51
|
Li JZ, Hao XH, Wu HP, Li M, Liu XM, Wu ZB. An enriched environment delays the progression from mild cognitive impairment to Alzheimer's disease in senescence-accelerated mouse prone 8 mice. Exp Ther Med 2021; 22:1320. [PMID: 34630674 PMCID: PMC8495563 DOI: 10.3892/etm.2021.10755] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Accepted: 10/16/2020] [Indexed: 12/19/2022] Open
Abstract
A previous study demonstrated that middle-aged (5-6 months of age) senescence-accelerated mouse prone 8 (SAMP8) mice can be used as animal models of mild cognitive impairment (MCI). An enriched environment (EE) can mitigate cognitive decline and decrease the pathological changes associated with various neurodegenerative diseases. In the present study, the learning-memory abilities of SAMP8 mice during the MCI phase (5 months of age) was evaluated and neuropathological changes in the hippocampus were examined after the mice were exposed to an EE for 60 days. In the Morris water maze test, EE-exposed mice demonstrated significantly decreased escape latency and increased time spent in the target quadrant and number of platform crossings compared with control mice. Terminal deoxynucleotidyl transferase dUTP nick end labeling and Nissl staining showed that EE-exposed mice had reduced neuronal apoptosis and increased number of surviving neurons compared with control mice. Golgi staining, transmission electron microscopy, and immunohistochemical staining demonstrated that EE-exposed mice exhibited increased dendritic spine densities among secondary and tertiary apical dendrites; increases in synaptic numerical density, synaptic surface density, and expression of synaptophysin; and reduced deposition of amyloid-β (Aβ) and expression of amyloid-precursor protein (APP) in the hippocampal CA1 region compared with control mice. These results demonstrate that EE exposure effectively decreases neuronal loss and regulates neuronal synaptic plasticity by reducing the expression of APP and the deposition of Aβ in the hippocampal CA1 region, thereby mitigating cognitive decline in SAMP8 mice during the MCI phase and delaying the progression from MCI to Alzheimer's disease.
Collapse
Affiliation(s)
- Jian-Zhong Li
- Department of Human Anatomy, Changzhi Medical College, Changzhi, Shangxi 046000, P.R. China
| | - Xing-Hua Hao
- Department of Clinical Psychology, Heping Hospital Affiliated to Changzhi Medical College, Changzhi, Shangxi 046000, P.R. China
| | - Hai-Ping Wu
- Department of Human Anatomy, Changzhi Medical College, Changzhi, Shangxi 046000, P.R. China
| | - Ming Li
- Department of Human Anatomy, Changzhi Medical College, Changzhi, Shangxi 046000, P.R. China
| | - Xue-Min Liu
- Department of Human Anatomy, Changzhi Medical College, Changzhi, Shangxi 046000, P.R. China
| | - Zhi-Bing Wu
- Department of Human Anatomy, Changzhi Medical College, Changzhi, Shangxi 046000, P.R. China
| |
Collapse
|
52
|
Different changes in pre- and postsynaptic components in the hippocampal CA1 subfield after transient global cerebral ischemia. Brain Struct Funct 2021; 227:345-360. [PMID: 34626230 DOI: 10.1007/s00429-021-02404-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 09/29/2021] [Indexed: 10/20/2022]
Abstract
To date, ischemia-induced damage to dendritic spines has attracted considerable attention, while the possible effects of ischemia on presynaptic components has received relatively less attention. To further examine ischemia-induced changes in pre- and postsynaptic specializations in the hippocampal CA1 subfield, we modeled global cerebral ischemia with two-stage 4-vessel-occlusion in rats, and found that three postsynaptic markers, microtubule-associated protein 2 (MAP2), postsynaptic density protein 95 (PSD95), and filamentous F-actin (F-actin), were all substantially decreased in the CA1 subfield after ischemia/reperfusion (I/R). Although no significant change was detected in synapsin I, a presynaptic marker, in the CA1 subfield at the protein level, confocal microscopy revealed that the number and size of synapsin I puncta were significantly changed in the CA1 stratum radiatum after I/R. The size of synapsin I puncta became slightly, but significantly reduced on Day 1.5 after I/R. From Days 2 to 7 after I/R, the number of synapsin I puncta became moderately decreased, while the size of synapsin I puncta was significantly increased. Interestingly, some enlarged puncta of synapsin I were observed in close proximity to the dendritic shafts of CA1 pyramidal cells. Due to the more substantial decrease in the number of F-actin puncta, the ratio of synapsin I/F-actin puncta was significantly increased after I/R. The decrease in synapsin I puncta size in the early stage of I/R may be the result of excessive neurotransmitter release due to I/R-induced hyperexcitability in CA3 pyramidal cells, while the increase in synapsin I puncta in the later stage of I/R may reflect a disability of synaptic vesicle release due to the loss of postsynaptic contacts.
Collapse
|
53
|
Fan Y, Shen M, Huo Y, Gao X, Li C, Zheng R, Zhang J. Total Cerebral Small Vessel Disease Burden on MRI Correlates With Medial Temporal Lobe Atrophy and Cognitive Performance in Patients of a Memory Clinic. Front Aging Neurosci 2021; 13:698035. [PMID: 34566621 PMCID: PMC8456168 DOI: 10.3389/fnagi.2021.698035] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Accepted: 08/09/2021] [Indexed: 11/13/2022] Open
Abstract
Background: Cerebral small vessel disease (cSVD) and neurodegeneration are the two main causes of dementia and are considered distinct pathological processes, while studies have shown overlaps and interactions between the two pathological pathways. Medial temporal atrophy (MTA) is considered a classic marker of neurodegeneration. We aimed to investigate the relationship of total cSVD burden and MTA on MRI using a total cSVD score and to explore the impact of the two MRI features on cognition. Methods: Patients in a memory clinic were enrolled, who underwent brain MRI scan and cognitive evaluation within 7 days after the first visit. MTA and total cSVD score were rated using validated visual scales. Cognitive function was assessed by using Mini-Mental State Examination (MMSE) and Montreal Cognitive Assessment (MoCA) scales. Spearman's correlation and regression models were used to test (i) the association between MTA and total cSVD score as well as each cSVD marker and (ii) the correlation of the MRI features and cognitive status. Results: A total of 312 patients were finally enrolled, with a median age of 75.0 (66.0-80.0) years and 40.7% (127/312) males. All of them finished MRI and MMSE, and 293 subjects finished MoCA. Of note, 71.8% (224/312) of the patients had at least one of the cSVD markers, and 48.7% (152/312) of them had moderate-severe MTA. The total cSVD score was independently associated with MTA levels, after adjusting for age, gender, years of education, and other vascular risk factors (OR 1.191, 95% CI 1.071-1.324, P = 0.001). In regard to individual markers, a significant association existed only between white matter hyperintensities and MTA after adjusting for the factors mentioned above (OR 1.338, 95% CI 1.050-1.704, P = 0.018). Both MTA and total cSVD score were independent risk factors for MMSE ≤ 26 (MTA: OR 1.877, 95% CI 1.407-2.503, P < 0.001; total cSVD score: OR 1.474, 95% CI 1.132-1.921, P = 0.004), and MoCA < 26 (MTA: OR 1.629, 95% CI 1.112-2.388, P = 0.012; total cSVD score: OR 1.520, 95% CI 1.068-2.162, P = 0.020). Among all the cSVD markers, microbleed was found significantly associated with MMSE ≤ 26, while no marker was demonstrated a relationship with MoCA < 26. Conclusion: Cerebral small vessel disease was related to MTA in patients of a memory clinic, and both the MRI features had a significant association with cognitive impairment.
Collapse
Affiliation(s)
- Yangyi Fan
- Department of Neurology, Peking University People's Hospital, Beijing, China
| | - Ming Shen
- Department of Neurology, Peking University People's Hospital, Beijing, China
| | - Yang Huo
- Department of Neurology, Peking University People's Hospital, Beijing, China
| | - Xuguang Gao
- Department of Neurology, Peking University People's Hospital, Beijing, China
| | - Chun Li
- Department of Rheumatology and Immunology, Peking University People's Hospital, Beijing, China
| | - Ruimao Zheng
- Neuroscience Research Institute, Peking University, Beijing, China
| | - Jun Zhang
- Department of Neurology, Peking University People's Hospital, Beijing, China
| |
Collapse
|
54
|
Golubiani G, Lagani V, Solomonia R, Müller M. Metabolomic Fingerprint of Mecp2-Deficient Mouse Cortex: Evidence for a Pronounced Multi-Facetted Metabolic Component in Rett Syndrome. Cells 2021; 10:cells10092494. [PMID: 34572143 PMCID: PMC8472238 DOI: 10.3390/cells10092494] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 09/17/2021] [Accepted: 09/18/2021] [Indexed: 01/10/2023] Open
Abstract
Using unsupervised metabolomics, we defined the complex metabolic conditions in the cortex of a mouse model of Rett syndrome (RTT). RTT, which represents a cause of mental and cognitive disabilities in females, results in profound cognitive impairment with autistic features, motor disabilities, seizures, gastrointestinal problems, and cardiorespiratory irregularities. Typical RTT originates from mutations in the X-chromosomal methyl-CpG-binding-protein-2 (Mecp2) gene, which encodes a transcriptional modulator. It then causes a deregulation of several target genes and metabolic alterations in the nervous system and peripheral organs. We identified 101 significantly deregulated metabolites in the Mecp2-deficient cortex of adult male mice; 68 were increased and 33 were decreased compared to wildtypes. Pathway analysis identified 31 mostly upregulated metabolic pathways, in particular carbohydrate and amino acid metabolism, key metabolic mitochondrial/extramitochondrial pathways, and lipid metabolism. In contrast, neurotransmitter-signaling is dampened. This metabolic fingerprint of the Mecp2-deficient cortex of severely symptomatic mice provides further mechanistic insights into the complex RTT pathogenesis. The deregulated pathways that were identified—in particular the markedly affected amino acid and carbohydrate metabolism—confirm a complex and multifaceted metabolic component in RTT, which in turn signifies putative therapeutic targets. Furthermore, the deregulated key metabolites provide a choice of potential biomarkers for a more detailed rating of disease severity and disease progression.
Collapse
Affiliation(s)
- Gocha Golubiani
- Institut für Neuro- und Sinnesphysiologie, Zentrum Physiologie und Pathophysiologie, Universitätsmedizin Göttingen, Georg-August-Universität Göttingen, D-37130 Göttingen, Germany;
- Institute of Chemical Biology, Ilia State University, 0162 Tbilisi, Georgia; (V.L.); (R.S.)
| | - Vincenzo Lagani
- Institute of Chemical Biology, Ilia State University, 0162 Tbilisi, Georgia; (V.L.); (R.S.)
| | - Revaz Solomonia
- Institute of Chemical Biology, Ilia State University, 0162 Tbilisi, Georgia; (V.L.); (R.S.)
| | - Michael Müller
- Institut für Neuro- und Sinnesphysiologie, Zentrum Physiologie und Pathophysiologie, Universitätsmedizin Göttingen, Georg-August-Universität Göttingen, D-37130 Göttingen, Germany;
- Correspondence: ; Tel.: +49-551-39-22933
| |
Collapse
|
55
|
Transient Oxygen-Glucose Deprivation Causes Region- and Cell Type-Dependent Functional Deficits in the Mouse Hippocampus In Vitro. eNeuro 2021; 8:ENEURO.0221-21.2021. [PMID: 34475264 PMCID: PMC8482850 DOI: 10.1523/eneuro.0221-21.2021] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 06/21/2021] [Accepted: 07/03/2021] [Indexed: 01/11/2023] Open
Abstract
Neurons are highly vulnerable to conditions of hypoxia-ischemia (HI) such as stroke or transient ischemic attacks. Recovery of cognitive and behavioral functions requires re-emergence of coordinated network activity, which, in turn, relies on the well-orchestrated interaction of pyramidal cells (PYRs) and interneurons. We therefore modelled HI in the mouse hippocampus, a particularly vulnerable region showing marked loss of PYR and fast-spiking interneurons (FSIs) after hypoxic-ischemic insults. Transient oxygen-glucose deprivation (OGD) in ex vivo hippocampal slices led to a rapid loss of neuronal activity and spontaneous network oscillations (sharp wave-ripple complexes; SPW-Rs), and to the occurrence of a spreading depolarization. Following reperfusion, both SPW-R and neuronal spiking resumed, but FSI activity remained strongly reduced compared with PYR. Whole-cell recordings in CA1 PYR revealed, however, a similar reduction of both EPSCs and IPSCs, leaving inhibition-excitation (I/E) balance unaltered. At the network level, SPW-R incidence was strongly reduced and the remaining network events showed region-specific changes including reduced ripple energy in CA3 and increased ripple frequency in CA1. Together, our data show that transient hippocampal energy depletion results in severe functional alterations at the cellular and network level. While I/E balance is maintained, synaptic activity, interneuron spiking and coordinated network patterns remain reduced. Such alterations may be network-level correlates of cognitive and functional deficits after cerebral HI.
Collapse
|
56
|
Li X, Wen W, Li P, Fu Y, Chen H, Wang F, Dai Y, Xu S. Mitochondrial Protection and Against Glutamate Neurotoxicity via Shh/Ptch1 Signaling Pathway to Ameliorate Cognitive Dysfunction by Kaixin San in Multi-Infarct Dementia Rats. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:5590745. [PMID: 34306310 PMCID: PMC8285175 DOI: 10.1155/2021/5590745] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Accepted: 06/17/2021] [Indexed: 12/05/2022]
Abstract
Multi-infarct dementia (MID), a prominent subtype of vascular dementia (VD), is responsible for at least 15 to 20 percent of dementia in the elderly. Mitochondrial dysfunctions and glutamate neurotoxicity due to chronic hypoperfusion and oxidative stress were regarded as the major risk factors in the pathogenesis. Kaixin San (KXS), a classic prescription of Beiji Qianjin Yaofang, was applied to treatment for "amnesia" and has been demonstrated to alleviate the cognitive deficit in a variety of dementias, including MID. However, little is known whether mitochondria and glutamate are associated with the protection of KXS in MID treatment. The aim of this study was to investigate the role of KXS in improving the cognitive function of MID rats through strengthening mitochondrial functions and antagonizing glutamate neurotoxicity via the Shh/Ptch1 signaling pathway. Our data showed that KXS significantly ameliorated memory impairment and hippocampal neuron damage in MID rats. Moreover, KXS improved hippocampal mitochondrial functions by reducing the degree of mitochondrial swelling, increasing the mitochondrial membrane potential (MMP), and elevating the energy charge (EC) and ATP content in MID rats. As expected, the concentration of glutamate and the expression of p-NMDAR1 were significantly reduced by KXS in the brain tissue of MID rats. Furthermore, our results showed that KXS noticeably activated the Shh/Ptch1 signaling pathway which was demonstrated by remarkable elevations of Ptch1, Smo, and Gli1 protein levels in the brain tissue of MID rats. Intriguingly, the inhibition of the Shh signaling pathway with cyclopamine significantly inhibited the protective effects of KXS on glutamate-induced neurotoxicity in PC12 cells. To sum up, these findings suggested that KXS protected MID rats from memory loss by rescuing mitochondrial functions as well as against glutamate neurotoxicity through activating Shh/Ptch1 signaling pathway.
Collapse
Affiliation(s)
- Xiaoqiong Li
- Institute of Material Medica Integration and Transformation for Brain Disorders, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 611137, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 611137, China
| | - Wen Wen
- Institute of Material Medica Integration and Transformation for Brain Disorders, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 611137, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 611137, China
| | - Ping Li
- Institute of Material Medica Integration and Transformation for Brain Disorders, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 611137, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 611137, China
| | - Ying Fu
- Institute of Material Medica Integration and Transformation for Brain Disorders, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 611137, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 611137, China
| | - Hao Chen
- Institute of Material Medica Integration and Transformation for Brain Disorders, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 611137, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 611137, China
| | - Fushun Wang
- Institute of Brain and Psychological Science, Sichuan Normal University, Chengdu 610060, China
| | - Yuan Dai
- Institute of Material Medica Integration and Transformation for Brain Disorders, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 611137, China
- School of Health Preservation and Rehabilitation, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 610075, China
| | - Shijun Xu
- Institute of Material Medica Integration and Transformation for Brain Disorders, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 611137, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 611137, China
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu, Sichuan 611137, China
| |
Collapse
|
57
|
Bormann D, Stojanovic T, Cicvaric A, Schuld GJ, Cabatic M, Ankersmit HJ, Monje FJ. miRNA-132/212 Gene-Deletion Aggravates the Effect of Oxygen-Glucose Deprivation on Synaptic Functions in the Female Mouse Hippocampus. Cells 2021; 10:1709. [PMID: 34359879 PMCID: PMC8306255 DOI: 10.3390/cells10071709] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 07/01/2021] [Accepted: 07/02/2021] [Indexed: 12/29/2022] Open
Abstract
Cerebral ischemia and its sequelae, which include memory impairment, constitute a leading cause of disability worldwide. Micro-RNAs (miRNA) are evolutionarily conserved short-length/noncoding RNA molecules recently implicated in adaptive/maladaptive neuronal responses to ischemia. Previous research independently implicated the miRNA-132/212 cluster in cholinergic signaling and synaptic transmission, and in adaptive/protective mechanisms of neuronal responses to hypoxia. However, the putative role of miRNA-132/212 in the response of synaptic transmission to ischemia remained unexplored. Using hippocampal slices from female miRNA-132/212 double-knockout mice in an established electrophysiological model of ischemia, we here describe that miRNA-132/212 gene-deletion aggravated the deleterious effect of repeated oxygen-glucose deprivation insults on synaptic transmission in the dentate gyrus, a brain region crucial for learning and memory functions. We also examined the effect of miRNA-132/212 gene-deletion on the expression of key mediators in cholinergic signaling that are implicated in both adaptive responses to ischemia and hippocampal neural signaling. miRNA-132/212 gene-deletion significantly altered hippocampal AChE and mAChR-M1, but not α7-nAChR or MeCP2 expression. The effects of miRNA-132/212 gene-deletion on hippocampal synaptic transmission and levels of cholinergic-signaling elements suggest the existence of a miRNA-132/212-dependent adaptive mechanism safeguarding the functional integrity of synaptic functions in the acute phase of cerebral ischemia.
Collapse
Affiliation(s)
- Daniel Bormann
- Center for Physiology and Pharmacology, Department of Neurophysiology and Neuropharmacology, Medical University of Vienna, Schwarzspanierstraße 17, 1090 Vienna, Austria; (D.B.); (T.S.); (G.J.S.); (M.C.)
- Laboratory for Cardiac and Thoracic Diagnosis, Department of Surgery, Regeneration and Applied Immunology, Medical University of Vienna, Research Laboratories Vienna General Hospital, Waehringer Guertel 18-20, 1090 Vienna, Austria;
- Division of Thoracic Surgery, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Tamara Stojanovic
- Center for Physiology and Pharmacology, Department of Neurophysiology and Neuropharmacology, Medical University of Vienna, Schwarzspanierstraße 17, 1090 Vienna, Austria; (D.B.); (T.S.); (G.J.S.); (M.C.)
| | - Ana Cicvaric
- Department of Psychiatry and Behavioral Sciences, Albert Einstein College of Medicine, New York, NY 10461, USA;
| | - Gabor J. Schuld
- Center for Physiology and Pharmacology, Department of Neurophysiology and Neuropharmacology, Medical University of Vienna, Schwarzspanierstraße 17, 1090 Vienna, Austria; (D.B.); (T.S.); (G.J.S.); (M.C.)
| | - Maureen Cabatic
- Center for Physiology and Pharmacology, Department of Neurophysiology and Neuropharmacology, Medical University of Vienna, Schwarzspanierstraße 17, 1090 Vienna, Austria; (D.B.); (T.S.); (G.J.S.); (M.C.)
| | - Hendrik Jan Ankersmit
- Laboratory for Cardiac and Thoracic Diagnosis, Department of Surgery, Regeneration and Applied Immunology, Medical University of Vienna, Research Laboratories Vienna General Hospital, Waehringer Guertel 18-20, 1090 Vienna, Austria;
- Division of Thoracic Surgery, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
- Aposcience AG, Dresdner Straße 87/A 21, 1200 Vienna, Austria
| | - Francisco J. Monje
- Center for Physiology and Pharmacology, Department of Neurophysiology and Neuropharmacology, Medical University of Vienna, Schwarzspanierstraße 17, 1090 Vienna, Austria; (D.B.); (T.S.); (G.J.S.); (M.C.)
| |
Collapse
|
58
|
Vockert N, Perosa V, Ziegler G, Schreiber F, Priester A, Spallazzi M, Garcia-Garcia B, Aruci M, Mattern H, Haghikia A, Düzel E, Schreiber S, Maass A. Hippocampal vascularization patterns exert local and distant effects on brain structure but not vascular pathology in old age. Brain Commun 2021; 3:fcab127. [PMID: 34222874 PMCID: PMC8249103 DOI: 10.1093/braincomms/fcab127] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Revised: 04/25/2021] [Accepted: 06/03/2021] [Indexed: 12/29/2022] Open
Abstract
The hippocampus within the medial temporal lobe is highly vulnerable to age-related pathology such as vascular disease. We examined hippocampal vascularization patterns by harnessing the ultra-high resolution of 7 Tesla magnetic resonance angiography. Dual-supply hemispheres with a contribution of the anterior choroidal artery to hippocampal blood supply were distinguished from single-supply ones with a sole dependence on the posterior cerebral artery. A recent study indicated that a dual vascular supply is related to preserved cognition and structural hippocampal integrity in old age and vascular disease. Here, we examined the regional specificity of these structural benefits at the level of medial temporal lobe sub-regions and hemispheres. In a cross-sectional study with an older cohort of 17 patients with cerebral small vessel disease (70.7 ± 9.0 years, 35.5% female) and 27 controls (71.1 ± 8.2 years, 44.4% female), we demonstrate that differences in grey matter volumes related to the hippocampal vascularization pattern were specifically observed in the anterior hippocampus and entorhinal cortex. These regions were especially bigger in dual-supply hemispheres, but also seemed to benefit from a contralateral dual supply. We further show that total grey matter volumes were greater in people with at least one dual-supply hemisphere, indicating that the hippocampal vascularization pattern has more far-reaching structural implications beyond the medial temporal lobe. A mediation analysis identified total grey matter as a mediator of differences in global cognition. However, our analyses on multiple neuroimaging markers for cerebral small vessel disease did not reveal any evidence that an augmented hippocampal vascularization conveys resistance nor resilience against vascular pathology. We propose that an augmented hippocampal vascularization might contribute to maintaining structural integrity in the brain and preserving cognition despite age-related degeneration. As such, the binary hippocampal vascularization pattern could have major implications for brain structure and function in ageing and dementia independent of vascular pathology, while presenting a simple framework with potential applicability to the clinical setting.
Collapse
Affiliation(s)
- Niklas Vockert
- German Center for Neurodegenerative Diseases, 39120 Magdeburg, Germany
| | - Valentina Perosa
- Department of Neurology, Otto von Guericke University Magdeburg, 39120 Magdeburg, Germany
- J. Philip Kistler Stroke Research Center, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Gabriel Ziegler
- German Center for Neurodegenerative Diseases, 39120 Magdeburg, Germany
- Institute of Cognitive Neurology and Dementia Research, Otto-von-Guericke University, 39120 Magdeburg, Germany
| | - Frank Schreiber
- German Center for Neurodegenerative Diseases, 39120 Magdeburg, Germany
- Department of Neurology, Otto von Guericke University Magdeburg, 39120 Magdeburg, Germany
| | - Anastasia Priester
- Department of Neuroradiology, University Hospital Heidelberg, 69120 Heidelberg, Germany
| | - Marco Spallazzi
- Department of Medicine and Surgery, Unit of Neurology, Azienda Ospedaliero- Universitaria, 43126 Parma, Italy
| | - Berta Garcia-Garcia
- German Center for Neurodegenerative Diseases, 39120 Magdeburg, Germany
- Department of Neurology, Otto von Guericke University Magdeburg, 39120 Magdeburg, Germany
| | - Merita Aruci
- German Center for Neurodegenerative Diseases, 39120 Magdeburg, Germany
| | - Hendrik Mattern
- Biomedical Magnetic Resonance, Otto von Guericke University Magdeburg, 39120 Magdeburg, Germany
| | - Aiden Haghikia
- German Center for Neurodegenerative Diseases, 39120 Magdeburg, Germany
- Department of Neurology, Otto von Guericke University Magdeburg, 39120 Magdeburg, Germany
| | - Emrah Düzel
- German Center for Neurodegenerative Diseases, 39120 Magdeburg, Germany
- Department of Neurology, Otto von Guericke University Magdeburg, 39120 Magdeburg, Germany
- Institute of Cognitive Neurology and Dementia Research, Otto-von-Guericke University, 39120 Magdeburg, Germany
- Institute of Cognitive Neuroscience, University College London, London WC1N 3AZ, UK
- Center for Behavioral Brain Sciences (CBBS), 39106 Magdeburg, Germany
| | - Stefanie Schreiber
- German Center for Neurodegenerative Diseases, 39120 Magdeburg, Germany
- Department of Neurology, Otto von Guericke University Magdeburg, 39120 Magdeburg, Germany
- Center for Behavioral Brain Sciences (CBBS), 39106 Magdeburg, Germany
| | - Anne Maass
- German Center for Neurodegenerative Diseases, 39120 Magdeburg, Germany
| |
Collapse
|
59
|
Lee TK, Lee JC, Kim DW, Kim B, Sim H, Kim JD, Ahn JH, Park JH, Lee CH, Won MH, Choi SY. Ischemia-Reperfusion under Hyperthermia Increases Heme Oxygenase-1 in Pyramidal Neurons and Astrocytes with Accelerating Neuronal Loss in Gerbil Hippocampus. Int J Mol Sci 2021; 22:ijms22083963. [PMID: 33921375 PMCID: PMC8068892 DOI: 10.3390/ijms22083963] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 04/03/2021] [Accepted: 04/06/2021] [Indexed: 01/14/2023] Open
Abstract
It has been studied that the damage or death of neurons in the hippocampus is different according to hippocampal subregions, cornu ammonis 1–3 (CA1–3), after transient ischemia in the forebrain, showing that pyramidal neurons located in the subfield CA1 (CA1) are most vulnerable to this ischemia. Hyperthermia is a proven risk factor for brain ischemia and can develop more severe and extensive brain damage related with mortality rate. It is well known that heme oxygenase-1 (HO-1) activity and expression is increased by various stimuli in the brain, including hyperthermia. HO-1 can be either protective or deleterious in the central nervous system, and its roles depend on the expression levels of enzymes. In this study, we investigated the effects of hyperthermia during ischemia on HO-1 expression and neuronal damage/death in the hippocampus to examine the relationship between HO-1 and neuronal damage/death following 5-min transient ischemia in the forebrain using gerbils. Gerbils were assigned to four groups: (1) sham-operated gerbils with normothermia (Normo + sham group); (2) ischemia-operated gerbils with normothermia (Normo + ischemia group); (3) sham-operated gerbils with hyperthermia (39.5 ± 0.2 °C) during ischemia (Hyper + sham group); and (4) ischemia-operated gerbils with hyperthermia during ischemia (Hyper + ischemia group). HO-1 expression levels in CA1–3 of the Hyper + ischemia group were significantly higher than those in the Normo + ischemia group. HO-1 immunoreactivity in the Hyper + ischemia group was significantly increased in pyramidal neurons and astrocytes with time after ischemia, and the immunoreactivity was significantly higher than that in the Normo + ischemia group. In the Normo + Ischemia group, neuronal death was shown in pyramidal neurons located only in CA1 at 5 days after ischemia. However, in the Hyper + ischemia group, pyramidal neuronal death occurred in CA1–3 at 2 days after ischemia. Taken together, our findings showed that brain ischemic insult during hyperthermic condition brings up earlier and severer neuronal damage/death in the hippocampus, showing that HO-1 expression in neurons and astrocytes is different according to brain subregions and temperature condition. Based on these findings, we suggest that hyperthermia in patients with ischemic stroke must be taken into the consideration in the therapy.
Collapse
Affiliation(s)
- Tae-Kyeong Lee
- Department of Biomedical Science and Research Institute for Bioscience and Biotechnology, Hallym University, Chuncheon 24252, Gangwon, Korea;
| | - Jae-Chul Lee
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon 24341, Gangwon, Korea; (J.-C.L.); (B.K.); (H.S.); (J.H.A.)
| | - Dae Won Kim
- Department of Biochemistry and Molecular Biology, Research Institute of Oral Sciences, College of Dentistry, Gangnung-Wonju National University, Gangneung 25457, Gangwon, Korea;
| | - Bora Kim
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon 24341, Gangwon, Korea; (J.-C.L.); (B.K.); (H.S.); (J.H.A.)
| | - Hyejin Sim
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon 24341, Gangwon, Korea; (J.-C.L.); (B.K.); (H.S.); (J.H.A.)
| | - Jong Dai Kim
- Division of Food Biotechnology, School of Biotechnology, Kangwon National University, Chuncheon 24341, Gangwon, Korea;
| | - Ji Hyeon Ahn
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon 24341, Gangwon, Korea; (J.-C.L.); (B.K.); (H.S.); (J.H.A.)
- Department of Physical Therapy, College of Health Science, Youngsan University, Yangsan 50510, Gyeongnam, Korea
| | - Joon Ha Park
- Department of Anatomy, College of Korean Medicine, Dongguk University, Gyeongju 38066, Gyeongbuk, Korea;
| | - Choong-Hyun Lee
- Department of Pharmacy, College of Pharmacy, Dankook University, Cheonan 31116, Chungnam, Korea;
| | - Moo-Ho Won
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon 24341, Gangwon, Korea; (J.-C.L.); (B.K.); (H.S.); (J.H.A.)
- Correspondence: (M.-H.W.); (S.Y.C.); Tel.: +82-33-250-8891 (M.-H.W.); +82-33-248-2112 (S.Y.C.); Fax: +82-33-256-1614 (M.-H.W.); +82-33-241-1463 (S.Y.C.)
| | - Soo Young Choi
- Department of Biomedical Science and Research Institute for Bioscience and Biotechnology, Hallym University, Chuncheon 24252, Gangwon, Korea;
- Correspondence: (M.-H.W.); (S.Y.C.); Tel.: +82-33-250-8891 (M.-H.W.); +82-33-248-2112 (S.Y.C.); Fax: +82-33-256-1614 (M.-H.W.); +82-33-241-1463 (S.Y.C.)
| |
Collapse
|
60
|
Ko IG, Jin JJ, Hwang L, Kim SH, Kim CJ, Jeon JW, Chung JY, Han JH. Adenosine A2A receptor agonist polydeoxyribonucleotide ameliorates short-term memory impairment by suppressing cerebral ischemia-induced inflammation via MAPK pathway. PLoS One 2021; 16:e0248689. [PMID: 33735236 PMCID: PMC7971468 DOI: 10.1371/journal.pone.0248689] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Accepted: 03/03/2021] [Indexed: 12/12/2022] Open
Abstract
Cerebral ischemia causes tissue death owing to occlusion of the cerebral blood vessels, and cerebral ischemia activates mitogen-activated protein kinase (MAPK) and induces secretion of pro-inflammatory cytokines. Adenosine A2A receptor agonist, polydeoxyribonucleotide (PDRN), suppresses the secretion of pro-inflammatory cytokines and exhibits anti-inflammatory effect. In the current study, the therapeutic effect of PDRN on cerebral ischemia was evaluated using gerbils. For the induction of cerebral ischemia, the common carotid arteries were exposed, and then aneurysm clips were used to occlude the common carotid arteries bilaterally for 7 minutes. In the PDRN-treated groups, the gerbils were injected intraperitoneally with 0.3 mL of saline containing 8 mg/kg PDRN, per a day for 7 days following cerebral ischemia induction. In order to confirm the participation of the adenosine A2A receptor in the effects mediated by PDRN, 8 mg/kg 7-dimethyl-1-propargylxanthine (DMPX), adenosine A2A receptor antagonist, was treated with PDRN. In the current study, induction of ischemia enhanced the levels of pro-inflammatory cytokines and increased phosphorylation of MAPK signaling factors in the hippocampus and basolateral amygdala. However, treatment with PDRN ameliorated short-term memory impairment by suppressing the production of pro-inflammatory cytokines and inactivation of MAPK signaling factors in cerebral ischemia. Furthermore, PDRN treatment enhanced the concentration of cyclic adenosine-3,5'-monophosphate (cAMP) as well as phosphorylation of cAMP response element-binding protein (p-CREB). Co-treatment of DMPX and PDRN attenuated the therapeutic effect of PDRN on cerebral ischemia. Based on these findings, PDRN may be developed as the primary treatment in cerebral ischemia.
Collapse
Affiliation(s)
- Il-Gyu Ko
- Department of Physiology, College of Medicine, Kyung Hee University, Seoul, Korea
| | - Jun-Jang Jin
- Department of Physiology, College of Medicine, Kyung Hee University, Seoul, Korea
| | - Lakkyong Hwang
- Department of Physiology, College of Medicine, Kyung Hee University, Seoul, Korea
| | - Sang-Hoon Kim
- Department of Physiology, College of Medicine, Kyung Hee University, Seoul, Korea
| | - Chang-Ju Kim
- Department of Physiology, College of Medicine, Kyung Hee University, Seoul, Korea
| | - Jung Won Jeon
- Department of Internal Medicine, Kyung Hee University Hospital at Gangdong, College of Medicine, Kyung Hee University, Seoul, Korea
| | - Jun-Young Chung
- Department of Anesthesiology and Pain Medicine, Kyung Hee University Hospital at Gangdong, College of Medicine, Kyung Hee University, Seoul, Korea
| | - Jin Hee Han
- Department of Anesthesiology and Pain Medicine, Kyung Hee Medical Center, College of Medicine, Kyung Hee University, Seoul, Korea
- * E-mail:
| |
Collapse
|
61
|
Sieben A, Van Langenhove T, Vermeiren Y, Gossye H, Praet M, Vanhauwaert D, Cousaert C, Engelborghs S, Raedt R, Boon P, Santens P, De Deyn PP, Bracke KR, De Meulemeester K, Van Broeckhoven C, Martin JJ, Bjerke M. Hippocampal Sclerosis in Frontotemporal Dementia: When Vascular Pathology Meets Neurodegeneration. J Neuropathol Exp Neurol 2021; 80:313-324. [PMID: 33638350 DOI: 10.1093/jnen/nlab010] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Hippocampal sclerosis (HS) is a common neuropathological finding and has been associated with advanced age, TDP-43 proteinopathy, and cerebrovascular pathology. We analyzed neuropathological data of an autopsy cohort of early-onset frontotemporal dementia patients. The study aimed to determine whether in this cohort HS was related to TDP-43 proteinopathy and whether additional factors could be identified. We examined the relationship between HS, proteinopathies in frontotemporal cortices and hippocampus, Alzheimer disease, cerebrovascular changes, and age. We confirmed a strong association between HS and hippocampal TDP-43, whereas there was a weaker association between HS and frontotemporal lobar degeneration with TDP-43 pathology (FTLD-TDP). Nearly all of the FTLD-TDP cases had TDP-43 pathology in the hippocampus. HS was present in all FTLD-TDP type D cases, in 50% of the FTLD-TDP A cohort and in 6% of the FTLD-TDP B cohort. Our data also showed a significant association between HS and vascular changes. We reviewed the literature on HS and discuss possible pathophysiological mechanisms between TDP-43 pathology, cerebrovascular disease, and HS. Additionally, we introduced a quantitative neuronal cell count in CA1 to objectify the semiquantitative visual appreciation of HS.
Collapse
Affiliation(s)
- Anne Sieben
- Institute Born-Bunge, Neuropathology and Laboratory of Neurochemistry and Behavior, University of Antwerp, Antwerp, Belgium.,Department of Neurology, Ghent University Hospital, Ghent, Belgium.,Neurodegenerative Brain Diseases Group, Center for Molecular Neurology, VIB, Antwerp, Belgium.,Department of Neurology, AZ Jan Palfijn, Ghent, Belgium
| | - Tim Van Langenhove
- Department of Neurology, Ghent University Hospital, Ghent, Belgium.,Neurodegenerative Brain Diseases Group, Center for Molecular Neurology, VIB, Antwerp, Belgium
| | - Yannick Vermeiren
- Institute Born-Bunge, Neuropathology and Laboratory of Neurochemistry and Behavior, University of Antwerp, Antwerp, Belgium.,Division of Human Nutrition and Health, Chair Group of Nutritional Biology, Wageningen University and Research, Wageningen, The Netherlands
| | - Helena Gossye
- Neurodegenerative Brain Diseases Group, Center for Molecular Neurology, VIB, Antwerp, Belgium.,Department of Neurology, Antwerp University Hospital, Edegem, Belgium.,Institute Born-Bunge, Laboratory of Neurogenetics, University of Antwerp, Antwerp, Belgium.,Department of Neurology and Center for Neurosciences (C4N), UZ Brussel and Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Marleen Praet
- Department of Pathology, Ghent University Hospital, Ghent, Belgium
| | | | | | - Sebastiaan Engelborghs
- Institute Born-Bunge, Neuropathology and Laboratory of Neurochemistry and Behavior, University of Antwerp, Antwerp, Belgium.,Department of Neurology and Center for Neurosciences (C4N), UZ Brussel and Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Robrecht Raedt
- Department of Neurology, Ghent University Hospital, Ghent, Belgium
| | - Paul Boon
- Department of Neurology, Ghent University Hospital, Ghent, Belgium
| | - Patrick Santens
- Department of Neurology, Ghent University Hospital, Ghent, Belgium
| | - Peter Paul De Deyn
- Institute Born-Bunge, Neuropathology and Laboratory of Neurochemistry and Behavior, University of Antwerp, Antwerp, Belgium.,Neurodegenerative Brain Diseases Group, Center for Molecular Neurology, VIB, Antwerp, Belgium.,Department of Neurology and Alzheimer Research Center, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands
| | - Ken R Bracke
- Department of Respiratory Medicine, Ghent University Hospital, Ghent, Belgium
| | | | - Christine Van Broeckhoven
- Neurodegenerative Brain Diseases Group, Center for Molecular Neurology, VIB, Antwerp, Belgium.,Laboratory of Neurogenetics, Institute Born-Bunge, Antwerp, Belgium.,Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Jean-Jacques Martin
- Institute Born-Bunge, Neuropathology and Laboratory of Neurochemistry and Behavior, University of Antwerp, Antwerp, Belgium
| | - Maria Bjerke
- Institute Born-Bunge, Neuropathology and Laboratory of Neurochemistry and Behavior, University of Antwerp, Antwerp, Belgium.,Neurochemistry Laboratory, Department of Clinical Biology and Center for Neurosciences, University hospital Brussels and Free University of Brussels, Brussels, Belgium
| |
Collapse
|
62
|
Abstract
The brain is one of the organs that are preferentially targeted by adenosine-to-inosine (A-to-I) RNA editing, a posttranscriptional modification. This chemical modification affects neuronal development and functions at multiple levels, leading to normal brain homeostasis by increasing the complexity of the transcriptome. This includes modulation of the properties of ion channel and neurotransmitter receptors by recoding, redirection of miRNA targets by changing sequence complementarity, and suppression of immune response by altering RNA structure. Therefore, from another perspective, it appears that the brain is highly vulnerable to dysregulation of A-to-I RNA editing. Here, we focus on how aberrant A-to-I RNA editing is involved in neurological and neurodegenerative diseases of humans including epilepsy, amyotrophic lateral sclerosis, psychiatric disorders, developmental disorders, brain tumors, and encephalopathy caused by autoimmunity. In addition, we provide information regarding animal models to better understand the mechanisms behind disease phenotype.
Collapse
Affiliation(s)
- Pedro Henrique Costa Cruz
- Department of RNA Biology and Neuroscience, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Yukio Kawahara
- Department of RNA Biology and Neuroscience, Graduate School of Medicine, Osaka University, Osaka, Japan.
| |
Collapse
|
63
|
Heit BS, Dykas P, Chu A, Sane A, Larson J. Synaptic and Network Contributions to Anoxic Depolarization in Mouse Hippocampal Slices. Neuroscience 2021; 461:102-117. [PMID: 33636244 DOI: 10.1016/j.neuroscience.2021.02.021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 01/25/2021] [Accepted: 02/16/2021] [Indexed: 01/14/2023]
Abstract
Ischemic stroke remains the third leading cause of death and leading cause of adult disability worldwide. A key event in the pathophysiology of stroke is the anoxic depolarization (AD) of neurons in the ischemic core. Previous studies have established that both the latency to AD and the time spent in AD prior to re-oxygenation are predictors of neuronal death. The present studies used hippocampal slices from male and female mice to investigate the electrophysiological events that affect latency to AD after oxygen deprivation. The results confirm that the epoch between AD and re-oxygenation largely determines the magnitude of synaptic recovery after anoxic challenge. Using a selective antagonist of adenosine A1 receptors, we also confirmed that adenosine released during anoxia (ANOX) suppresses synaptic glutamate release; however, this action has no effect on AD latency or the potential for post-anoxic recovery of synaptic transmission. In contrast, antagonism of AMPA- and NMDA-type glutamate receptors significantly prolongs the latency to AD and alters the speed and synchrony of associated depolarizing waves. Experiments using slices with fields Cornu ammonis 3 (CA3) and Cornu ammonis 1 (CA1) disconnected showed that AD latency is longer in CA1 than in CA3; however, the early AD in CA3 is propagated to CA1 in intact slices. Finally, AD latency in CA1 was found to be longer in slices from female mice than in those from age-matched male mice. The results have implications for stroke prevention and for understanding brain adaptations in hypoxia-tolerant animals.
Collapse
Affiliation(s)
- Bradley S Heit
- Graduate Program in Neuroscience, University of Illinois at Chicago, Chicago, IL 60612, United States; Department of Psychiatry, University of Illinois at Chicago, Chicago, IL 60612, United States
| | - Patricia Dykas
- Department of Psychiatry, University of Illinois at Chicago, Chicago, IL 60612, United States
| | - Alex Chu
- Department of Psychiatry, University of Illinois at Chicago, Chicago, IL 60612, United States
| | - Abhay Sane
- Department of Psychiatry, University of Illinois at Chicago, Chicago, IL 60612, United States
| | - John Larson
- Department of Psychiatry, University of Illinois at Chicago, Chicago, IL 60612, United States.
| |
Collapse
|
64
|
Wisse LEM, Chételat G, Daugherty AM, de Flores R, la Joie R, Mueller SG, Stark CEL, Wang L, Yushkevich PA, Berron D, Raz N, Bakker A, Olsen RK, Carr VA. Hippocampal subfield volumetry from structural isotropic 1 mm 3 MRI scans: A note of caution. Hum Brain Mapp 2021; 42:539-550. [PMID: 33058385 PMCID: PMC7775994 DOI: 10.1002/hbm.25234] [Citation(s) in RCA: 78] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 09/01/2020] [Accepted: 09/29/2020] [Indexed: 01/05/2023] Open
Abstract
Spurred by availability of automatic segmentation software, in vivo MRI investigations of human hippocampal subfield volumes have proliferated in the recent years. However, a majority of these studies apply automatic segmentation to MRI scans with approximately 1 × 1 × 1 mm3 resolution, a resolution at which the internal structure of the hippocampus can rarely be visualized. Many of these studies have reported contradictory and often neurobiologically surprising results pertaining to the involvement of hippocampal subfields in normal brain function, aging, and disease. In this commentary, we first outline our concerns regarding the utility and validity of subfield segmentation on 1 × 1 × 1 mm3 MRI for volumetric studies, regardless of how images are segmented (i.e., manually or automatically). This image resolution is generally insufficient for visualizing the internal structure of the hippocampus, particularly the stratum radiatum lacunosum moleculare, which is crucial for valid and reliable subfield segmentation. Second, we discuss the fact that automatic methods that are employed most frequently to obtain hippocampal subfield volumes from 1 × 1 × 1 mm3 MRI have not been validated against manual segmentation on such images. For these reasons, we caution against using volumetric measurements of hippocampal subfields obtained from 1 × 1 × 1 mm3 images.
Collapse
Affiliation(s)
- Laura E. M. Wisse
- Diagnostic RadiologyLund UniversityLundSweden
- Penn Image Computing and Science Laboratory, Department of RadiologyUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Penn Memory Center, Department of NeurologyUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Gaël Chételat
- Université Normandie, InsermUniversité de Caen‐Normandie, Inserm UMR‐S U1237CaenFrance
| | - Ana M. Daugherty
- Department of PsychologyWayne State UniversityDetroitMichiganUSA
- Institute of GerontologyWayne State UniversityDetroitMichiganUSA
- Department of Psychiatry and Behavioral NeurosciencesWayne State UniversityDetroitMichiganUSA
| | - Robin de Flores
- Université Normandie, InsermUniversité de Caen‐Normandie, Inserm UMR‐S U1237CaenFrance
| | - Renaud la Joie
- Memory and Aging CenterUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Susanne G. Mueller
- Department of RadiologyUniversity of California San FranciscoSan FranciscoCaliforniaUSA
- Center for Imaging of Neurodegenerative DiseasesSan Francisco VA Medical CenterSan FranciscoCaliforniaUSA
| | - Craig E. L. Stark
- Department of Neurobiology and BehaviorUniversity of California IrvineIrvineCaliforniaUSA
| | - Lei Wang
- Department of Psychiatry and Behavioral SciencesNorthwestern University Feinberg School of MedicineChicagoIllinoisUSA
- Department of RadiologyNorthwestern University Feinberg School of MedicineChicagoIllinoisUSA
| | - Paul A. Yushkevich
- Penn Image Computing and Science Laboratory, Department of RadiologyUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - David Berron
- Clinical Memory Research Unit, Department of Clinical Sciences MalmöLund UniversityLundSweden
| | - Naftali Raz
- Department of PsychologyWayne State UniversityDetroitMichiganUSA
- Institute of GerontologyWayne State UniversityDetroitMichiganUSA
- Center for Lifespan PsychologyMax Planck Institute for Human DevelopmentBerlinGermany
| | - Arnold Bakker
- Department of Psychiatry and Behavioral SciencesJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | | | - Valerie A. Carr
- Department of PsychologySan Jose State UniversitySan JoseCaliforniaUSA
| |
Collapse
|
65
|
Buonarati OR, Cook SG, Goodell DJ, Chalmers NE, Rumian NL, Tullis JE, Restrepo S, Coultrap SJ, Quillinan N, Herson PS, Bayer KU. CaMKII versus DAPK1 Binding to GluN2B in Ischemic Neuronal Cell Death after Resuscitation from Cardiac Arrest. Cell Rep 2021; 30:1-8.e4. [PMID: 31914378 PMCID: PMC6959131 DOI: 10.1016/j.celrep.2019.11.076] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 09/25/2019] [Accepted: 11/19/2019] [Indexed: 12/12/2022] Open
Abstract
DAPK1 binding to GluN2B was prominently reported to mediate ischemic cell death in vivo. DAPK1 and CaMKII bind to the same GluN2B region, and their binding is mutually exclusive. Here, we show that mutating the binding region on GluN2B (L1298A/ R1300Q) protected against neuronal cell death induced by cardiac arrest followed by resuscitation. Importantly, the GluN2B mutation selectively abolished only CaMKII, but not DAPK1, binding. During ischemic or excitotoxic insults, CaMKII further accumulated at excitatory synapses, and this accumulation was mediated by GluN2B binding. Interestingly, extra-synaptic GluN2B decreased after ischemia, but its relative association with DAPK1 increased. Thus, ischemic neuronal death requires CaMKII binding to synaptic GluN2B, whereas any potential role for DAPK1 binding is restricted to a different, likely extra-synaptic population of GluN2B. Ischemic insults cause excitotoxic neuronal cell death via NMDA receptor overstimulation. Buonarati et al. find that excitotoxic insults cause DAPK1 movement to extra-synaptic NMDA receptors and CaMKII movement to synaptic NMDA receptors; importantly, preventing this CaMKII movement protects neurons from ischemic death.
Collapse
Affiliation(s)
- Olivia R Buonarati
- Department of Pharmacology, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Sarah G Cook
- Department of Pharmacology, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Dayton J Goodell
- Department of Pharmacology, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA; Program in Neuroscience, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Nicholas E Chalmers
- Department of Anesthesiology, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Nicole L Rumian
- Department of Pharmacology, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA; Program in Neuroscience, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Jonathan E Tullis
- Department of Pharmacology, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Susana Restrepo
- Department of Pharmacology, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Steven J Coultrap
- Department of Pharmacology, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Nidia Quillinan
- Program in Neuroscience, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA; Department of Anesthesiology, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Paco S Herson
- Department of Pharmacology, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA; Program in Neuroscience, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA; Department of Anesthesiology, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA.
| | - K Ulrich Bayer
- Department of Pharmacology, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA; Program in Neuroscience, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA.
| |
Collapse
|
66
|
Ledo A, Lourenço CF, Cadenas E, Barbosa RM, Laranjinha J. The bioactivity of neuronal-derived nitric oxide in aging and neurodegeneration: Switching signaling to degeneration. Free Radic Biol Med 2021; 162:500-513. [PMID: 33186742 DOI: 10.1016/j.freeradbiomed.2020.11.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 11/02/2020] [Accepted: 11/05/2020] [Indexed: 12/22/2022]
Abstract
The small and diffusible free radical nitric oxide (•NO) has fascinated biological and medical scientists since it was promoted from atmospheric air pollutant to biological ubiquitous signaling molecule. Its unique physical chemical properties expand beyond its radical nature to include fast diffusion in aqueous and lipid environments and selective reactivity in a biological setting determined by bioavailability and reaction rate constants with biomolecules. In the brain, •NO is recognized as a key player in numerous physiological processes ranging from neurotransmission/neuromodulation to neurovascular coupling and immune response. Furthermore, changes in its bioactivity are central to the molecular pathways associated with brain aging and neurodegeneration. The understanding of •NO bioactivity in the brain, however, requires the knowledge of its concentration dynamics with high spatial and temporal resolution upon stimulation of its synthesis. Here we revise our current understanding of the role of neuronal-derived •NO in brain physiology, aging and degeneration, focused on changes in the extracellular concentration dynamics of this free radical and the regulation of bioenergetic metabolism and neurovascular coupling.
Collapse
Affiliation(s)
- A Ledo
- Center for Neuroscience and Cell Biology, University of Coimbra, Rua Larga, 3004-504, Coimbra, Portugal; University of Coimbra, Faculty of Pharmacy, Azinhaga de Santa Comba, 3000-548, Coimbra, Portugal.
| | - C F Lourenço
- Center for Neuroscience and Cell Biology, University of Coimbra, Rua Larga, 3004-504, Coimbra, Portugal; University of Coimbra, Faculty of Pharmacy, Azinhaga de Santa Comba, 3000-548, Coimbra, Portugal
| | - E Cadenas
- Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, 90089, CA, USA
| | - R M Barbosa
- Center for Neuroscience and Cell Biology, University of Coimbra, Rua Larga, 3004-504, Coimbra, Portugal; University of Coimbra, Faculty of Pharmacy, Azinhaga de Santa Comba, 3000-548, Coimbra, Portugal
| | - J Laranjinha
- Center for Neuroscience and Cell Biology, University of Coimbra, Rua Larga, 3004-504, Coimbra, Portugal; University of Coimbra, Faculty of Pharmacy, Azinhaga de Santa Comba, 3000-548, Coimbra, Portugal
| |
Collapse
|
67
|
Adebayo OL, Dewenter I, Rinne L, Golubiani G, Solomonia R, Müller M. Intensified mitochondrial hydrogen peroxide release occurs in all brain regions, affects male as well as female Rett mice, and constitutes a life-long burden. Arch Biochem Biophys 2020; 696:108666. [PMID: 33160914 DOI: 10.1016/j.abb.2020.108666] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 10/29/2020] [Accepted: 10/31/2020] [Indexed: 12/28/2022]
Abstract
The neurodevelopmental disorder Rett syndrome (RTT) affects mostly females. Upon an apparently normal initial development, cognitive impairment, irregular breathing, motor dysfunction, and epilepsy occur. The complex pathogenesis includes, among others, mitochondrial impairment, redox imbalance, and oxidative damage. As these arise already in neonatal Rett mice, they were proposed contributors of disease progression. Several mitochondrial studies in RTT used either full brains or selected brain regions only. Here, we mapped mitochondria-related ROS generation brain wide. Using sophisticated multi-sample spectrofluorimetry, H2O2 release by isolated mitochondria was quantified in a coupled reaction of Amplex UltraRed and horseradish peroxidase. All brain regions and the entire lifespan were characterized in male and female mice. In WT mice, mitochondrial H2O2 release was usually highest in cortex and lowest in hippocampus. Maximum rates occurred at postnatal day (PD) 10 and they slightly declined with further maturation. Already at PD 10, male and female Rett mice showed exaggerated mitochondrial H2O2 releases in first brain regions and persistent brain-wide increases from PD 50 on. Interestingly, female Rett mice were more intensely affected than male Rett mice, with their brainstem, midbrain and hippocampus being most severely struck. In conclusion, we used a reliable multi-sample cuvette-based assay on mitochondrial ROS release to perform brain-wide analyzes along the entire lifespan. Mitochondrial H2O2 release in Rett mice is intensified in all brain regions, affects hemizygous males and heterozygous females, and involves all maturational stages. Therefore, intensified mitochondrial H2O2 release seriously needs to be considered throughout RTT pathogenesis and may constitute a potential therapeutic target.
Collapse
Affiliation(s)
- Olusegun L Adebayo
- Georg-August-Universität Göttingen, Universitätsmedizin Göttingen, Zentrum Physiologie und Pathophysiologie, Institut für Neuro- und Sinnesphysiologie, Germany; Department of Biochemistry, Faculty of Basic Medical Sciences, Redeemer's University, P.M.B. 230, Ede, Osun State, Nigeria
| | - Ina Dewenter
- Georg-August-Universität Göttingen, Universitätsmedizin Göttingen, Zentrum Physiologie und Pathophysiologie, Institut für Neuro- und Sinnesphysiologie, Germany
| | - Lena Rinne
- Georg-August-Universität Göttingen, Universitätsmedizin Göttingen, Zentrum Physiologie und Pathophysiologie, Institut für Neuro- und Sinnesphysiologie, Germany
| | - Gocha Golubiani
- Georg-August-Universität Göttingen, Universitätsmedizin Göttingen, Zentrum Physiologie und Pathophysiologie, Institut für Neuro- und Sinnesphysiologie, Germany; Institute of Chemical Biology, Ilia State University, Tbilisi, Georgia
| | - Revaz Solomonia
- Institute of Chemical Biology, Ilia State University, Tbilisi, Georgia
| | - Michael Müller
- Georg-August-Universität Göttingen, Universitätsmedizin Göttingen, Zentrum Physiologie und Pathophysiologie, Institut für Neuro- und Sinnesphysiologie, Germany.
| |
Collapse
|
68
|
Wang F, Hua S, Zhang Y, Yu H, Zhang Z, Zhu J, Liu R, Jiang Z. Association Between Small Vessel Disease Markers, Medial Temporal Lobe Atrophy and Cognitive Impairment After Stroke: A Systematic Review and Meta-Analysis. J Stroke Cerebrovasc Dis 2020; 30:105460. [PMID: 33227579 DOI: 10.1016/j.jstrokecerebrovasdis.2020.105460] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 10/23/2020] [Accepted: 11/03/2020] [Indexed: 11/27/2022] Open
Abstract
OBJECTIVES Two-thirds of stroke survivors suffer from cognitive impairment, and up to one-third of them progress to dementia. However, the underlying pathogenesis is complex and controversial. Recent evidence has found that cerebral small vessel disease (SVD) markers and the Alzheimer's disease (AD) neuroimaging marker medial temporal lobe atrophy (MTLA), alone or in combination, contribute to the pathogenesis of poststroke cognitive impairment (PSCI). In the present systematic review and meta-analysis, we synthesized proof for these neuroimaging risk factors among stroke patients. MATERIALS AND METHODS PUBMED, MEDLINE, EMBASE and the Cochrane Library were searched for studies investigating imaging predictors of cognitive impairment or dementia following stroke. Meta-analysis was conducted to compute the odds ratios (ORs). RESULTS Thirteen studies were enrolled in the present study, and only ten of them, comprising 2713 stroke patients, were eligible for inclusion in the meta-analysis. MTLA was significantly correlated with PSCI (OR = 1.97, 95% CI: 1.48-2.62, I2 = 0.0%). In addition, white matter hyperintensities (WMH), as a neuroimaging marker of SVD, were associated with PSCI (OR = 1.17, 95% CI: 1.12-1.22, I2 = 0.0%). However, the presence of lacunar infarcts and enlarged perivascular spaces (EPVS) were not associated with the risk of PSCI. CONCLUSIONS The findings of the present study suggest that MTLA and WMH were associated with an increased risk of PSCI.
Collapse
Affiliation(s)
- Furu Wang
- Department of Radiology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Sunyu Hua
- Department of Radiology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Yue Zhang
- Department of Radiology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Hongchang Yu
- Department of Radiology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | | | - Jiangtao Zhu
- Department of Radiology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Rong Liu
- Department of Radiology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Zhen Jiang
- Department of Radiology, The Second Affiliated Hospital of Soochow University, Suzhou, China.
| |
Collapse
|
69
|
Lana D, Ugolini F, Giovannini MG. An Overview on the Differential Interplay Among Neurons-Astrocytes-Microglia in CA1 and CA3 Hippocampus in Hypoxia/Ischemia. Front Cell Neurosci 2020; 14:585833. [PMID: 33262692 PMCID: PMC7686560 DOI: 10.3389/fncel.2020.585833] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 10/09/2020] [Indexed: 12/13/2022] Open
Abstract
Neurons have been long regarded as the basic functional cells of the brain, whereas astrocytes and microglia have been regarded only as elements of support. However, proper intercommunication among neurons-astrocytes-microglia is of fundamental importance for the functional organization of the brain. Perturbation in the regulation of brain energy metabolism not only in neurons but also in astrocytes and microglia may be one of the pathophysiological mechanisms of neurodegeneration, especially in hypoxia/ischemia. Glial activation has long been considered detrimental for survival of neurons, but recently it appears that glial responses to an insult are not equal but vary in different brain areas. In this review, we first take into consideration the modifications of the vascular unit of the glymphatic system and glial metabolism in hypoxic conditions. Using the method of triple-labeling fluorescent immunohistochemistry coupled with confocal microscopy (TIC), we recently studied the interplay among neurons, astrocytes, and microglia in chronic brain hypoperfusion. We evaluated the quantitative and morpho-functional alterations of the neuron-astrocyte-microglia triads comparing the hippocampal CA1 area, more vulnerable to ischemia, to the CA3 area, less vulnerable. In these contiguous and interconnected areas, in the same experimental hypoxic conditions, astrocytes and microglia show differential, finely regulated, region-specific reactivities. In both areas, astrocytes and microglia form triad clusters with apoptotic, degenerating neurons. In the neuron-astrocyte-microglia triads, the cell body of a damaged neuron is infiltrated and bisected by branches of astrocyte that create a microscar around it while a microglial cell phagocytoses the damaged neuron. These coordinated actions are consistent with the scavenging and protective activities of microglia. In hypoxia, the neuron-astrocyte-microglia triads are more numerous in CA3 than in CA1, further indicating their protective effects. These data, taken from contiguous and interconnected hippocampal areas, demonstrate that glial response to the same hypoxic insult is not equal but varies significantly. Understanding the differences of glial reactivity is of great interest to explain the differential susceptibility of hippocampal areas to hypoxia/ischemia. Further studies may evidence the differential reactivity of glia in different brain areas, explaining the higher or lower sensitivity of these areas to different insults and whether glia may represent a target for future therapeutic interventions.
Collapse
Affiliation(s)
- Daniele Lana
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Florence, Italy
| | - Filippo Ugolini
- Department of Health Sciences, Section of Anatomopathology, University of Florence, Florence, Italy
| | - Maria G Giovannini
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Florence, Italy
| |
Collapse
|
70
|
Rogers Flattery CN, Rosen RF, Farberg AS, Dooyema JM, Hof PR, Sherwood CC, Walker LC, Preuss TM. Quantification of neurons in the hippocampal formation of chimpanzees: comparison to rhesus monkeys and humans. Brain Struct Funct 2020; 225:2521-2531. [PMID: 32909100 DOI: 10.1007/s00429-020-02139-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 08/29/2020] [Indexed: 12/15/2022]
Abstract
The hippocampal formation is important for higher brain functions such as spatial navigation and the consolidation of memory, and it contributes to abilities thought to be uniquely human, yet little is known about how the human hippocampal formation compares to that of our closest living relatives, the chimpanzees. To gain insight into the comparative organization of the hippocampal formation in catarrhine primates, we quantified neurons stereologically in its major subdivisions-the granular layer of the dentate gyrus, CA4, CA2-3, CA1, and the subiculum-in archival brain tissue from six chimpanzees ranging from 29 to 43 years of age. We also sought evidence of Aβ deposition and hyperphosphorylated tau in the hippocampus and adjacent neocortex. A 42-year-old animal had moderate cerebral Aβ-amyloid angiopathy and tauopathy, but Aβ was absent and tauopathy was minimal in the others. Quantitatively, granule cells of the dentate gyrus were most numerous, followed by CA1, subiculum, CA4, and CA2-3. In the context of prior investigations of rhesus monkeys and humans, our findings indicate that, in the hippocampal formation as a whole, the proportions of neurons in CA1 and the subiculum progressively increase, and the proportion of dentate granule cells decreases, from rhesus monkeys to chimpanzees to humans. Because CA1 and the subiculum engender key hippocampal projection pathways to the neocortex, and because the neocortex varies in volume and anatomical organization among these species, these findings suggest that differences in the proportions of neurons in hippocampal subregions of catarrhine primates may be linked to neocortical evolution.
Collapse
Affiliation(s)
| | - Rebecca F Rosen
- National Institute of Mental Health, National Institutes of Health, Bethesda, MD, 20814, USA
| | - Aaron S Farberg
- Yerkes National Primate Research Center, Emory University, Atlanta, GA, 30329, USA
| | - Jeromy M Dooyema
- Yerkes National Primate Research Center, Emory University, Atlanta, GA, 30329, USA
| | - Patrick R Hof
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Chet C Sherwood
- Department of Anthropology and Center for the Advanced Study of Human Paleobiology, The George Washington University, Washington, DC, 20052, USA
| | - Lary C Walker
- Yerkes National Primate Research Center, Emory University, Atlanta, GA, 30329, USA.,Department of Neurology, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Todd M Preuss
- Yerkes National Primate Research Center, Emory University, Atlanta, GA, 30329, USA.,Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, 30322, USA
| |
Collapse
|
71
|
Hedderich DM, Avram M, Menegaux A, Nuttall R, Zimmermann J, Schneider SC, Schmitz-Koep B, Daamen M, Scheef L, Boecker H, Zimmer C, Baumann N, Bartmann P, Wolke D, Bäuml JG, Sorg C. Hippocampal subfield volumes are nonspecifically reduced in premature-born adults. Hum Brain Mapp 2020; 41:5215-5227. [PMID: 32845045 PMCID: PMC7670635 DOI: 10.1002/hbm.25187] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 07/07/2020] [Accepted: 08/11/2020] [Indexed: 01/06/2023] Open
Abstract
Reduced global hippocampus volumes have been demonstrated in premature‐born individuals, from newborns to adults; however, it is unknown whether hippocampus subfield (HCSF) volumes are differentially affected by premature birth and how relevant they are for cognitive performance. To address these questions, we investigated magnetic resonance imaging (MRI)‐derived HCSF volumes in very premature‐born adults, and related them with general cognitive performance in adulthood. We assessed 103 very premature‐born (gestational age [GA] <32 weeks and/or birth weight <1,500 g) and 109 term‐born individuals with cognitive testing and structural MRI at 26 years of age. HCSFs were automatically segmented based on three‐dimensional T1‐ and T2‐weighted sequences and studied both individually and grouped into three functional units, namely hippocampus proper (HP), subicular complex (SC), and dentate gyrus (DG). Cognitive performance was measured using the Wechsler‐Adult‐Intelligence‐Scale (full‐scale intelligence quotient [FS‐IQ]) at 26 years. We observed bilateral volume reductions for almost all HCSF volumes in premature‐born adults and associations with GA and neonatal treatment intensity but not birth weight. Left‐sided HP, SC, and DG volumes were associated with adult FS‐IQ. Furthermore, left DG volume was a mediator of the association between GA and adult FS‐IQ in premature‐born individuals. Results demonstrate nonspecifically reduced HCSF volumes in premature‐born adults; but specific associations with cognitive outcome highlight the importance of the left DG. Data suggest that specific interventions toward hippocampus function might be promising to lower adverse cognitive effects of prematurity.
Collapse
Affiliation(s)
- Dennis M Hedderich
- Department of Neuroradiology, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany.,Technical University of Munich-NIC Neuroimaging Center, Munich, Germany
| | - Mihai Avram
- Department of Neuroradiology, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany.,Technical University of Munich-NIC Neuroimaging Center, Munich, Germany
| | - Aurore Menegaux
- Department of Neuroradiology, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany.,Technical University of Munich-NIC Neuroimaging Center, Munich, Germany
| | - Rachel Nuttall
- Department of Neuroradiology, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany.,Technical University of Munich-NIC Neuroimaging Center, Munich, Germany
| | - Juliana Zimmermann
- Department of Neuroradiology, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany.,Technical University of Munich-NIC Neuroimaging Center, Munich, Germany
| | - Sebastian C Schneider
- Department of Neuroradiology, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany.,Technical University of Munich-NIC Neuroimaging Center, Munich, Germany
| | - Benita Schmitz-Koep
- Department of Neuroradiology, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany.,Technical University of Munich-NIC Neuroimaging Center, Munich, Germany
| | - Marcel Daamen
- Functional Neuroimaging Group, Department of Radiology, University Hospital Bonn, Bonn, Germany.,Department of Neonatology, University Hospital Bonn, Bonn, Germany
| | - Lukas Scheef
- Functional Neuroimaging Group, Department of Radiology, University Hospital Bonn, Bonn, Germany
| | - Henning Boecker
- Functional Neuroimaging Group, Department of Radiology, University Hospital Bonn, Bonn, Germany
| | - Claus Zimmer
- Department of Neuroradiology, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany.,Technical University of Munich-NIC Neuroimaging Center, Munich, Germany
| | - Nicole Baumann
- Department of Psychology, University of Warwick, Coventry, UK.,Warwick Medical School, University of Warwick, Coventry, UK
| | - Peter Bartmann
- Department of Neonatology, University Hospital Bonn, Bonn, Germany
| | - Dieter Wolke
- Department of Psychology, University of Warwick, Coventry, UK.,Warwick Medical School, University of Warwick, Coventry, UK
| | - Josef G Bäuml
- Department of Neuroradiology, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany.,Technical University of Munich-NIC Neuroimaging Center, Munich, Germany
| | - Christian Sorg
- Department of Neuroradiology, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany.,Technical University of Munich-NIC Neuroimaging Center, Munich, Germany.,Department of Psychiatry, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany
| |
Collapse
|
72
|
Zhu T, Wang L, Tian F, Zhao X, Pu XP, Sun GB, Sun XB. Anti-ischemia/reperfusion injury effects of notoginsenoside R1 on small molecule metabolism in rat brain after ischemic stroke as visualized by MALDI-MS imaging. Biomed Pharmacother 2020; 129:110470. [PMID: 32768957 DOI: 10.1016/j.biopha.2020.110470] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 06/17/2020] [Accepted: 06/24/2020] [Indexed: 02/09/2023] Open
Abstract
Ischemic stroke is a syndrome of severe neurological responses that cause neuronal death, damage to the neurovascular unit and inflammation. Notoginsenoside R1 (NG-R1) is a neuroprotective drug that is commonly used to treat neurodegenerative and cerebrovascular diseases. However, its potential mechanisms on the regulation of small molecule metabolism in ischemic stroke are largely unknown. The aim of this study was to explore the potential mechanisms of NG-R1 on the regulation of small molecule metabolism after ischemic stroke. Here, we found that NG-R1 reduced infarct size and improved neurological deficits by ameliorating neuronal damage and inhibiting glial activation in MCAO/R rats. Furthermore, using matrix-assisted laser desorption/ionization mass spectrometry imaging (MALDI-MSI), we clarified that NG-R1 regulated ATP metabolism, the tricarboxylic acid (TCA) cycle, the malate-aspartate shuttle, antioxidant activity, and the homeostasis of iron and phospholipids in the striatum and hippocampus of middle cerebral artery occlusion/reperfusion (MCAO/R) rats. In general, NG-R1 is a promising compound for brain protection from ischemic/reperfusion injury, possibly through the regulation of brain small molecule metabolism.
Collapse
Affiliation(s)
- Ting Zhu
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, 100193, China; Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100193, China; Key Laboratory of New Drug Discovery Based on Classic Chinese Medicine Prescription, Chinese Academy of Medical Sciences, Beijing, 100193, China.
| | - Lei Wang
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, 100193, China; Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100193, China; Key Laboratory of New Drug Discovery Based on Classic Chinese Medicine Prescription, Chinese Academy of Medical Sciences, Beijing, 100193, China; Harbin University of Commerce, Harbin, Heilongjiang, 150000, China.
| | - Fang Tian
- National Key Research Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing, 100191, China.
| | - Xin Zhao
- National Key Research Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing, 100191, China.
| | - Xiao-Ping Pu
- National Key Research Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing, 100191, China.
| | - Gui-Bo Sun
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, 100193, China; Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100193, China; Key Laboratory of New Drug Discovery Based on Classic Chinese Medicine Prescription, Chinese Academy of Medical Sciences, Beijing, 100193, China.
| | - Xiao-Bo Sun
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, 100193, China; Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100193, China; Key Laboratory of New Drug Discovery Based on Classic Chinese Medicine Prescription, Chinese Academy of Medical Sciences, Beijing, 100193, China.
| |
Collapse
|
73
|
Van Etten EJ, Bharadwaj PK, Nguyen LA, Hishaw GA, Trouard TP, Alexander GE. Right hippocampal volume mediation of subjective memory complaints differs by hypertension status in healthy aging. Neurobiol Aging 2020; 94:271-280. [PMID: 32688134 DOI: 10.1016/j.neurobiolaging.2020.06.012] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 06/13/2020] [Accepted: 06/15/2020] [Indexed: 12/28/2022]
Abstract
Subjective memory complaints (SMCs) may be an important early indicator of cognitive aging and preclinical Alzheimer's disease risk. This study investigated whether age-related differences in right or left hippocampal volume underlie SMCs, if these relationships differ by hypertension status, and how they are related to objective memory performance in a group of 190 healthy older adults, 50-89 years of age. Analyses revealed a significant mediation of the relationship between age and mild SMCs by right hippocampal volume that was moderated by hypertension status. This moderated mediation effect was not observed with left hippocampal volume. Additionally, a moderated serial mediation model showed that age predicted right hippocampal volume, which predicted SMCs, and in turn predicted objective memory performance on several measures of verbal selective reminding in individuals with hypertension, but not in non-hypertensives. Together, these findings suggest that even mild SMCs, in the context of hypertension, provide an early indicator of cognitive aging, reflecting a potential link among vascular risk, SMCs, and the preclinical risk for Alzheimer's disease.
Collapse
Affiliation(s)
- Emily J Van Etten
- Department of Psychology, University of Arizona, Tucson, AZ, USA; Evelyn F. McKnight Brain Institute, University of Arizona, Tucson, AZ, USA
| | - Pradyumna K Bharadwaj
- Department of Psychology, University of Arizona, Tucson, AZ, USA; Evelyn F. McKnight Brain Institute, University of Arizona, Tucson, AZ, USA
| | - Lauren A Nguyen
- Department of Psychology, University of Arizona, Tucson, AZ, USA; Evelyn F. McKnight Brain Institute, University of Arizona, Tucson, AZ, USA
| | - Georg A Hishaw
- Department of Neurology, University of Arizona, Tucson, AZ, USA
| | - Theodore P Trouard
- Evelyn F. McKnight Brain Institute, University of Arizona, Tucson, AZ, USA; Department of Biomedical Engineering, University of Arizona, Tucson, AZ, USA; Arizona Alzheimer's Consortium, Phoenix, AZ, USA
| | - Gene E Alexander
- Department of Psychology, University of Arizona, Tucson, AZ, USA; Evelyn F. McKnight Brain Institute, University of Arizona, Tucson, AZ, USA; Arizona Alzheimer's Consortium, Phoenix, AZ, USA; Department of Psychiatry, University of Arizona, Tucson, AZ, USA; Neuroscience Graduate Interdisciplinary Program, University of Arizona, Tucson, AZ, USA; Physiological Sciences Graduate Interdisciplinary Program, University of Arizona, Tucson, AZ, USA.
| |
Collapse
|
74
|
Hajihasani MM, Soheili V, Zirak MR, Sahebkar A, Shakeri A. Natural products as safeguards against monosodium glutamate-induced toxicity. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2020; 23:416-430. [PMID: 32489556 PMCID: PMC7239414 DOI: 10.22038/ijbms.2020.43060.10123] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 01/04/2020] [Indexed: 12/17/2022]
Abstract
Monosodium glutamate is a sodium salt of a nonessential amino acid, L-glutamic acid, which is widely used in food industry. Glutamate plays an important role in principal brain functions including formation and stabilization of synapses, memory, cognition, learning, as well as cellular metabolism. However, ingestion of foodstuffs rich in monosodium glutamate can result in the outbreak of several health disorders such as neurotoxicity, hepatotoxicity, obesity and diabetes. The usage of medicinal plants and their natural products as a therapy against MSG used in food industry has been suggested to be protective. Calendula officinalis, Curcuma longa, Green Tea, Ginkgo biloba and vitamins are some of the main natural products with protective effect against mentioned monosodium glutamate toxicity through different mechanisms. This review provides a summary on the toxicity of monosodium glutamate and the protective effects of natural products against monosodium glutamate -induced toxicity.
Collapse
Affiliation(s)
- Mohammad Mahdi Hajihasani
- Department of Pharmaceutical Control, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Vahid Soheili
- Department of Pharmaceutical Control, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Reza Zirak
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amirhossein Sahebkar
- Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Abolfazl Shakeri
- Department of Pharmacognosy, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
75
|
Li Y, Ding R, Wang F, Guo C, Liu A, Wei L, Yuan S, Chen F, Hou S, Ma Z, Zhang Y, Cudmore RH, Wang X, Shen H. Transient ischemia-reperfusion induces cortical hyperactivity and AMPAR trafficking in the somatosensory cortex. Aging (Albany NY) 2020; 12:4299-4321. [PMID: 32155129 PMCID: PMC7093173 DOI: 10.18632/aging.102881] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Accepted: 02/05/2020] [Indexed: 01/20/2023]
Abstract
Brain ischemia results from cardiac arrest, stroke or head trauma. The structural basis of rescuing the synaptic impairment and cortical dysfunctions induced in the stage of ischemic-reperfusion can occur if therapeutic interventions are applied in time, but the functional basis for this resilience remains elusive. Here, we explore the changes in cortical activity and a-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid receptor (AMPAR) GluA1 subunit in spine (sGluA1) after transient ischemia-reperfusion in vivo for 28 days. Using in vivo two-photon microscopy in the mouse somatosensory cortex, we found that the average frequency of Ca2+ transients in the spine (there was an unusual synchrony) was higher after 15 min of ischemia-reperfusion. In addition, the transient ischemia-reperfusion caused a reflective enhancement of AMPARs, which eventually restored to normal. The cortical hyperactivity (Ca2+ transients) and the increase in AMPARs were successfully blocked by an NMDA receptor antagonist. Thus, the increase of AMPARs, cortical hyperactivity and the unusual synchrony might be the reason for reperfusion injury after short-term transient ischemia.
Collapse
Affiliation(s)
- Yuanyuan Li
- School of Biomedical Engineering, Tianjin Medical University, Tianjin, China
| | - Ran Ding
- Chinese Institute for Brain Research, Beijing (CIBR), Beijing, China
| | - Feifei Wang
- School of Biomedical Engineering, Tianjin Medical University, Tianjin, China
| | - Cuiping Guo
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Ministry of Education of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Aili Liu
- School of Biomedical Engineering, Tianjin Medical University, Tianjin, China
| | - Liangpeng Wei
- School of Biomedical Engineering, Tianjin Medical University, Tianjin, China
| | - Shiyang Yuan
- School of Biomedical Engineering, Tianjin Medical University, Tianjin, China
| | - Feng Chen
- School of Biomedical Engineering, Tianjin Medical University, Tianjin, China
| | - Shaowei Hou
- School of Biomedical Engineering, Tianjin Medical University, Tianjin, China
| | - Zengguang Ma
- School of Biomedical Engineering, Tianjin Medical University, Tianjin, China
| | - Yan Zhang
- Tianjin Key Laboratory of Retinal Function and Diseases, Tianjin Medical University Eye Hospital, Eye Institute and School of Optometry and Ophthalmology, Tianjin Medical University, Tianjin, China
| | - Robert H Cudmore
- Department of Physiology and Membrane Biology, University of California Davis School of Medicine, Sacramento, CA 95817, USA
| | - Xiaochuan Wang
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Ministry of Education of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Division of Neurodegenerative Disorders, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Hui Shen
- School of Biomedical Engineering, Tianjin Medical University, Tianjin, China.,Research Institute of Neurology, General Hospital, Tianjin Medical University, Tianjin, China
| |
Collapse
|
76
|
Song K, Liu X, Zheng Q, Zhang L, Zhang H, Yu H, Zhu Y, Huang LA, Chen Y. Secondary injury to distal regions after intracerebral hemorrhage influence neurological functional outcome. Aging (Albany NY) 2020; 12:4283-4298. [PMID: 32146443 PMCID: PMC7093199 DOI: 10.18632/aging.102880] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Accepted: 02/04/2020] [Indexed: 12/15/2022]
Abstract
Although many studies have focused on functional impairment after intracerebral hemorrhage, little is known about the relationship between secondary injuries to distal regions and neurological function. Our study aimed to evaluate the secondary injuries after intracerebral hemorrhage and explore their relationship to neurological functional outcome. Twenty-one patients with hemorrhages in supratentorial, deep locations and 10 healthy subjects were recruited. Longitudinal examinations of diffusion tensor imaging, hydrogen proton magnetic resonance spectroscopy imaging and neuropsychological assessment were performed after weeks 1 and 12 to elucidate the relationship between magnetic resonance imaging parameters and neurologic outcomes. By week 12, motor function had significantly improved, but cognitive function had deteriorated compared to week 1. Fractional anisotropy values for the ipsilateral cerebral peduncle correlated with motor function at week 1. No significant correlation between fractional anisotropy for the ipsilateral cerebral peduncle and the Fugl-Meyer Motor Scale was found at week 12. Fractional anisotropy values for the ipsilateral hippocampus were related to the Montreal Cognitive Assessment and Mini-Mental State Examination at weeks 1 and 12. Deep supratentorial hemorrhage may result in injury to distal regions, which correlate with impaired motor and cognitive function.
Collapse
Affiliation(s)
- Kangping Song
- Department of Neurology, Institute of Clinical Neuroscience, The First Affiliated Hospital, Jinan University, Guangzhou 510632, Guangdong, China.,Department of Neurology, Clinical Medical College, Yangzhou University, Yangzhou 225000, Jiangsu, China
| | - Xiaojie Liu
- Department of Neurology, Clinical Medical College, Yangzhou University, Yangzhou 225000, Jiangsu, China
| | - Qiuyue Zheng
- Department of Neurology, Clinical Medical College, Yangzhou University, Yangzhou 225000, Jiangsu, China
| | - Lingling Zhang
- Department of Neurology, Institute of Clinical Neuroscience, The First Affiliated Hospital, Jinan University, Guangzhou 510632, Guangdong, China
| | - Hongying Zhang
- Medical Imaging Center, Clinical Medical College, Yangzhou University, Yangzhou 225000, Jiangsu, China
| | - Hailong Yu
- Department of Neurology, Clinical Medical College, Yangzhou University, Yangzhou 225000, Jiangsu, China
| | - Yan Zhu
- Department of Neurology, Clinical Medical College, Yangzhou University, Yangzhou 225000, Jiangsu, China
| | - Li-An Huang
- Department of Neurology, Institute of Clinical Neuroscience, The First Affiliated Hospital, Jinan University, Guangzhou 510632, Guangdong, China
| | - Yingzhu Chen
- Department of Neurology, Clinical Medical College, Yangzhou University, Yangzhou 225000, Jiangsu, China
| |
Collapse
|
77
|
Dey D, Parihar VK, Szabo GG, Klein PM, Tran J, Moayyad J, Ahmed F, Nguyen QA, Murry A, Merriott D, Nguyen B, Goldman J, Angulo MC, Piomelli D, Soltesz I, Baulch JE, Limoli CL. Neurological Impairments in Mice Subjected to Irradiation and Chemotherapy. Radiat Res 2020; 193:407-424. [PMID: 32134362 DOI: 10.1667/rr15540.1] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Radiotherapy, surgery and the chemotherapeutic agent temozolomide (TMZ) are frontline treatments for glioblastoma multiforme (GBM). However beneficial, GBM treatments nevertheless cause anxiety or depression in nearly 50% of patients. To further understand the basis of these neurological complications, we investigated the effects of combined radiotherapy and TMZ chemotherapy (combined treatment) on neurological impairments using a mouse model. Five weeks after combined treatment, mice displayed anxiety-like behaviors, and at 15 weeks both anxiety- and depression-like behaviors were observed. Relevant to the known roles of the serotonin axis in mood disorders, we found that 5HT1A serotonin receptor levels were decreased by ∼50% in the hippocampus at both early and late time points, and a 37% decrease in serotonin levels was observed at 15 weeks postirradiation. Furthermore, chronic treatment with the selective serotonin reuptake inhibitor fluoxetine was sufficient for reversing combined treatment-induced depression-like behaviors. Combined treatment also elicited a transient early increase in activated microglia in the hippocampus, suggesting therapy-induced neuroinflammation that subsided by 15 weeks. Together, the results of this study suggest that interventions targeting the serotonin axis may help ameliorate certain neurological side effects associated with the clinical management of GBM to improve the overall quality of life for cancer patients.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Faizy Ahmed
- Departments of Anatomy and Neurobiology, University of California, Irvine, California 92697
| | | | | | | | | | | | | | - Daniele Piomelli
- Departments of Anatomy and Neurobiology, University of California, Irvine, California 92697
| | - Ivan Soltesz
- Departments of Neurology and Neurological Sciences, Stanford University, Palo Alto, California 94305
| | | | | |
Collapse
|
78
|
Zhao Y, Wu X, Chen X, Li J, Tian C, Chen J, Xiao C, Zhong G, He S. Calcineurin Signaling Mediates Disruption of the Axon Initial Segment Cytoskeleton after Injury. iScience 2020; 23:100880. [PMID: 32062456 PMCID: PMC7031317 DOI: 10.1016/j.isci.2020.100880] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2019] [Revised: 10/23/2019] [Accepted: 01/29/2020] [Indexed: 11/30/2022] Open
Abstract
The axon initial segment (AIS) cytoskeleton undergoes rapid and irreversible disruption prior to cell death after injury, and loss of AIS integrity can produce profound neurological effects on the nervous system. Here we described a previously unrecognized mechanism for ischemia-induced alterations in AIS integrity. We show that in hippocampal CA1 pyramidal neurons Nav1.6 mostly preserves at the AIS after disruption of the cytoskeleton in a mouse model of middle cerebral artery occlusion. Genetic removal of neurofascin-186 leads to rapid disruption of Nav1.6 following injury, indicating that neurofascin is required for Nav1.6 maintenance at the AIS after cytoskeleton collapse. Importantly, calcineurin inhibition with FK506 fully protects AIS integrity and sufficiently prevents impairments of spatial learning and memory from injury. This study provides evidence that calcineurin activation is primarily involved in initiating disassembly of the AIS cytoskeleton and that maintaining AIS integrity is crucial for therapeutic strategies to facilitate recovery from injury. Ion channels are mostly retained at the AIS after ischemic injury Neurofascin is required for clustering ion channels at the AIS after ischemia Calcineurin inhibition protects AIS structural integrity and function from ischemia Calcineurin inhibition protects cognitive function against impairment by ischemia
Collapse
Affiliation(s)
- Yanan Zhao
- School of Life Science and Technology, ShanghaiTech University, 393 Middle Huaxia Road, Pudong New District, Shanghai 201210, China
| | - Xuanyuan Wu
- School of Life Science and Technology, ShanghaiTech University, 393 Middle Huaxia Road, Pudong New District, Shanghai 201210, China
| | - Xin Chen
- School of Life Science and Technology, ShanghaiTech University, 393 Middle Huaxia Road, Pudong New District, Shanghai 201210, China
| | - Jianan Li
- School of Life Science and Technology, ShanghaiTech University, 393 Middle Huaxia Road, Pudong New District, Shanghai 201210, China
| | - Cuiping Tian
- iHuman Institute, ShanghaiTech University, 393 Middle Huaxia Road, Pudong New District, Shanghai 201210, China
| | - Jiangrui Chen
- School of Life Science and Technology, ShanghaiTech University, 393 Middle Huaxia Road, Pudong New District, Shanghai 201210, China; Institute of Neuroscience, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China; University of Chinese Academy of Sciences, Beijing, China
| | - Cheng Xiao
- School of Anesthesiology, Xuzhou Medical University, 209 Tongshan Road, KJL-D423, Xuzhou, Jiangsu Province 221004, China
| | - Guisheng Zhong
- School of Life Science and Technology, ShanghaiTech University, 393 Middle Huaxia Road, Pudong New District, Shanghai 201210, China; iHuman Institute, ShanghaiTech University, 393 Middle Huaxia Road, Pudong New District, Shanghai 201210, China.
| | - Shuijin He
- School of Life Science and Technology, ShanghaiTech University, 393 Middle Huaxia Road, Pudong New District, Shanghai 201210, China.
| |
Collapse
|
79
|
Meyer P, Grandgirard D, Lehner M, Haenggi M, Leib SL. Grafted Neural Progenitor Cells Persist in the Injured Site and Differentiate Neuronally in a Rodent Model of Cardiac Arrest-Induced Global Brain Ischemia. Stem Cells Dev 2020; 29:574-585. [PMID: 31964231 DOI: 10.1089/scd.2019.0190] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Hypoxic-ischemic brain injury is the leading cause of disability and death after successful resuscitation from cardiac arrest, and, to date, no specific treatment option is available to prevent subsequent neurofunctional impairments. The hippocampal cornu ammonis segment 1 (CA1) is one of the brain areas most affected by hypoxia, and its degeneration is correlated with memory deficits in patients and corresponding animal models. The aim of this work was to evaluate the feasibility of neural progenitor cell (NPC) transplantation into the hippocampus in a refined rodent cardiac arrest model. Adult rats were subjected to 12 min of potassium-induced cardiac arrest and followed up to 6 weeks. Histological analysis showed extensive neuronal cell death specifically in the hippocampal CA1 segment, without any spontaneous regeneration. Neurofunctional assessment revealed transient memory deficits in ischemic animals compared to controls, detectable after 4 weeks, but not after 6 weeks. Using stereotactic surgery, embryonic NPCs were transplanted in a subset of animals 1 week after cardiac arrest and their survival, migration, and differentiation were assessed histologically. Transplanted cells showed a higher persistence in the CA1 segment of animals after ischemia. Glia in the damaged CA1 segment expressed the chemotactic factor stromal cell-derived factor 1 (SDF-1), while transplanted NPCs expressed its receptor CXC chemokine receptor 4 (CXCR4), suggesting that the SDF-1/CXCR4 pathway, known to be involved in the migration of neural stem cells toward injured brain regions, directs the observed retention of cells in the damaged area. Using immunostaining, we could demonstrate that transplanted cells differentiated into mature neurons. In conclusion, our data document the survival, persistence in the injured area, and neuronal differentiation of transplanted NPCs, and thus their potential to support brain regeneration after hypoxic-ischemic injury. This may represent an option worth further investigation to improve the outcome of patients after cardiac arrest.
Collapse
Affiliation(s)
- Patricia Meyer
- Neuroinfection Laboratory, Institute for Infectious Diseases, University of Bern, Bern, Switzerland.,Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland.,Department of Intensive Care Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland.,Cluster for Regenerative Neuroscience, DBMR, University of Bern, Bern, Switzerland
| | - Denis Grandgirard
- Neuroinfection Laboratory, Institute for Infectious Diseases, University of Bern, Bern, Switzerland.,Cluster for Regenerative Neuroscience, DBMR, University of Bern, Bern, Switzerland
| | - Marika Lehner
- Neuroinfection Laboratory, Institute for Infectious Diseases, University of Bern, Bern, Switzerland.,Cluster for Regenerative Neuroscience, DBMR, University of Bern, Bern, Switzerland
| | - Matthias Haenggi
- Department of Intensive Care Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Stephen L Leib
- Neuroinfection Laboratory, Institute for Infectious Diseases, University of Bern, Bern, Switzerland.,Cluster for Regenerative Neuroscience, DBMR, University of Bern, Bern, Switzerland
| |
Collapse
|
80
|
Livingston JM, McDonald MW, Gagnon T, Jeffers MS, Gomez-Smith M, Antonescu S, Cron GO, Boisvert C, Lacoste B, Corbett D. Influence of metabolic syndrome on cerebral perfusion and cognition. Neurobiol Dis 2020; 137:104756. [PMID: 31978604 DOI: 10.1016/j.nbd.2020.104756] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Revised: 12/19/2019] [Accepted: 01/20/2020] [Indexed: 02/06/2023] Open
Abstract
Vascular cognitive impairment (VCI) is associated with chronic cerebral hypoperfusion (CCH) and memory deficits, and often occurs concurrently with metabolic syndrome (MetS). Despite their common occurrence, it is unknown whether CCH and MetS act synergistically to exacerbate VCI-associated pathology. Here, using male Sprague-Dawley rats, we examined the effects of a clinically relevant model of adolescent-onset MetS and adult-onset CCH on neuro-vascular outcomes, combining a cafeteria diet with a 2-vessel occlusion (2VO) model. Using longitudinal imaging, histology, and behavioural assessments, we identified several features of MetS and CCH including reduced cerebral blood volume, white matter atrophy, alterations in hippocampal cell density, and memory impairment. Furthermore, we identified a number of significant associations, potentially predictive of MetS and pathophysiological outcomes. White matter volume was positively correlated to HDL cholesterol; hippocampal cell density was negatively correlated to fasted blood glucose; cerebral blood flow and volume was negatively predicted by the combination of 2VO surgery and increased fasted blood glucose. These results emphasize the importance of including comorbid conditions when modeling VCI, and they outline a highly translational preclinical model that could be used to investigate potential interventions to mitigate VCI-associated pathology and cognitive decline.
Collapse
Affiliation(s)
- Jessica M Livingston
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Matthew W McDonald
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada; Canadian Partnership for Stroke Recovery, Ottawa, ON, Canada
| | - Therese Gagnon
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Matthew S Jeffers
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada; Canadian Partnership for Stroke Recovery, Ottawa, ON, Canada
| | - Mariana Gomez-Smith
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Sabina Antonescu
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Greg O Cron
- Ottawa Hospital Research Institute, Neuroscience Program, Ottawa, ON, Canada; Department of Medical Imaging, The Ottawa Hospital, Ottawa, ON, Canada; Department of Radiology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Carlie Boisvert
- Ottawa Hospital Research Institute, Neuroscience Program, Ottawa, ON, Canada
| | - Baptiste Lacoste
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada; Canadian Partnership for Stroke Recovery, Ottawa, ON, Canada; Ottawa Hospital Research Institute, Neuroscience Program, Ottawa, ON, Canada; University of Ottawa Brain and Mind Research Institute, Ottawa, ON, Canada
| | - Dale Corbett
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada; Canadian Partnership for Stroke Recovery, Ottawa, ON, Canada; University of Ottawa Brain and Mind Research Institute, Ottawa, ON, Canada.
| |
Collapse
|
81
|
Li H, Kittur FS, Hung CY, Li PA, Ge X, Sane DC, Xie J. Quantitative Proteomics Reveals the Beneficial Effects of Low Glucose on Neuronal Cell Survival in an in vitro Ischemic Penumbral Model. Front Cell Neurosci 2020; 14:272. [PMID: 33033473 PMCID: PMC7491318 DOI: 10.3389/fncel.2020.00272] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Accepted: 07/31/2020] [Indexed: 01/04/2023] Open
Abstract
Understanding proteomic changes in the ischemic penumbra are crucial to rescue those salvageable cells and reduce the damage of an ischemic stroke. Since the penumbra region is dynamic with heterogeneous cells/tissues, tissue sampling from animal models of stroke for the molecular study is a challenge. In this study, cultured hippocampal HT22 cells under hypoxia treatment for 17.5 h with 0.69 mM low glucose (H+LG) could mimic ischemic penumbral cells since they had much higher cell viability and viable cell number compared to hypoxia without glucose (H-G) treatment. To validate established cell-based ischemic penumbral model and understand the beneficial effects of low glucose (LG), quantitative proteomics analysis was performed on H+LG, H-G, and normoxia with normal 22 mM glucose (N+G) treated cells. We identified 427 differentially abundant proteins (DAPs) between H-G and N+G and further identified 105 DAPs between H+LG and H-G. Analysis of 105 DAPs revealed that LG promotes cell survival by activating HIF1α to enhance glycolysis; preventing the dysregulations of extracellular matrix remodeling, cell cycle and division, and antioxidant and detoxification; as well as attenuating inflammatory reaction response, protein synthesis and neurotransmission activity. Our results demonstrated that this established cell-based system could mimic penumbral conditions and can be used for molecular studies.
Collapse
Affiliation(s)
- Hua Li
- Department of Pharmaceutical Sciences, Biomanufacturing Research Institute and Technology Enterprise, North Carolina Central University, Durham, NC, United States
| | - Farooqahmed S Kittur
- Department of Pharmaceutical Sciences, Biomanufacturing Research Institute and Technology Enterprise, North Carolina Central University, Durham, NC, United States
| | - Chiu-Yueh Hung
- Department of Pharmaceutical Sciences, Biomanufacturing Research Institute and Technology Enterprise, North Carolina Central University, Durham, NC, United States
| | - P Andy Li
- Department of Pharmaceutical Sciences, Biomanufacturing Research Institute and Technology Enterprise, North Carolina Central University, Durham, NC, United States
| | - Xinghong Ge
- Department of Pharmaceutical Sciences, Biomanufacturing Research Institute and Technology Enterprise, North Carolina Central University, Durham, NC, United States.,Department of Dermatology, General Hospital of Ningxia Medical University, Yinchuan, China
| | - David C Sane
- Carilion Clinic, Virginia Tech Carilion School of Medicine, Roanoke, VA, United States
| | - Jiahua Xie
- Department of Pharmaceutical Sciences, Biomanufacturing Research Institute and Technology Enterprise, North Carolina Central University, Durham, NC, United States
| |
Collapse
|
82
|
Lee TK, Park JH, Ahn JH, Kim H, Song M, Lee JC, Kim JD, Jeon YH, Choi JH, Lee CH, Hwang IK, Yan BC, Won MH, Kang IJ. Pretreatment of Populus tomentiglandulosa protects hippocampal CA1 pyramidal neurons from ischemia-reperfusion injury in gerbils via increasing SODs expressions and maintaining BDNF and IGF-I expressions. Chin J Nat Med 2019; 17:424-434. [PMID: 31262455 DOI: 10.1016/s1875-5364(19)30050-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Indexed: 12/31/2022]
Abstract
To examine the effects of Populus tomentiglandulosa (PT) extract on the expressions of antioxidant enzymes and neurotrophic factors in the cornu ammonis 1 (CA1) region of the hippocampus at 5 min after inducing transient global cerebral ischemia (TGCI) in gerbils, TGCI was induced by occlusion of common carotid arteries for 5 min. Before ischemic surgery, 200 mg·kg-1 PT extract was orally administrated once daily for 7 d. We performed neuronal nuclear antigen immunohistochemistry and Fluoro-Jade B staining. Furthermore, we determined in situ production of superoxide anion radical, expression levels of SOD1 and SOD2 as antioxidant enzymes and brain-derived neurotrophic factor (BDNF) and insulin-like growth factor I (IGF-I) as neurotrophic factors. Pretreatment with 200 mg·kg-1 PT extract prevented neuronal death (loss). Furthermore, pretreatment with 200 mg·kg-1 PT extract significantly inhibited the production of superoxide anion radical, increased expressions of SODs and maintained expressions of BDNF and IGF-I. Such increased expressions of SODs were maintained in the neurons after IRI. In summary, pretreated PT extract can significantly increase levels of SODs and protect the neurons against TGCI, suggesting that PT can be a useful natural agent to protect against TGCI.
Collapse
Affiliation(s)
- Tae-Kyeong Lee
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon 24341, Republic of Korea
| | - Joon Ha Park
- Department of Biomedical Science, Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon 24252, Republic of Korea
| | - Ji Hyeon Ahn
- Department of Biomedical Science, Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon 24252, Republic of Korea
| | - Hyunjung Kim
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon 24341, Republic of Korea
| | - Minah Song
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon 24341, Republic of Korea
| | - Jae-Chul Lee
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon 24341, Republic of Korea
| | - Jong Dai Kim
- Division of Food Biotechnology, School of Biotechnology, Kangwon National University, Chuncheon 24341, Republic of Korea
| | - Yong Hwan Jeon
- Department of Radiology, School of Medicine, Kangwon National University, Chuncheon 24289, Republic of Korea
| | - Jung Hoon Choi
- Department of Anatomy, College of Veterinary Medicine, Kangwon National University, Chuncheon 24341, Republic of Korea
| | - Choong Hyun Lee
- Department of Pharmacy, College of Pharmacy, Dankook University, Cheonan 31116, Republic of Korea
| | - In Koo Hwang
- Department of Anatomy and Cell Biology, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul 08826, Republic of Korea
| | - Bing-Chun Yan
- Jiangsu Key Laboratory of Integrated Traditional Chinese, Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou 225001, China
| | - Moo-Ho Won
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon 24341, Republic of Korea.
| | - Il Jun Kang
- Department of Food Science and Nutrition, Hallym University, Chuncheon 24252, Republic of Korea.
| |
Collapse
|
83
|
Wang W, Liu X, Lu H, Liu L, Wang Y, Yu Y, Zhang T. A method for predicting the success of Pulsinell’s four-vessel occlusion rat model by LDF monitoring of cerebral blood flow decline. J Neurosci Methods 2019; 328:108439. [DOI: 10.1016/j.jneumeth.2019.108439] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 09/18/2019] [Accepted: 09/18/2019] [Indexed: 12/18/2022]
|
84
|
Ernst L, David B, Gaubatz J, Domínguez-Narciso I, Lüchters G, Becker AJ, Weber B, Hattingen E, Elger CE, Rüber T. Volumetry of Mesiotemporal Structures Reflects Serostatus in Patients with Limbic Encephalitis. AJNR Am J Neuroradiol 2019; 40:2081-2089. [PMID: 31727746 DOI: 10.3174/ajnr.a6289] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Accepted: 09/11/2019] [Indexed: 01/07/2023]
Abstract
BACKGROUND AND PURPOSE Limbic encephalitis is an autoimmune disease. A variety of autoantibodies have been associated with different subtypes of limbic encephalitis, whereas its MR imaging signature is uniformly characterized by mesiotemporal abnormalities across subtypes. Here, we hypothesized that patients with limbic encephalitis would show subtype-specific mesiotemporal structural correlates, which could be classified by supervised machine learning on an individual level. MATERIALS AND METHODS T1WI MPRAGE scans from 46 patients with antibodies against glutamic acid decarboxylase and 34 patients with antibodies against the voltage-gated potassium channel complex (including 10 patients with leucine-rich glioma-inactivated 1 autoantibodies) and 48 healthy controls were retrospectively ascertained. Parcellation of the amygdala, hippocampus, and hippocampal subfields was performed using FreeSurfer. Volumes were extracted and compared between groups using unpaired, 2-tailed t tests. The volumes of hippocampal subfields were analyzed using a multivariate linear model and a binary decision tree classifier. RESULTS Temporomesial volume alterations were most pronounced in an early stage and in the affected hemispheric side of patients. Statistical analysis revealed antibody-specific hippocampal fingerprints with a higher volume of CA1 in patients with glutamic acid decarboxylase-associated limbic encephalitis (P = .02), compared with controls, whereas CA1 did not differ from that in controls in patients with voltage-gated potassium channel complex autoantibodies. The classifier could successfully distinguish between patients with autoantibodies against leucine-rich glioma-inactivated 1 and glutamic acid decarboxylase with a specificity of 87% and a sensitivity of 80%. CONCLUSIONS Our results suggest stage-, side- and antibody-specific structural correlates of limbic encephalitis; thus, they create a perspective toward an MR imaging-based diagnosis.
Collapse
Affiliation(s)
- L Ernst
- From the Department of Epileptology (L.E., B.D., J.G., I.D.-N., C.E.E., T.R.)
| | - B David
- From the Department of Epileptology (L.E., B.D., J.G., I.D.-N., C.E.E., T.R.)
| | - J Gaubatz
- From the Department of Epileptology (L.E., B.D., J.G., I.D.-N., C.E.E., T.R.)
| | - I Domínguez-Narciso
- From the Department of Epileptology (L.E., B.D., J.G., I.D.-N., C.E.E., T.R.)
| | - G Lüchters
- Center for Development Research (G.L.), University of Bonn, Bonn, Germany
| | | | - B Weber
- Institute for Experimental Epileptology and Cognition Research (B.W.)
| | - E Hattingen
- Department of Radiology (E.H.), University of Bonn Medical Center, Bonn, Germany
- Department of Neuroradiology (E.H.), Goethe University Frankfurt, Frankfurt, Germany
| | - C E Elger
- From the Department of Epileptology (L.E., B.D., J.G., I.D.-N., C.E.E., T.R.)
| | - T Rüber
- From the Department of Epileptology (L.E., B.D., J.G., I.D.-N., C.E.E., T.R.)
- Epilepsy Center Frankfurt Rhine-Main (T.R.)
- Department of Neurology, and Center for Personalized Translational Epilepsy Research (T.R.), Goethe-University Frankfurt, Frankfurt am Main, Germany
| |
Collapse
|
85
|
Kim YS, Cho JH, Shin MC, Park Y, Park CW, Tae HJ, Cho JH, Kim IS, Lee TK, Park YE, Ahn JH, Park JH, Kim DW, Won MH, Lee JC. Effects of regional body temperature variation during asphyxial cardiac arrest on mortality and brain damage in a rat model. J Therm Biol 2019; 87:102466. [PMID: 31999601 DOI: 10.1016/j.jtherbio.2019.102466] [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: 09/10/2019] [Revised: 11/18/2019] [Accepted: 11/21/2019] [Indexed: 01/19/2023]
Abstract
To date, hypothermia has focused on improving rates of resuscitation to increase survival in patients sustaining cardiac arrest (CA). Towards this end, the role of body temperature in neuronal damage or death during CA needs to be determined. However, few studies have investigated the effect of regional temperature variation on survival rate and neurological outcomes. In this study, adult male rats (12 week-old) were used under the following four conditions: (i) whole-body normothermia (37 ± 0.5 °C) plus (+) no asphyxial CA, (ii) whole-body normothermia + CA, (iii) whole-body hypothermia (33 ± 0.5 °C)+CA, (iv) body hypothermia/brain normothermia + CA, and (v) brain hypothermia/body normothermia + CA. The survival rate after resuscitation was significantly elevated in groups exposed to whole-body hypothermia plus CA and body hypothermia/brain normothermia plus CA, but not in groups exposed to whole-body normothermia combined with CA and brain hypothermia/body normothermia plus CA. However, the group exposed to hypothermia/brain normothermia combined with CA exhibited higher neuroprotective effects against asphyxial CA injury, i.e. improved neurological deficit and neuronal death in the hippocampus compared with those involving whole-body normothermia combined with CA. In addition, neurological deficit and neuronal death in the group of rat exposed to brain hypothermia/body normothermia and CA were similar to those in the rats subjected to whole-body normothermia and CA. In brief, only brain hypothermia during CA was not associated with effective survival rate, neurological function or neuronal protection compared with those under body (but not brain) hypothermia during CA. Our present study suggests that regional temperature in patients during CA significantly affects the outcomes associated with survival rate and neurological recovery.
Collapse
Affiliation(s)
- Yoon Sung Kim
- Department of Emergency Medicine, School of Medicine, Kangwon National University, Chuncheon, Gangwon, 24341, Republic of Korea; Department of Emergency Medicine, Samcheok Medical Center, Samcheok, Kangwon, 25920, Republic of Korea
| | - Jun Hwi Cho
- Department of Emergency Medicine, School of Medicine, Kangwon National University, Chuncheon, Gangwon, 24341, Republic of Korea
| | - Myoung-Cheol Shin
- Department of Emergency Medicine, School of Medicine, Kangwon National University, Chuncheon, Gangwon, 24341, Republic of Korea
| | - Yoonsoo Park
- Department of Emergency Medicine, School of Medicine, Kangwon National University, Chuncheon, Gangwon, 24341, Republic of Korea
| | - Chan Woo Park
- Department of Emergency Medicine, School of Medicine, Kangwon National University, Chuncheon, Gangwon, 24341, Republic of Korea
| | - Hyun-Jin Tae
- Bio-Safety Research Institute, College of Veterinary Medicine, Chonbuk National University, Iksan, Chonbuk, 54596, Republic of Korea
| | - Jeong Hwi Cho
- Bio-Safety Research Institute, College of Veterinary Medicine, Chonbuk National University, Iksan, Chonbuk, 54596, Republic of Korea
| | - In-Shik Kim
- Bio-Safety Research Institute, College of Veterinary Medicine, Chonbuk National University, Iksan, Chonbuk, 54596, Republic of Korea
| | - Tae-Kyeong Lee
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, Gangwon, 24341, Republic of Korea
| | - Young Eun Park
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, Gangwon, 24341, Republic of Korea
| | - Ji Hyeon Ahn
- Department of Biomedical Science and Research Institute for Bioscience and Biotechnology, Hallym University, Chuncheon, Gangwon, 24252, Republic of Korea
| | - Joon Ha Park
- Department of Anatomy, College of Korean Medicine, Dongguk University, Gyeongju, Gyeongbuk, 38066, Republic of Korea
| | - Dae Won Kim
- Department of Biochemistry and Molecular Biology, Research Institute of Oral Sciences, College of Dentistry, Kangnung-Wonju National University, Gangneung, Gangwon, 25457, Republic of Korea
| | - Moo-Ho Won
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, Gangwon, 24341, Republic of Korea.
| | - Jae-Chul Lee
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, Gangwon, 24341, Republic of Korea.
| |
Collapse
|
86
|
Kreisman NR, Wooliscroft LB, Campbell CF, Dotiwala AK, Cox ML, Denson AC, Betancourt AM, Tomchuck SL. Preconditioning hippocampal slices with hypothermia promotes rapid tolerance to hypoxic depolarization and swelling: Mediation by erythropoietin. Brain Res 2019; 1726:146517. [PMID: 31634451 DOI: 10.1016/j.brainres.2019.146517] [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: 04/17/2019] [Revised: 08/06/2019] [Accepted: 10/17/2019] [Indexed: 12/26/2022]
Abstract
We suggested previously that hippocampal slices were protected from hypoxic depolarization and swelling by preincubating them at room temperature (Kreisman et al., 2000). We postulated that hypothermic preconditioning induced tolerance in our slices, which protected against hypoxic depolarization and swelling. Control hippocampal slices were incubated at 34-35 °C for two hours and the response to 10 min of severe hypoxia was compared to slices which were preconditioned for two hours at room temperature (22-23 °C) prior to warming to 34-35 °C. Recordings of the extracellular DC potential provided an index of tissue depolarization and changes in tissue light transmittance provided an index of swelling. Hypothermic preconditioning significantly reduced hypoxia-induced swelling, particularly in CA3 and the dentate inner blade. Since erythropoietin (EPO) had been shown to mediate hypoxic preconditioning, we tested whether EPO also mediated hypothermic preconditioning in our slices. Recombinant rat EPO (1-10 micromolar) mitigated hypoxia-induced swelling and depolarization in dentate inner blade of unconditioned slices in a dose-dependent manner. We also blocked the protective effects of hypothermic preconditioning on hypoxic depolarization and swelling in the inner blade of the dentate gyrus by administering soluble EPO receptor in the bath and treating slices with wortmannin to block phosphorylation of PI3 kinase, a critical step in the activation of the downstream neuroprotectant, Akt. These results suggest that EPO mediates tolerance to hypoxic depolarization and swelling induced by hypothermic preconditioning. They also emphasize that various preincubation protocols used in experiments with hippocampal slices may differentially affect basal electrophysiological and metabolic properties of those slices.
Collapse
Affiliation(s)
- Norman R Kreisman
- Department of Physiology, Tulane University School of Medicine, New Orleans, LA 70112, United States; Tulane Brain Institute, New Orleans, LA 70118, United States.
| | | | - Carolyn F Campbell
- Department of Physiology, Tulane University School of Medicine, New Orleans, LA 70112, United States; Department of Pharmacology, Tulane University School of Medicine, New Orleans, LA 70112, United States
| | - Ary K Dotiwala
- Tulane Brain Institute, New Orleans, LA 70118, United States
| | - Michael L Cox
- Department of Physiology, Tulane University School of Medicine, New Orleans, LA 70112, United States
| | - Aaron C Denson
- Department of Physiology, Tulane University School of Medicine, New Orleans, LA 70112, United States
| | - Aline M Betancourt
- Department of Medicine, Tulane University School of Medicine, New Orleans, LA 70112, United States
| | - Suzanne L Tomchuck
- Department of Microbiology & Immunology, Tulane University School of Medicine, New Orleans, LA 70112, United States
| |
Collapse
|
87
|
Neuroprotection mediated by remote preconditioning is associated with a decrease in systemic oxidative stress and changes in brain and blood glutamate concentration. Neurochem Int 2019; 129:104461. [DOI: 10.1016/j.neuint.2019.05.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 04/30/2019] [Accepted: 05/07/2019] [Indexed: 11/20/2022]
|
88
|
Leeuwen L, Schiller RM, Rietman AB, van Rosmalen J, Wildschut ED, Houmes RJM, Tibboel D, IJsselstijn H. Risk Factors of Impaired Neuropsychologic Outcome in School-Aged Survivors of Neonatal Critical Illness. Crit Care Med 2019; 46:401-410. [PMID: 29194146 DOI: 10.1097/ccm.0000000000002869] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
OBJECTIVE Until now, long-term outcome studies have focused on general cognitive functioning and its risk factors following neonatal extracorporeal membrane oxygenation and/or congenital diaphragmatic hernia. However, it is currently unknown which neuropsychological domains are most affected in these patients and which clinical variables can be used to predict specific neuropsychological problems. This study aimed to identify affected neuropsychological domains and its clinical determinants in survivors of neonatal extracorporeal membrane oxygenation and/or congenital diaphragmatic hernia. DESIGN Prospective follow-up study. SETTING Tertiary university hospital. PATIENTS Sixty-five 8-year-old survivors of neonatal extracorporeal membrane oxygenation and/or congenital diaphragmatic hernia. INTERVENTIONS None. MEASUREMENTS AND MAIN RESULTS Intelligence, attention, memory, executive functioning and visuospatial processing were evaluated using validated tests and compared with Dutch reference data. Assessed risk factors of outcome were illness severity indicators, number of anesthetic procedures in the first year of life, and growth at 1 year. Patients had average intelligence (mean intelligence quotient ± SD, 95 ± 16), but significantly poorer sustained attention (mean z score ± SD, -2.73 ± 2.57), verbal (immediate, -1.09 ± 1.27; delayed, -1.14 ± 1.86), and visuospatial memory (immediate, -1.48 ± 1.02; delayed, -1.57 ± 1.01; recognition, -1.07 ± 3.10) than the norm. Extracorporeal membrane oxygenation-treated congenital diaphragmatic hernia patients had significantly lower mean intelligence quotient (84 ± 12) than other neonatal extracorporeal membrane oxygenation patients (94 ± 10) and congenital diaphragmatic hernia patients not treated with extracorporeal membrane oxygenation (100 ± 20). Maximum vasoactive-inotropic score was negatively associated with delayed verbal (B = -0.02; 95% CI, -0.03 to -0.002; p = 0.026) and visuospatial memory (B = -0.01; 95% CI, -0.02 to -0.001; p = 0.024). CONCLUSIONS We found memory and attention deficits in 8-year-old neonatal extracorporeal membrane oxygenation and congenital diaphragmatic hernia survivors. The maximum dose of vasoactive medication was negatively associated with verbal and visuospatial memory, which may suggest an effect of early cerebral hypoperfusion in determining these abnormalities.
Collapse
Affiliation(s)
- Lisette Leeuwen
- Intensive Care and Department of Pediatric Surgery, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Raisa M Schiller
- Intensive Care and Department of Pediatric Surgery, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands
| | - André B Rietman
- Intensive Care and Department of Pediatric Surgery, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands
| | | | - Enno D Wildschut
- Intensive Care and Department of Pediatric Surgery, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Robert Jan M Houmes
- Intensive Care and Department of Pediatric Surgery, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Dick Tibboel
- Intensive Care and Department of Pediatric Surgery, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Hanneke IJsselstijn
- Intensive Care and Department of Pediatric Surgery, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands
| |
Collapse
|
89
|
Font‐Belmonte E, Ugidos IF, Santos‐Galdiano M, González‐Rodríguez P, Anuncibay‐Soto B, Pérez‐Rodríguez D, Gonzalo‐Orden JM, Fernández‐López A. Post‐ischemic salubrinal administration reduces necroptosis in a rat model of global cerebral ischemia. J Neurochem 2019; 151:777-794. [DOI: 10.1111/jnc.14789] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Revised: 05/24/2019] [Accepted: 05/29/2019] [Indexed: 02/06/2023]
Affiliation(s)
| | - Irene F. Ugidos
- Área de Biología Celular, Instituto de Biomedicina University of León León Spain
| | | | | | - Berta Anuncibay‐Soto
- Área de Biología Celular, Instituto de Biomedicina University of León León Spain
| | | | | | | |
Collapse
|
90
|
de Jong DLK, de Heus RAA, Rijpma A, Donders R, Olde Rikkert MGM, Günther M, Lawlor BA, van Osch MJP, Claassen JAHR. Effects of Nilvadipine on Cerebral Blood Flow in Patients With Alzheimer Disease. Hypertension 2019; 74:413-420. [PMID: 31203725 DOI: 10.1161/hypertensionaha.119.12892] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Cerebrovascular changes, including reduced cerebral blood flow (CBF), occur early in the development of Alzheimer disease and may accelerate disease progression. This randomized, double-blind, placebo-controlled study investigated how 6 months of treatment with the calcium antagonist nilvadipine would affect CBF in patients with mild-to-moderate Alzheimer disease. CBF was measured with magnetic resonance arterial spin labeling in whole-brain gray matter and in a priori defined regions of interest including the hippocampus. Fifty-eight patients were randomly assigned (29 in each group), of whom 22 in both groups had no magnetic resonance exclusion criteria and were medication compliant over 6 months. Mean age was 72.8±6.2 years, mean mini-mental state examination was 20.4±3.4. Nilvadipine treatment lowered systolic blood pressure (Δ=-11.5 [95% CI, -19.7 to -3.2] mm Hg; P<0.01), while whole-brain gray-matter CBF remained stable (Δ=5.4 [95% CI, -6.4 to 17.2] mL/100 g per minute; P=0.36). CBF in the hippocampus increased (left: Δ=24.4 [95% CI, 4.3-44.5] mL/100 g per minute; P=0.02; right: Δ=20.1 [95% CI, -0.6 to 40.8] mL/100 g per minute; P=0.06). There was no significant change in CBF in the posterior cingulate cortex (Δ=5.2 [95% CI, -16.5 to 27.0] mL/100 g per minute; P=0.63) or other regions of interest. In conclusion, nilvadipine reduced blood pressure and increased CBF in the hippocampus, whereas other regions showed stable or small nonsignificant increases in CBF. These findings not only indicate preserved cerebral autoregulation in Alzheimer disease but also point toward beneficial cerebrovascular effects of antihypertensive treatment. Clinical Trial Registration- URL: http://www.clinicaltrials.gov . Unique identifier: NCT02017340.
Collapse
Affiliation(s)
- Daan L K de Jong
- From the Department of Geriatric Medicine, Donders Institute for Brain Cognition and Behaviour (D.L.K.d.J., R.A.A.d.H., A.R., M.G.M.O.R., J.A.H.R.C.), Radboud University Medical Center, Nijmegen, the Netherlands.,Radboudumc Alzheimer Center, Nijmegen, the Netherlands (D.L.K.d.J., R.A.A.d.H., A.R., M.G.M.O.R., J.A.H.R.C.)
| | - Rianne A A de Heus
- From the Department of Geriatric Medicine, Donders Institute for Brain Cognition and Behaviour (D.L.K.d.J., R.A.A.d.H., A.R., M.G.M.O.R., J.A.H.R.C.), Radboud University Medical Center, Nijmegen, the Netherlands.,Radboudumc Alzheimer Center, Nijmegen, the Netherlands (D.L.K.d.J., R.A.A.d.H., A.R., M.G.M.O.R., J.A.H.R.C.)
| | - Anne Rijpma
- From the Department of Geriatric Medicine, Donders Institute for Brain Cognition and Behaviour (D.L.K.d.J., R.A.A.d.H., A.R., M.G.M.O.R., J.A.H.R.C.), Radboud University Medical Center, Nijmegen, the Netherlands.,Radboudumc Alzheimer Center, Nijmegen, the Netherlands (D.L.K.d.J., R.A.A.d.H., A.R., M.G.M.O.R., J.A.H.R.C.)
| | - Rogier Donders
- Department of Health Evidence (R.D.), Radboud University Medical Center, Nijmegen, the Netherlands
| | - Marcel G M Olde Rikkert
- From the Department of Geriatric Medicine, Donders Institute for Brain Cognition and Behaviour (D.L.K.d.J., R.A.A.d.H., A.R., M.G.M.O.R., J.A.H.R.C.), Radboud University Medical Center, Nijmegen, the Netherlands.,Radboudumc Alzheimer Center, Nijmegen, the Netherlands (D.L.K.d.J., R.A.A.d.H., A.R., M.G.M.O.R., J.A.H.R.C.)
| | - Matthias Günther
- Frauenhofer Institute for Medical Imaging Computing MEVIS, Bremen, Germany (M.G.)
| | - Brian A Lawlor
- Trinity College Institute of Neuroscience, Dublin, Ireland (B.A.L.)
| | - Matthias J P van Osch
- Department of Radiology, C.J. Gorter Center for High Field MRI, Leiden University Medical Center, the Netherlands (M.J.P.v.O.)
| | - Jurgen A H R Claassen
- From the Department of Geriatric Medicine, Donders Institute for Brain Cognition and Behaviour (D.L.K.d.J., R.A.A.d.H., A.R., M.G.M.O.R., J.A.H.R.C.), Radboud University Medical Center, Nijmegen, the Netherlands.,Radboudumc Alzheimer Center, Nijmegen, the Netherlands (D.L.K.d.J., R.A.A.d.H., A.R., M.G.M.O.R., J.A.H.R.C.)
| |
Collapse
|
91
|
Ahn JH, Ohk TG, Kim DW, Kim H, Song M, Lee TK, Lee JC, Yang GE, Shin MC, Cho JH, Choi SY, Won MH, Park JH. Fluoro-Jade B histofluorescence staining detects dentate granule cell death after repeated five-minute transient global cerebral ischemia. Metab Brain Dis 2019; 34:951-956. [PMID: 30830598 DOI: 10.1007/s11011-019-00404-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Accepted: 02/25/2019] [Indexed: 02/06/2023]
Abstract
Transient global cerebral ischemia (tGCI)-induced neuronal damage is variable according to its duration and degree. There are many studies on the damage or death of pyramidal cells of the hippocampus proper (CA1-3) in rodent models of tGCI. However, studies on the death of granule cells in the hippocampal dentate gyrus (DG) following tGCI have not yet been addressed. In this study, we examined the damage/death of granule cells in the gerbil DG at 5 days after various durations (5, 10, and 15 min) of single tGCI and repeated tGCI (two 5-min tGCI with 1-h interval) using cresyl violet staining, NeuN immunohistochemistry and Fluoro-Jade B (F-J B) histofluorescence staining. Neuronal death was observed only in the polymorphic layer in all single tGCI-operated groups. However, in the repeated tGCI-operated group, massive neuronal death was observed in the granule cell layer as well as in the polymorphic layer by using F-J B histofluorescence staining. In addition, microgliosis in the DG was significantly increased in the repeated tGCI-operated group compared to the 15-min tGCI-operated group. Taken together, these findings indicate that repeated brief tGCI causes granule cell death in the DG which could not occur by a longer duration of single tGCI.
Collapse
Affiliation(s)
- Ji Hyeon Ahn
- Department of Biomedical Science and Research Institute for Bioscience and Biotechnology, Hallym University, 1 Hallimdaehak-gil, Chuncheon, Gangwon, 24252, Republic of Korea
| | - Taek Geun Ohk
- Department of Emergency Medicine, School of Medicine, Kangwon National University, Chuncheon, Gangwon, 24341, Republic of Korea
| | - Dae Won Kim
- Department of Biochemistry and Molecular Biology, and Research Institute of Oral Sciences, College of Dentistry, Gangnung-Wonju National University, Gangneung, Gangwon, 25457, Republic of Korea
| | - Hyunjung Kim
- Department of Neurobiology, School of Medicine, Kangwon National University, 1 Kangwondaehak-gil, Chuncheon, Gangwon, 24341, Republic of Korea
| | - Minah Song
- Department of Neurobiology, School of Medicine, Kangwon National University, 1 Kangwondaehak-gil, Chuncheon, Gangwon, 24341, Republic of Korea
| | - Tae-Kyeong Lee
- Department of Neurobiology, School of Medicine, Kangwon National University, 1 Kangwondaehak-gil, Chuncheon, Gangwon, 24341, Republic of Korea
| | - Jae-Chul Lee
- Department of Neurobiology, School of Medicine, Kangwon National University, 1 Kangwondaehak-gil, Chuncheon, Gangwon, 24341, Republic of Korea
| | - Go Eun Yang
- Department of Radiology, Kangwon National University Hospital, Chuncheon, Gangwon, 24289, Republic of Korea
| | - Myoung Cheol Shin
- Department of Emergency Medicine, School of Medicine, Kangwon National University, Chuncheon, Gangwon, 24341, Republic of Korea
| | - Jun Hwi Cho
- Department of Emergency Medicine, School of Medicine, Kangwon National University, Chuncheon, Gangwon, 24341, Republic of Korea
| | - Soo Young Choi
- Department of Biomedical Science and Research Institute for Bioscience and Biotechnology, Hallym University, 1 Hallimdaehak-gil, Chuncheon, Gangwon, 24252, Republic of Korea
| | - Moo-Ho Won
- Department of Neurobiology, School of Medicine, Kangwon National University, 1 Kangwondaehak-gil, Chuncheon, Gangwon, 24341, Republic of Korea.
| | - Joon Ha Park
- Department of Biomedical Science and Research Institute for Bioscience and Biotechnology, Hallym University, 1 Hallimdaehak-gil, Chuncheon, Gangwon, 24252, Republic of Korea.
| |
Collapse
|
92
|
Blockade of Acid-Sensing Ion Channels Attenuates Recurrent Hypoglycemia-Induced Potentiation of Ischemic Brain Damage in Treated Diabetic Rats. Neuromolecular Med 2019; 21:454-466. [PMID: 31134484 DOI: 10.1007/s12017-019-08546-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Accepted: 05/17/2019] [Indexed: 12/18/2022]
Abstract
Diabetes is a chronic metabolic disease and cerebral ischemia is a serious complication of diabetes. Anti-diabetic therapy mitigates this complication but increases the risk of exposure to recurrent hypoglycemia (RH). We showed previously that RH exposure increases ischemic brain damage in insulin-treated diabetic (ITD) rats. The present study evaluated the hypothesis that increased intra-ischemic acidosis in RH-exposed ITD rats leads to pronounced post-ischemic hypoperfusion via activation of acid-sensing (proton-gated) ion channels (ASICs). Streptozotocin-diabetic rats treated with insulin were considered ITD rats. ITD rats were exposed to RH for 5 days and were randomized into Psalmotoxin1 (PcTx1, ASIC1a inhibitor), APETx2 (ASIC3 inhibitor), or vehicle groups. Transient global cerebral ischemia was induced overnight after RH. Cerebral blood flow was measured using laser Doppler flowmetry. Ischemic brain injury in hippocampus was evaluated using histopathology. Post-ischemic hypoperfusion in RH-exposed rats was of greater extent than that in control rats. Inhibition of ASICs prevented RH-induced increase in the extent of post-ischemic hypoperfusion and ischemic brain injury. Since ASIC activation-induced store-operated calcium entry (SOCE) plays a role in vascular tone, next we tested if acidosis activates SOCE via activating ASICs in vascular smooth muscle cells (VSMCs). We observed that SOCE in VSMCs at lower pH is ASIC3 dependent. The results show the role of ASIC in post-ischemic hypoperfusion and increased ischemic damage in RH-exposed ITD rats. Understanding the pathways mediating exacerbated ischemic brain injury in RH-exposed ITD rats may help lower diabetic aggravation of ischemic brain damage.
Collapse
|
93
|
Cheng M, Yang L, Dong Z, Wang M, Sun Y, Liu H, Wang X, Sai N, Huang G, Zhang X. Folic acid deficiency enhanced microglial immune response via the Notch1/nuclear factor kappa B p65 pathway in hippocampus following rat brain I/R injury and BV2 cells. J Cell Mol Med 2019; 23:4795-4807. [PMID: 31087489 PMCID: PMC6584545 DOI: 10.1111/jcmm.14368] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2018] [Revised: 03/13/2019] [Accepted: 04/14/2019] [Indexed: 12/22/2022] Open
Abstract
Recent studies revealed that folic acid deficiency (FD) increased the likelihood of stroke and aggravated brain injury after focal cerebral ischaemia. The microglia-mediated inflammatory response plays a crucial role in the complicated pathologies that lead to ischaemic brain injury. However, whether FD is involved in the activation of microglia and the neuroinflammation after experimental stroke and the underlying mechanism is still unclear. The aim of the present study was to assess whether FD modulates the Notch1/nuclear factor kappa B (NF-κB) pathway and enhances microglial immune response in a rat middle cerebral artery occlusion-reperfusion (MCAO) model and oxygen-glucose deprivation (OGD)-treated BV-2 cells. Our results exhibited that FD worsened neuronal cell death and exaggerated microglia activation in the hippocampal CA1, CA3 and Dentate gyrus (DG) subregions after cerebral ischaemia/reperfusion. The hippocampal CA1 region was more sensitive to ischaemic injury and FD treatment. The protein expressions of proinflammatory cytokines such as tumour necrosis factor-α, interleukin-1β and interleukin-6 were also augmented by FD treatment in microglial cells of the post-ischaemic hippocampus and in vitro OGD-stressed microglia model. Moreover, FD not only dramatically enhanced the protein expression levels of Notch1 and NF-κB p65 but also promoted the phosphorylation of pIkBα and the nuclear translocation of NF-κB p65. Blocking of Notch1 with N-[N-(3, 5-difluorophenacetyl)-l-alanyl]-S-phenylglycine t-butyl ester partly attenuated the nuclear translocation of NF-κB p65 and the protein expression of neuroinflammatory cytokines in FD-treated hypoxic BV-2 microglia. These results suggested that Notch1/NF-κB p65 pathway-mediated microglial immune response may be a molecular mechanism underlying cerebral ischaemia-reperfusion injury worsened by FD treatment.
Collapse
Affiliation(s)
- Man Cheng
- Department of Nutrition and Food Science, School of Public Health, Tianjin Medical University, Tianjin, China.,Tianjin Key Laboratory of Environment, Nutrition and Public Health, Tianjin, China
| | - Liu Yang
- Department of Nutrition and Food Science, School of Public Health, Tianjin Medical University, Tianjin, China.,Tianjin Key Laboratory of Environment, Nutrition and Public Health, Tianjin, China
| | - Zhiping Dong
- Department of Nutrition and Food Science, School of Public Health, Tianjin Medical University, Tianjin, China.,Tianjin Key Laboratory of Environment, Nutrition and Public Health, Tianjin, China
| | - Mengying Wang
- Department of Nutrition and Food Science, School of Public Health, Tianjin Medical University, Tianjin, China.,Tianjin Key Laboratory of Environment, Nutrition and Public Health, Tianjin, China
| | - Yan Sun
- School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Huan Liu
- Department of Nutrition and Food Science, School of Public Health, Tianjin Medical University, Tianjin, China.,Tianjin Key Laboratory of Environment, Nutrition and Public Health, Tianjin, China
| | - Xuan Wang
- Department of Nutrition and Food Science, School of Public Health, Tianjin Medical University, Tianjin, China.,Tianjin Key Laboratory of Environment, Nutrition and Public Health, Tianjin, China
| | - Na Sai
- Department of Nutrition and Food Science, School of Public Health, Tianjin Medical University, Tianjin, China.,Tianjin Key Laboratory of Environment, Nutrition and Public Health, Tianjin, China
| | - Guowei Huang
- Department of Nutrition and Food Science, School of Public Health, Tianjin Medical University, Tianjin, China.,Tianjin Key Laboratory of Environment, Nutrition and Public Health, Tianjin, China
| | - Xumei Zhang
- Department of Nutrition and Food Science, School of Public Health, Tianjin Medical University, Tianjin, China.,Tianjin Key Laboratory of Environment, Nutrition and Public Health, Tianjin, China
| |
Collapse
|
94
|
Aanes S, Bjuland KJ, Sripada K, Sølsnes AE, Grunewaldt KH, Håberg A, Løhaugen GC, Skranes J. Reduced hippocampal subfield volumes and memory function in school-aged children born preterm with very low birthweight (VLBW). Neuroimage Clin 2019; 23:101857. [PMID: 31136968 PMCID: PMC6536855 DOI: 10.1016/j.nicl.2019.101857] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 05/07/2019] [Accepted: 05/09/2019] [Indexed: 01/31/2023]
Abstract
BACKGROUND The hippocampus, an essential structure for learning and memory, has a reduced volume in preterm born (gestational age < 37 weeks) individuals with very low birth weight (VLBW: birth weight < 1500 g), which may affect memory function. However, the hippocampus is a complex structure with distinct subfields related to specific memory functions. These subfields are differentially affected by a variety of neuropathological conditions, but it remains unclear how these subfields may be affected by medical complications following preterm birth which may cause aberrant brain development, and the consequences of this on learning and memory function in children with VLBW. METHODS Children born preterm with VLBW (n = 34) and term-born controls from the Norwegian Mother and Child Cohort Study (MoBa) (n = 104) underwent structural MRI and a neuropsychological assessment of memory function at primary school age. FreeSurfer 6.0 was used to analyze the volumes of hippocampal subfields which were compared between groups, as was memory performance. Correlations between abnormal hippocampal subfields and memory performance were explored in the VLBW group. RESULTS All absolute hippocampal subfield volumes were lower in the children with VLBW compared to MoBa term-born controls, and the volumes of the left and right dentate gyrus and the right subiculum remained significantly lower after correcting for total intracranial volume. The VLBW group had inferior working memory performance and the score on the subtest Spatial Span backwards was positively correlated to the volume of the right dentate gyrus. CONCLUSIONS Hippocampal subfield volumes seem to be differently affected by early brain development related to preterm birth. The dentate gyrus appears particularly susceptible to adverse effects of preterm birth. Reduced working memory function among children with VLBW was associated with smaller volume of right dentate gyrus. This finding demonstrates alterations in hippocampal structure-function relationships associated with early brain development related to preterm birth.
Collapse
Affiliation(s)
- Synne Aanes
- Department of Clinical and Molecular Medicine, Norwegian University of Science & Technology, Trondheim, Norway.
| | | | - Kam Sripada
- Department of Clinical and Molecular Medicine, Norwegian University of Science & Technology, Trondheim, Norway
| | - Anne Elisabeth Sølsnes
- Department of Clinical and Molecular Medicine, Norwegian University of Science & Technology, Trondheim, Norway
| | - Kristine H Grunewaldt
- Department of Clinical and Molecular Medicine, Norwegian University of Science & Technology, Trondheim, Norway; Department of Pediatrics, St Olav University Hospital, Trondheim, Norway
| | - Asta Håberg
- Department of Neuromedicine and Movement Science, Norwegian University of Science & Technology, Trondheim, Norway
| | - Gro C Løhaugen
- Department of Pediatrics, Sørlandet Hospital, Arendal, Norway
| | - Jon Skranes
- Department of Clinical and Molecular Medicine, Norwegian University of Science & Technology, Trondheim, Norway; Department of Pediatrics, Sørlandet Hospital, Arendal, Norway
| |
Collapse
|
95
|
Fontes K, Rohlicek CV, Saint-Martin C, Gilbert G, Easson K, Majnemer A, Marelli A, Chakravarty MM, Brossard-Racine M. Hippocampal alterations and functional correlates in adolescents and young adults with congenital heart disease. Hum Brain Mapp 2019; 40:3548-3560. [PMID: 31070841 DOI: 10.1002/hbm.24615] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Revised: 03/30/2019] [Accepted: 04/24/2019] [Indexed: 01/18/2023] Open
Abstract
There is a high prevalence of neurodevelopmental impairments in individuals living with congenital heart disease (CHD) and the neural correlates of these impairments are not yet fully understood. Recent studies have shown that hippocampal volume and shape differences may provide unique biomarkers for neurodevelopmental disorders. The hippocampus is vulnerable to early life injury, especially in populations at risk for hypoxemia or hemodynamic instability such as in neonates with CHD. We compared hippocampal gray and white matter volume and morphometry between youth born with CHD (n = 50) aged 16-24 years and healthy peers (n = 48). We also explored whether hippocampal gray and white matter volume and morphometry are associated with executive function and self-regulation deficits. To do so, participants underwent 3T brain magnetic resonance imaging and completed the self-reported Behavior Rating Inventory of Executive Function-Adult version. We found that youth with CHD had smaller hippocampal volumes (all statistics corrected for false discovery rate; q < 0.05) as compared to controls. We also observed significant smaller surface area bilaterally and inward displacement on the left hippocampus predominantly on the ventral side (q < 0.10) in the CHD group that were not present in the controls. Left CA1 and CA2/3 were negatively associated with working memory (p < .05). Here, we report, for the first-time, hippocampal morphometric alterations in youth born with CHD when compared to healthy peers, as well as, structure-function relationships between hippocampal volumes and executive function. These differences may reflect long lasting alterations in brain development specific to individual with CHD.
Collapse
Affiliation(s)
- Kimberly Fontes
- Advances in Brain and Child Health Development Research Laboratory, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| | - Charles V Rohlicek
- Department of Pediatrics, Division of Cardiology, Montreal Children's Hospital, McGill University Health Centre, Montreal, Quebec, Canada
| | - Christine Saint-Martin
- Department of Medical Imaging, Division of Pediatric Radiology, Montreal Children's Hospital, McGill University Health Centre, Montreal, Quebec, Canada
| | | | - Kaitlyn Easson
- Advances in Brain and Child Health Development Research Laboratory, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| | - Annette Majnemer
- School of Physical and Occupational Therapy, McGill University, Montreal, Quebec, Canada
| | - Ariane Marelli
- McGill Adult Unit for Congenital Heart Disease Excellence, McGill University, Montreal, Quebec, Canada
| | - M Mallar Chakravarty
- Computational Brain Anatomy Laboratory, Cerebral Imaging Centre - Douglas Mental Health University Institute, Verdun, Quebec, Canada.,Department of Psychiatry, McGill University, Montreal, Quebec, Canada.,Department of Biological and Biomedical Engineering, McGill University, Montreal, Quebec, Canada
| | - Marie Brossard-Racine
- Advances in Brain and Child Health Development Research Laboratory, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada.,School of Physical and Occupational Therapy, McGill University, Montreal, Quebec, Canada.,Department of Pediatrics, Division of Neonatology, Montreal Children's Hospital, McGill University Health Centre, Montreal, Quebec, Canada
| |
Collapse
|
96
|
Restoration of brain circulation and cellular functions hours post-mortem. Nature 2019; 568:336-343. [PMID: 30996318 DOI: 10.1038/s41586-019-1099-1] [Citation(s) in RCA: 162] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Accepted: 03/01/2019] [Indexed: 11/08/2022]
Abstract
The brains of humans and other mammals are highly vulnerable to interruptions in blood flow and decreases in oxygen levels. Here we describe the restoration and maintenance of microcirculation and molecular and cellular functions of the intact pig brain under ex vivo normothermic conditions up to four hours post-mortem. We have developed an extracorporeal pulsatile-perfusion system and a haemoglobin-based, acellular, non-coagulative, echogenic, and cytoprotective perfusate that promotes recovery from anoxia, reduces reperfusion injury, prevents oedema, and metabolically supports the energy requirements of the brain. With this system, we observed preservation of cytoarchitecture; attenuation of cell death; and restoration of vascular dilatory and glial inflammatory responses, spontaneous synaptic activity, and active cerebral metabolism in the absence of global electrocorticographic activity. These findings demonstrate that under appropriate conditions the isolated, intact large mammalian brain possesses an underappreciated capacity for restoration of microcirculation and molecular and cellular activity after a prolonged post-mortem interval.
Collapse
|
97
|
Umukoro S, Oghwere EE, Ben-Azu B, Owoeye O, Ajayi AM, Omorogbe O, Okubena O. Jobelyn® ameliorates neurological deficits in rats with ischemic stroke through inhibition of release of pro-inflammatory cytokines and NF-κB signaling pathway. PATHOPHYSIOLOGY 2019; 26:77-88. [DOI: 10.1016/j.pathophys.2018.10.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 09/28/2018] [Accepted: 10/16/2018] [Indexed: 12/13/2022] Open
|
98
|
Shigiyama F, Hamanoue M, Kobayashi M, Takamatsu K. Cell-permeable p38 MAP kinase protects adult hippocampal neurons from cell death. Neurosci Lett 2019; 699:115-121. [PMID: 30735722 DOI: 10.1016/j.neulet.2019.02.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 01/18/2019] [Accepted: 02/02/2019] [Indexed: 10/27/2022]
Abstract
p38 mitogen-activated protein (MAP) kinase (p38) is a member of the MAP kinase family. Previous reports using p38 chemical inhibitors have suggested that its activation contributes to hippocampal neuronal cell death rather than cell survival. In this study, we used both a cell-permeable p38 protein containing the HIV protein transduction domain (PTD) and cultured adult hippocampal neurons, which were differentiated from cultured adult hippocampal neural stem/progenitor cells (NPCs), to evaluate the direct function of p38 on adult hippocampal neurons. Our immunocytochemical experiments demonstrated that wild-type cell-permeable p38 protein prevents cell death of adult hippocampal neurons induced by a low glucose condition. Our findings indicate that cell-permeable p38 protein may be useful in preventing the degeneration of higher brain function occurring through hippocampal neuronal cell death, and furthermore, that the maintenance of intracellular p38 levels could be another therapeutic target for neurodegenerative diseases such as Alzheimer's disease (AD).
Collapse
Affiliation(s)
- Fumiko Shigiyama
- Department of Physiology, Toho University Graduate School of Medicine, 143-8540, Tokyo, Japan
| | - Makoto Hamanoue
- Department of Physiology, Toho University Graduate School of Medicine, 143-8540, Tokyo, Japan; Department of Physiology, Toho University School of Medicine, 143-8540, Tokyo, Japan.
| | - Masaaki Kobayashi
- Department of Physiology, Toho University Graduate School of Medicine, 143-8540, Tokyo, Japan; Department of Physiology, Toho University School of Medicine, 143-8540, Tokyo, Japan
| | - Ken Takamatsu
- Department of Physiology, Toho University Graduate School of Medicine, 143-8540, Tokyo, Japan; Department of Physiology, Toho University School of Medicine, 143-8540, Tokyo, Japan
| |
Collapse
|
99
|
Abstract
BACKGROUND The small molecule pifithrin-μ reversibility inhibits the mitochondrial pathway of apoptosis. The neuronal effects of pifithrin-μ applied after cardiac arrest are unknown. We hypothesized that pifithrin-μ reduces neuronal damage in the most vulnerable brain region, the hippocampus, after cardiac arrest. METHODS In two randomized controlled series we administered pifithrin-μ or control in 109 rats resuscitated after 8 or 10 min of cardiac arrest. Neuronal damage was blindly assessed with histology (Fluoro Jade B: FJB, cresyl violet: CV) in the most vulnerable brain region (CA1 segment of hippocampus) and with a series of neurobehavioral tests (Open Field Task, Tape-Removal Test, Morris Water Maze test). Mixed ANOVA was used to combine both series, simple comparisons were done with t tests or Mann-Whitney U test. RESULTS Pifithrin-μ reduced the number of degenerating, FJB-positive neurons by 25% (mixed ANOVA p group = 0.014). This was more prominent after 8 min cardiac arrest (8 min arrest pifithrin-μ 94 ± 47 vs control 128 ± 37; n = 11 each; 10 min arrest pifithrin-μ 78 ± 44, n = 15 vs control 101 ± 31, n = 18; p group* arrest length interaction = 0.622). The reduction of ischemic CV-positive neurons in pifithrin-μ animals was not significant (ANOVA p group = 0.063). No significant group differences were found in neurobehavioral testing. CONCLUSION Temporarily inhibition of apoptosis with pifithrin-μ after cardiac arrest decreases the number of injured neurons in the CA1 segment of hippocampus in a cardiac arrest rat model, without clinical correlate. Further studies should elucidate the role of this neuroprotective agent in different settings and with longer cardiac arrest.
Collapse
|
100
|
Deep Brain Stimulation Rescues Memory and Synaptic Activity in a Rat Model of Global Ischemia. J Neurosci 2019; 39:2430-2440. [PMID: 30696731 DOI: 10.1523/jneurosci.1222-18.2019] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Revised: 01/07/2019] [Accepted: 01/11/2019] [Indexed: 12/18/2022] Open
Abstract
Deep brain stimulation (DBS) is remarkably effective in treating Parkinson's disease and is currently under investigation for the treatment of neuropsychiatric disorders including Alzheimer's disease. Until now, DBS has not been examined for its cognitive benefits in the context of hypoxic-ischemic injuries. Here, we investigated the effect of DBS in a rat model of global ischemia (GI) that mimics the neurological consequences occurring after a cardiac arrest. We show that DBS rescues memory deficits induced by GI and produces changes in synaptic activity in the hippocampus. Novel approaches to improve neurological outcomes after stroke are urgently needed; therefore, the present study highlights a possible role for DBS in the treatment of cognitive impairment associated with ischemia.
Collapse
|