BACKGROUND Neuroimaging of cerebral blood flow (CBF) can inform our understanding of the pathophysiology of bipolar disorder (BD) as there is increasing support for the concept that BD is in part a vascular disease. Despite numerous studies examining CBF in BD, there has not yet been a review of the literature on the topic of CBF in BD. METHODS A systematic review of the literature on CBF in BD was performed using the Preferred Reporting Items for Systematic Reviews and Meta-Analysis (PRISMA). Studies included measured CBF by single-photon emission computerized tomography (SPECT), positron emission tomography (PET), arterial spin labelling (ASL) or perfusion weighted imaging (PWI) in a group of BD patients. RESULTS Thirty-three studies with a total of 508 subjects with BD and 538 controls were included (n = 15 SPECT; n = 8 PET; n = 7 ASL; n = 1 PWI; n = 2 other). The majority of studies in BD depression and mania reported widespread resting hypoperfusion in cingulate gyrus, frontal, and anterior temporal regions in comparison to healthy controls (HC). Findings in euthymic BD subjects and in symptomatically heterogeneous groups were less consistent. Studies that examined CBF responses to cognitive or emotional stimuli in BD subjects have reported hypoperfusion or different regions involved in comparison to HC. LIMITATIONS Important methodological heterogeneity between studies, and small number of subjects per study. CONCLUSIONS The most consistent findings to date are hypoperfusion in BD mood episodes, and hypoactive CBF responses to emotional or cognitive challenges. Future studies examining CBF are warranted, including prospective studies, studies examining CBF as a treatment target, and multimodal imaging studies.
Collapse
Affiliation(s)
- Simina Toma
- Centre for Youth Bipolar Disorder, Sunnybrook Health Sciences Centre, Toronto, Canada; Department of Psychiatry, University of Toronto, Toronto, Canada
| | - Bradley J MacIntosh
- Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, Canada; Heart and Stroke Foundation Canadian Partnership for Stroke Recovery, Sunnybrook Health Sciences Centre, Toronto, Canada; Department of Medical Biophysics, University of Toronto, Toronto, Canada; Department of Physical Sciences, Sunnybrook Health Sciences Centre, Toronto, Canada
| | - Walter Swardfager
- Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, Canada; Heart and Stroke Foundation Canadian Partnership for Stroke Recovery, Sunnybrook Health Sciences Centre, Toronto, Canada; Department of Pharmacology, University of Toronto, Toronto, Canada
| | - Benjamin I Goldstein
- Centre for Youth Bipolar Disorder, Sunnybrook Health Sciences Centre, Toronto, Canada; Department of Psychiatry, University of Toronto, Toronto, Canada; Department of Pharmacology, University of Toronto, Toronto, Canada.
| |
Collapse
|
42
|
Teixeira SC, Madureira JB, Azevedo EI, Castro PM. Ageing affects the balance between central and peripheral mechanisms of cerebrovascular regulation with increasing influence of systolic blood pressure levels. Eur J Appl Physiol 2018; 119:519-529. [PMID: 30467594 DOI: 10.1007/s00421-018-4036-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Accepted: 11/15/2018] [Indexed: 01/02/2023]
Abstract
BACKGROUND Arterial baroreflex (BR) and cerebral autoregulation (CA) are two major regulatory mechanisms that maintain constant cerebral perfusion. Little is known about the interplay between these mechanisms, particularly when considering the effects of ageing or sex. PURPOSE We studied the relationship between dynamic CA and BR sensitivity (BRS) in healthy subjects by sex and in different age strata. METHODS 95 healthy adults (52% female), 20-80 years-old, were recruited. Arterial blood pressure (Finometer), 3-lead electrocardiogram and cerebral blood flow velocity in middle cerebral arteries (transcranial Doppler) were monitored. We assessed CA by transfer function analysis and BRS in frequency and time domain. RESULTS With increasing age, BRS diminished (ANCOVA R2 = 0.281, p < 0.001) but CA parameters did not change significantly (p > 0.05). Overall, there was an inverse relationship between the efficacy of BRS and CA low-frequency gain [multivariate linear regression β = 0.41 (0.31; 0.61), p < 0.001]. However, this association suffers changes with ageing: in older subjects BRS and CA were not correlated [β = 0.10 (- 0.41; 0.62), p = 0.369]. Instead, decreasing systolic blood pressure correlated with less efficient CA [lower CA low-frequency gain β = - 0.02 (- 0.03; - 0.02), p = 0.003]. Sex did not affect BRS and CA relationship. CONCLUSIONS Cerebral blood supply is governed by a tuned balance between BR and CA which is lost with age as BRS decreases dramatically. Low systolic blood pressure values might be harmful to older subjects as they might reduce the ability to keep cerebral blood flow tightly controlled.
Collapse
Affiliation(s)
- Sofia Cunha Teixeira
- Department of Clinical Neurosciences and Mental Health, Faculty of Medicine, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319, Porto, Portugal.
| | - João Brandão Madureira
- Department of Clinical Neurosciences and Mental Health, Faculty of Medicine, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319, Porto, Portugal
| | - Elsa Irene Azevedo
- Department of Clinical Neurosciences and Mental Health, Faculty of Medicine, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319, Porto, Portugal.,Cardiovascular Research and Development Centre, Faculty of Medicine, University of Porto, Porto, Portugal.,Department of Neurology, São João Hospital Centre, Porto, Portugal
| | - Pedro Miguel Castro
- Department of Clinical Neurosciences and Mental Health, Faculty of Medicine, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319, Porto, Portugal.,Cardiovascular Research and Development Centre, Faculty of Medicine, University of Porto, Porto, Portugal.,Department of Neurology, São João Hospital Centre, Porto, Portugal
| |
Collapse
|
43
|
Quinn KM, Billaut F, Bulmer AC, Minahan CL. Cerebral oxygenation declines but does not impair peak oxygen uptake during incremental cycling in women using oral contraceptives. Eur J Appl Physiol 2018; 118:2417-2427. [PMID: 30167957 DOI: 10.1007/s00421-018-3968-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Accepted: 08/11/2018] [Indexed: 12/31/2022]
Abstract
PURPOSE To compare prefrontal cortex oxygenation in recreationally-active women using oral contraceptives (WomenOC; n = 8) to women with a natural menstrual cycle (WomenNC; n = 8) during incremental exercise to exhaustion. METHODS Participants performed incremental cycling to exhaustion to determine lactate threshold 1 (LT1) and 2 (LT2) and peak oxygen uptake (VO2peak). Prefrontal cortex oxygenation was monitored via near-infrared spectroscopy through concentration changes in oxy-haemoglobin (Δ[HbO2]), deoxy-haemoglobin (Δ[HHb]), total-haemoglobin (Δ[tHb]) and tissue saturation index (TSI). RESULTS 17β-oestradiol and progesterone were lower in WomenOC (35 ± 26; 318 ± 127 pmol·L-1, respectively) than WomenNC (261 ± 156; 858 ± 541 pmol·L-1, respectively). There were no differences in full blood examination results or serum nitric oxide (p > 0.05). However, WomenOC presented lower concentrations in ferric-reducing ability of plasma (- 8%; effect size; ES - 0.52 ± 0.61), bilirubin (- 32%; ES - 0.56 ± 0.62) and uric acid (- 17%; ES - 0.53 ± 0.61). Cardiopulmonary parameters were similar between groups during cycling, including VO2peak (p = 0.99). While there was a significant effect of time on all parameters measured by near-infrared spectroscopy during incremental cycling, there was no effect of OC at LT1, LT2 or exhaustion calculated as a change from baseline (TSI; p = 0.096, Δ[HbO2]; p = 0.143, Δ[HHb]; p = 0.085 and Δ[tHb]; p = 0.226). The change in TSI from LT1 to LT2 was significantly different between groups (WomenNC; mean difference + 2.06%, WomenOC; mean difference - 1.73%; p = 0.003). CONCLUSION Prefrontal tissue oxygenation declined at a lower relative exercise intensity in WomenOC as compared to WomenNC, however, this did not influence VO2peak. The results provide the first evidence for variance in the cerebral oxygenation response to exercise, which may be associated with female sex hormones.
Collapse
Affiliation(s)
- Karlee M Quinn
- Griffith Sports Physiology and Performance, Gold Coast campus, School of Allied Health Sciences, Griffith University, Gold Coast, QLD, 4222, Australia. .,Menzies Health Institute Queensland, Griffith University, Gold Coast, QLD, Australia. .,Queensland Academy of Sport, Nathan, QLD, Australia.
| | - François Billaut
- Department of Kinesiology, Faculty of Medicine, Laval University, Quebec, Canada
| | - Andrew C Bulmer
- Menzies Health Institute Queensland, Griffith University, Gold Coast, QLD, Australia.,School of Medical Science, Griffith University, Gold Coast, QLD, Australia
| | - Clare L Minahan
- Griffith Sports Physiology and Performance, Gold Coast campus, School of Allied Health Sciences, Griffith University, Gold Coast, QLD, 4222, Australia.,Menzies Health Institute Queensland, Griffith University, Gold Coast, QLD, Australia
| |
Collapse
|
44
|
Xu XH, Ye XH, Cai JS, Gao T, Zhao GH, Zhang WJ, Tong LS, Gao F. Association of Renal Dysfunction With Remote Diffusion-Weighted Imaging Lesions and Total Burden of Cerebral Small Vessel Disease in Patients With Primary Intracerebral Hemorrhage. Front Aging Neurosci 2018; 10:171. [PMID: 29930507 PMCID: PMC6001158 DOI: 10.3389/fnagi.2018.00171] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2017] [Accepted: 05/18/2018] [Indexed: 01/13/2023] Open
Abstract
Objective: Remote diffusion-weighted imaging (DWI) lesions (R-DWIL) found in intracerebral hemorrhage (ICH) patients are considered as an additional marker of cerebral small vessel disease (cSVD). This study aimed to investigate the association of renal dysfunction and R-DWIL, as well as the total burden of cSVD on magnetic resonance imaging among patients with primary ICH. Methods: One hundred and twenty-six consecutive patients were prospectively enrolled. R-DWIL on DWI, as well as other imaging markers of cSVD, including lacunes, white matter lesions, cerebral microbleeds, and enlarged perivascular spaces were rated using validated scales. Renal dysfunction was evaluated either by reduced estimated glomerular filtration rate (eGFR) or the presence of proteinuria or increased cystatin C. Results: After adjustments for potential confounders by logistic regression, impaired eGFR [odds ratio (OR) 6.00, 95% confidence interval (CI) 1.73-20.78], proteinuria (OR 3.07, 95% CI 1.25-7.54) and increased cystatin C (OR 2.73, 95% CI 1.11-6.72) were correlated with presence of R-DWIL. A similar association was also found between cystatin C levels (OR 3.16, 95% CI 1.39-7.19), proteinuria (OR 2.79, 95% CI 1.34-5.83) and the comprehensive cSVD burden. Conclusions: Renal dysfunction are associated with the presence of R-DWIL, and total burden of cSVD in patients with primary ICH.
Collapse
Affiliation(s)
- Xu-Hua Xu
- Department of Neurology, The Fourth Affiliated Hospital, School of Medicine, Zhejiang University, Yiwu, China.,School of Medicine, Zhejiang University, Hangzhou, China
| | - Xiang-Hua Ye
- Department of Neurology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Jin-Song Cai
- Department of Radiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Ting Gao
- Department of Neurology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Guo-Hua Zhao
- Department of Neurology, The Fourth Affiliated Hospital, School of Medicine, Zhejiang University, Yiwu, China.,Department of Neurology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Wen-Ji Zhang
- Department of Radiology, The Fourth Affiliated Hospital, School of Medicine, Zhejiang University, Yiwu, China
| | - Lu-Sha Tong
- Department of Neurology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Feng Gao
- School of Medicine, Zhejiang University, Hangzhou, China.,Department of Neurology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
45
|
Blackburn SL, Kumar PT, McBride D, Zeineddine HA, Leclerc J, Choi HA, Dash PK, Grotta J, Aronowski J, Cardenas JC, Doré S. Unique Contribution of Haptoglobin and Haptoglobin Genotype in Aneurysmal Subarachnoid Hemorrhage. Front Physiol 2018; 9:592. [PMID: 29904350 PMCID: PMC5991135 DOI: 10.3389/fphys.2018.00592] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Accepted: 05/02/2018] [Indexed: 01/12/2023] Open
Abstract
Survivors of cerebral aneurysm rupture are at risk for significant morbidity and neurological deficits. Much of this is related to the effects of blood in the subarachnoid space which induces an inflammatory cascade with numerous downstream consequences. Recent clinical trials have not been able to reduce the toxic effects of free hemoglobin or improve clinical outcome. One reason for this may be the inability to identify patients at high risk for neurologic decline. Recently, haptoglobin genotype has been identified as a pertinent factor in diabetes, sickle cell, and cardiovascular disease, with the Hp 2-2 genotype contributing to increased complications. Haptoglobin is a protein synthesized by the liver that binds free hemoglobin following red blood cell lysis, and in doing so, prevents hemoglobin induced toxicity and facilitates clearance. Clinical studies in patients with subarachnoid hemorrhage indicate that Hp 2-2 patients may be a high-risk group for hemorrhage related complications and poor outcome. We review the relevance of haptoglobin in subarachnoid hemorrhage and discuss the effects of genotype and expression levels on the known mechanisms of early brain injury (EBI) and cerebral ischemia after aneurysm rupture. A better understanding of haptoglobin and its role in preventing hemoglobin related toxicity should lead to novel therapeutic avenues.
Collapse
Affiliation(s)
- Spiros L Blackburn
- Department of Neurosurgery, The University of Texas Houston Health Sciences Center, Houston, TX, United States
| | - Peeyush T Kumar
- Department of Neurosurgery, The University of Texas Houston Health Sciences Center, Houston, TX, United States
| | - Devin McBride
- Department of Neurosurgery, The University of Texas Houston Health Sciences Center, Houston, TX, United States
| | - Hussein A Zeineddine
- Department of Neurosurgery, The University of Texas Houston Health Sciences Center, Houston, TX, United States
| | - Jenna Leclerc
- Department of Anesthesiology, University of Florida, College of Medicine, Gainesville, FL, United States
| | - H Alex Choi
- Department of Neurosurgery, The University of Texas Houston Health Sciences Center, Houston, TX, United States
| | - Pramod K Dash
- Department of Neurosurgery, The University of Texas Houston Health Sciences Center, Houston, TX, United States
| | - James Grotta
- Department of Neurology, The University of Texas Health Sciences Center, Houston, TX, United States
| | - Jaroslaw Aronowski
- Department of Neurology, The University of Texas Health Sciences Center, Houston, TX, United States
| | - Jessica C Cardenas
- Department of Surgery, Division of Acute Care Surgery and Center for Translational Injury Research, The University of Texas Health Science Center, Houston, TX, United States
| | - Sylvain Doré
- Department of Anesthesiology, University of Florida, College of Medicine, Gainesville, FL, United States.,Departments of Neurology, Psychiatry, Psychology, Pharmaceutics, and Neuroscience, University of Florida, McKnight Brain Institute, Gainesville, FL, United States
| |
Collapse
|
46
|
Age-related changes in cerebrovascular reactivity and their relationship to cognition: A four-year longitudinal study. Neuroimage 2018; 174:257-262. [PMID: 29567504 DOI: 10.1016/j.neuroimage.2018.03.033] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Revised: 02/07/2018] [Accepted: 03/16/2018] [Indexed: 11/20/2022] Open
Abstract
Although cerebrovascular factors are the second leading cause of cognitive impairment and dementia in elderly, the precise spatial and temporal trajectories of vascular decline in aging have not been fully characterized. With an advanced cerebrovascular reactivity (CVR) MRI technique that specifically informs vascular stiffness and dilatory ability of cerebral vessels, we present four-year longitudinal CVR data measured in 116 healthy individuals (20-88 years of age). Our data revealed a spatial heterogeneity in vascular decline in aging (p = 0.003), in that temporal lobe showed the fastest rate of longitudinal CVR decline, followed by parietal and frontal lobes. The rate of CVR decline was also age-dependent. Middle age, not older age, manifested the fastest rate of longitudinal CVR decline (p < 0.05). Longitudinal changes in CVR were associated with changes in processing speed (p = 0.031) and episodic memory (p = 0.022), but not with working memory or reasoning. The rate of longitudinal CVR change was not different between hypertensive and normotensive participants. However, cross-sectionally, individuals with hypertension revealed in a lower CVR compared to normotensive participants (p = 0.016). These findings help elucidate age-related decline in brain hemodynamics and support CVR as a non-invasive biomarker in evaluating cerebrovascular conditions in elderly individuals.
Collapse
|
47
|
Császár N, Scholkmann F, Salari V, Szőke H, Bókkon I. Phosphene perception is due to the ultra-weak photon emission produced in various parts of the visual system: glutamate in the focus. Rev Neurosci 2018; 27:291-9. [PMID: 26544101 DOI: 10.1515/revneuro-2015-0039] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2015] [Accepted: 10/11/2015] [Indexed: 12/24/2022]
Abstract
Phosphenes are experienced sensations of light, when there is no light causing them. The physiological processes underlying this phenomenon are still not well understood. Previously, we proposed a novel biopsychophysical approach concerning the cause of phosphenes based on the assumption that cellular endogenous ultra-weak photon emission (UPE) is the biophysical cause leading to the sensation of phosphenes. Briefly summarized, the visual sensation of light (phosphenes) is likely to be due to the inherent perception of UPE of cells in the visual system. If the intensity of spontaneous or induced photon emission of cells in the visual system exceeds a distinct threshold, it is hypothesized that it can become a conscious light sensation. Discussing several new and previous experiments, we point out that the UPE theory of phosphenes should be really considered as a scientifically appropriate and provable mechanism to explain the physiological basis of phosphenes. In the present paper, we also present our idea that some experiments may support that the cortical phosphene lights are due to the glutamate-related excess UPE in the occipital cortex.
Collapse
|
48
|
Fateeva VV, Vorobyova OV. [Nitric oxide: from the mechanism of action to pharmacological effects in cerebrovascular diseases]. Zh Nevrol Psikhiatr Im S S Korsakova 2017; 117:131-135. [PMID: 29171501 DOI: 10.17116/jnevro2017117101131-135] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The article presents the data of studies of nitric oxide (NO) in the pathogenesis of cerebrovascular diseases. It is emphasized that endothelial dysfunction contributes to the formation of cerebrovascular diseases. Generalized data on preparations with endothelioprotective effect, as well as own data on the use of the preparation 'Divaza' in patients of middle and advanced age with chronic cerebrovascular disease are given.
Collapse
Affiliation(s)
- V V Fateeva
- Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation, Moscow, Russia
| | - O V Vorobyova
- Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation, Moscow, Russia
| |
Collapse
|
49
|
Villéga F, Delpech JC, Griton M, André C, Franconi JM, Miraux S, Konsman JP. Circulating bacterial lipopolysaccharide-induced inflammation reduces flow in brain-irrigating arteries independently from cerebrovascular prostaglandin production. Neuroscience 2017; 346:160-172. [DOI: 10.1016/j.neuroscience.2017.01.018] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Revised: 01/07/2017] [Accepted: 01/11/2017] [Indexed: 12/24/2022]
|
50
|
Nguyen CTO, Hui F, Charng J, Velaedan S, van Koeverden AK, Lim JKH, He Z, Wong VHY, Vingrys AJ, Bui BV, Ivarsson M. Retinal biomarkers provide "insight" into cortical pharmacology and disease. Pharmacol Ther 2017; 175:151-177. [PMID: 28174096 DOI: 10.1016/j.pharmthera.2017.02.009] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
The retina is an easily accessible out-pouching of the central nervous system (CNS) and thus lends itself to being a biomarker of the brain. More specifically, the presence of neuronal, vascular and blood-neural barrier parallels in the eye and brain coupled with fast and inexpensive methods to quantify retinal changes make ocular biomarkers an attractive option. This includes its utility as a biomarker for a number of cerebrovascular diseases as well as a drug pharmacology and safety biomarker for the CNS. It is a rapidly emerging field, with some areas well established, such as stroke risk and multiple sclerosis, whereas others are still in development (Alzheimer's, Parkinson's, psychological disease and cortical diabetic dysfunction). The current applications and future potential of retinal biomarkers, including potential ways to improve their sensitivity and specificity are discussed. This review summarises the existing literature and provides a perspective on the strength of current retinal biomarkers and their future potential.
Collapse
Affiliation(s)
- Christine T O Nguyen
- Department of Optometry and Vision Sciences, University of Melbourne, Parkville, 3010, Victoria, Australia.
| | - Flora Hui
- Department of Optometry and Vision Sciences, University of Melbourne, Parkville, 3010, Victoria, Australia
| | - Jason Charng
- Department of Optometry and Vision Sciences, University of Melbourne, Parkville, 3010, Victoria, Australia
| | - Shajan Velaedan
- Department of Optometry and Vision Sciences, University of Melbourne, Parkville, 3010, Victoria, Australia
| | - Anna K van Koeverden
- Department of Optometry and Vision Sciences, University of Melbourne, Parkville, 3010, Victoria, Australia
| | - Jeremiah K H Lim
- Department of Optometry and Vision Sciences, University of Melbourne, Parkville, 3010, Victoria, Australia
| | - Zheng He
- Department of Optometry and Vision Sciences, University of Melbourne, Parkville, 3010, Victoria, Australia
| | - Vickie H Y Wong
- Department of Optometry and Vision Sciences, University of Melbourne, Parkville, 3010, Victoria, Australia
| | - Algis J Vingrys
- Department of Optometry and Vision Sciences, University of Melbourne, Parkville, 3010, Victoria, Australia
| | - Bang V Bui
- Department of Optometry and Vision Sciences, University of Melbourne, Parkville, 3010, Victoria, Australia
| | - Magnus Ivarsson
- Department of Optometry and Vision Sciences, University of Melbourne, Parkville, 3010, Victoria, Australia
| |
Collapse
|