1
|
El Amine B, Fournier J, Minoves M, Baillieul S, Roche F, Perek N, Pépin JL, Tamisier R, Khouri C, Rome C, Briançon-Marjollet A. Cerebral oxidative stress, inflammation and apoptosis induced by intermittent hypoxia: a systematic review and meta-analysis of rodent data. Eur Respir Rev 2024; 33:240162. [PMID: 39694586 DOI: 10.1183/16000617.0162-2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 09/21/2024] [Indexed: 12/20/2024] Open
Abstract
Obstructive sleep apnoea (OSA) contributes to cerebrovascular diseases and cognitive decline. Preclinical studies support the deleterious impact on the brain of intermittent hypoxia (IH), one of the main components of OSA, but heterogeneity in rodent species and brain regions studied, or induced by IH paradigms, can challenge interpretation of the studies. Hence, we conducted a systematic review and meta-analysis to evaluate the impact of IH on rodent brain oxidative stress, inflammation, apoptosis and the expression of brain-derived neurotrophic factor (BDNF) and hypoxia-inducible factor 1 (HIF-1). PubMed and Web of Science searches identified 663 articles related to IH exposure, of which 60 were included. The examined outcomes were oxidative stress, inflammation, apoptosis, HIF-1 or BDNF in brains. Standardised mean difference was used to compare studies. Metaregressions were performed to clarify the impact of IH exposure parameters, rodent characteristics or cerebral localisation on these outcomes. IH-induced oxidative stress (increased malondialdehyde (MDA) and NADPH oxidase (NOX) and decreased superoxide dismutase), increased inflammation (tumour necrosis factor-α, NF-κB and inducible nitric oxide synthase), HIF-1 and apoptosis evaluated by terminal deoxynucleotidyl transferase dUTP nick-end labelling and cleaved caspase-3. In contrast, B-cell lymphoma 2 (BCL2) and BDNF expression were not significantly modified. Metaregressions showed that MDA, NOX and BDNF were associated with determinants of IH cycles (inspired oxygen fraction and duration of hypoxia) and some parameters depended on localisation. Rodent characteristics had little impact on the outcomes. Our meta-analysis robustly establishes that IH, independently of other confounders, has a strong effect on the brain by inducing oxidative stress, inflammation and apoptosis in rodent models. Our findings support the interest of considering and treating cerebral consequences of OSA in clinical practice.
Collapse
Affiliation(s)
- Bayan El Amine
- Univ. Grenoble Alpes, Inserm U1300, CHU Grenoble Alpes, HP2 Laboratory, Grenoble, France
- Univ. Grenoble Alpes, Inserm U1216, Grenoble Institut Neurosciences, Grenoble, France
| | - Joey Fournier
- Univ. Grenoble Alpes, Inserm CIC1406, CHU de Grenoble, Grenoble, France
| | - Mélanie Minoves
- Univ. Grenoble Alpes, Inserm U1300, CHU Grenoble Alpes, HP2 Laboratory, Grenoble, France
| | - Sébastien Baillieul
- Univ. Grenoble Alpes, Inserm U1300, CHU Grenoble Alpes, HP2 Laboratory, Grenoble, France
| | - Frédéric Roche
- Université Jean Monnet, Inserm U1059 Sainbiose, Saint Etienne, France
- Physiologie Clinique et de l'Exercice, CHU, Saint Etienne, France
| | - Nathalie Perek
- Université Jean Monnet, Inserm U1059 Sainbiose, Saint Etienne, France
| | - Jean-Louis Pépin
- Univ. Grenoble Alpes, Inserm U1300, CHU Grenoble Alpes, HP2 Laboratory, Grenoble, France
| | - Renaud Tamisier
- Univ. Grenoble Alpes, Inserm U1300, CHU Grenoble Alpes, HP2 Laboratory, Grenoble, France
| | - Charles Khouri
- Univ. Grenoble Alpes, Inserm U1300, CHU Grenoble Alpes, HP2 Laboratory, Grenoble, France
- Univ. Grenoble Alpes, Inserm CIC1406, CHU de Grenoble, Grenoble, France
| | - Claire Rome
- Univ. Grenoble Alpes, Inserm U1216, Grenoble Institut Neurosciences, Grenoble, France
| | | |
Collapse
|
2
|
Frias-Anaya E, Gallego-Gutierrez H, Gongol B, Weinsheimer S, Lai CC, Orecchioni M, Sriram A, Bui CM, Nelsen B, Hale P, Pham A, Shenkar R, DeBiasse D, Lightle R, Girard R, Li Y, Srinath A, Daneman R, Nudleman E, Sun H, Guma M, Dubrac A, Mesarwi OA, Ley K, Kim H, Awad IA, Ginsberg MH, Lopez-Ramirez MA. Mild Hypoxia Accelerates Cerebral Cavernous Malformation Disease Through CX3CR1-CX3CL1 Signaling. Arterioscler Thromb Vasc Biol 2024; 44:1246-1264. [PMID: 38660801 PMCID: PMC11111348 DOI: 10.1161/atvbaha.123.320367] [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: 11/01/2023] [Accepted: 04/05/2024] [Indexed: 04/26/2024]
Abstract
BACKGROUND Heterogeneity in the severity of cerebral cavernous malformations (CCMs) disease, including brain bleedings and thrombosis that cause neurological disabilities in patients, suggests that environmental, genetic, or biological factors act as disease modifiers. Still, the underlying mechanisms are not entirely understood. Here, we report that mild hypoxia accelerates CCM disease by promoting angiogenesis, neuroinflammation, and vascular thrombosis in the brains of CCM mouse models. METHODS We used genetic studies, RNA sequencing, spatial transcriptome, micro-computed tomography, fluorescence-activated cell sorting, multiplex immunofluorescence, coculture studies, and imaging techniques to reveal that sustained mild hypoxia via the CX3CR1-CX3CL1 (CX3C motif chemokine receptor 1/chemokine [CX3C motif] ligand 1) signaling pathway influences cell-specific neuroinflammatory interactions, contributing to heterogeneity in CCM severity. RESULTS Histological and expression profiles of CCM neurovascular lesions (Slco1c1-iCreERT2;Pdcd10fl/fl; Pdcd10BECKO) in male and female mice found that sustained mild hypoxia (12% O2, 7 days) accelerates CCM disease. Our findings indicate that a small reduction in oxygen levels can significantly increase angiogenesis, neuroinflammation, and thrombosis in CCM disease by enhancing the interactions between endothelium, astrocytes, and immune cells. Our study indicates that the interactions between CX3CR1 and CX3CL1 are crucial in the maturation of CCM lesions and propensity to CCM immunothrombosis. In particular, this pathway regulates the recruitment and activation of microglia and other immune cells in CCM lesions, which leads to lesion growth and thrombosis. We found that human CX3CR1 variants are linked to lower lesion burden in familial CCMs, proving it is a genetic modifier in human disease and a potential marker for aggressiveness. Moreover, monoclonal blocking antibody against CX3CL1 or reducing 1 copy of the Cx3cr1 gene significantly reduces hypoxia-induced CCM immunothrombosis. CONCLUSIONS Our study reveals that interactions between CX3CR1 and CX3CL1 can modify CCM neuropathology when lesions are accelerated by environmental hypoxia. Moreover, a hypoxic environment or hypoxia signaling caused by CCM disease influences the balance between neuroinflammation and neuroprotection mediated by CX3CR1-CX3CL1 signaling. These results establish CX3CR1 as a genetic marker for patient stratification and a potential predictor of CCM aggressiveness.
Collapse
MESH Headings
- Animals
- Female
- Humans
- Male
- Mice
- Chemokine CX3CL1/metabolism
- Chemokine CX3CL1/genetics
- CX3C Chemokine Receptor 1/genetics
- CX3C Chemokine Receptor 1/metabolism
- Disease Models, Animal
- Hemangioma, Cavernous, Central Nervous System/genetics
- Hemangioma, Cavernous, Central Nervous System/metabolism
- Hemangioma, Cavernous, Central Nervous System/pathology
- Hypoxia/metabolism
- Hypoxia/complications
- Mice, Inbred C57BL
- Mice, Knockout
- Neovascularization, Pathologic/metabolism
- Neuroinflammatory Diseases/metabolism
- Neuroinflammatory Diseases/pathology
- Neuroinflammatory Diseases/genetics
- Signal Transduction
Collapse
Affiliation(s)
- Eduardo Frias-Anaya
- Department of Medicine (E.F.-A., H.G.-G., C.C.L., C.M.B., B.N., P.H., A.P., H.S., M.G., O.A.M., M.H.G., M.A.L.-R.), University of California San Diego, La Jolla
| | - Helios Gallego-Gutierrez
- Department of Medicine (E.F.-A., H.G.-G., C.C.L., C.M.B., B.N., P.H., A.P., H.S., M.G., O.A.M., M.H.G., M.A.L.-R.), University of California San Diego, La Jolla
| | - Brendan Gongol
- Department of Health Sciences, Victor Valley College, Victorville, CA (B.G.)
- Institute for Integrative Genome Biology, 1207F Genomics Building, University of California, Riverside (B.G.)
| | - Shantel Weinsheimer
- Department of Anesthesia and Perioperative Care, Institute for Human Genetics, University of California, San Francisco (S.W., A.S., H.K.)
| | - Catherine Chinhchu Lai
- Department of Medicine (E.F.-A., H.G.-G., C.C.L., C.M.B., B.N., P.H., A.P., H.S., M.G., O.A.M., M.H.G., M.A.L.-R.), University of California San Diego, La Jolla
| | - Marco Orecchioni
- Division of Inflammation Biology, La Jolla Institute for Immunology, CA (M.O., K.L.)
| | - Aditya Sriram
- Department of Anesthesia and Perioperative Care, Institute for Human Genetics, University of California, San Francisco (S.W., A.S., H.K.)
| | - Cassandra M Bui
- Department of Medicine (E.F.-A., H.G.-G., C.C.L., C.M.B., B.N., P.H., A.P., H.S., M.G., O.A.M., M.H.G., M.A.L.-R.), University of California San Diego, La Jolla
| | - Bliss Nelsen
- Department of Medicine (E.F.-A., H.G.-G., C.C.L., C.M.B., B.N., P.H., A.P., H.S., M.G., O.A.M., M.H.G., M.A.L.-R.), University of California San Diego, La Jolla
| | - Preston Hale
- Department of Medicine (E.F.-A., H.G.-G., C.C.L., C.M.B., B.N., P.H., A.P., H.S., M.G., O.A.M., M.H.G., M.A.L.-R.), University of California San Diego, La Jolla
| | - Angela Pham
- Department of Medicine (E.F.-A., H.G.-G., C.C.L., C.M.B., B.N., P.H., A.P., H.S., M.G., O.A.M., M.H.G., M.A.L.-R.), University of California San Diego, La Jolla
| | - Robert Shenkar
- Neurovascular Surgery Program, Department of Neurological Surgery, The University of Chicago Medicine and Biological Sciences, IL (R.S., D.D., R.L., R.G., Y.L., A.S., I.A.A.)
| | - Dorothy DeBiasse
- Neurovascular Surgery Program, Department of Neurological Surgery, The University of Chicago Medicine and Biological Sciences, IL (R.S., D.D., R.L., R.G., Y.L., A.S., I.A.A.)
| | - Rhonda Lightle
- Neurovascular Surgery Program, Department of Neurological Surgery, The University of Chicago Medicine and Biological Sciences, IL (R.S., D.D., R.L., R.G., Y.L., A.S., I.A.A.)
| | - Romuald Girard
- Neurovascular Surgery Program, Department of Neurological Surgery, The University of Chicago Medicine and Biological Sciences, IL (R.S., D.D., R.L., R.G., Y.L., A.S., I.A.A.)
| | - Ying Li
- Neurovascular Surgery Program, Department of Neurological Surgery, The University of Chicago Medicine and Biological Sciences, IL (R.S., D.D., R.L., R.G., Y.L., A.S., I.A.A.)
| | - Abhinav Srinath
- Neurovascular Surgery Program, Department of Neurological Surgery, The University of Chicago Medicine and Biological Sciences, IL (R.S., D.D., R.L., R.G., Y.L., A.S., I.A.A.)
| | - Richard Daneman
- Department of Pharmacology (R.D., M.A.L.-R.), University of California San Diego, La Jolla
| | - Eric Nudleman
- Department of Ophthalmology (E.N.), University of California San Diego, La Jolla
| | - Hao Sun
- Department of Medicine (E.F.-A., H.G.-G., C.C.L., C.M.B., B.N., P.H., A.P., H.S., M.G., O.A.M., M.H.G., M.A.L.-R.), University of California San Diego, La Jolla
| | - Monica Guma
- Department of Medicine (E.F.-A., H.G.-G., C.C.L., C.M.B., B.N., P.H., A.P., H.S., M.G., O.A.M., M.H.G., M.A.L.-R.), University of California San Diego, La Jolla
| | - Alexandre Dubrac
- Centre de Recherche, CHU St. Justine, Montréal, Quebec, Canada. Département de Pathologie et Biologie Cellulaire, Université de Montréal, Quebec, Canada (A.D.)
| | - Omar A Mesarwi
- Department of Medicine (E.F.-A., H.G.-G., C.C.L., C.M.B., B.N., P.H., A.P., H.S., M.G., O.A.M., M.H.G., M.A.L.-R.), University of California San Diego, La Jolla
| | - Klaus Ley
- Division of Inflammation Biology, La Jolla Institute for Immunology, CA (M.O., K.L.)
| | - Helen Kim
- Department of Anesthesia and Perioperative Care, Institute for Human Genetics, University of California, San Francisco (S.W., A.S., H.K.)
| | - Issam A Awad
- Neurovascular Surgery Program, Department of Neurological Surgery, The University of Chicago Medicine and Biological Sciences, IL (R.S., D.D., R.L., R.G., Y.L., A.S., I.A.A.)
| | - Mark H Ginsberg
- Department of Medicine (E.F.-A., H.G.-G., C.C.L., C.M.B., B.N., P.H., A.P., H.S., M.G., O.A.M., M.H.G., M.A.L.-R.), University of California San Diego, La Jolla
| | - Miguel Alejandro Lopez-Ramirez
- Department of Medicine (E.F.-A., H.G.-G., C.C.L., C.M.B., B.N., P.H., A.P., H.S., M.G., O.A.M., M.H.G., M.A.L.-R.), University of California San Diego, La Jolla
- Department of Pharmacology (R.D., M.A.L.-R.), University of California San Diego, La Jolla
| |
Collapse
|
3
|
Sarchi PV, Gomez Cuautle D, Rossi A, Ramos AJ. Participation of the spleen in the neuroinflammation after pilocarpine-induced status epilepticus: implications for epileptogenesis and epilepsy. Clin Sci (Lond) 2024; 138:555-572. [PMID: 38602323 DOI: 10.1042/cs20231621] [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: 12/11/2023] [Revised: 03/28/2024] [Accepted: 04/11/2024] [Indexed: 04/12/2024]
Abstract
Epilepsy, a chronic neurological disorder characterized by recurrent seizures, affects millions of individuals worldwide. Despite extensive research, the underlying mechanisms leading to epileptogenesis, the process by which a normal brain develops epilepsy, remain elusive. We, here, explored the immune system and spleen responses triggered by pilocarpine-induced status epilepticus (SE) focusing on their role in the epileptogenesis that follows SE. Initial examination of spleen histopathology revealed transient disorganization of white pulp, in animals subjected to SE. This disorganization, attributed to immune activation, peaked at 1-day post-SE (1DPSE) but returned to control levels at 3DPSE. Alterations in peripheral blood lymphocyte populations, demonstrated a decrease following SE, accompanied by a reduction in CD3+ T-lymphocytes. Further investigations uncovered an increased abundance of T-lymphocytes in the piriform cortex and choroid plexus at 3DPSE, suggesting a specific mobilization toward the Central Nervous System. Notably, splenectomy mitigated brain reactive astrogliosis, neuroinflammation, and macrophage infiltration post-SE, particularly in the hippocampus and piriform cortex. Additionally, splenectomized animals exhibited reduced lymphatic follicle size in the deep cervical lymph nodes. Most significantly, splenectomy correlated with improved neuronal survival, substantiated by decreased neuronal loss and reduced degenerating neurons in the piriform cortex and hippocampal CA2-3 post-SE. Overall, these findings underscore the pivotal role of the spleen in orchestrating immune responses and neuroinflammation following pilocarpine-induced SE, implicating the peripheral immune system as a potential therapeutic target for mitigating neuronal degeneration in epilepsy.
Collapse
Affiliation(s)
- Paula Virginia Sarchi
- Laboratorio de Neuropatología Molecular, Instituto de Biología Celular y Neurociencia "Prof. E. De Robertis", Facultad de Medicina, Universidad de Buenos Aires-CONICET, Paraguay 2155 3er piso (1121) Ciudad de Buenos Aires, Argentina
| | - Dante Gomez Cuautle
- Laboratorio de Neuropatología Molecular, Instituto de Biología Celular y Neurociencia "Prof. E. De Robertis", Facultad de Medicina, Universidad de Buenos Aires-CONICET, Paraguay 2155 3er piso (1121) Ciudad de Buenos Aires, Argentina
| | - Alicia Rossi
- Laboratorio de Neuropatología Molecular, Instituto de Biología Celular y Neurociencia "Prof. E. De Robertis", Facultad de Medicina, Universidad de Buenos Aires-CONICET, Paraguay 2155 3er piso (1121) Ciudad de Buenos Aires, Argentina
| | - Alberto Javier Ramos
- Laboratorio de Neuropatología Molecular, Instituto de Biología Celular y Neurociencia "Prof. E. De Robertis", Facultad de Medicina, Universidad de Buenos Aires-CONICET, Paraguay 2155 3er piso (1121) Ciudad de Buenos Aires, Argentina
| |
Collapse
|
4
|
Liu F, Yan W, Chen C, Zeng Y, Kong Y, He X, Pei P, Wang S, Zhang T. Acetylome analyses provide novel insights into the effects of chronic intermittent hypoxia on hippocampus-dependent cognitive impairment. Front Mol Neurosci 2024; 17:1324458. [PMID: 38455734 PMCID: PMC10917988 DOI: 10.3389/fnmol.2024.1324458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 02/08/2024] [Indexed: 03/09/2024] Open
Abstract
Introduction Chronic intermittent hypoxia (CIH) can negatively affect hippocampal function through various molecular mechanisms. Protein acetylation, a frequently occurring modification, plays crucial roles in synaptic plasticity and cognitive processes. However, the global protein acetylation induced by CIH in the hippocampus and its specific effects on hippocampal function and behavior remain poorly understood. Methods To address this gap, we conducted a study using liquid chromatography-tandem mass spectrometry to analyze the lysine acetylome and proteome of the hippocampus in healthy adult mice exposed to intermittent hypoxia for 4 weeks (as a CIH model) compared to normoxic mice (as a control). Results We identified and quantified a total of 2,184 lysine acetylation sites in 1,007 proteins. Analysis of these acetylated proteins revealed disturbances primarily in oxidative phosphorylation, the tricarboxylic acid (TCA) cycle, and glycolysis, all of which are localized exclusively to mitochondria. Additionally, we observed significant changes in the abundance of 21 proteins, some of which are known to be associated with cognitive impairments. Discussion This study helps to elucidate the molecular mechanisms underlying CIH-induced changes in protein acetylation in the hippocampus. By providing valuable insights into the pathophysiological processes associated with CIH and their impacts on hippocampal function, our findings contribute to a better understanding of the consequences of CIH-induced changes in protein acetylation in the hippocampus and the potential role of CIH in cognitive impairment.
Collapse
Affiliation(s)
- Fan Liu
- Children’s Hospital Capital Institute of Pediatrics, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Beijing Municipal Key Laboratory of Child Development and Nutriomics, Capital Institute of Pediatrics, Beijing, China
- Graduate School of Peking Union Medical College, Beijing, China
| | - Weiheng Yan
- Children’s Hospital Capital Institute of Pediatrics, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Chen Chen
- Beijing Municipal Key Laboratory of Child Development and Nutriomics, Capital Institute of Pediatrics, Beijing, China
| | - Yubing Zeng
- Children’s Hospital Capital Institute of Pediatrics, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Beijing Municipal Key Laboratory of Child Development and Nutriomics, Capital Institute of Pediatrics, Beijing, China
| | - Yaru Kong
- Children’s Hospital Capital Institute of Pediatrics, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xuejia He
- Beijing Municipal Key Laboratory of Child Development and Nutriomics, Capital Institute of Pediatrics-Peking University Teaching Hospital, Beijing, China
| | - Pei Pei
- Beijing Municipal Key Laboratory of Child Development and Nutriomics, Capital Institute of Pediatrics, Beijing, China
| | - Shan Wang
- Children’s Hospital Capital Institute of Pediatrics, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Beijing Municipal Key Laboratory of Child Development and Nutriomics, Capital Institute of Pediatrics, Beijing, China
- Graduate School of Peking Union Medical College, Beijing, China
- Beijing Municipal Key Laboratory of Child Development and Nutriomics, Capital Institute of Pediatrics-Peking University Teaching Hospital, Beijing, China
| | - Ting Zhang
- Children’s Hospital Capital Institute of Pediatrics, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Beijing Municipal Key Laboratory of Child Development and Nutriomics, Capital Institute of Pediatrics, Beijing, China
- Graduate School of Peking Union Medical College, Beijing, China
- Beijing Municipal Key Laboratory of Child Development and Nutriomics, Capital Institute of Pediatrics-Peking University Teaching Hospital, Beijing, China
| |
Collapse
|
5
|
Cieri MB, Villarreal A, Gomez-Cuautle DD, Mailing I, Ramos AJ. Progression of reactive gliosis and astroglial phenotypic changes following stab wound-induced traumatic brain injury in mice. J Neurochem 2023; 167:183-203. [PMID: 37592830 DOI: 10.1111/jnc.15941] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 06/29/2023] [Accepted: 08/03/2023] [Indexed: 08/19/2023]
Abstract
Astrocytes are the main homeostatic cells in the central nervous system (CNS) and they have an essential role in preserving neuronal physiology. After brain injury, astrocytes become reactive, and that involves a profound change in the astroglial gene expression program as well as intense cytoskeleton remodeling that has been classically shown by the up-regulation of glial fibrillary acidic protein (GFAP), a pan-reactive gene over-expressed in reactive astrocytes, independently of the type of injury. Using the stab wound rodent model of penetrating traumatic injury in the cortex, we here studied the reactive astroglial morphology and reactive microgliosis in detail at 1, 3, 7, 14, and 28 days post-injury (dpi). By combining immunohistochemistry, morphometrical parameters, and Sholl analysis, we segmented the astroglial cell population into clusters of reactive astrocytes that were localized in the core, penumbra, and distal regions of the stab wound. Specifically, highly reactive clusters with more complex morphology, increased C3, decreased aquaporin-4 (AQP4), and glutamine synthetase (GS) expression, were enriched at 7 dpi when behavioral alterations, microgliosis, and neuronal alterations in injured mice were most significant. While pro-inflammatory gain of function with peripheral lipopolysaccharide (LPS) administration immediately after a stab wound expanded these highly reactive astroglial clusters, the treatment with the NF-κB inhibitor sulfasalazine reduced the abundance of this highly reactive cluster. Increased neuronal loss and exacerbated reactive microgliosis at 7 dpi were associated with the expansion of the highly reactive astroglial cluster. We conclude that highly reactive astrocytes found in stab wound injury, but expanded in pro-inflammatory conditions, are a population of astrocytes that become engaged in pathological remodeling with a pro-inflammatory gain of function and loss of homeostatic capacity. Controlling this astroglial population may be a tempting strategy to reduce neuronal loss and neuroinflammation in the injured brain.
Collapse
Affiliation(s)
- Maria Belen Cieri
- Laboratorio de Neuropatología Molecular, Instituto de Biología Celular y Neurociencia "Prof. E. De Robertis", UBA-CONICET, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Alejandro Villarreal
- Laboratorio de Neuropatología Molecular, Instituto de Biología Celular y Neurociencia "Prof. E. De Robertis", UBA-CONICET, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Dante Daniel Gomez-Cuautle
- Laboratorio de Neuropatología Molecular, Instituto de Biología Celular y Neurociencia "Prof. E. De Robertis", UBA-CONICET, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Ingrid Mailing
- Laboratorio de Neuropatología Molecular, Instituto de Biología Celular y Neurociencia "Prof. E. De Robertis", UBA-CONICET, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Alberto Javier Ramos
- Laboratorio de Neuropatología Molecular, Instituto de Biología Celular y Neurociencia "Prof. E. De Robertis", UBA-CONICET, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
6
|
Guo W, Mao X, Han D, Wang H, Zhang W, Zhang G, Zhang N, Nie B, Li H, Song Y, Wu Y, Chang L. Sleep deprivation aggravated amyloid β oligomers-induced damage to the cerebellum of rats: Evidence from magnetic resonance imaging. AGING BRAIN 2023; 4:100091. [PMID: 37600754 PMCID: PMC10432242 DOI: 10.1016/j.nbas.2023.100091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Revised: 07/29/2023] [Accepted: 08/01/2023] [Indexed: 08/22/2023] Open
Abstract
For quite a long time, researches on Alzheimer's disease (AD) primarily focused on the cortex and hippocampus, while the cerebellum has been ignored because of its abnormalities considered to appear in the late stage of AD. In recent years, increasing evidence suggest that the cerebellar pathological changes possibly occur in the preclinical phase of AD, which is also associated with sleep disorder. Sleep disturbance is a high risk factor of AD. However, the changes and roles of cerebellum has rarely been reported under conditions of AD accompanied with sleep disorders. In this study, using an amyloid-β oligomers (AβO)-induced rat model of AD subjected to sleep deprivation, combining with a 7.0 T animals structural magnetic resonance imaging (MRI), we assessed structural changes of cerebellum in MRI. Our results showed that sleep deprivation combined with AβO led to an increased FA value in the anterior lobe of cerebellum, decreased ADC value in the cerebellar lobes and cerebellar nuclei, and increased cerebellum volume. Besides that, sleep deprivation exacerbated the damage of AβO to the cerebellar structural network. This study demonstrated that sleep deprivation could aggravate the damage to cerebellum induced by AβO. The present findings provide supporting evidence for the involvement of cerebellum in the early pathology of AD and sleep loss. Our data would contribute to advancing the understanding of the mysterious role of cerebellum in AD and sleep disorders, as well as would be helpful for developing non-invasive MRI biomarkers for screening early AD patients with self-reported sleep disturbances.
Collapse
Affiliation(s)
- Wensheng Guo
- Department of Anatomy, School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Beijing Institute of Brain Disorders, Capital Medical University, Beijing, China
| | - Xin Mao
- Department of Anatomy, School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Beijing Institute of Brain Disorders, Capital Medical University, Beijing, China
| | - Ding Han
- Department of Anatomy, School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Beijing Institute of Brain Disorders, Capital Medical University, Beijing, China
| | - Hongqi Wang
- Department of Anatomy, School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Beijing Institute of Brain Disorders, Capital Medical University, Beijing, China
| | - Wanning Zhang
- Department of Anatomy, School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Beijing Institute of Brain Disorders, Capital Medical University, Beijing, China
| | - Guitao Zhang
- Department of Anatomy, School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Beijing Institute of Brain Disorders, Capital Medical University, Beijing, China
| | - Ning Zhang
- Department of Neuropsychiatry and Behavioral Neurology and Clinical Psychology, China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Binbin Nie
- Beijing Engineering Research Center of Radiographic Techniques and Equipment, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, China
| | - Hui Li
- Department of Anatomy, School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Beijing Institute of Brain Disorders, Capital Medical University, Beijing, China
| | - Yizhi Song
- Department of Anatomy, School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Beijing Institute of Brain Disorders, Capital Medical University, Beijing, China
| | - Yan Wu
- Department of Anatomy, School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Beijing Institute of Brain Disorders, Capital Medical University, Beijing, China
| | - Lirong Chang
- Department of Anatomy, School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Beijing Institute of Brain Disorders, Capital Medical University, Beijing, China
| |
Collapse
|
7
|
Yu C, Fu Y, Lu Y, Huang Y, Chen F, Wei J, Li L, Ampadu JA, Wang Y, Zheng W, Jiang C, Li W, Lui S, Cai X. Alterations of brain gray matter volume in children with obstructive sleep apnea. Front Neurol 2023; 14:1107086. [PMID: 37265465 PMCID: PMC10230248 DOI: 10.3389/fneur.2023.1107086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Accepted: 04/24/2023] [Indexed: 06/03/2023] Open
Abstract
Objective Obstructive sleep apnea (OSA) seriously affects the children's cognitive functions, but the neuroimaging mechanism of cognitive impairment is still unclear. The purpose of our study was to explore the difference in brain local gray matter volume (GMV) between children with OSA and non-OSA, and the correlation between the difference regions of brain gray matter volume and cognitive, the severity of OSA. Method Eighty-three children aged 8-13 years were recruited in our study, 52 children were diagnosed as OSA by polysomnography, and 31 as the non-OSA. All the subjects were underwent high-resolution 3-dimensional T1-weighted magnetic resonance images. The voxel-based morphometry (VBM) was be used to analyse the local GMV. The Das-Naglieri cognitive assessment system (DN: CAS) was used to assess the subjects' cognitive. The difference of local GMV between the two groups was analyzed by two-sample T-test. The PSG variables and the scores of DN: CAS between the OSA group and non-OSA group were compared by independent samples t-tests. Pearson correlation was used to calculate the association between the difference areas of gray matter volumes in brain and DN: CAS scores, obstructive apnea/hypopnea index (OAHI, an index of the severity of OSA). Results The gray matter volume of the right Middle Frontal Gyrus (MFG_R) in OSA children were larger than the non-OSA children, and the OSA children had lower scores of the Word Series in DN: CAS. There was negative correlation between the scores of Expressive Attention in DN: CAS and the gray matter volume of the right middle frontal gyrus, and it was no significantly correlation between OAHI and the gray matter volume of the right middle frontal gyrus. Conclusion Our results suggest that the development of gray matter volume in frontal cortex, which associated with attention, were sensitive to the effects of OSA, provides neuroimaging evidence for cognitive impairment in children with OSA.
Collapse
Affiliation(s)
- Chenyi Yu
- Department of Pediatrics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yuchuan Fu
- Department of Radiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yi Lu
- Department of Radiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yinyin Huang
- Department of Radiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Fangfang Chen
- Department of Radiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jiayun Wei
- Department of Pediatrics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Lingling Li
- Department of Pediatrics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Janet Akoto Ampadu
- Department of Pediatrics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yu Wang
- Department of Radiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Weikun Zheng
- Department of Pediatrics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Changcan Jiang
- Department of Otorhinolaryngology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Weiyuan Li
- Department of Pneumology, Yuxi Children's Hospital, Yuxi, China
| | - Su Lui
- Department of Radiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
- Department of Radiology, West China Hospital of Sichuan University, Chengdu, China
| | - Xiaohong Cai
- Department of Pediatrics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
8
|
Feng J, Men W, Yu X, Liu W, Zhang S, Liu J, Ma L. High-altitude exposure duration dependent global and regional gray matter volume decrease in healthy immigrants: a cross-sectional study. Acta Radiol 2023; 64:751-759. [PMID: 35369766 DOI: 10.1177/02841851221091674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND The correlation between brain injury and high-altitude (HA) exposure duration (Dur_HA) as well as peripheral oxygen saturation (SpO2) remains unclear. PURPOSE To evaluate the global and regional brain volume differences between HA immigrants and sea-level residents, and the relationship between brain volume with Dur_HA and SpO2. MATERIAL AND METHODS Structural magnetic resonance imaging (MRI) scans were acquired in 33 healthy male HA immigrants (HA group) and 33 matched sea-level male residents (SL group). Differences in global gray matter volume (GMV), white matter volume (WMV), brain parenchyma volume (BV), total intracranial volume (TIV), and the volume-fraction (the ratio of GMV/TIV, WMV/TIV, BV/TIV) were assessed. Regional gray matter differences were investigated using voxel-based morphology analysis. The volume of clusters with GM loss were calculated as the volume of volume of interest (V_VOI). Student's t-test and partial correlation were adopted for statistic calculation. RESULTS Compared to the SL group, the HA immigrants had larger WMV (P = 0.015), smaller ratio of GMV/WMV (P = 0.022), and regional gray matter loss in bilateral basal ganglion, limbic system, midbrain, and vermis (cluster size >100 voxels, family-wise error corrected at P = 0.01). The global GMV, BV, and V_VOI confined to vermis had negative correlations with the Dur_HA (r = -0.369, P = 0.049; r = -0.380, P = 0.042; and r = -0.471, P = 0.010. Neither global nor regional brain volume correlated with SpO2. CONCLUSION Global and regional brain are affected by long-term HA exposure, and global and regional gray matter have a time-dependent volume loss.
Collapse
Affiliation(s)
- Jie Feng
- 104607Medical School of Chinese People's Liberation Army, Beijing, PR China
- Department of Radiology, The First Medical Center of Chinese People's Liberation Army General Hospital, Beijing, PR China
- Department of Radiology, Corps Hospital of Shanxi Province of Chinese People's Armed Police Force, Taiyuan, PR China
| | - Weiwei Men
- Center for MRI Research, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, PR China
- Beijing City Key Lab for Medical Physics and Engineering, Institute of Heavy Ion Physics, School of Physics, Peking University, Beijing, PR China
| | - Xiao Yu
- 104607Medical School of Chinese People's Liberation Army, Beijing, PR China
- Department of Radiology, The First Medical Center of Chinese People's Liberation Army General Hospital, Beijing, PR China
| | - Wenjia Liu
- Department of Radiology, The First Medical Center of Chinese People's Liberation Army General Hospital, Beijing, PR China
| | - Shiyu Zhang
- 104607Medical School of Chinese People's Liberation Army, Beijing, PR China
- Department of Radiology, Beijing Friendship Hospital, 535066Capital Medical University, Beijing, PR China
| | - Jie Liu
- Department of Radiology, General Hospital of Tibet Military Region, Lhasa, Tibet, PR China
| | - Lin Ma
- 104607Medical School of Chinese People's Liberation Army, Beijing, PR China
- Department of Radiology, The First Medical Center of Chinese People's Liberation Army General Hospital, Beijing, PR China
| |
Collapse
|
9
|
Lee SA, Im K, Seo JY, Jung M. Association between sleep apnea severity and symptoms of depression and anxiety among individuals with obstructive sleep apnea. Sleep Med 2023; 101:11-18. [PMID: 36332382 DOI: 10.1016/j.sleep.2022.09.023] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Revised: 09/20/2022] [Accepted: 09/26/2022] [Indexed: 01/09/2023]
Abstract
OBJECTIVE We determined whether the severity of sleep apnea is associated with symptoms of depression and anxiety in patients with obstructive sleep apnea (OSA) and whether symptoms of OSA, other than respiratory events, are associated with depression and anxiety symptoms. METHODS Depressive and anxiety symptoms were defined as a Patient Health Questionnaire-9 score of ≥10 and a Generalized Anxiety Disorder-7 score of ≥8, respectively. Sleep apnea severity and rapid eye movement-related OSA were evaluated using the apnea-hypopnea index (AHI). Subjective symptoms of OSA were assessed using the Sleep Apnea Severity Questionnaire (SASQ). We conducted multivariate logistic regression analyses. RESULTS We included 1390 adult patients with OSA (80.4% men) and 125 control subjects with primary snoring. Symptoms of depression and anxiety were present in 15.5% and 14.4% of patients with OSA, respectively. The prevalence of depressive and anxiety symptoms did not differ between OSA and control subjects after controlling for age and sex. Severe OSA, defined as an AHI score of ≥30, was significantly less likely than mild OSA to be associated with the presence of depression and anxiety symptoms in both the unadjusted and adjusted models (models were adjusted by age, sex, medical comorbidities, history of psychiatric disorders, and daytime sleepiness). By contrast, OSA symptoms assessed using the SASQ were positively correlated with the presence of depression and anxiety symptoms in both the unadjusted and adjusted models. Specifically, nocturnal awakening and morning waking symptoms, but not nocturnal breathing difficulties, were positively correlated with the presence of depression and anxiety symptoms. Subjects with rapid eye movement-related OSA were more likely to have depressive symptoms, but not anxiety, than those without. CONCLUSIONS Symptoms of depression and anxiety were negatively correlated with the severity of sleep apnea but positively correlated with nocturnal awakening and early morning symptoms of OSA.
Collapse
Affiliation(s)
- Sang-Ahm Lee
- Department of Neurology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.
| | - Kayeong Im
- Department of Neurology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Jin Young Seo
- Department of Neurology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Mina Jung
- Department of Neurology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
10
|
Nacafaliyev V, Ortan P, Sayin SS. Relationship between obstructive sleep apnoea syndrome and silent brain infarction. Postgrad Med J 2022:7148069. [PMID: 37130819 DOI: 10.1136/pmj-2022-141911] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 09/14/2022] [Indexed: 11/03/2022]
Abstract
BACKGROUND The relationship between obstructive sleep apnea syndrome (OSAS) and ischaemic stroke is less known. OBJECTIVES This study aimed to investigate the relationship between OSAS and silent brain infarcts (SBI). METHODS Patients who applied to our clinic with the complaint of snoring, respiratory arrest during sleep, that underwent polysomnography were included. All patients were undergone cranial magnetic resonance imaging to detect SBI. RESULTS SBI was found in 176 (51.5%) of 270 patients in the group with OSAS and 94 (34.8%) patients without OSAS. The patients were evaluated according to their Apnea-Hypopnea Index(AHI) ratio, and those with were found to be significant in terms of SBI. SBI was detected in 56.56% in the moderate and severe (AHI ˃15) stage group and 39.94% in the normal and mild (AHI ≤15) OSAS group (p=0.009). CONCLUSIONS SBI was found to be significantly higher in patients with moderate and severe stage OSAS compared to the normal and mild OSAS group. Desaturations during sleep may influence the formation of these infarcts. Therefore, this study reported that patients with moderate and severe sleep apnea syndrome may have a higher risk of developing ischaemic cerebrovascular disease and that the treatment of these patients should be planned in this respect.
Collapse
Affiliation(s)
- Vusal Nacafaliyev
- Department of Neurology, University of Health Sciences Izmir Bozyaka Education and Research Hospital, Izmir, Turkey
| | - Pınar Ortan
- Neurology, University of Health Sciences Izmir Bozyaka Education and Research Hospital, Izmir, Turkey
| | - Sevgi Sidika Sayin
- Department of Neurology, University of Health Sciences Izmir Bozyaka Education and Research Hospital, Izmir, Turkey
| |
Collapse
|
11
|
Enhanced Ocular Surface and Intraoral Nociception via a Transient Receptor Potential Vanilloid 1 Mechanism in a Rat Model of Obstructive Sleep Apnea. Neuroscience 2021; 483:66-81. [PMID: 34883200 DOI: 10.1016/j.neuroscience.2021.12.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 11/29/2021] [Accepted: 12/01/2021] [Indexed: 12/28/2022]
Abstract
Obstructive sleep apnea (OSA), characterized by low arterial oxygen saturation during sleep, is associated with an increased risk of orofacial pain. In this study, we simulated chronic intermittent hypoxia (CIH) during the sleep/rest phase (light phase) to determine the role of transient receptor potential vanilloid 1 (TRPV1) in mediating enhanced orofacial nocifensive behavior and trigeminal spinal subnucleus caudalis (Vc) neuronal responses to capsaicin (a TRPV1 agonist) stimulation in a rat model of OSA. Rats were subjected to CIH (nadir O2, 5%) during the light phase for 8 or 16 consecutive days. CIH yielded enhanced behavioral responses to capsaicin after application to the ocular surface and intraoral mucosa, which was reversed under normoxic conditions. The percentage of TRPV1-immunoreactive trigeminal ganglion neurons was greater in CIH rats than in normoxic rats and recovered under normoxic conditions after CIH. The ratio of large-sized TRPV1-immunoreactive trigeminal ganglion neurons increased in CIH rats. The density of TRPV1 positive primary afferent terminals in the superficial laminae of Vc was higher in CIH rats. Phosphorylated extracellular signal-regulated kinase (pERK)-immunoreactive cells intermingled with the central terminal of TRPV1-positive afferents in the Vc. The number of pERK-immunoreactive cells following low-dose capsaicin (0.33 µM) application to the tongue was significantly greater in the middle portion of the Vc of CIH rats than of normoxic rats and recovered under normoxic conditions after CIH. These data suggest that CIH during the sleep (light) phase is sufficient to transiently enhance pain on the ocular surface and intraoral mucosa via TRPV1-dependent mechanisms.
Collapse
|
12
|
Bonilla-Jaime H, Zeleke H, Rojas A, Espinosa-Garcia C. Sleep Disruption Worsens Seizures: Neuroinflammation as a Potential Mechanistic Link. Int J Mol Sci 2021; 22:12531. [PMID: 34830412 PMCID: PMC8617844 DOI: 10.3390/ijms222212531] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2021] [Revised: 11/16/2021] [Accepted: 11/18/2021] [Indexed: 12/13/2022] Open
Abstract
Sleep disturbances, such as insomnia, obstructive sleep apnea, and daytime sleepiness, are common in people diagnosed with epilepsy. These disturbances can be attributed to nocturnal seizures, psychosocial factors, and/or the use of anti-epileptic drugs with sleep-modifying side effects. Epilepsy patients with poor sleep quality have intensified seizure frequency and disease progression compared to their well-rested counterparts. A better understanding of the complex relationship between sleep and epilepsy is needed, since approximately 20% of seizures and more than 90% of sudden unexpected deaths in epilepsy occur during sleep. Emerging studies suggest that neuroinflammation, (e.g., the CNS immune response characterized by the change in expression of inflammatory mediators and glial activation) may be a potential link between sleep deprivation and seizures. Here, we review the mechanisms by which sleep deprivation induces neuroinflammation and propose that neuroinflammation synergizes with seizure activity to worsen neurodegeneration in the epileptic brain. Additionally, we highlight the relevance of sleep interventions, often overlooked by physicians, to manage seizures, prevent epilepsy-related mortality, and improve quality of life.
Collapse
Affiliation(s)
- Herlinda Bonilla-Jaime
- Departamento de Biología de la Reproducción, Área de Biología Conductual y Reproductiva, Universidad Autónoma Metropolitana-Iztapalapa, Ciudad de Mexico CP 09340, Mexico;
| | - Helena Zeleke
- Neuroscience and Behavioral Biology Program, College of Arts and Sciences, Emory University, Atlanta, GA 30322, USA;
| | - Asheebo Rojas
- Department of Pharmacology and Chemical Biology, School of Medicine, Emory University, Atlanta, GA 30322, USA
| | - Claudia Espinosa-Garcia
- Department of Pharmacology and Chemical Biology, School of Medicine, Emory University, Atlanta, GA 30322, USA
| |
Collapse
|
13
|
Czornyj L, Auzmendi J, Lazarowski A. Transporter hypothesis in pharmacoresistant epilepsies Is it at the central or peripheral level? Epilepsia Open 2021; 7 Suppl 1:S34-S46. [PMID: 34542938 PMCID: PMC9340303 DOI: 10.1002/epi4.12537] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 08/30/2021] [Accepted: 08/31/2021] [Indexed: 12/22/2022] Open
Abstract
The multidrug‐resistance (MDR) phenotype is typically observed in patients with refractory epilepsy (RE) whose seizures are not controlled despite receiving several combinations of more than two antiseizure medications (ASMs) directed against different ion channels or neurotransmitter receptors. Since the use of bromide in 1860, more than 20 ASMs have been developed; however, historically ~30% of cases of RE with MDR phenotype remains unchanged. Irrespective of metabolic biotransformation, the biodistribution of ASMs and their metabolites depends on the functional expression of some ATP‐binding cassette transporters (ABC‐t) in different organs, such as the blood‐brain barrier (BBB), bowel, liver, and kidney, among others. ABC‐t, such as P‐glycoprotein (P‐gp), multidrug resistance–associated protein (MRP‐1), and breast cancer–resistance protein (BCRP), are mainly expressed in excretory organs and play a critical role in the pharmacokinetics (PK) of all drugs. The transporter hypothesis can explain pharmacoresistance to a broad spectrum of ASMs, even when administered simultaneously. Since ABC‐t expression can be induced by hypoxia, inflammation, or seizures, a high frequency of uncontrolled seizures increases the risk of RE. These stimuli can induce ABC‐t expression in excretory organs and in previously non‐expressing (electrically responsive) cells, such as neurons or cardiomyocytes. In this regard, an alternative mechanism to the classical pumping function of P‐gp indicates that P‐gp activity can also produce a significant reduction in resting membrane potential (ΔΨ0 = −60 to −10 mV). P‐gp expression in neurons and cardiomyocytes can produce membrane depolarization and participate in epileptogenesis, heart failure, and sudden unexpected death in epilepsy. On this basis, ABC‐t play a peripheral role in controlling the PK of ASMs and their access to the brain and act at a central level, favoring neuronal depolarization by mechanisms independent of ion channels or neurotransmitters that current ASMs cannot control.
Collapse
Affiliation(s)
- Liliana Czornyj
- Neurology Service, "Juan P. Garrahan" National Children's Hospital, Buenos Aires, Argentina
| | - Jerónimo Auzmendi
- Institute for Research in Physiopathology and Clinical Biochemistry (INFIBIOC), Clinical Biochemistry Department, School of Pharmacy and Biochemistry, University of Buenos Aires, Buenos Aires, Argentina.,National Council for Scientific and Technical Research (CONICET), Buenos Aires, Argentina
| | - Alberto Lazarowski
- Institute for Research in Physiopathology and Clinical Biochemistry (INFIBIOC), Clinical Biochemistry Department, School of Pharmacy and Biochemistry, University of Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
14
|
Intermittent Hypoxia and Effects on Early Learning/Memory: Exploring the Hippocampal Cellular Effects of Pediatric Obstructive Sleep Apnea. Anesth Analg 2021; 133:93-103. [PMID: 33234943 DOI: 10.1213/ane.0000000000005273] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
This review provides an update on the neurocognitive phenotype of pediatric obstructive sleep apnea (OSA). Pediatric OSA is associated with neurocognitive deficits involving memory, learning, and executive functioning. Adenotonsillectomy (AT) is presently accepted as the first-line surgical treatment for pediatric OSA, but the executive function deficits do not resolve postsurgery, and the timeline for recovery remains unknown. This finding suggests that pediatric OSA potentially causes irreversible damage to multiple areas of the brain. The focus of this review is the hippocampus, 1 of the 2 major sites of postnatal neurogenesis, where new neurons are formed and integrated into existing circuitry and the mammalian center of learning/memory functions. Here, we review the clinical phenotype of pediatric OSA, and then discuss existing studies of OSA on different cell types in the hippocampus during critical periods of development. This will set the stage for future study using preclinical models to understand the pathogenesis of persistent neurocognitive dysfunction in pediatric OSA.
Collapse
|
15
|
Villarreal A, Vidos C, Monteverde Busso M, Cieri MB, Ramos AJ. Pathological Neuroinflammatory Conversion of Reactive Astrocytes Is Induced by Microglia and Involves Chromatin Remodeling. Front Pharmacol 2021; 12:689346. [PMID: 34234677 PMCID: PMC8255379 DOI: 10.3389/fphar.2021.689346] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 05/25/2021] [Indexed: 01/06/2023] Open
Abstract
Following brain injury or in neurodegenerative diseases, astrocytes become reactive and may suffer pathological remodeling, features of which are the loss of their homeostatic functions and a pro-inflammatory gain of function that facilitates neurodegeneration. Pharmacological intervention to modulate this astroglial response and neuroinflammation is an interesting new therapeutic research strategy, but it still requires a deeper understanding of the underlying cellular and molecular mechanisms of the phenomenon. Based on the known microglial–astroglial interaction, the prominent role of the nuclear factor kappa B (NF-κB) pathway in mediating astroglial pathological pro-inflammatory gain of function, and its ability to recruit chromatin-remodeling enzymes, we first explored the microglial role in the initiation of astroglial pro-inflammatory conversion and then monitored the progression of epigenetic changes in the astrocytic chromatin. Different configurations of primary glial culture were used to modulate microglia–astrocyte crosstalk while inducing pro-inflammatory gain of function by lipopolysaccharide (LPS) exposure. In vivo, brain ischemia by cortical devascularization (pial disruption) was performed to verify the presence of epigenetic marks in reactive astrocytes. Our results showed that 1) microglia is required to initiate the pathological conversion of astrocytes by triggering the NF-κB signaling pathway; 2) this interaction is mediated by soluble factors and induces stable astroglial phenotypic changes; 3) the pathological conversion promotes chromatin remodeling with stable increase in H3K9K14ac, temporary increase in H3K27ac, and temporary reduction in heterochromatin mark H3K9me3; and 4) in vivo reactive astrocytes show increased H3K27ac mark in the neuroinflammatory milieu from the ischemic penumbra. Our findings indicate that astroglial pathological pro-inflammatory gain of function is associated with profound changes in the configuration of astrocytic chromatin, which in turn are initiated by microglia-derived cues. These results open a new avenue in the study of potential pharmacological interventions that modify the initiation and stabilization of astroglial pathological remodeling, which would be useful in acute and chronic CNS injury. Epigenetic changes represent a plausible pharmacological target to interfere with the stabilization of the pathological astroglial phenotype.
Collapse
Affiliation(s)
- Alejandro Villarreal
- Laboratorio de Neuropatología Molecular, Instituto de Biología Celular y Neurociencia "Prof. E. De Robertis" UBA-CONICET, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Camila Vidos
- Laboratorio de Neuropatología Molecular, Instituto de Biología Celular y Neurociencia "Prof. E. De Robertis" UBA-CONICET, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Matías Monteverde Busso
- Laboratorio de Neuropatología Molecular, Instituto de Biología Celular y Neurociencia "Prof. E. De Robertis" UBA-CONICET, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina.,Primera Unidad Académica de Histología, Embriología, Biología Celular y Genética, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - María Belén Cieri
- Laboratorio de Neuropatología Molecular, Instituto de Biología Celular y Neurociencia "Prof. E. De Robertis" UBA-CONICET, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina.,Primera Unidad Académica de Histología, Embriología, Biología Celular y Genética, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Alberto Javier Ramos
- Laboratorio de Neuropatología Molecular, Instituto de Biología Celular y Neurociencia "Prof. E. De Robertis" UBA-CONICET, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina.,Primera Unidad Académica de Histología, Embriología, Biología Celular y Genética, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
16
|
Hofmann GC, Hasser EM, Kline DD. Unilateral vagotomy alters astrocyte and microglial morphology in the nucleus tractus solitarii of the rat. Am J Physiol Regul Integr Comp Physiol 2021; 320:R945-R959. [PMID: 33978480 PMCID: PMC8285617 DOI: 10.1152/ajpregu.00019.2021] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 04/09/2021] [Accepted: 05/04/2021] [Indexed: 12/11/2022]
Abstract
The nucleus tractus solitarii (nTS) is the initial site of integration of sensory information from the cardiorespiratory system and contributes to reflex responses to hypoxia. Afferent fibers of the bilateral vagus nerves carry input from the heart, lungs, and other organs to the nTS where it is processed and modulated. Vagal afferents and nTS neurons are integrally associated with astrocytes and microglia that contribute to neuronal activity and influence cardiorespiratory control. We hypothesized that vagotomy would alter glial morphology and cardiorespiratory responses to hypoxia. Unilateral vagotomy (or sham surgery) was performed in rats. Prior to and seven days after surgery, baseline and hypoxic cardiorespiratory responses were monitored in conscious and anesthetized animals. The brainstem was sectioned and caudal, mid-area postrema (mid-AP), and rostral sections of the nTS were prepared for immunohistochemistry. Vagotomy increased immunoreactivity (-IR) of astrocytic glial fibrillary acidic protein (GFAP), specifically at mid-AP in the nTS. Similar results were found in the dorsal motor nucleus of the vagus (DMX). Vagotomy did not alter nTS astrocyte number, yet increased astrocyte branching and altered morphology. In addition, vagotomy both increased nTS microglia number and produced morphologic changes indicative of activation. Cardiorespiratory baseline parameters and hypoxic responses remained largely unchanged, but vagotomized animals displayed fewer augmented breaths (sighs) in response to hypoxia. Altogether, vagotomy alters nTS glial morphology, indicative of functional changes in astrocytes and microglia that may affect cardiorespiratory function in health and disease.
Collapse
Affiliation(s)
- Gabrielle C Hofmann
- Comparative Medicine, University of Missouri, Columbia, Missouri
- Area Pathobiology, University of Missouri, Columbia, Missouri
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri
| | - Eileen M Hasser
- Area Pathobiology, University of Missouri, Columbia, Missouri
- Department of Biomedical Sciences, University of Missouri, Columbia, Missouri
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri
| | - David D Kline
- Area Pathobiology, University of Missouri, Columbia, Missouri
- Department of Biomedical Sciences, University of Missouri, Columbia, Missouri
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri
| |
Collapse
|
17
|
Kaliszewska A, Allison J, Martini M, Arias N. Improving Age-Related Cognitive Decline through Dietary Interventions Targeting Mitochondrial Dysfunction. Int J Mol Sci 2021; 22:ijms22073574. [PMID: 33808221 PMCID: PMC8036520 DOI: 10.3390/ijms22073574] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 03/24/2021] [Accepted: 03/25/2021] [Indexed: 12/13/2022] Open
Abstract
Aging is inevitable and it is one of the major contributors to cognitive decline. However, the mechanisms underlying age-related cognitive decline are still the object of extensive research. At the biological level, it is unknown how the aging brain is subjected to progressive oxidative stress and neuroinflammation which determine, among others, mitochondrial dysfunction. The link between mitochondrial dysfunction and cognitive impairment is becoming ever more clear by the presence of significant neurological disturbances in human mitochondrial diseases. Possibly, the most important lifestyle factor determining mitochondrial functioning is nutrition. Therefore, with the present work, we review the latest findings disclosing a link between nutrition, mitochondrial functioning and cognition, and pave new ways to counteract cognitive decline in late adulthood through diet.
Collapse
Affiliation(s)
- Aleksandra Kaliszewska
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, Denmark Hill, London SE5 8AF, UK; (A.K.); (J.A.)
| | - Joseph Allison
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, Denmark Hill, London SE5 8AF, UK; (A.K.); (J.A.)
| | - Matteo Martini
- Department of Psychology, University of East London, London E154LZ, UK;
| | - Natalia Arias
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, Denmark Hill, London SE5 8AF, UK; (A.K.); (J.A.)
- Instituto de Neurociencias del Principado de Asturias (INEUROPA), 33005 Oviedo, Spain
- Correspondence:
| |
Collapse
|
18
|
Ferini-Strambi L, Hensley M, Salsone M. Decoding Causal Links Between Sleep Apnea and Alzheimer’s Disease. J Alzheimers Dis 2021; 80:29-40. [DOI: 10.3233/jad-201066] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Obstructive sleep apnea (OSA) and Alzheimer’s disease (AD) are two common chronic diseases with a well-documented association. Whether the association is causal has been highlighted by recent evidence reporting a neurobiological link between these disorders. This narrative review discusses the brain regions and networks involved in OSA as potential vulnerable areas for the development of AD neuropathology with a particular focus on gender-related implications. Using a neuroimaging perspective supported by neuropathological investigations, we provide a new model of neurodegeneration common to OSA and AD, that we have called OSA-AD neurodegeneration in order to decode the causal links between these two chronic conditions.
Collapse
Affiliation(s)
| | - Michael Hensley
- John Hunter Hospital and The University of Newcastle, Newcastle, Australia
| | - Maria Salsone
- IRCCS San Raffaele Scientific Institute, Department of Clinical Neurosciences, Neurology-Sleep Disorder Center, Milan, Italy
- Institute of Molecular Bioimaging and Physiology, National Research Council, Catanzaro, Italy
| |
Collapse
|
19
|
André C, Rehel S, Kuhn E, Landeau B, Moulinet I, Touron E, Ourry V, Le Du G, Mézenge F, Tomadesso C, de Flores R, Bejanin A, Sherif S, Delcroix N, Manrique A, Abbas A, Marchant NL, Lutz A, Klimecki OM, Collette F, Arenaza-Urquijo EM, Poisnel G, Vivien D, Bertran F, de la Sayette V, Chételat G, Rauchs G. Association of Sleep-Disordered Breathing With Alzheimer Disease Biomarkers in Community-Dwelling Older Adults: A Secondary Analysis of a Randomized Clinical Trial. JAMA Neurol 2021; 77:716-724. [PMID: 32202593 DOI: 10.1001/jamaneurol.2020.0311] [Citation(s) in RCA: 75] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Importance Increasing evidence suggests that sleep-disordered breathing (SDB) increases the risk of developing Alzheimer clinical syndrome. However, the brain mechanisms underlying the link between SDB and Alzheimer disease are still unclear. Objective To determine which brain changes are associated with the presence of SDB in older individuals who are cognitively unimpaired, including changes in amyloid deposition, gray matter volume, perfusion, and glucose metabolism. Design, Setting, and Participants This cross-sectional study was conducted using data from the Age-Well randomized clinical trial of the Medit-Ageing European project, acquired between 2016 and 2018 at Cyceron Center in Caen, France. Community-dwelling older adults were assessed for eligibility and were enrolled in the Age-Well clinical trial if they did not meet medical or cognitive exclusion criteria and were willing to participate. Participants who completed a detailed neuropsychological assessment, polysomnography, a magnetic resonance imaging, and florbetapir and fluorodeoxyglucose positron emission tomography scans were included in the analyses. Main Outcomes and Measures Based on an apnea-hypopnea index cutoff of 15 events per hour, participants were classified as having SDB or not. Voxelwise between-group comparisons were performed for each neuroimaging modality, and secondary analyses aimed at identifying which SDB parameter (sleep fragmentation, hypoxia severity, or frequency of respiratory disturbances) best explained the observed brain changes and assessing whether SDB severity and/or SDB-associated brain changes are associated with cognitive and behavioral changes. Results Of 157 participants initially assessed, 137 were enrolled in the Age-Well clinical trial, and 127 were analyzed in this study. The mean (SD) age of the 127 participants was 69.1 (3.9) years, and 80 (63.0%) were women. Participants with SDB showed greater amyloid burden (t114 = 4.51; familywise error-corrected P = .04; Cohen d, 0.83), gray matter volume (t119 = 4.12; familywise error-corrected P = .04; Cohen d, 0.75), perfusion (t116 = 4.62; familywise error-corrected P = .001; Cohen d, 0.86), and metabolism (t79 = 4.63; familywise error-corrected P = .001; Cohen d, 1.04), overlapping mainly over the posterior cingulate cortex and precuneus. No association was found with cognition, self-reported cognitive and sleep difficulties, or excessive daytime sleepiness symptoms. Conclusions and Relevance The SDB-associated brain changes in older adults who are cognitively unimpaired include greater amyloid deposition and neuronal activity in Alzheimer disease-sensitive brain regions, notably the posterior cingulate cortex and precuneus. These results support the need to screen and treat for SDB, especially in asymptomatic older populations, to reduce Alzheimer disease risk. Trial Registration ClinicalTrials.gov Identifier: NCT02977819.
Collapse
Affiliation(s)
- Claire André
- Normandie Université, Université de Caen, Institut National de la Santé et de la Recherche Médicale, Unité 1237 "Physiopathology and Imaging of Neurological Disorders," Institut Blood and Brain @ Caen-Normandie, GIP Cyceron, Caen, France.,Normandie Université, Université de Caen, Paris Sciences & Lettres Université, École Pratique des Hautes Études, Institut National de la Santé et de la Recherche Médicale, Unité 1077 "Neuropsychologie et Imagerie de la Mémoire Humaine," Centre Hospitalier Universitaire de Caen, GIP Cyceron, Caen, France
| | - Stéphane Rehel
- Normandie Université, Université de Caen, Institut National de la Santé et de la Recherche Médicale, Unité 1237 "Physiopathology and Imaging of Neurological Disorders," Institut Blood and Brain @ Caen-Normandie, GIP Cyceron, Caen, France.,Normandie Université, Université de Caen, Paris Sciences & Lettres Université, École Pratique des Hautes Études, Institut National de la Santé et de la Recherche Médicale, Unité 1077 "Neuropsychologie et Imagerie de la Mémoire Humaine," Centre Hospitalier Universitaire de Caen, GIP Cyceron, Caen, France
| | - Elizabeth Kuhn
- Normandie Université, Université de Caen, Institut National de la Santé et de la Recherche Médicale, Unité 1237 "Physiopathology and Imaging of Neurological Disorders," Institut Blood and Brain @ Caen-Normandie, GIP Cyceron, Caen, France
| | - Brigitte Landeau
- Normandie Université, Université de Caen, Institut National de la Santé et de la Recherche Médicale, Unité 1237 "Physiopathology and Imaging of Neurological Disorders," Institut Blood and Brain @ Caen-Normandie, GIP Cyceron, Caen, France
| | - Inès Moulinet
- Normandie Université, Université de Caen, Institut National de la Santé et de la Recherche Médicale, Unité 1237 "Physiopathology and Imaging of Neurological Disorders," Institut Blood and Brain @ Caen-Normandie, GIP Cyceron, Caen, France
| | - Edelweiss Touron
- Normandie Université, Université de Caen, Institut National de la Santé et de la Recherche Médicale, Unité 1237 "Physiopathology and Imaging of Neurological Disorders," Institut Blood and Brain @ Caen-Normandie, GIP Cyceron, Caen, France
| | - Valentin Ourry
- Normandie Université, Université de Caen, Paris Sciences & Lettres Université, École Pratique des Hautes Études, Institut National de la Santé et de la Recherche Médicale, Unité 1077 "Neuropsychologie et Imagerie de la Mémoire Humaine," Centre Hospitalier Universitaire de Caen, GIP Cyceron, Caen, France
| | - Gwendoline Le Du
- Normandie Université, Université de Caen, Institut National de la Santé et de la Recherche Médicale, Unité 1237 "Physiopathology and Imaging of Neurological Disorders," Institut Blood and Brain @ Caen-Normandie, GIP Cyceron, Caen, France
| | - Florence Mézenge
- Normandie Université, Université de Caen, Institut National de la Santé et de la Recherche Médicale, Unité 1237 "Physiopathology and Imaging of Neurological Disorders," Institut Blood and Brain @ Caen-Normandie, GIP Cyceron, Caen, France
| | - Clémence Tomadesso
- Normandie Université, Université de Caen, Institut National de la Santé et de la Recherche Médicale, Unité 1237 "Physiopathology and Imaging of Neurological Disorders," Institut Blood and Brain @ Caen-Normandie, GIP Cyceron, Caen, France
| | - Robin de Flores
- Normandie Université, Université de Caen, Institut National de la Santé et de la Recherche Médicale, Unité 1237 "Physiopathology and Imaging of Neurological Disorders," Institut Blood and Brain @ Caen-Normandie, GIP Cyceron, Caen, France
| | - Alexandre Bejanin
- Normandie Université, Université de Caen, Institut National de la Santé et de la Recherche Médicale, Unité 1237 "Physiopathology and Imaging of Neurological Disorders," Institut Blood and Brain @ Caen-Normandie, GIP Cyceron, Caen, France
| | - Siya Sherif
- Normandie Université, Université de Caen, Institut National de la Santé et de la Recherche Médicale, Unité 1237 "Physiopathology and Imaging of Neurological Disorders," Institut Blood and Brain @ Caen-Normandie, GIP Cyceron, Caen, France
| | - Nicolas Delcroix
- Centre National de la Recherche Scientifique, Unité Mixte de Service 3048, GIP Cyceron, Caen, France
| | - Alain Manrique
- Normandie Université, Université de Caen, EA 4650 "Signalisation, Électrophysiologie et Imagerie des Lésions d'Ischémie-Reperfusion Myocardique", GIP Cyceron, Caen, France
| | - Ahmed Abbas
- Normandie Université, Université de Caen, Paris Sciences & Lettres Université, École Pratique des Hautes Études, Institut National de la Santé et de la Recherche Médicale, Unité 1077 "Neuropsychologie et Imagerie de la Mémoire Humaine," Centre Hospitalier Universitaire de Caen, GIP Cyceron, Caen, France
| | - Natalie L Marchant
- Division of Psychiatry, University College London, London, United Kingdom
| | - Antoine Lutz
- Lyon Neuroscience Research Center, Institut National de la Santé et de la Recherche Médicale Unité 1028, Centre National de la Recherche Scientifique Unité Mixte de Recherche 5292, Lyon University, Lyon, France
| | - Olga M Klimecki
- Swiss Center for Affective Sciences, Department of Medicine, University of Geneva, Geneva, Switzerland
| | - Fabienne Collette
- GIGA-Cyclotron Research Centre, In Vivo Imaging and Psychology and Cognitive Neuroscience Unit, Liège University, Liège, Belgium
| | - Eider M Arenaza-Urquijo
- Normandie Université, Université de Caen, Institut National de la Santé et de la Recherche Médicale, Unité 1237 "Physiopathology and Imaging of Neurological Disorders," Institut Blood and Brain @ Caen-Normandie, GIP Cyceron, Caen, France
| | - Géraldine Poisnel
- Normandie Université, Université de Caen, Institut National de la Santé et de la Recherche Médicale, Unité 1237 "Physiopathology and Imaging of Neurological Disorders," Institut Blood and Brain @ Caen-Normandie, GIP Cyceron, Caen, France
| | - Denis Vivien
- Normandie Université, Université de Caen, Institut National de la Santé et de la Recherche Médicale, Unité 1237 "Physiopathology and Imaging of Neurological Disorders," Institut Blood and Brain @ Caen-Normandie, GIP Cyceron, Caen, France.,Département de Recherche Clinique, Centre Hospitalier Universitaire de Caen-Normandie, Caen, France
| | - Françoise Bertran
- Unité d'Exploration et de Traitement des Troubles du Sommeil, Centre Hospitalier Universitaire de Caen, Caen, France
| | - Vincent de la Sayette
- Normandie Université, Université de Caen, Paris Sciences & Lettres Université, École Pratique des Hautes Études, Institut National de la Santé et de la Recherche Médicale, Unité 1077 "Neuropsychologie et Imagerie de la Mémoire Humaine," Centre Hospitalier Universitaire de Caen, GIP Cyceron, Caen, France.,Service de Neurologie, Centre Hospitalier Universitaire de Caen, Caen, France
| | - Gaël Chételat
- Normandie Université, Université de Caen, Institut National de la Santé et de la Recherche Médicale, Unité 1237 "Physiopathology and Imaging of Neurological Disorders," Institut Blood and Brain @ Caen-Normandie, GIP Cyceron, Caen, France
| | - Géraldine Rauchs
- Normandie Université, Université de Caen, Paris Sciences & Lettres Université, École Pratique des Hautes Études, Institut National de la Santé et de la Recherche Médicale, Unité 1077 "Neuropsychologie et Imagerie de la Mémoire Humaine," Centre Hospitalier Universitaire de Caen, GIP Cyceron, Caen, France
| | | |
Collapse
|
20
|
Baril AA, Martineau-Dussault MÈ, Sanchez E, André C, Thompson C, Legault J, Gosselin N. Obstructive Sleep Apnea and the Brain: a Focus on Gray and White Matter Structure. Curr Neurol Neurosci Rep 2021; 21:11. [PMID: 33586028 DOI: 10.1007/s11910-021-01094-2] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/13/2021] [Indexed: 12/24/2022]
Abstract
PURPOSE OF REVIEW Obstructive sleep apnea is extremely prevalent in the elderly and may precipitate dementia. We review recent advances on gray and white matter structure in obstructive sleep apnea, the impact of treatment, and potential pathological and neurodegenerative processes underlying brain structural changes. RECENT FINDINGS Two opposite patterns are observed in neuroimaging studies of obstructive sleep apnea. One may indicate cellular damage (gray matter atrophy, higher white matter hyperintensity burden, lower white matter fractional anisotropy, higher water diffusivities), while the other (gray matter hypertrophy, restricted white matter diffusivities) may reflect transitory responses, such as intracellular edema, reactive gliosis or compensatory structural changes. Treating obstructive sleep apnea could partly reverse these structural changes. Structural alterations related to obstructive sleep apnea may follow a multi-determined biphasic pattern depending on numerous factors (e.g. severity, symptomatology, age) that could tip the scale toward neurodegeneration and need to be investigated by longitudinal studies.
Collapse
Affiliation(s)
- Andrée-Ann Baril
- The Framingham Heart Study, Boston University School of Medicine, Boston, MA, USA
| | - Marie-Ève Martineau-Dussault
- Center for Advanced Research in Sleep Medicine, Centre Intégré Universitaire de Santé et de Services Sociaux du Nord-de-l'Île-de-Montréal, 5400 boul. Gouin Ouest, local J-5135, Montréal, Québec, H4J 1C5, Canada.,Department of Psychology, Université de Montréal, Montréal, Canada
| | - Erlan Sanchez
- Center for Advanced Research in Sleep Medicine, Centre Intégré Universitaire de Santé et de Services Sociaux du Nord-de-l'Île-de-Montréal, 5400 boul. Gouin Ouest, local J-5135, Montréal, Québec, H4J 1C5, Canada.,Department of Neuroscience, Université de Montréal, Montréal, Canada
| | - Claire André
- Physiopathology and Imaging of Neurological Disorders, Institut National de la Santé et de la Recherche Médicale, Institut Blood and Brain, Université de Caen, Normandie Université, GIP Cyceron, Caen, France.,Neuropsychologie et Imagerie de la Mémoire Humain, Institut National de la Santé et de la Recherche Médicale, Centre Hospitalier Universitaire de Caen, Université de Caen, Normandie Université, Paris Sciences & Lettres Université, École Pratique des Hautes Études, GIP Cyceron, Caen, France
| | - Cynthia Thompson
- Center for Advanced Research in Sleep Medicine, Centre Intégré Universitaire de Santé et de Services Sociaux du Nord-de-l'Île-de-Montréal, 5400 boul. Gouin Ouest, local J-5135, Montréal, Québec, H4J 1C5, Canada
| | - Julie Legault
- Center for Advanced Research in Sleep Medicine, Centre Intégré Universitaire de Santé et de Services Sociaux du Nord-de-l'Île-de-Montréal, 5400 boul. Gouin Ouest, local J-5135, Montréal, Québec, H4J 1C5, Canada.,Department of Psychology, Université de Montréal, Montréal, Canada
| | - Nadia Gosselin
- Center for Advanced Research in Sleep Medicine, Centre Intégré Universitaire de Santé et de Services Sociaux du Nord-de-l'Île-de-Montréal, 5400 boul. Gouin Ouest, local J-5135, Montréal, Québec, H4J 1C5, Canada. .,Department of Psychology, Université de Montréal, Montréal, Canada.
| |
Collapse
|
21
|
Auzmendi J, Palestro P, Blachman A, Gavernet L, Merelli A, Talevi A, Calabrese GC, Ramos AJ, Lazarowski A. Cannabidiol (CBD) Inhibited Rhodamine-123 Efflux in Cultured Vascular Endothelial Cells and Astrocytes Under Hypoxic Conditions. Front Behav Neurosci 2020; 14:32. [PMID: 32256321 PMCID: PMC7090129 DOI: 10.3389/fnbeh.2020.00032] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Accepted: 02/17/2020] [Indexed: 12/14/2022] Open
Abstract
Despite the constant development of new antiepileptic drugs (AEDs), more than 30% of patients develop refractory epilepsy (RE) characterized by a multidrug-resistant (MDR) phenotype. The “transporters hypothesis” indicates that the mechanism of this MDR phenotype is the overexpression of ABC transporters such as P-glycoprotein (P-gp) in the neurovascular unit cells, limiting access of the AEDs to the brain. Recent clinical trials and basic studies have shown encouraging results for the use of cannabinoids in RE, although its mechanisms of action are still not fully understood. Here, we have employed astrocytes and vascular endothelial cell cultures subjected to hypoxia, to test the effect of cannabidiol (CBD) on the P-gp-dependent Rhodamine-123 (Rho-123) efflux. Results show that during hypoxia, intracellular Rho-123 accumulation after CBD treatment is similar to that induced by the P-gp inhibitor Tariquidar (Tq). Noteworthy, this inhibition is like that registered in non-hypoxia conditions. Additionally, docking studies predicted that CBD could behave as a P-gp substrate by the interaction with several residues in the α-helix of the P-gp transmembrane domain. Overall, these findings suggest a direct effect of CBD on the Rho-123 P-gp-dependent efflux activity, which might explain why the CBD add-on treatment regimen in RE patients results in a significant reduction in seizure frequency.
Collapse
Affiliation(s)
- Jerónimo Auzmendi
- Instituto de Fisiopatología y Bioquímica Clínica, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina.,Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
| | - Pablo Palestro
- Laboratorio de Investigaciones Bioactivas y Desarrollo, Departamento de Ciencias Biológicas, Facultad de Ciencias Exactas, Universidad de La Plata, La Plata, Argentina
| | - Agustín Blachman
- Cátedra de Biología Celular y Molecular, Departamento de Ciencias Biológicas, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Luciana Gavernet
- Laboratorio de Investigaciones Bioactivas y Desarrollo, Departamento de Ciencias Biológicas, Facultad de Ciencias Exactas, Universidad de La Plata, La Plata, Argentina
| | - Amalia Merelli
- Instituto de Fisiopatología y Bioquímica Clínica, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Alan Talevi
- Laboratorio de Investigaciones Bioactivas y Desarrollo, Departamento de Ciencias Biológicas, Facultad de Ciencias Exactas, Universidad de La Plata, La Plata, Argentina
| | - Graciela Cristina Calabrese
- Cátedra de Biología Celular y Molecular, Departamento de Ciencias Biológicas, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Alberto Javier Ramos
- Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina.,Laboratorio de Neuropatología Molecular, Instituto de Biología Celular y Neurociencia "Prof. E. De Robertis," Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Alberto Lazarowski
- Instituto de Fisiopatología y Bioquímica Clínica, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
22
|
Baril AA, Gagnon K, Descoteaux M, Bedetti C, Chami S, Sanchez E, Montplaisir J, De Beaumont L, Gilbert D, Poirier J, Pelleieux S, Osorio RS, Carrier J, Gosselin N. Cerebral white matter diffusion properties and free-water with obstructive sleep apnea severity in older adults. Hum Brain Mapp 2020; 41:2686-2701. [PMID: 32166865 PMCID: PMC7294053 DOI: 10.1002/hbm.24971] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 01/25/2020] [Accepted: 02/16/2020] [Indexed: 12/13/2022] Open
Abstract
Characterizing the effects of obstructive sleep apnea (OSA) on the aging brain could be key in our understanding of neurodegeneration in this population. Our objective was to assess white matter properties in newly diagnosed and untreated adults with mild to severe OSA. Sixty‐five adults aged 55 to 85 were recruited and divided into three groups: control (apnea‐hypopnea index ≤5/hr; n = 18; 65.2 ± 7.2 years old), mild (>5 to ≤15 hr; n = 27; 64.2 ± 5.3 years old) and moderate to severe OSA (>15/hr; n = 20; 65.2 ± 5.5 years old). Diffusion tensor imaging metrics (fractional anisotropy (FA), axial diffusivity (AD), radial diffusivity, and mean diffusivity) were compared between groups with Tract‐Based Spatial Statistics within the white matter skeleton created by the technique. Groups were also compared for white matter hyperintensities volume and the free‐water (FW) fraction. Compared with controls, mild OSA participants showed widespread areas of lower diffusivity (p < .05 corrected) and lower FW fraction (p < .05). Participants with moderate to severe OSA showed lower AD in the corpus callosum compared with controls (p < .05 corrected). No between‐group differences were observed for FA or white matter hyperintensities. Lower white matter diffusivity metrics is especially marked in mild OSA, suggesting that even the milder form may lead to detrimental outcomes. In moderate to severe OSA, competing pathological responses might have led to partial normalization of diffusion metrics.
Collapse
Affiliation(s)
- Andrée-Ann Baril
- Research Centre, Hôpital du Sacré-Coeur de Montréal, Montréal, Québec, Canada.,Faculty of Medicine, Université de Montréal, Montréal, Québec, Canada.,The Framingham Heart Study, Boston University School of Medicine, Boston, Massachussetts
| | - Katia Gagnon
- Research Centre, Hôpital du Sacré-Coeur de Montréal, Montréal, Québec, Canada.,Department of Psychology, Université du Québec à Montréal, Montréal, Québec, Canada.,Research Centre, Hôpital Rivière-des-Prairies, Montréal, Québec, Canada.,Department of Psychology, Université de Montréal, Montréal, Québec, Canada
| | - Maxime Descoteaux
- Research Centre, Centre hospitalier universitaire de Sherbrooke, Sherbrooke, Québec, Canada.,Computer Science Department, Faculty of Sciences, Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Christophe Bedetti
- Research Centre, Hôpital du Sacré-Coeur de Montréal, Montréal, Québec, Canada.,Research Centre, Institut universitaire de gériatrie de Montréal, Montréal, Québec, Canada
| | - Sirin Chami
- Research Centre, Hôpital du Sacré-Coeur de Montréal, Montréal, Québec, Canada.,Faculty of Medicine, Université de Montréal, Montréal, Québec, Canada
| | - Erlan Sanchez
- Research Centre, Hôpital du Sacré-Coeur de Montréal, Montréal, Québec, Canada.,Faculty of Medicine, Université de Montréal, Montréal, Québec, Canada
| | - Jacques Montplaisir
- Research Centre, Hôpital du Sacré-Coeur de Montréal, Montréal, Québec, Canada.,Faculty of Medicine, Université de Montréal, Montréal, Québec, Canada
| | - Louis De Beaumont
- Research Centre, Hôpital du Sacré-Coeur de Montréal, Montréal, Québec, Canada.,Faculty of Medicine, Université de Montréal, Montréal, Québec, Canada
| | - Danielle Gilbert
- Research Centre, Hôpital du Sacré-Coeur de Montréal, Montréal, Québec, Canada
| | - Judes Poirier
- Centre for Studies on Prevention of Alzheimer's disease, Douglas Institute, Verdun, Québec, Canada.,Department of Psychiatry and Medicine, McGill University, Montréal, Québec, Canada
| | - Sandra Pelleieux
- Centre for Studies on Prevention of Alzheimer's disease, Douglas Institute, Verdun, Québec, Canada.,Department of Psychiatry and Medicine, McGill University, Montréal, Québec, Canada
| | - Ricardo S Osorio
- Department of Psychiatry, Center for Brain Health, NYU Langone Medical Center, New York, New York, USA
| | - Julie Carrier
- Research Centre, Hôpital du Sacré-Coeur de Montréal, Montréal, Québec, Canada.,Research Centre, Institut universitaire de gériatrie de Montréal, Montréal, Québec, Canada.,Department of Psychology, Université de Montréal, Montréal, Québec, Canada
| | - Nadia Gosselin
- Research Centre, Hôpital du Sacré-Coeur de Montréal, Montréal, Québec, Canada.,Department of Psychology, Université de Montréal, Montréal, Québec, Canada
| |
Collapse
|
23
|
Li W, Yu Y, Li D, Xu N, Fang J, Sun Y, Xu M, Wang X, Han X, Zhang X, Lv C, Han F. TLR2 deficiency attenuated chronic intermittent hypoxia-induced neurocognitive deficits. Int Immunopharmacol 2020; 81:106284. [PMID: 32058931 DOI: 10.1016/j.intimp.2020.106284] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 01/22/2020] [Accepted: 02/02/2020] [Indexed: 12/17/2022]
Abstract
Chronic intermittent hypoxia (CIH) is the main symptom of obstructive sleep apnea syndrome (OSAS) and causes neural damage and cognitive deficits via neuroinflammation. Toll-like receptors (TLRs), especially TLR2, play an important role in neuroinflammation. However, the mechanisms by which TLR2 participates in CIH-induced cognitive deficits remain unclear. In this study, wild-type (WT) and TLR2 knock out (KO) mice were exposed to CIH for 8 weeks, and their social novelty discrimination, spatial learning and memory were severely compromised. Additionally, seriously damaged neurons and abnormally activated glia were observed in the CA1 and dentate gyrus (DG) areas of the hippocampus. Mechanistically, knocking out the TLR2 gene significantly alleviated these pathological changes and improved the behavioral performance. Together, these findings demonstrate that the TLR2-MyD88 signaling pathway might play an important role in CIH-induced cognitive deficits.
Collapse
Affiliation(s)
- Wanting Li
- Binzhou Medical University, 346 Guanhai Road, YanTai 264003, China
| | - Yan Yu
- Binzhou Medical University, 346 Guanhai Road, YanTai 264003, China.
| | - Dongze Li
- Binzhou Medical University, 346 Guanhai Road, YanTai 264003, China
| | - Na Xu
- Binzhou Medical University, 346 Guanhai Road, YanTai 264003, China
| | - Jidong Fang
- Binzhou Medical University, 346 Guanhai Road, YanTai 264003, China
| | - Yeying Sun
- Binzhou Medical University, 346 Guanhai Road, YanTai 264003, China
| | - Maolei Xu
- Binzhou Medical University, 346 Guanhai Road, YanTai 264003, China
| | - Xi Wang
- Yantai Affiliated Hospital of Binzhou Medical University, 717 Jinbu Street, YanTai 264199, China
| | - Xin Han
- Binzhou Medical University, 346 Guanhai Road, YanTai 264003, China
| | - Xiuli Zhang
- Binzhou Medical University, 346 Guanhai Road, YanTai 264003, China
| | - Changjun Lv
- Binzhou Medical University, 346 Guanhai Road, YanTai 264003, China.
| | - Fang Han
- Binzhou Medical University, 346 Guanhai Road, YanTai 264003, China.
| |
Collapse
|
24
|
Lee SA, Kim HJ, Lee Y. Subjective nocturnal symptoms have different associations with depressive symptoms and anxiety than with daytime sleepiness in patients with obstructive sleep apnea. Sleep Med 2020; 69:58-64. [PMID: 32045855 DOI: 10.1016/j.sleep.2019.12.019] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 11/29/2019] [Accepted: 12/23/2019] [Indexed: 11/30/2022]
Abstract
BACKGROUND We determined the relationships among the subjective symptoms of sleep apnea and daytime sleepiness, depressive symptoms, and anxiety in adults with obstructive sleep apnea (OSA). METHODS We developed the Subjective Apnea Severity Questionnaire (SASQ) to measure subjective OSA symptoms during the night and on waking in the morning. Construct validity and reliability were assessed. The Epworth Sleepiness Scale (ESS), Beck Depression Inventory (BDI), and State Scale of State Trait Anxiety Inventory (STAI-S) were applied. Multiple linear regression analyses were performed, and the results were adjusted for several confounders. RESULTS A total of 337 OSA patients were included. The SASQ consists of eight items with three domains. Cronbach's α for the SASQ was 0.657. The mean SASQ score was 1.35 ± 0.59. Symptoms related to nocturnal breathing difficulties were associated with polysomnographic (PSG) respiratory parameters. In the adjusted models, total SASQ scores were associated with ESS scores but not with BDI or STAI-S scores. Unlike other symptom groups, nocturnal breathing difficulties tended toward a positive relationship with ESS scores (p = 0.076), but were negatively related to BDI scores (p = 0.003) and STAI-S scores (p = 0.012). Symptoms related to nocturnal awakening or morning waking were either positively related or unrelated to ESS, BDI, and STAI-S scores. CONCLUSIONS The subjective OSA symptoms measured via the SASQ were associated with daytime sleepiness in adults with OSA, but not with depressive symptoms or anxiety. Nocturnal breathing difficulties were positively related to daytime sleepiness, but negatively related to depressive symptoms and anxiety.
Collapse
Affiliation(s)
- Sang-Ahm Lee
- Department of Neurology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.
| | - Hyo Jae Kim
- Department of Neurology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Yoojin Lee
- Department of Neurology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
25
|
Macefield VG, Henderson LA. Identifying Increases in Activity of the Human RVLM Through MSNA-Coupled fMRI. Front Neurosci 2020; 13:1369. [PMID: 32038124 PMCID: PMC6985468 DOI: 10.3389/fnins.2019.01369] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2019] [Accepted: 12/04/2019] [Indexed: 11/13/2022] Open
Abstract
AIM We initially developed concurrent recording of muscle sympathetic nerve activity (MSNA) and functional magnetic resonance imaging (fMRI) of the brain to functionally identify the human homolog of the rostral ventrolateral medulla (RVLM). Here we summarize the cortical and subcortical connections to the RVLM, as identified using MSNA-coupled fMRI. METHODS MSNA was recorded via tungsten microelectrodes inserted into the peroneal nerve. Gradient echo, echo-planar fMRI was performed at 3T (Philips Achieva). 200 volumes (46 axial slices (TR = 8 s, TE = 4 s, flip angle = 90°, raw voxel size = 1.5 × 1.5 × 2.75 mm) were collected in a 4 s-ON, 4 s-OFF sparse sampling protocol and MSNA measured in each 1 s epoch in the 4-s period between scans. Blood oxygen level dependent (BOLD) signal intensity was measured in the corresponding 1 s epoch 4 s later to account for peripheral neural conduction and central neurovascular coupling delays. RESULTS BOLD signal intensity was positively related to bursts of MSNA in the RVLM, dorsomedial hypothalamus (DMH), ventromedial hypothalamus (VMH), insula, dorsolateral prefrontal cortex (dlPFC), posterior cingulate cortex (PCC), and precuneus, and negatively related in the caudal ventrolateral medulla (CVLM), nucleus tractus solitarius (NTS), and the midbrain periaqueductal gray (PAG). During physiological increases in MSNA (tonic muscle pain), MSNA-coupled BOLD signal intensity was greater in RVLM, NTS, PAG, DMH, dlPFC, medial prefrontal cortex (mPFC), precuneus, and anterior cingulate cortex (ACC) than at rest. During pathophysiological increases in MSNA [obstructive sleep apnoea (OSA)] signal intensity was also higher in dlPFC, mPFC, ACC, and precuneus than in controls. Conversely, signal intensity was lower in RVLM in OSA than in controls, which we interpret as reflecting a withdrawal of active inhibition of the RVLM. CONCLUSION These results suggest that multiple cortical and subcortical areas are functionally coupled to the RVLM, which in turn is functionally coupled to the generation of spontaneous bursts of MSNA and their augmentation during physiological and pathophysiological increase in vasoconstrictor drive.
Collapse
Affiliation(s)
- Vaughan G. Macefield
- Human Autonomic Neurophysiology Laboratory, School of Medicine, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
- Department of Physiology, School of Biomedical Sciences, The University of Melbourne, Melbourne, VIC, Australia
| | - Luke A. Henderson
- Discipline of Anatomy and Histology, School of Medical Sciences, The University of Sydney, Sydney, NSW, Australia
- Brain and Mind Centre, The University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
26
|
Flores KR, Viccaro F, Aquilini M, Scarpino S, Ronchetti F, Mancini R, Di Napoli A, Scozzi D, Ricci A. Protective role of brain derived neurotrophic factor (BDNF) in obstructive sleep apnea syndrome (OSAS) patients. PLoS One 2020; 15:e0227834. [PMID: 31951637 PMCID: PMC6968866 DOI: 10.1371/journal.pone.0227834] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Accepted: 12/30/2019] [Indexed: 02/06/2023] Open
Abstract
Obstructive sleep apnea syndrome (OSAS) is a common disorder characterized by repeated episodes of upper airways collapse during the sleep. The following intermittent hypoxia triggers a state of chronic inflammation, which also interests the nervous system leading to neuronal damage and increased risk of cognitive impairment. Brain derived neurotrophic factor (BDNF) is a growth factor often associated with neuroplasticity and neuroprotection whose levels increase in several condition associated with neuronal damage. However, whether patients affected by OSAS have altered BDNF levels and whether such alteration may be reflective of their cognitive impairment is still controversial. Here we show that, when compared to healthy control volunteers, OSAS patients have increased serum levels of BDNF. Moreover, OSAS patients with the higher levels of BDNF also have reduced neurocognitive impairment as measured by The Montreal Cognitive Assessment (MoCA) questionnaire. Treatment with standard non-invasive mechanical ventilation (CPAP) also was able to ameliorate the level of cognitive impairment. Altogether our results indicate that BDNF levels represent a neuroprotective response to intermittent hypoxia in OSAS patients.
Collapse
Affiliation(s)
- Krisstopher Richard Flores
- Department of Clinical and Molecular Medicine, Division of Respiratory Diseases, Sant’Andrea Hospital, Sapienza University, Rome, Italy
| | - Fausta Viccaro
- Department of Clinical and Molecular Medicine, Division of Respiratory Diseases, Sant’Andrea Hospital, Sapienza University, Rome, Italy
| | - Mauro Aquilini
- Department of Clinical and Molecular Medicine, Division of Respiratory Diseases, Sant’Andrea Hospital, Sapienza University, Rome, Italy
| | - Stefania Scarpino
- Department of Clinical and Molecular Medicine, Division of Respiratory Diseases, Sant’Andrea Hospital, Sapienza University, Rome, Italy
| | - Francesco Ronchetti
- Department of Clinical and Molecular Medicine, Division of Respiratory Diseases, Sant’Andrea Hospital, Sapienza University, Rome, Italy
| | - Rita Mancini
- Department of Clinical and Molecular Medicine, Division of Respiratory Diseases, Sant’Andrea Hospital, Sapienza University, Rome, Italy
| | - Arianna Di Napoli
- Department of Clinical and Molecular Medicine, Division of Respiratory Diseases, Sant’Andrea Hospital, Sapienza University, Rome, Italy
| | - Davide Scozzi
- Department of Surgery, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Alberto Ricci
- Department of Clinical and Molecular Medicine, Division of Respiratory Diseases, Sant’Andrea Hospital, Sapienza University, Rome, Italy
- * E-mail:
| |
Collapse
|
27
|
Owen JE, BenediktsdÓttir B, Gislason T, Robinson SR. Neuropathological investigation of cell layer thickness and myelination in the hippocampus of people with obstructive sleep apnea. Sleep 2019; 42:5139668. [PMID: 30346595 DOI: 10.1093/sleep/zsy199] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Indexed: 12/11/2022] Open
Abstract
Obstructive sleep apnea (OSA) is commonly associated with memory impairments. Although MRI studies have found volumetric differences in the hippocampus of people with OSA compared with controls, MRI lacks the spatial resolution to detect changes in the specific regions of the hippocampus that process different types of memory. The present study performed histopathological investigations on autopsy brain tissue from 32 people with OSA (17 females and 15 males) to examine whether the thickness and myelination of the hippocampus and entorhinal cortex (EC) vary as a function of OSA severity. Increasing OSA severity was found to be related to cortical thinning in the molecular layer of the dentate gyrus (r2 = 0.136, p = 0.038), the CA1 (overall, r2 = 0.135, p = 0.039; layer 1, r2 = 0.157, p = 0.025; layer 2, r2 = 0.255, p = 0.003; and layer 3, r2 = 0.185, p = 0.014) and in some layers of the EC (layer 1, r2 = 0.186, p = 0.028; trend in layer 3, r2 = 0.124, p = 0.078). OSA severity was also related to decreased myelin in the deep layers but not the superficial layers of the EC (layer 6, r2 = 0.282, p = 0.006; deep white matter, r2 = 0.390, p = 0.001). Patients known to have used continuous positive airway pressure (CPAP) treatment showed no significant reductions in cortical thickness when compared with controls, suggesting that CPAP had a protective effect. However, CPAP did not protect against myelin loss. The regions of decreased cortical thickness and demyelination are locations of synaptic connections in both the polysynaptic (episodic and spatial) and direct (semantic) memory pathways and may underpin the impairments observed in episodic, semantic, and spatial memory in people with OSA.
Collapse
Affiliation(s)
- Jessica E Owen
- School of Health and Biomedical Sciences, RMIT University, Bundoora, Victoria, Australia
| | | | - Thorarinn Gislason
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland.,Department of Sleep Medicine, Landspitali - The National University Hospital of Iceland, Reykjavik, Iceland
| | - Stephen R Robinson
- School of Health and Biomedical Sciences, RMIT University, Bundoora, Victoria, Australia
| |
Collapse
|
28
|
Macey PM. Damage to the hippocampus in obstructive sleep apnea: a link no longer missing. Sleep 2019; 42:5280062. [PMID: 30615182 DOI: 10.1093/sleep/zsy266] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Affiliation(s)
- Paul M Macey
- UCLA School of Nursing, University of California at Los Angeles, Los Angeles, CA
| |
Collapse
|
29
|
Predicting Reactive Astrogliosis Propagation by Bayesian Computational Modeling: the Repeater Stations Model. Mol Neurobiol 2019; 57:879-895. [PMID: 31522382 DOI: 10.1007/s12035-019-01749-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Accepted: 08/26/2019] [Indexed: 10/26/2022]
Abstract
Reactive astrogliosis occurs upon focal brain injury and in neurodegenerative diseases. The mechanisms that propagate reactive astrogliosis to distal parts of the brain, in a rapid wave that activates astrocytes and other cell types along the way, are not completely understood. It is proposed that damage-associated molecular patterns (DAMP) released by necrotic cells from the injury core have a major role in the reactive astrogliosis initiation but whether they also participate in reactive astrogliosis propagation remains to be determined. We here developed a Bayesian computational model to define the most probable model for reactive astrogliosis propagation. Starting with experimental data from GFAP-immunostained reactive astrocytes, we defined five types of astrocytes based on morphometrical cues and registered the position of each reactive astrocyte cell type in the hemisphere ipsilateral to the injured site after 3 and 7 days post-ischemia. We developed equations for the changes in DAMP concentration (due to diffusion, binding to receptors or degradation), soluble mediators secretion, and for the evolution reactive astrogliosis. We tested four predefined models based on abovementioned previous hypothesis and modifications to it. Our results showed that DAMP diffusion alone has not justified the reactive astrogliosis propagation as previously assumed. Only two models succeeded in accurately reproducing the experimentally measured data and they highlighted the role of microglia and the glial secretion of soluble mediators to sustain the reactive signal and activating neighboring astrocytes. Thus, our in silico analysis proposes that glial cells behave as repeater stations of the injury signal in order to propagate reactive astrogliosis.
Collapse
|
30
|
Rosciszewski G, Cadena V, Auzmendi J, Cieri MB, Lukin J, Rossi AR, Murta V, Villarreal A, Reinés A, Gomes FCA, Ramos AJ. Detrimental Effects of HMGB-1 Require Microglial-Astroglial Interaction: Implications for the Status Epilepticus -Induced Neuroinflammation. Front Cell Neurosci 2019; 13:380. [PMID: 31507379 PMCID: PMC6718475 DOI: 10.3389/fncel.2019.00380] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 08/02/2019] [Indexed: 01/23/2023] Open
Abstract
Temporal Lobe Epilepsy (TLE) is the most common form of human epilepsy and available treatments with antiepileptic drugs are not disease-modifying therapies. The neuroinflammation, neuronal death and exacerbated plasticity that occur during the silent period, following the initial precipitating event (IPE), seem to be crucial for epileptogenesis. Damage Associated Molecular Patterns (DAMP) such as HMGB-1, are released early during this period concomitantly with a phenomenon of reactive gliosis and neurodegeneration. Here, using a combination of primary neuronal and glial cell cultures, we show that exposure to HMGB-1 induces dendrite loss and neurodegeneration in a glial-dependent manner. In glial cells, loss of function studies showed that HMGB-1 exposure induces NF-κB activation by engaging a signaling pathway that involves TLR2, TLR4, and RAGE. In the absence of glial cells, HMGB-1 failed to induce neurodegeneration of primary cultured cortical neurons. Moreover, purified astrocytes were unable to fully respond to HMGB-1 with NF-κB activation and required microglial cooperation. In agreement, in vivo HMGB-1 blockage with glycyrrhizin, immediately after pilocarpine-induced status epilepticus (SE), reduced neuronal degeneration, reactive astrogliosis and microgliosis in the long term. We conclude that microglial-astroglial cooperation is required for astrocytes to respond to HMGB-1 and to induce neurodegeneration. Disruption of this HMGB-1 mediated signaling pathway shows beneficial effects by reducing neuroinflammation and neurodegeneration after SE. Thus, early treatment strategies during the latency period aimed at blocking downstream signaling pathways activated by HMGB-1 are likely to have a significant effect in the neuroinflammation and neurodegeneration that are proposed as key factors in epileptogenesis.
Collapse
Affiliation(s)
- Gerardo Rosciszewski
- Laboratorio de Neuropatología Molecular, Instituto de Biología Celular y Neurociencia "Prof. E. De Robertis" UBA-CONICET, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Vanesa Cadena
- Laboratorio de Neuropatología Molecular, Instituto de Biología Celular y Neurociencia "Prof. E. De Robertis" UBA-CONICET, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Jerónimo Auzmendi
- Laboratorio de Neuropatología Molecular, Instituto de Biología Celular y Neurociencia "Prof. E. De Robertis" UBA-CONICET, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - María Belén Cieri
- Laboratorio de Neuropatología Molecular, Instituto de Biología Celular y Neurociencia "Prof. E. De Robertis" UBA-CONICET, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Jerónimo Lukin
- Laboratorio de Neuropatología Molecular, Instituto de Biología Celular y Neurociencia "Prof. E. De Robertis" UBA-CONICET, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Alicia R Rossi
- Laboratorio de Neuropatología Molecular, Instituto de Biología Celular y Neurociencia "Prof. E. De Robertis" UBA-CONICET, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Veronica Murta
- Laboratorio de Neuropatología Molecular, Instituto de Biología Celular y Neurociencia "Prof. E. De Robertis" UBA-CONICET, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Alejandro Villarreal
- Laboratorio de Neuropatología Molecular, Instituto de Biología Celular y Neurociencia "Prof. E. De Robertis" UBA-CONICET, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Analia Reinés
- Laboratorio de Neurofarmacología, Instituto de Biología Celular y Neurociencia "Prof. E. De Robertis" UBA-CONICET, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Flávia C A Gomes
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Alberto Javier Ramos
- Laboratorio de Neuropatología Molecular, Instituto de Biología Celular y Neurociencia "Prof. E. De Robertis" UBA-CONICET, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
31
|
Merelli A, Ramos AJ, Lazarowski A, Auzmendi J. Convulsive Stress Mimics Brain Hypoxia and Promotes the P-Glycoprotein (P-gp) and Erythropoietin Receptor Overexpression. Recombinant Human Erythropoietin Effect on P-gp Activity. Front Neurosci 2019; 13:750. [PMID: 31379495 PMCID: PMC6652211 DOI: 10.3389/fnins.2019.00750] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Accepted: 07/05/2019] [Indexed: 12/19/2022] Open
Abstract
Erythropoietin (EPO) is not only a hormone that promotes erythropoiesis but also has a neuroprotective effect on neurons attributed to its known anti-apoptotic action. Previously, our group has demonstrated that recombinant-human EPO (rHu-EPO) can protect neurons and recovery motor activity in a chemical focal brain hypoxia model (Merelli et al., 2011). We and others also have reported that repetitive seizures can mimic a hypoxic- like condition by HIF-1α nuclear translocation and high neuronal expression P-gp. Here, we report that a single 20-min status epilepticus (SE) induces P-gp and EPO-R expression in cortical pyramidal neurons and only P-gp expression in astrocytes. In vitro, excitotoxic stress (300 μM glutamate, 5 min), can also induce the expression of EPO-R and P-gp simultaneously with both HIF-1α and NFkB nuclear translocation in primary cortical neurons. Primary astrocytes exposed to chemical hypoxia with CoCl2 (0.3 mM, 6 h) increased P-gp expression as well as an increased efflux of Rhodamine 123 (Rho123) that is a P-gp substrate. Tariquidar, a specific 3er generation P-gp-blocker was used as an efflux inhibitor control. Astrocytes treated with rHu-EPO showed a significant recovery of the Rho123 retention in a similar way as seen by Tariquidar, demonstrating for first time that rHu-EPO can inhibit the P-gp-dependent efflux activity. Taking together, these data suggest that stimulation of EPO depending signaling system could not only play a central role in brain cell protection, but this system could be a new tool for reverse the pharmacoresistant phenotype in refractory epilepsy as well as in other pharmacoresistant hypoxic brain diseases expressing P-gp.
Collapse
Affiliation(s)
- Amalia Merelli
- Departamento de Bioquímica Clínica, Instituto de Investigaciones en Fisiopatología y Bioquímica Clínica (INFIBIOC), Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Alberto Javier Ramos
- Laboratorio de Neuropatología Molecular, Instituto de Biología Celular y Neurociencia "Prof. E. De Robertis" IBCN-UBA-CONICET, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Alberto Lazarowski
- Departamento de Bioquímica Clínica, Instituto de Investigaciones en Fisiopatología y Bioquímica Clínica (INFIBIOC), Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Jeronimo Auzmendi
- Laboratorio de Neuropatología Molecular, Instituto de Biología Celular y Neurociencia "Prof. E. De Robertis" IBCN-UBA-CONICET, Universidad de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
32
|
Is severe obstructive sleep apnea associated with less depressive symptoms? J Psychosom Res 2019; 122:6-12. [PMID: 31126412 DOI: 10.1016/j.jpsychores.2019.04.017] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2018] [Revised: 04/26/2019] [Accepted: 04/26/2019] [Indexed: 11/21/2022]
Abstract
PURPOSE To investigate the relationship between obstructive sleep apnea (OSA) and symptoms of depression and anxiety in OSA patients. METHODS Symptoms were assessed using the Beck Depression Inventory (BDI) and the state part of the State-Trait Anxiety Inventory (STAI-S). BDI scores of ≥10 and STAI-S scores of ≥40 were considered to indicate the presence of depression and anxiety, respectively. Apnea severities measured using polysomnography were categorized into mild, moderate, and severe subgroups bounded by the 33rd and 66th percentiles of each polysomnographic parameter. Data were stratified by age, gender, and level of daytime sleepiness. RESULTS The study population comprised 795 adult patients (86.9% men). Symptoms of depression and anxiety were present in 46.2% and 49.2% of patients, respectively. Excessive daytime sleepiness was present in 40.0% of patients and did not differ depending on the level of apnea severity. Results of crude logistic regression analyses indicated that depressive symptoms were more prevalent in patients with mild OSA than those with severe OSA, regardless of the categorizing method. These results remained statistically significant following adjustment for several confounding factors. These relationships were similar but less prominent in measures of anxiety. In the sub-analyses, such negative associations between severity of OSA and depressive symptoms tended to be observed only in patients with daytime sleepiness. CONCLUSIONS Symptoms of depression and anxiety were found to be more prevalent in patients with mild OSA than those with severe OSA. Excessive daytime sleepiness was shown to affect the severity of depressive symptoms.
Collapse
|
33
|
Andrade AG, Bubu OM, Varga AW, Osorio RS. The Relationship between Obstructive Sleep Apnea and Alzheimer's Disease. J Alzheimers Dis 2019; 64:S255-S270. [PMID: 29782319 DOI: 10.3233/jad-179936] [Citation(s) in RCA: 131] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Obstructive sleep apnea (OSA) and Alzheimer's disease (AD) are highly prevalent conditions with growing impact on our aging society. While the causes of OSA are now better characterized, the mechanisms underlying AD are still largely unknown, challenging the development of effective treatments. Cognitive impairment, especially affecting attention and executive functions, is a recognized clinical consequence of OSA. A deeper contribution of OSA to AD pathogenesis is now gaining support from several lines of research. OSA is intrinsically associated with disruptions of sleep architecture, intermittent hypoxia and oxidative stress, intrathoracic and hemodynamic changes as well as cardiovascular comorbidities. All of these could increase the risk for AD, rendering OSA as a potential modifiable target for AD prevention. Evidence supporting the relevance of each of these mechanisms for AD risk, as well as a possible effect of AD in OSA expression, will be explored in this review.
Collapse
Affiliation(s)
- Andreia G Andrade
- Department of Neurology, Alzheimer's Disease Center, NYU Langone Medical Center, New York, NY, USA.,Department of Psychiatry, Center for Brain Health, NYU Langone Medical Center, New York, NY, USA
| | - Omonigho M Bubu
- Department of Epidemiology and Biostatistics, College of Public Health, University of South Florida, Tampa, FL, USA
| | - Andrew W Varga
- Division of Pulmonary, Critical Care and Sleep Medicine at the Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ricardo S Osorio
- Department of Psychiatry, Center for Brain Health, NYU Langone Medical Center, New York, NY, USA.,Nathan S. Kline Institute for Psychiatric Research, Orangeburg, New York, NY, USA
| |
Collapse
|
34
|
Zárate SC, Traetta ME, Codagnone MG, Seilicovich A, Reinés AG. Humanin, a Mitochondrial-Derived Peptide Released by Astrocytes, Prevents Synapse Loss in Hippocampal Neurons. Front Aging Neurosci 2019; 11:123. [PMID: 31214013 PMCID: PMC6555273 DOI: 10.3389/fnagi.2019.00123] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Accepted: 05/09/2019] [Indexed: 01/13/2023] Open
Abstract
Astroglial cells are crucial for central nervous system (CNS) homeostasis. They undergo complex morpho-functional changes during aging and in response to hormonal milieu. Ovarian hormones positively affect different astroglia parameters, including regulation of cell morphology and release of neurotrophic and neuroprotective factors. Thus, ovarian hormone loss during menopause has profound impact in astroglial pathophysilogy and has been widely associated to the process of brain aging. Humanin (HN) is a secreted mitochondrial-encoded peptide with neuroprotective effects. It is localized in several tissues with high metabolic rate and its expression decreases with age. In the brain, humanin has been found in glial cells in physiological conditions. We previously reported that surgical menopause induces hippocampal mitochondrial dysfunction that mimics an aging phenotype. However, the effect of ovarian hormone deprivation on humanin expression in this area has not been studied. Also, whether astrocytes express and release humanin and the regulation of such processes by ovarian hormones remain elusive. Although humanin has also proven to be beneficial in ameliorating cognitive impairment induced by different insults, its putative actions on structural synaptic plasticity have not been fully addressed. In a model of surgical menopause in rats, we studied hippocampal humanin expression and localization by real-time quantitative polymerase chain reaction (RT-qPCR) and double immunohistochemistry, respectively. Humanin production and release and ovarian hormone regulation of such processes were studied in cultured astrocytes by flow cytometry and ELISA, respectively. Humanin effects on glutamate-induced structural synaptic alterations were determined in primary cultures of hippocampal neurons by immunocytochemistry. Humanin expression was lower in the hippocampus of ovariectomized rats and its immunoreactivity colocalized with astroglial markers. Chronic ovariectomy also promoted the presence of less complex astrocytes in this area. Ovarian hormones increased humanin intracellular content and release by cultured astrocytes. Humanin prevented glutamate-induced dendritic atrophy and reduction in puncta number and total puncta area for pre-synaptic marker synaptophysin in cultured hippocampal neurons. In conclusion, astroglial functional and morphological alterations induced by chronic ovariectomy resemble an aging phenotype and could affect astroglial support to neuronal function by altering synaptic connectivity and functionality. Reduced astroglial-derived humanin may represent an underlying mechanism for synaptic dysfunction and cognitive decline after menopause.
Collapse
Affiliation(s)
- Sandra Cristina Zárate
- Instituto de Investigaciones Biomédicas (INBIOMED, UBA-CONICET), Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina.,Departamento de Histología, Embriología, Biología Celular y Genética, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Marianela Evelyn Traetta
- Instituto de Biología Celular y Neurociencias "Prof. E. De Robertis" (IBCN, UBA-CONICET), Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina.,Departamento de Farmacología, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Martín Gabriel Codagnone
- Instituto de Biología Celular y Neurociencias "Prof. E. De Robertis" (IBCN, UBA-CONICET), Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina.,Departamento de Farmacología, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Adriana Seilicovich
- Instituto de Investigaciones Biomédicas (INBIOMED, UBA-CONICET), Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina.,Departamento de Histología, Embriología, Biología Celular y Genética, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Analía Gabriela Reinés
- Instituto de Biología Celular y Neurociencias "Prof. E. De Robertis" (IBCN, UBA-CONICET), Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina.,Departamento de Farmacología, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
35
|
Allen LA, Harper RM, Lhatoo S, Lemieux L, Diehl B. Neuroimaging of Sudden Unexpected Death in Epilepsy (SUDEP): Insights From Structural and Resting-State Functional MRI Studies. Front Neurol 2019; 10:185. [PMID: 30891003 PMCID: PMC6413533 DOI: 10.3389/fneur.2019.00185] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Accepted: 02/13/2019] [Indexed: 01/31/2023] Open
Abstract
The elusive nature of sudden unexpected death in epilepsy (SUDEP) has led to investigations of mechanisms and identification of biomarkers of this fatal scenario that constitutes the leading cause of premature death in epilepsy. In this short review, we compile evidence from structural and functional neuroimaging that demonstrates alterations to brain structures and networks involved in central autonomic and respiratory control in SUDEP and those at elevated risk. These findings suggest that compromised central control of vital regulatory processes may contribute to SUDEP. Both structural changes and dysfunctional interactions indicate potential mechanisms underlying the fatal event; contributions to individual risk prediction will require further study. The nature and sites of functional disruptions suggest potential non-invasive interventions to overcome failing processes.
Collapse
Affiliation(s)
- Luke A. Allen
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, London, United Kingdom
- Epilepsy Society MRI Unit, Chalfont St Peter, London, United Kingdom
- The Center for SUDEP Research, National Institute of Neurological Disorders and Stroke, Bethesda, MD, United States
| | - Ronald M. Harper
- The Center for SUDEP Research, National Institute of Neurological Disorders and Stroke, Bethesda, MD, United States
- Department of Neurobiology, David Geffen School of Medicine at UCLA, University of California, Los Angeles, Los Angeles, CA, United States
- Brain Research Institute, University of California, Los Angeles, Los Angeles, CA, United States
| | - Samden Lhatoo
- The Center for SUDEP Research, National Institute of Neurological Disorders and Stroke, Bethesda, MD, United States
- Department of Neurology, University of Texas Health Sciences Center at Houston, Houston, TX, United States
| | - Louis Lemieux
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, London, United Kingdom
- Epilepsy Society MRI Unit, Chalfont St Peter, London, United Kingdom
| | - Beate Diehl
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, London, United Kingdom
- Epilepsy Society MRI Unit, Chalfont St Peter, London, United Kingdom
- The Center for SUDEP Research, National Institute of Neurological Disorders and Stroke, Bethesda, MD, United States
| |
Collapse
|
36
|
Macheda T, Roberts K, Lyons DN, Higgins E, Ritter KJ, Lin AL, Alilain WJ, Bachstetter AD. Chronic Intermittent Hypoxia Induces Robust Astrogliosis in an Alzheimer's Disease-Relevant Mouse Model. Neuroscience 2018; 398:55-63. [PMID: 30529693 DOI: 10.1016/j.neuroscience.2018.11.040] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Revised: 11/19/2018] [Accepted: 11/27/2018] [Indexed: 12/17/2022]
Abstract
Sleep disturbances are a common early symptom of neurodegenerative diseases, including Alzheimer's disease (AD) and other age-related dementias, and emerging evidence suggests that poor sleep may be an important contributor to development of amyloid pathology. Of the causes of sleep disturbances, it is estimated that 10-20% of adults in the United States have sleep-disordered breathing (SDB) disorder, with obstructive sleep apnea accounting for the majority of the SBD cases. The clinical and epidemiological data clearly support a link between sleep apnea and AD; yet, almost no experimental research is available exploring the mechanisms associated with this correlative link. Therefore, we exposed an AD-relevant mouse model (APP/PS1 KI) to chronic intermittent hypoxia (IH) (an experimental model of sleep apnea) to begin to describe one of the potential mechanisms by which SDB could increase the risk of dementia. Previous studies have found that astrogliosis is a contributor to neuropathology in models of chronic IH and AD; therefore, we hypothesized that a reactive astrocyte response might be a contributing mechanism in the neuroinflammation associated with sleep apnea. To test this hypothesis, 10-11-month-old wild-type (WT) and APP/PS1 KI mice were exposed to 10 hours of IH, daily for four weeks. At the end of four weeks brains were analyzed from amyloid burden and astrogliosis. No effect was found for chronic IH exposure on amyloid-beta levels or plaque load in the APP/PS1 KI mice. A significant increase in GFAP staining was found in the APP/PS1 KI mice following chronic IH exposure, but not in the WT mice. Profiling of genes associated with different phenotypes of astrocyte activation identified GFAP, CXCL10, and Ggta1 as significant responses activated in the APP/PS1 KI mice exposed to chronic IH.
Collapse
Affiliation(s)
- Teresa Macheda
- Spinal Cord & Brain Injury Research Center, University of Kentucky, Lexington, KY, United States; Department of Neuroscience, University of Kentucky, Lexington, KY, United States
| | - Kelly Roberts
- Spinal Cord & Brain Injury Research Center, University of Kentucky, Lexington, KY, United States; Department of Neuroscience, University of Kentucky, Lexington, KY, United States
| | - Danielle N Lyons
- Spinal Cord & Brain Injury Research Center, University of Kentucky, Lexington, KY, United States; Department of Neuroscience, University of Kentucky, Lexington, KY, United States
| | - Emma Higgins
- Spinal Cord & Brain Injury Research Center, University of Kentucky, Lexington, KY, United States; Department of Neuroscience, University of Kentucky, Lexington, KY, United States
| | - Kyle J Ritter
- Spinal Cord & Brain Injury Research Center, University of Kentucky, Lexington, KY, United States; Department of Neuroscience, University of Kentucky, Lexington, KY, United States
| | - Ai-Ling Lin
- Sanders Brown Center on Aging, University of Kentucky, Lexington, KY, United States; Department of Nutrition and Pharmacology, University of Kentucky, Lexington, KY, United States
| | - Warren J Alilain
- Spinal Cord & Brain Injury Research Center, University of Kentucky, Lexington, KY, United States; Department of Neuroscience, University of Kentucky, Lexington, KY, United States
| | - Adam D Bachstetter
- Spinal Cord & Brain Injury Research Center, University of Kentucky, Lexington, KY, United States; Department of Neuroscience, University of Kentucky, Lexington, KY, United States.
| |
Collapse
|
37
|
Wang B, Li W, Jin H, Nie X, Shen H, Li E, Wang W. Curcumin attenuates chronic intermittent hypoxia-induced brain injuries by inhibiting AQP4 and p38 MAPK pathway. Respir Physiol Neurobiol 2018; 255:50-57. [DOI: 10.1016/j.resp.2018.05.006] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Revised: 05/09/2018] [Accepted: 05/10/2018] [Indexed: 11/28/2022]
|
38
|
Hypoxia-Induced Neuroinflammation and Learning-Memory Impairments in Adult Zebrafish Are Suppressed by Glucosamine. Mol Neurobiol 2018; 55:8738-8753. [PMID: 29589284 DOI: 10.1007/s12035-018-1017-9] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2017] [Accepted: 03/16/2018] [Indexed: 10/17/2022]
Abstract
This study investigated changes in neuroinflammation and cognitive function in adult zebrafish exposed to acute hypoxia and protective effects of glucosamine (GlcN) against hypoxia-induced brain damage. The survival rate of zebrafish following exposure to hypoxia was improved by GlcN pretreatment. Moreover, hypoxia-induced upregulation of neuroglobin, NOS2α, glial fibrillary acidic protein, and S100β in zebrafish was suppressed by GlcN. Hypoxia stimulated cell proliferation in the telencephalic ventral domain and in cerebellum subregions. GlcN decreased the number of bromodeoxyuridine (BrdU)-positive cells in the telencephalon region, but not in cerebellum regions. Transient motor neuron defects, assessed by measuring the locomotor and exploratory activity of zebrafish exposed to hypoxia recovered quickly. GlcN did not affect hypoxia-induced motor activity changes. In passive avoidance tests, hypoxia impaired learning and memory ability, deficits that were rescued by GlcN. A learning stimulus increased the nuclear translocation of phosphorylated cAMP response element binding protein (p-CREB), an effect that was greatly inhibited by hypoxia. GlcN restored nuclear p-CREB after a learning trial in hypoxia-exposed zebrafish. The neurotransmitters, γ-aminobutyric acid and glutamate, were increased after hypoxia in the zebrafish brain, and GlcN further increased their levels. In contrast, acetylcholine levels were reduced by hypoxia and restored by GlcN. Acetylcholinesterase inhibitor physostigmine partially reversed the impaired learning and memory of hypoxic zebrafish. This study represents the first examination of the molecular mechanisms underlying hypoxia-induced memory and learning defects in a zebrafish model. Our results further suggest that GlcN-associated hexosamine metabolic pathway could be an important therapeutic target for hypoxic brain damage.
Collapse
|
39
|
Auzmendi J, Buchholz B, Salguero J, Cañellas C, Kelly J, Men P, Zubillaga M, Rossi A, Merelli A, Gelpi RJ, Ramos AJ, Lazarowski A. Pilocarpine-Induced Status Epilepticus Is Associated with P-Glycoprotein Induction in Cardiomyocytes, Electrocardiographic Changes, and Sudden Death. Pharmaceuticals (Basel) 2018; 11:ph11010021. [PMID: 29462915 PMCID: PMC5874717 DOI: 10.3390/ph11010021] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2018] [Revised: 02/07/2018] [Accepted: 02/13/2018] [Indexed: 02/06/2023] Open
Abstract
Sudden unexpected death in epilepsy (SUDEP) is the major cause of death in those patients suffering from refractory epilepsy (RE), with a 24-fold higher risk relative to the normal population. SUDEP risk increases with seizure frequency and/or seizure-duration as in RE and Status Epilepticus (SE). P-glycoprotein (P-gp), the product of the multidrug resistant ABCB1-MDR-1 gene, is a detoxifying pump that extrudes drugs out of the cells and can confer pharmacoresistance to the expressing cells. Neurons and cardiomyocytes normally do not express P-gp, however, it is overexpressed in the brain of patients or in experimental models of RE and SE. P-gp was also detected after brain or cardiac hypoxia. We have previously demonstrated that repetitive pentylenetetrazole (PTZ)-induced seizures increase P-gp expression in the brain, which is associated with membrane depolarization in the hippocampus, and in the heart, which is associated with fatal SE. SE can produce hypoxic-ischemic altered cardiac rhythm (HIACR) and severe arrhythmias, and both are related with SUDEP. Here, we investigate whether SE induces the expression of hypoxia-inducible transcription factor (HIF)-1α and P-gp in cardiomyocytes, which is associated with altered heart rhythm, and if these changes are related with the spontaneous death rate. SE was induced in Wistar rats once a week for 3 weeks, by lithium-pilocarpine-paradigm. Electrocardiograms, HIF-1α, and P-gp expression in cardiomyocytes, were evaluated in basal conditions and 72 h after SE. All spontaneous deaths occurred 48 h after each SE was registered. We observed that repeated SE induced HIF-1α and P-gp expression in cardiomyocytes, electrocardiographic (ECG) changes, and a high rate of spontaneous death. Our results suggest that the highly accumulated burden of convulsive stress results in a hypoxic heart insult, where P-gp expression may play a depolarizing role in cardiomyocyte membranes and in the development of the ECG changes, such as QT interval prolongation, that could be related with SUDEP. We postulate that this mechanism could explain, in part, the higher SUDEP risk in patients with RE or SE.
Collapse
Affiliation(s)
- Jerónimo Auzmendi
- Laboratorio de Neuropatología Molecular, Instituto de Biología Celular y Neurociencia "Profesor E. De Robertis" IBCN UBA-CONICET, Buenos Aires CP1121, Argentina.
| | - Bruno Buchholz
- Departamento de Patología, Instituto de Fisiopatología Cardiovascular (INFICA), Universidad de Buenos Aires, Facultad de Medicina, Buenos Aires C1121ABG, Argentina.
| | - Jimena Salguero
- Departamento de Fisicomatemática, Laboratorio de Radioisótopos, Cátedra de Física, Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Junín 956, Buenos Aires C1113AAD, Argentina.
| | - Carlos Cañellas
- Laboratorio Tecnonuclear SA, Arias 4176, Buenos Aires C1430CRP, Argentina.
| | - Jazmín Kelly
- Departamento de Patología, Instituto de Fisiopatología Cardiovascular (INFICA), Universidad de Buenos Aires, Facultad de Medicina, Buenos Aires C1121ABG, Argentina.
| | - Paula Men
- Departamento de Bioquímica Clínica, Instituto de Investigaciones en Fisiopatología y Bioquímica Clínica (INFIBIOC), Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Junín 956, Buenos Aires C1113AAD, Argentina.
| | - Marcela Zubillaga
- Departamento de Fisicomatemática, Laboratorio de Radioisótopos, Cátedra de Física, Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Junín 956, Buenos Aires C1113AAD, Argentina.
| | - Alicia Rossi
- Laboratorio de Neuropatología Molecular, Instituto de Biología Celular y Neurociencia "Profesor E. De Robertis" IBCN UBA-CONICET, Buenos Aires CP1121, Argentina.
| | - Amalia Merelli
- Departamento de Bioquímica Clínica, Instituto de Investigaciones en Fisiopatología y Bioquímica Clínica (INFIBIOC), Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Junín 956, Buenos Aires C1113AAD, Argentina.
| | - Ricardo J Gelpi
- Departamento de Patología, Instituto de Fisiopatología Cardiovascular (INFICA), Universidad de Buenos Aires, Facultad de Medicina, Buenos Aires C1121ABG, Argentina.
| | - Alberto J Ramos
- Laboratorio de Neuropatología Molecular, Instituto de Biología Celular y Neurociencia "Profesor E. De Robertis" IBCN UBA-CONICET, Buenos Aires CP1121, Argentina.
| | - Alberto Lazarowski
- Departamento de Bioquímica Clínica, Instituto de Investigaciones en Fisiopatología y Bioquímica Clínica (INFIBIOC), Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Junín 956, Buenos Aires C1113AAD, Argentina.
| |
Collapse
|
40
|
Murta V, Schilrreff P, Rosciszewski G, Morilla MJ, Ramos AJ. G5G2.5 core-shell tecto-dendrimer specifically targets reactive glia in brain ischemia. J Neurochem 2018; 144:748-760. [DOI: 10.1111/jnc.14286] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Revised: 11/18/2017] [Accepted: 12/11/2017] [Indexed: 01/02/2023]
Affiliation(s)
- Veronica Murta
- Departamento de Histología, Embriología, Biología Celular y Genética; Facultad de Medicina; Universidad de Buenos Aires; Buenos Aires Argentina
- Laboratorio de Neuropatología Molecular; Instituto de Biología Celular y Neurociencia “Prof. E. De Robertis” IBCN UBA-CONICET; Buenos Aires Argentina
| | - Priscila Schilrreff
- Programa de Nanomedicinas; Departamento de Ciencia y Tecnología; Universidad Nacional de Quilmes; Bernal Argentina
| | - Gerardo Rosciszewski
- Departamento de Histología, Embriología, Biología Celular y Genética; Facultad de Medicina; Universidad de Buenos Aires; Buenos Aires Argentina
- Laboratorio de Neuropatología Molecular; Instituto de Biología Celular y Neurociencia “Prof. E. De Robertis” IBCN UBA-CONICET; Buenos Aires Argentina
| | - Maria Jose Morilla
- Programa de Nanomedicinas; Departamento de Ciencia y Tecnología; Universidad Nacional de Quilmes; Bernal Argentina
| | - Alberto Javier Ramos
- Departamento de Histología, Embriología, Biología Celular y Genética; Facultad de Medicina; Universidad de Buenos Aires; Buenos Aires Argentina
- Laboratorio de Neuropatología Molecular; Instituto de Biología Celular y Neurociencia “Prof. E. De Robertis” IBCN UBA-CONICET; Buenos Aires Argentina
| |
Collapse
|
41
|
Merelli A, Rodríguez JCG, Folch J, Regueiro MR, Camins A, Lazarowski A. Understanding the Role of Hypoxia Inducible Factor During Neurodegeneration for New Therapeutics Opportunities. Curr Neuropharmacol 2018; 16:1484-1498. [PMID: 29318974 PMCID: PMC6295932 DOI: 10.2174/1570159x16666180110130253] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Revised: 11/24/2017] [Accepted: 01/08/2018] [Indexed: 12/14/2022] Open
Abstract
Neurodegeneration (NDG) is linked with the progressive loss of neural function with intellectual and/or motor impairment. Several diseases affecting older individuals, including Alzheimer's disease, Amyotrophic Lateral Sclerosis, Huntington's disease, Parkinson's disease, stroke, Multiple Sclerosis and many others, are the most relevant disorders associated with NDG. Since other pathologies such as refractory epilepsy, brain infections, or hereditary diseases such as "neurodegeneration with brain iron accumulation", also lead to chronic brain inflammation with loss of neural cells, NDG can be said to affect all ages. Owing to an energy and/or oxygen supply imbalance, different signaling mechanisms including MAPK/PI3K-Akt signaling pathways, glutamatergic synapse formation, and/or translocation of phosphatidylserine, might activate some central executing mechanism common to all these pathologies and also related to oxidative stress. Hypoxia inducible factor 1-α (HIF-1α) plays a twofold role through gene activation, in the sense that this factor has to "choose" whether to protect or to kill the affected cells. Most of the afore-mentioned processes follow a protracted course and are accompanied by progressive iron accumulation in the brain. We hypothesize that the neuroprotective effects of iron chelators are acting against the generation of free radicals derived from iron, and also induce sufficient -but not excessive- activation of HIF-1α, so that only the hypoxia-rescue genes will be activated. In this regard, the expression of the erythropoietin receptor in hypoxic/inflammatory neurons could be the cellular "sign" to act upon by the nasal administration of pharmacological doses of Neuro-EPO, inducing not only neuroprotection, but eventually, neurorepair as well.
Collapse
Affiliation(s)
| | | | | | | | | | - Alberto Lazarowski
- Address correspondence to this author at the Clinical Biochemistry Department, School of Pharmacy and Biochemistry, University of Buenos Aires-Argentina, Junín 954, Buenos Aires-Argentina; Tel: +54-11-5950-8674;, E-mail:
| |
Collapse
|
42
|
Rosenzweig I, Morrell MJ. Hypotrophy versus Hypertrophy: It's Not Black or White with Gray Matter. Am J Respir Crit Care Med 2017; 195:1416-1418. [PMID: 28569579 DOI: 10.1164/rccm.201701-0109ed] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Affiliation(s)
- Ivana Rosenzweig
- 1 Institute of Psychiatry, Psychology, and Neuroscience King's College London London, United Kingdom.,2 Sleep Disorders Centre Guy's and St. Thomas' NHS Foundation Trust London, United Kingdom
| | - Mary J Morrell
- 3 National Heart and Lung Institute Imperial College London London, United Kingdom and.,4 NIHR Respiratory Disease Biomedical Research Unit Royal Brompton and Harefield NHS Foundation Trust and Imperial College London London, United Kingdom
| |
Collapse
|
43
|
Rossi A, Murta V, Auzmendi J, Ramos AJ. Early Gabapentin Treatment during the Latency Period Increases Convulsive Threshold, Reduces Microglial Activation and Macrophage Infiltration in the Lithium-Pilocarpine Model of Epilepsy. Pharmaceuticals (Basel) 2017; 10:ph10040093. [PMID: 29182533 PMCID: PMC5748648 DOI: 10.3390/ph10040093] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Revised: 11/14/2017] [Accepted: 11/19/2017] [Indexed: 12/20/2022] Open
Abstract
The lithium-pilocarpine model of epilepsy reproduces several features of temporal lobe epilepsy in humans, including the chronological timeline of an initial latency period followed by the development of spontaneous seizures. Epilepsy therapies in humans are implemented, as a rule, after the onset of the spontaneous seizures. We here studied the potential effect on epileptogenesis of starting an early treatment during the latency period, in order to prevent the development of spontaneous seizures. Adult male Wistar rats were treated with 3 mEq/kg LiCl, and 20 h later 30 mg/kg pilocarpine. Once status epilepticus (SE) was achieved, it was allowed to last for 20 min, and then motor seizures were controlled with the administration of 20 mg/kg diazepam. At 1DPSE (DPSE, days post-status epilepticus), animals started to receive 400 mg/kg/day gabapentin or saline for 4 days. At 5DPSE, we observed that SE induced an early profuse microglial and astroglial reactivity, increased synaptogenic trombospondin-1 expression and reduced AQP4 expression in astroglial ending feet. Blood brain barrier (BBB) integrity seemed to be compromised, as infiltrating NG2+ macrophages and facilitated access to the CNS was observed by transplanting eGFP+ blood cells and bone marrow-derived progenitors in the SE animals. The early 4-day gabapentin treatment successfully reduced microglial cell reactivity and blood-borne cell infiltration, without significantly altering the mRNA of proinflammatory cytokines IL-1β and TNFα immediately after the treatment. After 21DSPE, another group of animals that developed SE and received 4 days of gabapentin treatment, were re-exposed to subconvulsive accumulative doses of pilocarpine (10 mg/kg/30 min) and were followed by recording the Racine scale reached. Early 4-day gabapentin treatment reduced the Racine scale reached by the animals, reduced animal mortality, and reduced the number of animals that achieved SE (34% vs. 72%). We conclude that early gabapentin treatment following SE, during the latency period, is able to reduce neuroinflammation and produces a persistent effect that limits seizures and increases convulsive threshold, probably by restricting microglial reactivity and spurious synaptogenesis.
Collapse
Affiliation(s)
- Alicia Rossi
- Departamento de Histología, Embriología, Biología Celular y Genética, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires CP1121, Argentina.
- Laboratorio de Neuropatología Molecular, Instituto de Biología Celular y Neurociencia "Profesor E. De Robertis" IBCN UBA-CONICET, Buenos Aires CP1121, Argentina.
| | - Veronica Murta
- Laboratorio de Neuropatología Molecular, Instituto de Biología Celular y Neurociencia "Profesor E. De Robertis" IBCN UBA-CONICET, Buenos Aires CP1121, Argentina.
| | - Jerónimo Auzmendi
- Laboratorio de Neuropatología Molecular, Instituto de Biología Celular y Neurociencia "Profesor E. De Robertis" IBCN UBA-CONICET, Buenos Aires CP1121, Argentina.
| | - Alberto Javier Ramos
- Departamento de Histología, Embriología, Biología Celular y Genética, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires CP1121, Argentina.
- Laboratorio de Neuropatología Molecular, Instituto de Biología Celular y Neurociencia "Profesor E. De Robertis" IBCN UBA-CONICET, Buenos Aires CP1121, Argentina.
| |
Collapse
|
44
|
Macey PM, Sarma MK, Prasad JP, Ogren JA, Aysola R, Harper RM, Thomas MA. Obstructive sleep apnea is associated with altered midbrain chemical concentrations. Neuroscience 2017; 363:76-86. [PMID: 28893651 PMCID: PMC5983363 DOI: 10.1016/j.neuroscience.2017.09.001] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Revised: 08/25/2017] [Accepted: 09/03/2017] [Indexed: 12/28/2022]
Abstract
Obstructive sleep apnea (OSA) is accompanied by altered structure and function in cortical, limbic, brainstem, and cerebellar regions. The midbrain is relatively unexamined, but contains many integrative nuclei which mediate physiological functions that are disrupted in OSA. We therefore assessed the chemistry of the midbrain in OSA in this exploratory study. We used a recently developed accelerated 2D magnetic resonance spectroscopy (2D-MRS) technique, compressed sensing-based 4D echo-planar J-resolved spectroscopic imaging (4D-EP-JRESI), to measure metabolites in the midbrain of 14 OSA (mean age±SD:54.6±10.6years; AHI:35.0±19.4; SAO2 min:83±7%) and 26 healthy control (50.7±8.5years) subjects. High-resolution T1-weighted scans allowed voxel localization. MRS data were processed with custom MATLAB-based software, and metabolite ratios calculated with respect to the creatine peak using a prior knowledge fitting (ProFit) algorithm. The midbrain in OSA showed decreased N-acetylaspartate (NAA; OSA:1.24±0.43, Control:1.47±0.41; p=0.03; independent samples t-test), a marker of neuronal viability. Increased levels in OSA over control subjects appeared in glutamate (Glu; OSA:1.23±0.57, Control:0.98±0.33; p=0.03), ascorbate (Asc; OSA:0.56±0.28, Control:0.42±0.20; (50.7±8.5years; p=0.03), and myo-inositol (mI; OSA:0.96±0.48, Control:0.72±0.35; p=0.03). No differences between groups appeared in γ-aminobutyric acid (GABA) or taurine. The midbrain in OSA patients shows decreased NAA, indicating neuronal injury or dysfunction. Higher Glu levels may reflect excitotoxic processes and astrocyte activation, and higher mI is also consistent with glial activation. Higher Asc levels may result from oxidative stress induced by intermittent hypoxia in OSA. Additionally, Asc and Glu are involved with glutamatergic processes, which are likely upregulated in the midbrain nuclei of OSA patients. The altered metabolite levels help explain dysfunction and structural deficits in the midbrain of OSA patients.
Collapse
Key Words
- Asc, ascorbate
- Asp, aspartate
- Ch, choline
- GABA, gamma-aminobutyric acid
- GPC, glycerophosphorylcholine
- GSH, glutathione
- Gln, glutamine
- Glu, glutamate
- Gly, glycine
- NAA, N-acetylaspartate
- NAAG, N-acetylaspartate glutamate
- PCh, phosphocholine
- PE, phosphoethanolamine
- Scy, scyllo-inositol
- Tau, taurine
- Thr, threonine
- autonomic
- intermittent hypoxia
- mI, myo-inositol
- magnetic resonance spectroscopy
- periaqueductal gray
- respiration
- sleep-disordered breathing
Collapse
Affiliation(s)
- Paul M Macey
- School of Nursing, David Geffen School of Medicine at UCLA, University of California at Los Angeles, Los Angeles, CA 90095, United States; Brain Research Institute, David Geffen School of Medicine at UCLA, University of California at Los Angeles, Los Angeles, CA 90095, United States.
| | - Manoj K Sarma
- Department of Radiological Sciences, David Geffen School of Medicine at UCLA, University of California at Los Angeles, Los Angeles, CA 90095, United States
| | - Janani P Prasad
- School of Nursing, David Geffen School of Medicine at UCLA, University of California at Los Angeles, Los Angeles, CA 90095, United States
| | - Jennifer A Ogren
- Department of Neurobiology, David Geffen School of Medicine at UCLA, University of California at Los Angeles, Los Angeles, CA 90095, United States
| | - Ravi Aysola
- Department of Medicine (Division of Pulmonary and Critical Care), David Geffen School of Medicine at UCLA, University of California at Los Angeles, Los Angeles, CA 90095, United States
| | - Ronald M Harper
- Brain Research Institute, David Geffen School of Medicine at UCLA, University of California at Los Angeles, Los Angeles, CA 90095, United States; Department of Neurobiology, David Geffen School of Medicine at UCLA, University of California at Los Angeles, Los Angeles, CA 90095, United States
| | - M Albert Thomas
- Department of Radiological Sciences, David Geffen School of Medicine at UCLA, University of California at Los Angeles, Los Angeles, CA 90095, United States
| |
Collapse
|
45
|
Randall SR, Warton CMR, Holmes MJ, Cotton MF, Laughton B, van der Kouwe AJW, Meintjes EM. Larger Subcortical Gray Matter Structures and Smaller Corpora Callosa at Age 5 Years in HIV Infected Children on Early ART. Front Neuroanat 2017; 11:95. [PMID: 29163068 PMCID: PMC5673662 DOI: 10.3389/fnana.2017.00095] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2017] [Accepted: 10/16/2017] [Indexed: 11/13/2022] Open
Abstract
Sub-Saharan Africa is home to 90% of HIV infected (HIV+) children. Since the advent of antiretroviral therapy (ART), HIV/AIDS has transitioned to a chronic condition where central nervous system (CNS) damage may be ongoing. Although, most guidelines recommend early ART to reduce CNS viral reservoirs, the brain may be more vulnerable to potential neurotoxic effects of ART during the rapid development phase in the first years of life. Here we investigate differences in subcortical volumes between 5-year-old HIV+ children who received early ART (before age 18 months) and uninfected children using manual tracing of Magnetic Resonance Images. Participants included 61 Xhosa children (43 HIV+/18 uninfected, mean age = 5.4 ± 0.3 years, 25 male) from the children with HIV early antiretroviral (CHER) trial; 27 children initiated ART before 12 weeks of age (ART-Before12Wks) and 16 after 12 weeks (ART-After12Wks). Structural images were acquired on a 3T Allegra MRI in Cape Town and manually traced using MultiTracer. Volumetric group differences (HIV+ vs. uninfected; ART-Before12Wks vs. ART-After12Wks) were examined for the caudate, nucleus accumbens (NA), putamen (Pu), globus pallidus (GP), and corpus callosum (CC), as well as associations within infected children of structure volumes with age at ART initiation and CD4/CD8 as a proxy for immune health. HIV+ children had significantly larger NA and Pu volumes bilaterally and left GP volumes than controls, whilst CC was smaller. Bilateral Pu was larger in both treatment groups compared to controls, while left GP and bilateral NA were enlarged only in ART-After12Wks children. CC was smaller in both treatment groups compared to controls, and smaller in ART-After12Wks compared to ART-Before12Wks. Within infected children, delayed ART initiation was associated with larger Pu volumes, effects that remained significant when controlling for sex and duration of treatment interruption (left β = 0.447, p = 0.005; right β = 0.325, p = 0.051), and lower CD4/CD8 with larger caudates controlling for sex (left β = -0.471, p = 0.002; right β = -0.440, p = 0.003). Volumetric differences were greater in children who initiated ART after 12 weeks. Results suggest damage is ongoing despite early ART and viral load suppression; however, earlier treatment is neuroprotective.
Collapse
Affiliation(s)
- Steven R. Randall
- Department of Human Biology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Christopher M. R. Warton
- Department of Human Biology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Martha J. Holmes
- MRC/UCT Medical Imaging Research Unit, Division of Biomedical Engineering, Department of Human Biology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Mark F. Cotton
- Children's Infectious Diseases Clinical Research Unit, Department of Paediatrics and Child Health, Tygerberg Children's Hospital & Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Barbara Laughton
- Children's Infectious Diseases Clinical Research Unit, Department of Paediatrics and Child Health, Tygerberg Children's Hospital & Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Andre J. W. van der Kouwe
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, United States
| | - Ernesta M. Meintjes
- MRC/UCT Medical Imaging Research Unit, Division of Biomedical Engineering, Department of Human Biology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
46
|
Baril AA, Gagnon K, Brayet P, Montplaisir J, De Beaumont L, Carrier J, Lafond C, L'Heureux F, Gagnon JF, Gosselin N. Gray Matter Hypertrophy and Thickening with Obstructive Sleep Apnea in Middle-aged and Older Adults. Am J Respir Crit Care Med 2017; 195:1509-1518. [PMID: 28060546 DOI: 10.1164/rccm.201606-1271oc] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
RATIONALE Obstructive sleep apnea causes intermittent hypoxemia, hemodynamic fluctuations, and sleep fragmentation, all of which could damage cerebral gray matter that can be indirectly assessed by neuroimaging. OBJECTIVES To investigate whether markers of obstructive sleep apnea severity are associated with gray matter changes among middle-aged and older individuals. METHODS Seventy-one subjects (ages, 55-76 yr; apnea-hypopnea index, 0.2-96.6 events/h) were evaluated by magnetic resonance imaging. Two techniques were used: (1) voxel-based morphometry, which measures gray matter volume and concentration; and (2) FreeSurfer (an open source software suite) automated segmentation, which estimates the volume of predefined cortical/subcortical regions and cortical thickness. Regression analyses were performed between gray matter characteristics and markers of obstructive sleep apnea severity (hypoxemia, respiratory disturbances, and sleep fragmentation). MEASUREMENTS AND MAIN RESULTS Subjects had few symptoms, that is, sleepiness, depression, anxiety, and cognitive deficits. Although no association was found with voxel-based morphometry, FreeSurfer revealed increased gray matter with obstructive sleep apnea. Higher levels of hypoxemia correlated with increased volume and thickness of the left lateral prefrontal cortex as well as increased thickness of the right frontal pole, the right lateral parietal lobules, and the left posterior cingulate cortex. Respiratory disturbances positively correlated with right amygdala volume, and more severe sleep fragmentation was associated with increased thickness of the right inferior frontal gyrus. CONCLUSIONS Gray matter hypertrophy and thickening were associated with hypoxemia, respiratory disturbances, and sleep fragmentation. These structural changes in a group of middle-aged and older individuals may represent adaptive/reactive brain mechanisms attributed to a presymptomatic stage of obstructive sleep apnea.
Collapse
Affiliation(s)
- Andrée-Ann Baril
- 1 Centre d'études avancées en médecine du sommeil, Hôpital du Sacré-Cœur de Montréal, Montreal, Quebec, Canada.,2 Département de psychiatrie
| | - Katia Gagnon
- 1 Centre d'études avancées en médecine du sommeil, Hôpital du Sacré-Cœur de Montréal, Montreal, Quebec, Canada.,3 Département de psychologie, Université du Québec à Montréal, Montreal, Quebec, Canada
| | - Pauline Brayet
- 1 Centre d'études avancées en médecine du sommeil, Hôpital du Sacré-Cœur de Montréal, Montreal, Quebec, Canada.,3 Département de psychologie, Université du Québec à Montréal, Montreal, Quebec, Canada
| | - Jacques Montplaisir
- 1 Centre d'études avancées en médecine du sommeil, Hôpital du Sacré-Cœur de Montréal, Montreal, Quebec, Canada.,2 Département de psychiatrie
| | - Louis De Beaumont
- 1 Centre d'études avancées en médecine du sommeil, Hôpital du Sacré-Cœur de Montréal, Montreal, Quebec, Canada.,4 Département de chirurgie
| | - Julie Carrier
- 1 Centre d'études avancées en médecine du sommeil, Hôpital du Sacré-Cœur de Montréal, Montreal, Quebec, Canada.,5 Département de psychologie, and
| | - Chantal Lafond
- 1 Centre d'études avancées en médecine du sommeil, Hôpital du Sacré-Cœur de Montréal, Montreal, Quebec, Canada
| | - Francis L'Heureux
- 1 Centre d'études avancées en médecine du sommeil, Hôpital du Sacré-Cœur de Montréal, Montreal, Quebec, Canada.,6 Département de Neurosciences, Université de Montréal, Montreal, Quebec, Canada; and
| | - Jean-François Gagnon
- 1 Centre d'études avancées en médecine du sommeil, Hôpital du Sacré-Cœur de Montréal, Montreal, Quebec, Canada.,3 Département de psychologie, Université du Québec à Montréal, Montreal, Quebec, Canada
| | - Nadia Gosselin
- 1 Centre d'études avancées en médecine du sommeil, Hôpital du Sacré-Cœur de Montréal, Montreal, Quebec, Canada.,5 Département de psychologie, and
| |
Collapse
|
47
|
Rosciszewski G, Cadena V, Murta V, Lukin J, Villarreal A, Roger T, Ramos AJ. Toll-Like Receptor 4 (TLR4) and Triggering Receptor Expressed on Myeloid Cells-2 (TREM-2) Activation Balance Astrocyte Polarization into a Proinflammatory Phenotype. Mol Neurobiol 2017; 55:3875-3888. [PMID: 28547529 DOI: 10.1007/s12035-017-0618-z] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Accepted: 05/12/2017] [Indexed: 01/07/2023]
Abstract
Astrocytes react to brain injury with a generic response known as reactive gliosis, which involves activation of multiple intracellular pathways including several that may be beneficial for neuronal survival. However, by unknown mechanisms, reactive astrocytes can polarize into a proinflammatory phenotype that induces neurodegeneration. In order to study reactive gliosis and astroglial polarization into a proinflammatory phenotype, we used cortical devascularization-induced brain ischemia in Wistar rats and primary astroglial cell cultures exposed to oxygen-glucose deprivation (OGD). We analyzed the profile of TLR4 expression and the consequences of its activation by gain- and loss-of-function studies, and the effects produced by the activation of triggering receptor expressed on myeloid cells-2 (TREM-2), a negative regulator of TLR4 signaling. Both OGD exposure on primary astroglial cell cultures and cortical devascularization brain ischemia in rats induced TLR4 expression in astrocytes. In vivo, astroglial TLR4 expression was specifically observed in the ischemic penumbra surrounding necrotic core. Functional studies showed that OGD increased the astroglial response to the TLR4 agonist lipopolysaccharide (LPS), and conversely, TLR4 knockout primary astrocytes had impaired nuclear factor kappa-B (NF-κB) activation when exposed to LPS. In gain-of-function studies, plasmid-mediated TLR4 over-expression exacerbated astroglial response to LPS as shown by sustained NF-κB activation and increased expression of proinflammatory cytokines IL-1β and TNFα. TREM-2 expression, although present in naïve primary astrocytes, was induced by OGD, LPS, or high-mobility group box 1 protein (HMGB-1) exposure. TREM-2 activation by antibody cross-linking or the overexpression of TREM-2 intracellular adaptor, DAP12, partially suppressed LPS-induced NF-κB activation in purified astrocytic cultures. In vivo, TREM-2 expression was observed in macrophages and astrocytes located in the ischemic penumbra. While TREM-2+ macrophages were abundant at 3 days post-lesion (DPL) in the ischemic core, TREM-2+ astrocytes persisted in the penumbra until 14DPL. This study demonstrates that TLR4 expression increases astroglial sensitivity to ligands facilitating astrocyte conversion towards a proinflammatory phenotype, and that astroglial TREM-2 modulates this response reducing the downstream NF-κB activation. Therefore, the availability of TLR4 and TREM-2 ligands in the ischemic environment may control proinflammatory astroglial conversion to the neurodegenerative phenotype.
Collapse
Affiliation(s)
- Gerardo Rosciszewski
- Universidad de Buenos Aires, Facultad de Medicina, Departamento de Histología, Embriología, Biología Celular y Genética, Buenos Aires, Argentina.,Laboratorio de Neuropatología Molecular, Instituto de Biología Celular y Neurociencia "Prof. E. De Robertis" Facultad de Medicina, Universidad de Buenos Aires, Calle Paraguay 2155 3er piso (1121),, Ciudad de Buenos Aires, Argentina
| | - Vanesa Cadena
- Universidad de Buenos Aires, Facultad de Medicina, Departamento de Histología, Embriología, Biología Celular y Genética, Buenos Aires, Argentina.,Laboratorio de Neuropatología Molecular, Instituto de Biología Celular y Neurociencia "Prof. E. De Robertis" Facultad de Medicina, Universidad de Buenos Aires, Calle Paraguay 2155 3er piso (1121),, Ciudad de Buenos Aires, Argentina
| | - Veronica Murta
- Universidad de Buenos Aires, Facultad de Medicina, Departamento de Histología, Embriología, Biología Celular y Genética, Buenos Aires, Argentina.,Laboratorio de Neuropatología Molecular, Instituto de Biología Celular y Neurociencia "Prof. E. De Robertis" Facultad de Medicina, Universidad de Buenos Aires, Calle Paraguay 2155 3er piso (1121),, Ciudad de Buenos Aires, Argentina
| | - Jeronimo Lukin
- Universidad de Buenos Aires, Facultad de Medicina, Departamento de Histología, Embriología, Biología Celular y Genética, Buenos Aires, Argentina.,Laboratorio de Neuropatología Molecular, Instituto de Biología Celular y Neurociencia "Prof. E. De Robertis" Facultad de Medicina, Universidad de Buenos Aires, Calle Paraguay 2155 3er piso (1121),, Ciudad de Buenos Aires, Argentina
| | - Alejandro Villarreal
- Universidad de Buenos Aires, Facultad de Medicina, Departamento de Histología, Embriología, Biología Celular y Genética, Buenos Aires, Argentina.,Department of Molecular Embryology, Institute for Anatomy and Cell Biology, University of Freiburg, 79104, Freiburg, Germany
| | - Thierry Roger
- Infectious Diseases Service, Department of Medicine, Lausanne University Hospital, Chemin des Boveresses 155, 1066, Epalinges, Switzerland
| | - Alberto Javier Ramos
- Universidad de Buenos Aires, Facultad de Medicina, Departamento de Histología, Embriología, Biología Celular y Genética, Buenos Aires, Argentina. .,Laboratorio de Neuropatología Molecular, Instituto de Biología Celular y Neurociencia "Prof. E. De Robertis" Facultad de Medicina, Universidad de Buenos Aires, Calle Paraguay 2155 3er piso (1121),, Ciudad de Buenos Aires, Argentina.
| |
Collapse
|
48
|
Cahill LS, Gazdzinski LM, Tsui AK, Zhou YQ, Portnoy S, Liu E, Mazer CD, Hare GM, Kassner A, Sled JG. Functional and anatomical evidence of cerebral tissue hypoxia in young sickle cell anemia mice. J Cereb Blood Flow Metab 2017; 37:994-1005. [PMID: 27165012 PMCID: PMC5363475 DOI: 10.1177/0271678x16649194] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Cerebral ischemia is a significant source of morbidity in children with sickle cell anemia; however, the mechanism of injury is poorly understood. Increased cerebral blood flow and low hemoglobin levels in children with sickle cell anemia are associated with increased stroke risk, suggesting that anemia-induced tissue hypoxia may be an important factor contributing to subsequent morbidity. To better understand the pathophysiology of brain injury, brain physiology and morphology were characterized in a transgenic mouse model, the Townes sickle cell model. Relative to age-matched controls, sickle cell anemia mice demonstrated: (1) decreased brain tissue pO2 and increased expression of hypoxia signaling protein in the perivascular regions of the cerebral cortex; (2) elevated basal cerebral blood flow , consistent with adaptation to anemia-induced tissue hypoxia; (3) significant reduction in cerebrovascular blood flow reactivity to a hypercapnic challenge; (4) increased diameter of the carotid artery; and (5) significant volume changes in white and gray matter regions in the brain, as assessed by ex vivo magnetic resonance imaging. Collectively, these findings support the hypothesis that brain tissue hypoxia contributes to adaptive physiological and anatomic changes in Townes sickle cell mice. These findings may help define the pathophysiology for stroke in children with sickle cell anemia.
Collapse
Affiliation(s)
- Lindsay S Cahill
- 1 Mouse Imaging Centre, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Lisa M Gazdzinski
- 1 Mouse Imaging Centre, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Albert Ky Tsui
- 2 Keenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute of St. Michael's Hospital, Department of Anesthesia, St. Michael's Hospital, University of Toronto, Toronto, Ontario, Canada
| | - Yu-Qing Zhou
- 1 Mouse Imaging Centre, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Sharon Portnoy
- 1 Mouse Imaging Centre, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Elaine Liu
- 2 Keenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute of St. Michael's Hospital, Department of Anesthesia, St. Michael's Hospital, University of Toronto, Toronto, Ontario, Canada
| | - C David Mazer
- 2 Keenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute of St. Michael's Hospital, Department of Anesthesia, St. Michael's Hospital, University of Toronto, Toronto, Ontario, Canada.,3 Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Gregory Mt Hare
- 2 Keenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute of St. Michael's Hospital, Department of Anesthesia, St. Michael's Hospital, University of Toronto, Toronto, Ontario, Canada.,3 Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Andrea Kassner
- 4 Department of Medical Imaging, University of Toronto and The Hospital for Sick Children, Toronto, Ontario, Canada
| | - John G Sled
- 1 Mouse Imaging Centre, The Hospital for Sick Children, Toronto, Ontario, Canada.,5 Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
49
|
Brabazon F, Wilson CM, Shukla DK, Mathur S, Jaiswal S, Bermudez S, Byrnes KR, Selwyn R. [18F]FDG-PET Combined with MRI Elucidates the Pathophysiology of Traumatic Brain Injury in Rats. J Neurotrauma 2017; 34:1074-1085. [DOI: 10.1089/neu.2016.4540] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Fiona Brabazon
- Neuroscience Program, Uniformed Services University of the Health Sciences, Bethesda, Maryland
| | - Colin M. Wilson
- Department of Radiology, Uniformed Services University of the Health Sciences, Bethesda, Maryland
- Center for Neuroscience and Regenerative Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland
| | - Dinesh K Shukla
- Department of Radiology, Uniformed Services University of the Health Sciences, Bethesda, Maryland
- Center for Neuroscience and Regenerative Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland
| | - Sanjeev Mathur
- Department of Radiology, Uniformed Services University of the Health Sciences, Bethesda, Maryland
- Center for Neuroscience and Regenerative Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland
| | - Shalini Jaiswal
- Department of Radiology, Uniformed Services University of the Health Sciences, Bethesda, Maryland
- Center for Neuroscience and Regenerative Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland
| | - Sara Bermudez
- Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, Bethesda, Maryland
| | - Kimberly R. Byrnes
- Neuroscience Program, Uniformed Services University of the Health Sciences, Bethesda, Maryland
- Center for Neuroscience and Regenerative Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland
- Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, Bethesda, Maryland
| | - Reed Selwyn
- Department of Radiology, Uniformed Services University of the Health Sciences, Bethesda, Maryland
- Center for Neuroscience and Regenerative Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland
- Department of Radiology, University of New Mexico, Albuquerque, New Mexico
| |
Collapse
|
50
|
Yagishita S, Suzuki S, Yoshikawa K, Iida K, Hirata A, Suzuki M, Takashima A, Maruyama K, Hirasawa A, Awaji T. Treatment of intermittent hypoxia increases phosphorylated tau in the hippocampus via biological processes common to aging. Mol Brain 2017; 10:2. [PMID: 28057021 PMCID: PMC5217192 DOI: 10.1186/s13041-016-0282-7] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Accepted: 12/15/2016] [Indexed: 12/17/2022] Open
Abstract
Sleep-disordered breathing produces cognitive impairments, and is possibly associated with Alzheimer disease (AD). Intermittent hypoxia treatment (IHT), an experimental model for sleep-disordered breathing, results in cognitive impairments in animals via unknown mechanisms. Here, we exposed mice to IHT protocols, and performed biochemical analyses and microarray analyses regarding their hippocampal samples. In particular, we performed gene ontology (GO)-based microarray analysis to elucidate effects of IHT on hippocampal functioning, which were compared with the effects of various previously-reported experimental conditions on that (ref. Gene Expression Omnibus, The National Center for Biotechnology Information). Our microarray analyses revealed that IHT and aging shared alterations in some common GO, which were also observed with kainic acid treatment, Dicer ablation, or moderate glutamate excess. Mapping the altered genes using the Kyoto Encyclopedia of Genes and Genomes PATHWAY database indicated that IHT and aging affected several pathways including “MAPK signaling pathway”, “PI3K-Akt signaling pathway”, and “glutamatergic synapse”. Consistent with the gene analyses, in vivo analyses revealed that IHT increased phosphorylated tau, reflecting an imbalance of kinases and/or phosphatases, and reduced proteins relevant to glutamatergic synapses. In addition, IHT increased phosphorylated p70 S6 kinase, indicating involvement of the mammalian target of rapamycin signaling pathway. Furthermore, IHT mice demonstrated hyperactivity in Y-maze tests, which was also observed in AD models. We obtained important data or something from the massive amount of microarray data, and confirmed the validity by in vivo analyses: the IHT-induced cognitive impairment may be partially explained by the fact that IHT increases phosphorylated tau via biological processes common to aging. Moreover, as aging is a major risk factor for AD, IHT is a novel model for investigating the pathological processes contributing to AD onset.
Collapse
Affiliation(s)
- Sosuke Yagishita
- Department of Pharmacology, Faculty of Medicine, Saitama Medical University, 38 Moro-hongo, Moroyama-machi, Iruma-gun, Saitama, 350-0495, Japan. .,Present address: Department of Peripheral Nervous System Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, 4-1-1 Ogawa-higashi, Kodaira, Tokyo, 187-8502, Japan.
| | - Seiya Suzuki
- Department of Pharmacology, Faculty of Medicine, Saitama Medical University, 38 Moro-hongo, Moroyama-machi, Iruma-gun, Saitama, 350-0495, Japan.,Faculty of Health and Medical Care, Saitama Medical University, 1397-1 Yamane, Hidaka-shi, Saitama, 350-1241, Japan
| | - Keisuke Yoshikawa
- Department of Pharmacology, Faculty of Medicine, Saitama Medical University, 38 Moro-hongo, Moroyama-machi, Iruma-gun, Saitama, 350-0495, Japan
| | - Keiko Iida
- Department of Genomic Drug Discovery Science, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida Shimoadachi-cho, Sakyo-ku, Kyoto, 606-8501, Japan
| | - Ayako Hirata
- Department of Genomic Drug Discovery Science, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida Shimoadachi-cho, Sakyo-ku, Kyoto, 606-8501, Japan
| | - Masahiko Suzuki
- Department of Pharmacology, Faculty of Medicine, Saitama Medical University, 38 Moro-hongo, Moroyama-machi, Iruma-gun, Saitama, 350-0495, Japan.,Faculty of Health and Medical Care, Saitama Medical University, 1397-1 Yamane, Hidaka-shi, Saitama, 350-1241, Japan
| | - Akihiko Takashima
- Department of Life Science, Faculty of Science, Gakushuin University, 1-5-1 Mejiro, Toshima-ku, Tokyo, 171-8588, Japan
| | - Kei Maruyama
- Department of Pharmacology, Faculty of Medicine, Saitama Medical University, 38 Moro-hongo, Moroyama-machi, Iruma-gun, Saitama, 350-0495, Japan
| | - Akira Hirasawa
- Department of Genomic Drug Discovery Science, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida Shimoadachi-cho, Sakyo-ku, Kyoto, 606-8501, Japan.
| | - Takeo Awaji
- Department of Pharmacology, Faculty of Medicine, Saitama Medical University, 38 Moro-hongo, Moroyama-machi, Iruma-gun, Saitama, 350-0495, Japan
| |
Collapse
|