1
|
Gupta P, Gupta RK, Gandhi BS, Singh P. Differential binding of CREB and REST/NRSF to NMDAR1 promoter is associated with the sex-selective cognitive deficit following postnatal PBDE-209 exposure in mice. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2024; 31:38710-38722. [PMID: 37002525 DOI: 10.1007/s11356-023-26107-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 02/20/2023] [Indexed: 06/19/2023]
Abstract
Neonatal exposure to decabromodiphenyl ether (PBDE-209), a widely used flame retardant, affects cognitive performances in the later stage of life in a sex-dependent manner. PBDE-209 interferes with glutamatergic signaling and N-methyl-D-aspartate receptor (NMDAR) subunits with unresolved regulatory mechanisms. This study exposed male and female mice pups through postnatal day (PND) 3-10 to PBDE-209 (oral dose: 0, 6, or 20 mg/kg body weight). The frontal cortex and hippocampus, collected from neonate (PND 11) and young (PND 60) mice, were analyzed for cAMP response element-binding protein (CREB) and RE1-silencing transcription factor/ Neuron-restrictive silencer factor (REST/NRSF) binding to NMDAR1 promoter and expression of NMDAR1 gene by electrophoretic mobility shift assay and semi-quantitative RT-PCR respectively. Behavioral changes were assessed using spontaneous alternation behavior and novel object recognition tests in young mice. In neonates, the binding of CREB was increased, while REST/NRSF was decreased significantly to their cognate NMDAR1 promoter sequences at the high dose of PBDE-209 in both the sexes. This reciprocal pattern of CREB and REST/NRSF interactions correlates with the up-regulation of NMDAR1 expression. Young males followed a similar pattern of CREB and REST/NRSF binding and NMDAR1 expression as in neonates. Surprisingly, young females did not show any alteration when compared to age-matched controls. Also, we found that only young males showed working and recognition memory deficits. These results indicate that early exposure to PBDE-209 interferes with CREB- and REST/NRSF-dependent regulation of the NMDAR1 gene in an acute setting. However, long-term effects persist only in young males that could be associated with cognitive impairment.
Collapse
Affiliation(s)
- Priya Gupta
- Department of Zoology, Women's College, Banaras Hindu University, Varanasi, UP, India
| | - Rajaneesh K Gupta
- Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi, UP, India
| | - Behrose S Gandhi
- Department of Zoology, Women's College, Banaras Hindu University, Varanasi, UP, India
| | - Poonam Singh
- Department of Zoology, Women's College, Banaras Hindu University, Varanasi, UP, India.
| |
Collapse
|
2
|
Ding F, Sun Q, Long C, Rasmussen RN, Peng S, Xu Q, Kang N, Song W, Weikop P, Goldman SA, Nedergaard M. Dysregulation of extracellular potassium distinguishes healthy ageing from neurodegeneration. Brain 2024; 147:1726-1739. [PMID: 38462589 PMCID: PMC11068329 DOI: 10.1093/brain/awae075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 02/15/2024] [Accepted: 02/18/2024] [Indexed: 03/12/2024] Open
Abstract
Progressive neuronal loss is a hallmark feature distinguishing neurodegenerative diseases from normal ageing. However, the underlying mechanisms remain unknown. Extracellular K+ homeostasis is a potential mediator of neuronal injury as K+ elevations increase excitatory activity. The dysregulation of extracellular K+ and potassium channel expressions during neurodegeneration could contribute to this distinction. Here we measured the cortical extracellular K+ concentration ([K+]e) in awake wild-type mice as well as murine models of neurodegeneration using K+-sensitive microelectrodes. Unexpectedly, aged wild-type mice exhibited significantly lower cortical [K+]e than young mice. In contrast, cortical [K+]e was consistently elevated in Alzheimer's disease (APP/PS1), amyotrophic lateral sclerosis (ALS) (SOD1G93A) and Huntington's disease (R6/2) models. Cortical resting [K+]e correlated inversely with neuronal density and the [K+]e buffering rate but correlated positively with the predicted neuronal firing rate. Screening of astrocyte-selective genomic datasets revealed a number of potassium channel genes that were downregulated in these disease models but not in normal ageing. In particular, the inwardly rectifying potassium channel Kcnj10 was downregulated in ALS and Huntington's disease models but not in normal ageing, while Fxyd1 and Slc1a3, each of which acts as a negative regulator of potassium uptake, were each upregulated by astrocytes in both Alzheimer's disease and ALS models. Chronic elevation of [K+]e in response to changes in gene expression and the attendant neuronal hyperexcitability may drive the neuronal loss characteristic of these neurodegenerative diseases. These observations suggest that the dysregulation of extracellular K+ homeostasis in a number of neurodegenerative diseases could be due to aberrant astrocytic K+ buffering and as such, highlight a fundamental role for glial dysfunction in neurodegeneration.
Collapse
Affiliation(s)
- Fengfei Ding
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY 14642, USA
- Department of Pharmacology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Qian Sun
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY 14642, USA
- Department of Pharmacology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Carter Long
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Rune Nguyen Rasmussen
- Center for Basic and Translational Neuroscience, Faculty of Health and Medical Sciences, Neurology Department, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Sisi Peng
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Qiwu Xu
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Ning Kang
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Wei Song
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Pia Weikop
- Center for Basic and Translational Neuroscience, Faculty of Health and Medical Sciences, Neurology Department, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Steven A Goldman
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY 14642, USA
- Center for Basic and Translational Neuroscience, Faculty of Health and Medical Sciences, Neurology Department, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Maiken Nedergaard
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY 14642, USA
- Center for Basic and Translational Neuroscience, Faculty of Health and Medical Sciences, Neurology Department, University of Copenhagen, 2200 Copenhagen, Denmark
| |
Collapse
|
3
|
Lapshina KV, Ekimova IV. Aquaporin-4 and Parkinson's Disease. Int J Mol Sci 2024; 25:1672. [PMID: 38338949 PMCID: PMC10855351 DOI: 10.3390/ijms25031672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 01/15/2024] [Accepted: 01/26/2024] [Indexed: 02/12/2024] Open
Abstract
The water-selective channel aquaporin-4 (AQP4) is implicated in water homeostasis and the functioning of the glymphatic system, which eliminates various metabolites from the brain tissue, including amyloidogenic proteins. Misfolding of the α-synuclein protein and its post-translational modifications play a crucial role in the development of Parkinson's disease (PD) and other synucleopathies, leading to the formation of cytotoxic oligomers and aggregates that cause neurodegeneration. Human and animal studies have shown an interconnection between AQP4 dysfunction and α-synuclein accumulation; however, the specific role of AQP4 in these mechanisms remains unclear. This review summarizes the current knowledge on the role of AQP4 dysfunction in the progression of α-synuclein pathology, considering the possible effects of AQP4 dysregulation on brain molecular mechanisms that can impact α-synuclein modification, accumulation and aggregation. It also highlights future directions that can help study the role of AQP4 in the functioning of the protective mechanisms of the brain during the development of PD and other neurodegenerative diseases.
Collapse
Affiliation(s)
- Ksenia V. Lapshina
- Laboratory of Comparative Thermophysiology, Sechenov Institute of Evolutionary Physiology and Biochemistry of RAS, 194223 Saint Petersburg, Russia;
| | | |
Collapse
|
4
|
Alkhalifa AE, Al-Ghraiybah NF, Odum J, Shunnarah JG, Austin N, Kaddoumi A. Blood-Brain Barrier Breakdown in Alzheimer's Disease: Mechanisms and Targeted Strategies. Int J Mol Sci 2023; 24:16288. [PMID: 38003477 PMCID: PMC10671257 DOI: 10.3390/ijms242216288] [Citation(s) in RCA: 52] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 11/07/2023] [Accepted: 11/09/2023] [Indexed: 11/26/2023] Open
Abstract
The blood-brain barrier (BBB) is a unique and selective feature of the central nervous system's vasculature. BBB dysfunction has been observed as an early sign of Alzheimer's Disease (AD) before the onset of dementia or neurodegeneration. The intricate relationship between the BBB and the pathogenesis of AD, especially in the context of neurovascular coupling and the overlap of pathophysiology in neurodegenerative and cerebrovascular diseases, underscores the urgency to understand the BBB's role more deeply. Preserving or restoring the BBB function emerges as a potentially promising strategy for mitigating the progression and severity of AD. Molecular and genetic changes, such as the isoform ε4 of apolipoprotein E (ApoEε4), a significant genetic risk factor and a promoter of the BBB dysfunction, have been shown to mediate the BBB disruption. Additionally, receptors and transporters like the low-density lipoprotein receptor-related protein 1 (LRP1), P-glycoprotein (P-gp), and the receptor for advanced glycation end products (RAGEs) have been implicated in AD's pathogenesis. In this comprehensive review, we endeavor to shed light on the intricate pathogenic and therapeutic connections between AD and the BBB. We also delve into the latest developments and pioneering strategies targeting the BBB for therapeutic interventions, addressing its potential as a barrier and a carrier. By providing an integrative perspective, we anticipate paving the way for future research and treatments focused on exploiting the BBB's role in AD pathogenesis and therapy.
Collapse
Affiliation(s)
| | | | | | | | | | - Amal Kaddoumi
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, 720 S. Donahue Dr., Auburn, AL 36849, USA; (A.E.A.); (N.F.A.-G.); (J.O.); (J.G.S.); (N.A.)
| |
Collapse
|
5
|
Oudart M, Avila-Gutierrez K, Moch C, Dossi E, Milior G, Boulay AC, Gaudey M, Moulard J, Lombard B, Loew D, Bemelmans AP, Rouach N, Chapat C, Cohen-Salmon M. The ribosome-associated protein RACK1 represses Kir4.1 translation in astrocytes and influences neuronal activity. Cell Rep 2023; 42:112456. [PMID: 37126448 DOI: 10.1016/j.celrep.2023.112456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 02/10/2023] [Accepted: 04/16/2023] [Indexed: 05/02/2023] Open
Abstract
The regulation of translation in astrocytes, the main glial cells in the brain, remains poorly characterized. We developed a high-throughput proteomics screen for polysome-associated proteins in astrocytes and focused on ribosomal protein receptor of activated protein C kinase 1 (RACK1), a critical factor in translational regulation. In astrocyte somata and perisynaptic astrocytic processes (PAPs), RACK1 preferentially binds to a number of mRNAs, including Kcnj10, encoding the inward-rectifying potassium (K+) channel Kir4.1. By developing an astrocyte-specific, conditional RACK1 knockout mouse model, we show that RACK1 represses production of Kir4.1 in hippocampal astrocytes and PAPs. Upregulation of Kir4.1 in the absence of RACK1 increases astrocytic Kir4.1-mediated K+ currents and volume. It also modifies neuronal activity attenuating burst frequency and duration. Reporter-based assays reveal that RACK1 controls Kcnj10 translation through the transcript's 5' untranslated region. Hence, translational regulation by RACK1 in astrocytes represses Kir4.1 expression and influences neuronal activity.
Collapse
Affiliation(s)
- Marc Oudart
- Center for Interdisciplinary Research in Biology, College de France, CNRS, INSERM, Université PSL, Labex Memolife, Paris, France
| | - Katia Avila-Gutierrez
- Center for Interdisciplinary Research in Biology, College de France, CNRS, INSERM, Université PSL, Labex Memolife, Paris, France
| | - Clara Moch
- Laboratoire de Biochimie, Ecole Polytechnique, CNRS, Université Paris-Saclay, Palaiseau, France
| | - Elena Dossi
- Center for Interdisciplinary Research in Biology, College de France, CNRS, INSERM, Université PSL, Labex Memolife, Paris, France
| | - Giampaolo Milior
- Center for Interdisciplinary Research in Biology, College de France, CNRS, INSERM, Université PSL, Labex Memolife, Paris, France
| | - Anne-Cécile Boulay
- Center for Interdisciplinary Research in Biology, College de France, CNRS, INSERM, Université PSL, Labex Memolife, Paris, France
| | - Mathis Gaudey
- Center for Interdisciplinary Research in Biology, College de France, CNRS, INSERM, Université PSL, Labex Memolife, Paris, France
| | - Julien Moulard
- Center for Interdisciplinary Research in Biology, College de France, CNRS, INSERM, Université PSL, Labex Memolife, Paris, France
| | - Bérangère Lombard
- CurieCoreTech Spectrométrie de Masse Protéomique, Institut Curie, University PSL, Paris, France
| | - Damarys Loew
- CurieCoreTech Spectrométrie de Masse Protéomique, Institut Curie, University PSL, Paris, France
| | - Alexis-Pierre Bemelmans
- CEA, Institut de Biologie François Jacob, Molecular Imaging Research Center (MIRCen), CNRS, Université Paris-Sud, Université Paris-Saclay, Fontenay-aux-Roses, France
| | - Nathalie Rouach
- Center for Interdisciplinary Research in Biology, College de France, CNRS, INSERM, Université PSL, Labex Memolife, Paris, France
| | - Clément Chapat
- Laboratoire de Biochimie, Ecole Polytechnique, CNRS, Université Paris-Saclay, Palaiseau, France
| | - Martine Cohen-Salmon
- Center for Interdisciplinary Research in Biology, College de France, CNRS, INSERM, Université PSL, Labex Memolife, Paris, France.
| |
Collapse
|
6
|
Obenaus A, Rodriguez-Grande B, Lee JB, Dubois CJ, Fournier ML, Cador M, Caille S, Badaut J. A single mild juvenile TBI in male mice leads to regional brain tissue abnormalities at 12 months of age that correlate with cognitive impairment at the middle age. Acta Neuropathol Commun 2023; 11:32. [PMID: 36859364 PMCID: PMC9976423 DOI: 10.1186/s40478-023-01515-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Accepted: 01/12/2023] [Indexed: 03/03/2023] Open
Abstract
Traumatic brain injury (TBI) has the highest incidence amongst the pediatric population and its mild severity represents the most frequent cases. Moderate and severe injuries as well as repetitive mild TBI result in lasting morbidity. However, whether a single mild TBI sustained during childhood can produce long-lasting modifications within the brain is still debated. We aimed to assess the consequences of a single juvenile mild TBI (jmTBI) at 12 months post-injury in a mouse model. Non-invasive diffusion tensor imaging (DTI) revealed significant microstructural alterations in the hippocampus and the in the substantia innominata/nucleus basalis (SI/NB), structures known to be involved in spatial learning and memory. DTI changes paralled neuronal loss, increased astrocytic AQP4 and microglial activation in the hippocampus. In contrast, decreased astrocytic AQP4 expression and microglia activation were observed in SI/NB. Spatial learning and memory were impaired and correlated with alterations in DTI-derived derived fractional ansiotropy (FA) and axial diffusivity (AD). This study found that a single juvenile mild TBI leads to significant region-specific DTI microstructural alterations, distant from the site of impact, that correlated with cognitive discriminative novel object testing and spatial memory impairments at 12 months after a single concussive injury. Our findings suggest that exposure to jmTBI leads to a chronic abnormality, which confirms the need for continued monitoring of symptoms and the development of long-term treatment strategies to intervene in children with concussions.
Collapse
Affiliation(s)
- Andre Obenaus
- Department of Pediatrics, University of California, Irvine, CA, USA
- Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, USA
| | | | - Jeong Bin Lee
- Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, USA
| | - Christophe J Dubois
- CNRS UMR 5536 RMSB, University of Bordeaux, 146 Rue Léo Saignat, 33076, Bordeaux Cedex, France
| | | | - Martine Cador
- CNRS, EPHE, INCIA UMR5287, University of Bordeaux, F33000, Bordeaux, France
| | - Stéphanie Caille
- CNRS, EPHE, INCIA UMR5287, University of Bordeaux, F33000, Bordeaux, France
| | - Jerome Badaut
- Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, USA.
- CNRS, EPHE, INCIA UMR5287, University of Bordeaux, F33000, Bordeaux, France.
- CNRS UMR 5536 RMSB, University of Bordeaux, 146 Rue Léo Saignat, 33076, Bordeaux Cedex, France.
| |
Collapse
|
7
|
Uddin MS, Lim LW. Glial cells in Alzheimer's disease: From neuropathological changes to therapeutic implications. Ageing Res Rev 2022; 78:101622. [PMID: 35427810 DOI: 10.1016/j.arr.2022.101622] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 04/04/2022] [Accepted: 04/08/2022] [Indexed: 12/20/2022]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder that usually develops slowly and progressively worsens over time. Although there has been increasing research interest in AD, its pathogenesis is still not well understood. Although most studies primarily focus on neurons, recent research findings suggest that glial cells (especially microglia and astrocytes) are associated with AD pathogenesis and might provide various possible therapeutic targets. Growing evidence suggests that microglia can provide protection against AD pathogenesis, as microglia with weakened functions and impaired responses to Aβ proteins are linked with elevated AD risk. Interestingly, numerous findings also suggest that microglial activation can be detrimental to neurons. Indeed, microglia can induce synapse loss via the engulfment of synapses, possibly through a complement-dependent process. Furthermore, they can worsen tau pathology and release inflammatory factors that cause neuronal damage directly or through the activation of neurotoxic astrocytes. Astrocytes play a significant role in various cerebral activities. Their impairment can mediate neurodegeneration and ultimately the retraction of synapses, resulting in AD-related cognitive deficits. Deposition of Aβ can result in astrocyte reactivity, which can further lead to neurotoxic effects and elevated secretion of inflammatory mediators and cytokines. Moreover, glial-induced inflammation in AD can exert both beneficial and harmful effects. Understanding the activities of astrocytes and microglia in the regulation of AD pathogenesis would facilitate the development of novel therapies. In this article, we address the implications of microglia and astrocytes in AD pathogenesis. We also discuss the mechanisms of therapeutic agents that exhibit anti-inflammatory effects against AD.
Collapse
Affiliation(s)
- Md Sahab Uddin
- Neuromodulation Laboratory, School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Lee Wei Lim
- Neuromodulation Laboratory, School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China.
| |
Collapse
|
8
|
Ford JN, Zhang Q, Sweeney EM, Merkler AE, de Leon MJ, Gupta A, Nguyen TD, Ivanidze J. Quantitative Water Permeability Mapping of Blood-Brain-Barrier Dysfunction in Aging. Front Aging Neurosci 2022; 14:867452. [PMID: 35462701 PMCID: PMC9024318 DOI: 10.3389/fnagi.2022.867452] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Accepted: 03/10/2022] [Indexed: 11/13/2022] Open
Abstract
Blood-brain-barrier (BBB) dysfunction is a hallmark of aging and aging-related disorders, including cerebral small vessel disease and Alzheimer's disease. An emerging biomarker of BBB dysfunction is BBB water exchange rate (kW) as measured by diffusion-weighted arterial spin labeling (DW-ASL) MRI. We developed an improved DW-ASL sequence for Quantitative Permeability Mapping and evaluated whole brain and region-specific kW in a cohort of 30 adults without dementia across the age spectrum. In this cross-sectional study, we found higher kW values in the cerebral cortex (mean = 81.51 min-1, SD = 15.54) compared to cerebral white matter (mean = 75.19 min-1, SD = 13.85) (p < 0.0001). We found a similar relationship for cerebral blood flow (CBF), concordant with previously published studies. Multiple linear regression analysis with kW as an outcome showed that age was statistically significant in the cerebral cortex (p = 0.013), cerebral white matter (p = 0.033), hippocampi (p = 0.043), orbitofrontal cortices (p = 0.042), and precunei cortices (p = 0.009), after adjusting for sex and number of vascular risk factors. With CBF as an outcome, age was statistically significant only in the cerebral cortex (p = 0.026) and precunei cortices (p = 0.020). We further found moderate negative correlations between white matter hyperintensity (WMH) kW and WMH volume (r = -0.51, p = 0.02), and normal-appearing white matter (NAWM) and WMH volume (r = -0.44, p = 0.05). This work illuminates the relationship between BBB water exchange and aging and may serve as the basis for BBB-targeted therapies for aging-related brain disorders.
Collapse
Affiliation(s)
- Jeremy N. Ford
- Department of Radiology, Massachusetts General Hospital, Boston, MA, United States,Department of Radiology, Weill Cornell Medicine, New York, NY, United States
| | - Qihao Zhang
- Department of Radiology, Weill Cornell Medicine, New York, NY, United States
| | - Elizabeth M. Sweeney
- Department of Biostatistics, University of Pennsylvania, Philadelphia, PA, United States
| | | | - Mony J. de Leon
- Department of Radiology, Weill Cornell Medicine, New York, NY, United States
| | - Ajay Gupta
- Department of Radiology, Weill Cornell Medicine, New York, NY, United States
| | - Thanh D. Nguyen
- Department of Radiology, Weill Cornell Medicine, New York, NY, United States
| | - Jana Ivanidze
- Department of Radiology, Weill Cornell Medicine, New York, NY, United States,*Correspondence: Jana Ivanidze,
| |
Collapse
|
9
|
Price BR, Johnson LA, Norris CM. Reactive astrocytes: The nexus of pathological and clinical hallmarks of Alzheimer's disease. Ageing Res Rev 2021; 68:101335. [PMID: 33812051 PMCID: PMC8168445 DOI: 10.1016/j.arr.2021.101335] [Citation(s) in RCA: 79] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 01/21/2021] [Accepted: 03/20/2021] [Indexed: 02/06/2023]
Abstract
Astrocyte reactivity is a hallmark of neuroinflammation that arises with Alzheimer’s disease (AD) and nearly every other neurodegenerative condition. While astrocytes certainly contribute to classic inflammatory processes (e.g. cytokine release, waste clearance, and tissue repair), newly emerging technologies for measuring and targeting cell specific activities in the brain have uncovered essential roles for astrocytes in synapse function, brain metabolism, neurovascular coupling, and sleep/wake patterns. In this review, we use a holistic approach to incorporate, and expand upon, classic neuroinflammatory concepts to consider how astrocyte dysfunction/reactivity modulates multiple pathological and clinical hallmarks of AD. Our ever-evolving understanding of astrocyte signaling in neurodegeneration is not only revealing new drug targets and treatments for dementia but is suggesting we reimagine AD pathophysiological mechanisms.
Collapse
Affiliation(s)
- Brittani R Price
- Department of Neuroscience, Tufts University School of Medicine, 136 Harrison Ave., Boston, MA, 02111, USA
| | - Lance A Johnson
- Sanders-Brown Center on Aging, University of Kentucky, 800 S. Limestone St., Lexington, KY, 40356, USA; Department of Physiology, University of Kentucky, College of Medicine, UK Medical Center MN 150, Lexington, KY, 40536, USA
| | - Christopher M Norris
- Sanders-Brown Center on Aging, University of Kentucky, 800 S. Limestone St., Lexington, KY, 40356, USA; Department of Pharmacology and Nutritional Sciences, University of Kentucky, College of Medicine, UK Medical Center MN 150, Lexington, KY, 40536, USA.
| |
Collapse
|
10
|
Blood-brain barrier dysfunction as a potential therapeutic target for neurodegenerative disorders. Arch Pharm Res 2021; 44:487-498. [PMID: 34028650 DOI: 10.1007/s12272-021-01332-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Accepted: 05/10/2021] [Indexed: 12/12/2022]
Abstract
The blood-brain barrier (BBB) is composed of specific tight junction proteins and transporters expressed on the lining of endothelial cells of the vasculature in the brain. The structural and functional integrity of the BBB is one of the most critical factors for maintaining brain homeostasis and is mainly regulated by complex interactions between various cell types, such as endothelial cells, pericytes, and astrocytes, which are shaped by their differential responses to changes in microenvironments. Alterations in these cellular components have been implicated in neurodegenerative disorders. Although it has long been considered that BBB dysfunction is a mere ramification of pathological phenomena, emerging evidence supports its critical role in the pathogenesis of various disorders. In epilepsy, heightened BBB permeability has been found to be associated with increased occurrence of spontaneous seizures. Additionally, exaggerated inflammatory responses significantly correlate with increased BBB permeability during healthy aging. Furthermore, it has been previously reported that BBB disruption can be an early marker for predicting cognitive impairment in the progression of Alzheimer's disease. We herein review a potential role of the major cellular components of the BBB, with a focus on the contribution of BBB disruption, in neurodegenerative disease progression.
Collapse
|
11
|
Dai D, He L, Chen Y, Zhang C. Astrocyte responses to nanomaterials: Functional changes, pathological changes and potential applications. Acta Biomater 2021; 122:66-81. [PMID: 33326883 DOI: 10.1016/j.actbio.2020.12.013] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 11/30/2020] [Accepted: 12/08/2020] [Indexed: 12/15/2022]
Abstract
Astrocytes are responsible for regulating and optimizing the functional environment of neurons in the brain and can reduce the adverse impacts of external factors by protecting neurons. However, excessive astrocyte activation upon stimulation may alter their initial protective effect and actually lead to aggravation of injury. Similar to the dual effects of astrocytes in the response to injury within the central nervous system (CNS), nanomaterials (NMs) can have either toxic or beneficial effects on astrocytes, serving to promote injury or inhibit tumors. As the important physiological functions of astrocytes have been gradually revealed, the effects of NMs on astrocytes and the underlying mechanisms have become a new frontier in nanomedicine and neuroscience. This review summarizes the in vitro and in vivo findings regarding the effects of various NMs on astrocytes, focusing on functional alterations and pathological processes in astrocytes, as well as the possible underlying mechanisms. We also emphasize the importance of co-culture models in studying the interaction between NMs and cells of the CNS. Finally, we discuss NMs that have shown promise for application in astrocyte-related diseases and propose some challenges and suggestions for further investigations, with the aim of providing guidance for the widespread application of NMs in the CNS.
Collapse
Affiliation(s)
- Danni Dai
- Stomatological Hospital, Southern Medical University, Guangzhou 510280, China
| | - Longwen He
- Stomatological Hospital, Southern Medical University, Guangzhou 510280, China
| | - Yuming Chen
- Stomatological Hospital, Southern Medical University, Guangzhou 510280, China
| | - Chao Zhang
- Stomatological Hospital, Southern Medical University, Guangzhou 510280, China.
| |
Collapse
|
12
|
Silva I, Silva J, Ferreira R, Trigo D. Glymphatic system, AQP4, and their implications in Alzheimer's disease. Neurol Res Pract 2021; 3:5. [PMID: 33499944 PMCID: PMC7816372 DOI: 10.1186/s42466-021-00102-7] [Citation(s) in RCA: 108] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 01/11/2021] [Indexed: 12/11/2022] Open
Abstract
Lacking conventional lymphatic system, the central nervous system requires alternative clearance systems, such as the glymphatic system, which promotes clearance of interstitial solutes. Aquaporin-4 water channels (AQP4) are an integral part of this system and related to neuropathologies, such as Alzheimer's disease (AD). The clearance of Alzheimer's associated proteins amyloid β and tau is diminished by glymphatic system impairment, due to lack of AQP4. Even though AQP4 mislocalisation (which affects its activity) is a phenotype of AD, it remains a controversial topic, as it is still unclear if it is a phenotype-promoting factor or a consequence of this pathology. This review provides important and updated knowledge about glymphatic system, AQP4 itself, and their link with Alzheimer's disease. Finally, AQP4 as a therapeutic target is proposed to ameliorate Alzheimer's Disease and other neuropathologies AQP4-related.
Collapse
Affiliation(s)
- Inês Silva
- Medical Sciences Department, University of Aveiro, 3810-193, Aveiro, Portugal
| | - Jéssica Silva
- Medical Sciences Department, University of Aveiro, 3810-193, Aveiro, Portugal
| | - Rita Ferreira
- Medical Sciences Department, University of Aveiro, 3810-193, Aveiro, Portugal
| | - Diogo Trigo
- Medical Sciences Department, University of Aveiro, 3810-193, Aveiro, Portugal.
- Neuroscience and Signalling Laboratory, Institute of Biomedicine (iBiMED), Department of Medical Sciences, University of Aveiro, 3810-193, Aveiro, Portugal.
| |
Collapse
|
13
|
Pro-Inflammatory Role of AQP4 in Mice Subjected to Intrastriatal Injections of the Parkinsonogenic Toxin MPP. Cells 2020; 9:cells9112418. [PMID: 33167342 PMCID: PMC7694382 DOI: 10.3390/cells9112418] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 11/01/2020] [Accepted: 11/02/2020] [Indexed: 12/28/2022] Open
Abstract
Aquaporin-4 (AQP4) is critically involved in brain water and volume homeostasis and has been implicated in a wide range of pathological conditions. Notably, evidence has been accrued to suggest that AQP4 plays a proinflammatory role by promoting release of astrocytic cytokines that activate microglia and other astrocytes. Neuroinflammation is a hallmark of Parkinson’s disease (PD), and we have previously shown that astrocytes in substantia nigra (SN) are enriched in AQP4 relative to cortical astrocytes, and that their complement of AQP4 is further increased following treatment with the parkinsonogenic toxin MPTP (1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine). Here, we investigated the effect of Aqp4 deletion on microglial activation in mice subjected to unilateral intrastriatal injection of 1-methyl-4-phenylpyridinium (MPP+, the toxic metabolite of MPTP). Our results show that MPP+ injections lead to a pronounced increase in the expression level of microglial activating genes in the ventral mesencephalon of wild type (WT) mice, but not Aqp4−/− mice. We also show, in WT mice, that MPP+ injections cause an upregulation of nigral AQP4 and swelling of astrocytic endfeet. These findings are consistent with the idea that AQP4 plays a pro-inflammatory role in Parkinson’s disease, secondary to the dysregulation of astrocytic volume homeostasis.
Collapse
|
14
|
Ohene Y, Harrison IF, Evans PG, Thomas DL, Lythgoe MF, Wells JA. Increased blood-brain barrier permeability to water in the aging brain detected using noninvasive multi-TE ASL MRI. Magn Reson Med 2020; 85:326-333. [PMID: 32910547 PMCID: PMC8432141 DOI: 10.1002/mrm.28496] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 07/27/2020] [Accepted: 08/03/2020] [Indexed: 11/25/2022]
Abstract
Purpose A fundamental goal in the drive to understand and find better treatments for dementia is the identification of the factors that render the aging brain vulnerable to neurodegenerative disease. Recent evidence indicates the integrity of the blood–brain barrier (BBB) to be an important component of functional failure underlying age‐related cognitive decline. Practical and sensitive measurement is necessary, therefore, to support diagnostic and therapeutic strategies targeted at maintaining BBB integrity in aging patients. Here, we investigated changes in BBB permeability to endogenous blood water in the aging brain. Methods A multiple‐echo‐time arterial spin‐labeling MRI technique, implemented on a 9.4T Bruker imaging system, was applied to 7‐ and 27‐month‐old mice to measure changes in water permeability across the BBB with aging. Results We observed that BBB water permeability was 32% faster in aged mice. This occurred along with a 2.1‐fold increase in mRNA expression of aquaporin‐4 water channels and a 7.1‐fold decrease in mRNA expression of α‐syntrophin protein, which anchors aquaporin‐4 to the BBB. Conclusion Age‐related changes to water permeability across the BBB can be captured using noninvasive noncontrast MRI techniques. Click here for author‐reader discussions
Collapse
Affiliation(s)
- Yolanda Ohene
- UCL Centre for Advanced Biomedical Imaging, Division of Medicine, University College London, London, United Kingdom
| | - Ian F Harrison
- UCL Centre for Advanced Biomedical Imaging, Division of Medicine, University College London, London, United Kingdom
| | - Phoebe G Evans
- UCL Centre for Advanced Biomedical Imaging, Division of Medicine, University College London, London, United Kingdom
| | - David L Thomas
- Neuroradiological Academic Unit, Department of Brain Repair and Rehabilitation, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom.,Dementia Research Centre, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom.,Wellcome Centre for Human Neuroimaging, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom
| | - Mark F Lythgoe
- UCL Centre for Advanced Biomedical Imaging, Division of Medicine, University College London, London, United Kingdom
| | - Jack A Wells
- UCL Centre for Advanced Biomedical Imaging, Division of Medicine, University College London, London, United Kingdom
| |
Collapse
|
15
|
Valenza M, Facchinetti R, Steardo L, Scuderi C. Altered Waste Disposal System in Aging and Alzheimer's Disease: Focus on Astrocytic Aquaporin-4. Front Pharmacol 2020; 10:1656. [PMID: 32063858 PMCID: PMC7000422 DOI: 10.3389/fphar.2019.01656] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Accepted: 12/17/2019] [Indexed: 12/15/2022] Open
Abstract
Among the diverse cell types included in the general population named glia, astrocytes emerge as being the focus of a growing body of research aimed at characterizing their heterogeneous and complex functions. Alterations of both their morphology and activities have been linked to a variety of neurological diseases. One crucial physiological need satisfied by astrocytes is the cleansing of the cerebral tissue from waste molecules. Several data demonstrate that aquaporin-4 (AQP-4), a protein expressed by astrocytes, is crucially important for facilitating the removal of waste products from the brain. Aquaporins are water channels found in all district of the human organism and the most abundant isoform in the brain is AQP-4. This protein is involved in a myriad of astrocytic activities, including calcium signal transduction, potassium buffering, synaptic plasticity, astrocyte migration, glial scar formation and neuroinflammation. The highest density of AQP-4 is found at the astrocytic domains closest to blood vessels, the endfeet that envelop brain vessels, with low to zero expression in other astrocytic membrane regions. Increased AQP-4 expression and loss of polarization have recently been documented in altered physiological conditions. Here we review the latest findings related to aging and Alzheimer’s disease (AD) on this topic, as well as the available knowledge on pharmacological tools to target AQP-4.
Collapse
Affiliation(s)
- Marta Valenza
- Department Physiology and Pharmacology "V. Erspamer", Sapienza University of Rome, Rome, Italy.,Epitech Group SpA, Saccolongo, Italy
| | - Roberta Facchinetti
- Department Physiology and Pharmacology "V. Erspamer", Sapienza University of Rome, Rome, Italy
| | - Luca Steardo
- Università Telematica Giustino Fortunato, Benevento, Italy
| | - Caterina Scuderi
- Department Physiology and Pharmacology "V. Erspamer", Sapienza University of Rome, Rome, Italy
| |
Collapse
|
16
|
Astrocyte Support for Oligodendrocyte Differentiation can be Conveyed via Extracellular Vesicles but Diminishes with Age. Sci Rep 2020; 10:828. [PMID: 31964978 PMCID: PMC6972737 DOI: 10.1038/s41598-020-57663-x] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 01/06/2020] [Indexed: 01/06/2023] Open
Abstract
The aging brain is associated with significant changes in physiology that alter the tissue microenvironment of the central nervous system (CNS). In the aged CNS, increased demyelination has been associated with astrocyte hypertrophy and aging has been implicated as a basis for these pathological changes. Aging tissues accumulate chronic cellular stress, which can lead to the development of a pro-inflammatory phenotype that can be associated with cellular senescence. Herein, we provide evidence that astrocytes aged in culture develop a spontaneous pro-inflammatory and senescence-like phenotype. We found that extracellular vesicles (EVs) from young astrocyte were sufficient to convey support for oligodendrocyte differentiation while this support was lost by EVs from aged astrocytes. Importantly, the negative influence of culture age on astrocytes, and their cognate EVs, could be countered by treatment with rapamycin. Comparative proteomic analysis of EVs from young and aged astrocytes revealed peptide repertoires unique to each age. Taken together, these findings provide new information on the contribution of EVs as potent mediators by which astrocytes can extert changing influence in either the disease or aged brain.
Collapse
|
17
|
Bronzuoli MR, Facchinetti R, Valenza M, Cassano T, Steardo L, Scuderi C. Astrocyte Function Is Affected by Aging and Not Alzheimer's Disease: A Preliminary Investigation in Hippocampi of 3xTg-AD Mice. Front Pharmacol 2019; 10:644. [PMID: 31244658 PMCID: PMC6562169 DOI: 10.3389/fphar.2019.00644] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Accepted: 05/17/2019] [Indexed: 01/12/2023] Open
Abstract
Old age is a risk factor for Alzheimer's disease (AD), which is characterized by hippocampal impairment together with substantial changes in glial cell functions. Are these alterations due to the disease progression or are they a consequence of aging? To start addressing this issue, we studied the expression of specific astrocytic and microglial structural and functional proteins in a validated transgenic model of AD (3×Tg-AD). These mice develop both amyloid plaques and neurofibrillary tangles, and initial signs of the AD-like pathology have been documented as early as three months of age. We compared male 3×Tg-AD mice at 6 and 12 months of age with their wild-type age-matched counterparts. We also investigated neurons by examining the expression of both the microtubule-associated protein 2 (MAP2), a neuronal structural protein, and the brain-derived neurotrophic factor (BDNF). The latter is indeed a crucial indicator for synaptic plasticity and neurogenesis/neurodegeneration. Our results show that astrocytes are more susceptible to aging than microglia, regardless of mouse genotype. Moreover, we discovered significant age-dependent alterations in the expression of proteins responsible for astrocyte-astrocyte and astrocyte-neuron communication, as well as a significant age-dependent decline in BDNF expression. Our data promote further research on the unexplored role of astroglia in both physiological and pathological aging.
Collapse
Affiliation(s)
- Maria Rosanna Bronzuoli
- Department of Physiology and Pharmacology "V. Erspamer," Sapienza University of Rome, Rome, Italy
| | - Roberta Facchinetti
- Department of Physiology and Pharmacology "V. Erspamer," Sapienza University of Rome, Rome, Italy
| | - Marta Valenza
- Department of Physiology and Pharmacology "V. Erspamer," Sapienza University of Rome, Rome, Italy.,Epitech Group SpA, Saccolongo, Italy
| | - Tommaso Cassano
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Luca Steardo
- Department of Physiology and Pharmacology "V. Erspamer," Sapienza University of Rome, Rome, Italy
| | - Caterina Scuderi
- Department of Physiology and Pharmacology "V. Erspamer," Sapienza University of Rome, Rome, Italy
| |
Collapse
|
18
|
Susceptibility of the cerebral cortex to spreading depolarization in neurological disease states: The impact of aging. Neurochem Int 2018; 127:125-136. [PMID: 30336178 DOI: 10.1016/j.neuint.2018.10.010] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Revised: 10/10/2018] [Accepted: 10/13/2018] [Indexed: 12/17/2022]
Abstract
Secondary injury following acute brain insults significantly contributes to poorer neurological outcome. The spontaneous, recurrent occurrence of spreading depolarization events (SD) has been recognized as a potent secondary injury mechanism in subarachnoid hemorrhage, malignant ischemic stroke and traumatic brain injury. In addition, SD is the underlying mechanism of the aura symptoms of migraineurs. The susceptibility of the nervous tissue to SD is subject to the metabolic status of the tissue, the ionic composition of the extracellular space, and the functional status of ion pumps, voltage-gated and other cation channels, glutamate receptors and excitatory amino acid transporters. All these mechanisms tune the excitability of the nervous tissue. Aging has also been found to alter SD susceptibility, which appears to be highest at young adulthood, and decline over the aging process. The lower susceptibility of the cerebral gray matter to SD in the old brain may be caused by the age-related impairment of mechanisms implicated in ion translocations between the intra- and extracellular compartments, glutamate signaling and surplus potassium and glutamate clearance. Even though the aging nervous tissue is thus less able to sustain SD, the consequences of SD recurrence in the old brain have proven to be graver, possibly leading to accelerated lesion maturation. Taken that recurrent SDs may pose an increased burden in the aging injured brain, the benefit of therapeutic approaches to restrict SD generation and propagation may be particularly relevant for elderly patients.
Collapse
|
19
|
Hayoz S, Pettis J, Bradley V, Segal SS, Jackson WF. Increased amplitude of inward rectifier K + currents with advanced age in smooth muscle cells of murine superior epigastric arteries. Am J Physiol Heart Circ Physiol 2017; 312:H1203-H1214. [PMID: 28432059 PMCID: PMC6146378 DOI: 10.1152/ajpheart.00679.2016] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Revised: 04/18/2017] [Accepted: 04/18/2017] [Indexed: 01/15/2023]
Abstract
Inward rectifier K+ channels (KIR) may contribute to skeletal muscle blood flow regulation and adapt to advanced age. Using mouse abdominal wall superior epigastric arteries (SEAs) from either young (3-6 mo) or old (24-26 mo) male C57BL/6 mice, we investigated whether SEA smooth muscle cells (SMCs) express functional KIR channels and how aging may affect KIR function. Freshly dissected SEAs were either enzymatically dissociated to isolate SMCs for electrophysiological recording (perforated patch) and mRNA expression or used intact for pressure myography. With 5 mM extracellular K+ concentration ([K+]o), exposure of SMCs to the KIR blocker Ba2+ (100 μM) had no significant effect (P > 0.05) on whole cell currents elicited by membrane potentials spanning -120 to -30 mV. Raising [K+]o to 15 mM activated Ba2+-sensitive KIR currents between -120 and -30 mV, which were greater in SMCs from old mice than in SMCs from young mice (P < 0.05). Pressure myography of SEAs revealed that while aging decreased maximum vessel diameter by ~8% (P < 0.05), it had no significant effect on resting diameter, myogenic tone, dilation to 15 mM [K+]o, Ba2+-induced constriction in 5 mM [K+]o, or constriction induced by 15 mM [K+]o in the presence of Ba2+ (P > 0.05). Quantitative RT-PCR revealed SMC expression of KIR2.1 and KIR2.2 mRNA that was not affected by age. Barium-induced constriction of SEAs from young and old mice suggests an integral role for KIR in regulating resting membrane potential and vasomotor tone. Increased functional expression of KIR channels during advanced age may compensate for other age-related changes in SEA function.NEW & NOTEWORTHY Ion channels are integral to blood flow regulation. We found greater functional expression of inward rectifying K+ channels in smooth muscle cells of resistance arteries of mouse skeletal muscle with advanced age. This adaptation to aging may contribute to the maintenance of vasomotor tone and blood flow regulation during exercise.
Collapse
Affiliation(s)
- Sebastien Hayoz
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan
| | - Jessica Pettis
- College of Veterinary Medicine, Michigan State University, East Lansing, Michigan
| | - Vanessa Bradley
- College of Veterinary Medicine, Michigan State University, East Lansing, Michigan
| | - Steven S Segal
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri; and
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri
| | - William F Jackson
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan;
| |
Collapse
|
20
|
Onoda A, Takeda K, Umezawa M. Dose-dependent induction of astrocyte activation and reactive astrogliosis in mouse brain following maternal exposure to carbon black nanoparticle. Part Fibre Toxicol 2017; 14:4. [PMID: 28148272 PMCID: PMC5289048 DOI: 10.1186/s12989-017-0184-6] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Accepted: 01/20/2017] [Indexed: 11/24/2022] Open
Abstract
Background Recent studies indicate that maternal exposure to ambient ultrafine particles and nanoparticles has adverse effects of on the central nervous system. Quantitative dose–response data is required to better understand the developmental neurotoxicity of nanoparticles. The present study investigated dose-dependent effects of maternal exposure to carbon black nanoparticle (CB-NP) on astrocyte in the brains of mouse offspring. Methods A CB-NP suspension (2.9, 15, or 73 μg/kg) was intranasally administered to pregnant ICR mice on gestational days 5 and 9. Cerebral cortex samples were collected from 6-week-old offspring and examined by Western blotting, immunostaining, microarray analysis, and quantitative reverse transcriptase-polymerase chain reaction. Placentae were collected from pregnant dams on gestational day 13 and examined by microarray analysis. Results Maternal exposure to CB-NP induced a dose-dependent increase in glial fibrillary acidic protein (GFAP) expression in the cerebral cortex; this increase was particularly observed in astrocytic end-feet attached to denatured perivascular macrophages. Moreover, maternal CB-NP exposure dose-dependently increased aquaporin-4 expression in the brain parenchyma region around blood vessels. The changes in the expression profiles of GFAP and Aqp4 in offspring after maternal CB-NP exposure were similar to those observed in mice of a more advanced age. The expression levels of mRNAs associated with angiogenesis, cell migration, proliferation, chemotaxis, and growth factor production were also altered in the cerebral cortex of offspring after maternal CB-NP exposure. Differentially expressed genes in placental tissues after CB-NP exposure did not populate any specific gene ontology category. Conclusions Maternal CB-NP exposure induced long-term activation of astrocytes resulting in reactive astrogliosis in the brains of young mice. Our observations suggest a potentially increased risk of the onset of age-related neurodegenerative diseases by maternal NP exposure. In this study, we report for the first time a quantitative dose–response relationship between maternal NP exposure and phenotypic changes in the central nervous system of the offspring. Moreover, our findings indicate that cortical GFAP and Aqp4 are useful biomarkers that can be employed in further studies aiming to elucidate the underlying mechanism of nanoparticle-mediated developmental neurotoxicity.
Collapse
Affiliation(s)
- Atsuto Onoda
- Department of Hygienic Chemistry, Graduate School of Pharmaceutical Sciences, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba, 278-8510, Japan. .,The Center for Environmental Health Science for the Next Generation, Research Institute for Science and Technology, Organization for Research Advancement, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba, 278-8510, Japan. .,Research Fellow of Japan Society for the Promotion of Science, 5-3-1 Kouji-machi, Chiyoda-ku, Tokyo, 102-0083, Japan.
| | - Ken Takeda
- The Center for Environmental Health Science for the Next Generation, Research Institute for Science and Technology, Organization for Research Advancement, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba, 278-8510, Japan
| | - Masakazu Umezawa
- The Center for Environmental Health Science for the Next Generation, Research Institute for Science and Technology, Organization for Research Advancement, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba, 278-8510, Japan.,Department of Materials Science and Technology, Faculty of Industrial Science and Technology, Tokyo University of Science, 6-3-1 Niijuku, Katsushika, Tokyo, 125-8585, Japan
| |
Collapse
|
21
|
Sapmaz E, Uysal M, Tumer MK, Sapmaz HI, Somuk BT, Arici A, Tas U. Investigation of age-related changes in the expression of aquaporin-1 and aquaporin-5 in the salivary glands of mice. Acta Otolaryngol 2016; 136:937-43. [PMID: 27067150 DOI: 10.3109/00016489.2016.1165353] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
CONCLUSION The increased AQP5 expression associated with ageing in glands, which mainly secreted a serous solution, suggests a compensation for the decreased amount of saliva secretion associated with age progression. OBJECTIVE To investigate the change in aquaporin-1 (AQP1) and aquaporin-5 (AQP5) expression in the salivary glands in young and elder mice. MATERIALS AND METHOD Twelve female mice from the Balb/C genus (30-50 g) were used. The mice were separated into two groups: Group I had 2-month-old mice and Group II had 18-month-old mice. Salivary glands (glandula parotidea, glandula sublungualis, glandula submaxillaris) were excised and examined immunohistochemically and histopathologically. AQP1 and AQP5 expression of young and elder mice was evaluated using the H-score. A p-value less than 0.05 was considered statistically significant. RESULTS Upon histopathological examination, the acini of glands were found to be atrophic in elder mice. The number and diameter of intercalated ducts were increased. Indeed, the amount of adipose tissue in the gland was increased. Upon immunohistochemical examination, both AQP1 and AQP5 levels in sublingual glands of elder mice were increased (p < 0.01 and p < 0.001, respectively). However, only AQP5 levels were increased in the parotid gland of elder mice (p < 0.01).
Collapse
Affiliation(s)
- Emrah Sapmaz
- a Department of Otorhinolaryngology, Faculty of Medicine , Gaziosmanpaşa University , Tokat , Turkey
| | - Murat Uysal
- b Department of Anatomy, Faculty of Medicine , Gaziosmanpaşa University , Tokat , Turkey
| | - Mehmet Kemal Tumer
- c Department of Oral and Maxillofacial Surgery, Faculty of Dentistry , Gaziosmanpaşa University , Tokat , Turkey
| | - Hilal Irmak Sapmaz
- b Department of Anatomy, Faculty of Medicine , Gaziosmanpaşa University , Tokat , Turkey
| | - Battal Tahsin Somuk
- a Department of Otorhinolaryngology, Faculty of Medicine , Gaziosmanpaşa University , Tokat , Turkey
| | - Akgul Arici
- d Department of Pathology, Faculty of Medicine , Gaziosmanpaşa University , Tokat , Turkey
| | - Ufuk Tas
- b Department of Anatomy, Faculty of Medicine , Gaziosmanpaşa University , Tokat , Turkey
| |
Collapse
|
22
|
Investigation of the effects of aging on the expression of aquaporin 1 and aquaporin 4 protein in heart tissue. Anatol J Cardiol 2016; 17:18-23. [PMID: 27443479 PMCID: PMC5324856 DOI: 10.14744/anatoljcardiol.2016.7033] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Objective: Aquaporin (AQP) 1 and AQP 4 are expressed in human heart and several studies have been focused on these two aquaporins. For this purpose, the present study is aimed to research the effects of aging on AQP 1 and AQP 4 in heart tissue. Methods: In this study, 14 Balb/C type white mice were used. Animals were divided into two equal groups. Group I consisted of 2-month-old young animals (n=7), and group II consisted of 18-month-old animals (n=7). To determine the AQP1 and AQP4 expression in the myocardium, the heart tissue was removed to perform western blotting and immunohistochemical and histopathological evaluations. Results: Muscle fibers of the heart in aged animals were more irregular and loosely organized in hematoxylin–eosin (H&E) stained sections. H-score analysis revealed that AQP1 and AQP4 immunoreactivity significantly increased in heart tissues of old mice compared with those of young mice (p<0.001). In addition, AQP1 and AQP4 protein expressions in the tissues of old animals were increased significantly according to western blot analysis (p=0.018 and p<0.001 for AQP1 and AQP4, respectively). Conclusion: Increased AQP1 and AQP4 levels in the heart tissue may be correlated with the maintenance of water and electrolytes balance, which decreases with aging. In this context, it might be the result of a compensatory response to decreased AQP4 functions. In addition, this increase with aging as demonstrated in our study might be one of the factors that increases the tendency of ischemia in elder people.
Collapse
|
23
|
The Neuroprotective Effect of the Association of Aquaporin-4/Glutamate Transporter-1 against Alzheimer's Disease. Neural Plast 2016; 2016:4626593. [PMID: 27057365 PMCID: PMC4736756 DOI: 10.1155/2016/4626593] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2015] [Revised: 11/21/2015] [Accepted: 11/30/2015] [Indexed: 11/21/2022] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder that is characterized by memory loss and cognitive dysfunction. Aquaporin-4 (AQP4), which is primarily expressed in astrocytes, is the major water channel expressed in the central nervous system (CNS). This protein plays an important role in water and ion homeostasis in the normal brain and in various brain pathological conditions. Emerging evidence suggests that AQP4 deficiency impairs learning and memory and that this may be related to the expression of glutamate transporter-1 (GLT-1). Moreover, the colocalization of AQP4 and GLT-1 has long been studied in brain tissue; however, far less is known about the potential influence that the AQP4/GLT-1 complex may have on AD. Research on the functional interaction of AQP4 and GLT-1 has been demonstrated to be of great significance in the study of AD. Here, we review the interaction of AQP4 and GLT-1 in astrocytes, which might play a pivotal role in the regulation of distinct cellular responses that involve neuroprotection against AD. The association of AQP4 and GLT-1 could greatly supplement previous research regarding neuroprotection against AD.
Collapse
|
24
|
Larson VA, Zhang Y, Bergles DE. Electrophysiological properties of NG2(+) cells: Matching physiological studies with gene expression profiles. Brain Res 2015; 1638:138-160. [PMID: 26385417 DOI: 10.1016/j.brainres.2015.09.010] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Revised: 08/31/2015] [Accepted: 09/08/2015] [Indexed: 01/11/2023]
Abstract
NG2(+) glial cells are a dynamic population of non-neuronal cells that give rise to myelinating oligodendrocytes in the central nervous system. These cells express numerous ion channels and neurotransmitter receptors, which endow them with a complex electrophysiological profile that is unique among glial cells. Despite extensive analysis of the electrophysiological properties of these cells, relatively little was known about the molecular identity of the channels and receptors that they express. The generation of new RNA-Seq datasets for NG2(+) cells has provided the means to explore how distinct genes contribute to the physiological properties of these progenitors. In this review, we systematically compare the results obtained through RNA-Seq transcriptional analysis of purified NG2(+) cells to previous physiological and molecular studies of these cells to define the complement of ion channels and neurotransmitter receptors expressed by NG2(+) cells in the mammalian brain and discuss the potential significance of the unique physiological properties of these cells. This article is part of a Special Issue entitled SI:NG2-glia(Invited only).
Collapse
Affiliation(s)
- Valerie A Larson
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Ye Zhang
- Department of Neurobiology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Dwight E Bergles
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|
25
|
CDRI-08 Attenuates REST/NRSF-Mediated Expression of NMDAR1 Gene in PBDE-209-Exposed Mice Brain. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2015; 2015:403840. [PMID: 26413122 PMCID: PMC4564648 DOI: 10.1155/2015/403840] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/17/2014] [Revised: 02/16/2015] [Accepted: 02/22/2015] [Indexed: 11/17/2022]
Abstract
CDRI-08 is a standardized bacoside enriched ethanolic extract of Bacopa monnieri, a nootropic plant. We reported that CDRI-08 attenuated oxidative stress and memory impairment in mice, induced by a flame retardant, PBDE-209. In order to explore the mechanism, present study was designed to examine the role of CDRI-08 on the expression of NMDAR1 (NR1) and the binding of REST/NRSF to NR1 promoter against postnatal exposure of PBDE-209. Male mice pups were orally supplemented with CDRI-08 at the doses of 40, 80, or 120 mg/kg along with PBDE-209 (20 mg/kg) during PND 3–10 and frontal cortex and hippocampus were collected at PND 11 and 60 to study the expression and regulation of NR1 by RT-PCR and electrophoretic mobility shift assay, respectively. The findings showed upregulated expression of NR1 and decreased binding of REST/NRSF to NR1 promoter after postnatal exposure of PBDE-209. Interestingly, supplementation with CDRI-08 significantly restored the expression of NR1 and binding of REST/NRSF to NR1 promoter near to the control value at the dose of 120 mg/kg. In conclusion, the results suggest that CDRI-08 possibly acts on glutamatergic system through expression and regulation of NR1 and may restore memory, impaired by PBDE-209 as reported in our previous study.
Collapse
|
26
|
Sfera A, Cummings M, Osorio C. Non-Neuronal Acetylcholine: The Missing Link Between Sepsis, Cancer, and Delirium? Front Med (Lausanne) 2015; 2:56. [PMID: 26347869 PMCID: PMC4543923 DOI: 10.3389/fmed.2015.00056] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Accepted: 07/29/2015] [Indexed: 12/11/2022] Open
Abstract
The interaction between living organisms and the environment requires a balancing act between genomic and epigenomic forces. Inflammation and cellular proliferation are kept in check by the genes, which code for their components and the microRNAs, which are capable of silencing the transcription of these genes. Acetylcholine (ACh) may play a unique role in the maintenance of this equilibrium, as the epigenomic inhibition of the gene coding for nicotinic receptors, and disinhibits the gene causing anergia in immune cells. We hypothesize that age-induced ACh deficiency is the result of an epigenomic dysfunction of microRNA-6775 (miR-6775), which silences the transcription of CHRNA7 gene [coding for alpha 7 nicotinic cholinergic receptors (nAChRs)]. When silenced, this gene induces decreased expression of alpha 7 nAChRs, which may predispose elderly individuals to inflammation, neuroinflammation, and delirium. We hypothesize further that miR-6775-induced hypocholinergia augments the expression of RNF 128, the gene related to anergy in lymphocytes (GRAIL). This gene favors regulatory T cells (Tregs), promoters of immunologic tolerance, which may predispose to both cancer and sepsis-induced immunosuppression.
Collapse
Affiliation(s)
- Adonis Sfera
- Psychiatry, Patton State Hospital , Patton, CA , USA
| | | | | |
Collapse
|
27
|
Gupta RK, Prasad S. Age-Dependent Alterations in the Interactions of NF-κB and N-myc with GLT-1/EAAT2 Promoter in the Pericontusional Cortex of Mice Subjected to Traumatic Brain Injury. Mol Neurobiol 2015; 53:3377-3388. [PMID: 26081154 DOI: 10.1007/s12035-015-9287-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2015] [Accepted: 06/03/2015] [Indexed: 01/09/2023]
Abstract
Traumatic brain injury (TBI) is one of the major risk factors of dementia, aging, and cognitive impairments, etc. We have previously reported that expression of the astrocytic glutamate transporter GLT-1/EAAT2 is downregulated in the pericontusional cortex of adult and old mice in post-TBI time-dependent manner, and the process of decline starts before in old than in adult TBI mice. However, relationship between age- and TBI-dependent alterations in GLT-1/EAAT2 expression and interactions of transcription factors NF-κB and N-myc with their cognate GLT-1/EAAT2 promoter sequences, an important step of its transcriptional control, is not known. To understand this, we developed TBI mouse model by modified chronic head injury (CHI) method, analyzed expression of GFAP, TNF-α, and AQP4 by RT-PCR for its validation, and analyzed interactions of NF-κB and N-myc with GLT-1/EAAT2 promoter sequences by electrophoretic mobility shift assay (EMSA). Our EMSA data revealed that interactions of NF-κB and N-myc with GLT-1/EAAT2 promoter sequences was significantly elevated in the ipsi-lateral cortex of both adult and old TBI mice in post-TBI time-dependent manner; however, these interactions started immediately in the old compared to that in adult TBI mice, which could be attributed to our previously reported age- and post-TBI time-dependent differential expression of GLT-1/EAAT2 in the pericontusional cortex.
Collapse
Affiliation(s)
- Rajaneesh K Gupta
- Biochemistry and Molecular Biology Laboratory, Department of Zoology, Banaras Hindu University, Varanasi, 221005, UP, India
| | - S Prasad
- Biochemistry and Molecular Biology Laboratory, Department of Zoology, Banaras Hindu University, Varanasi, 221005, UP, India.
| |
Collapse
|
28
|
Hanson E, Armbruster M, Cantu D, Andresen L, Taylor A, Danbolt NC, Dulla CG. Astrocytic glutamate uptake is slow and does not limit neuronal NMDA receptor activation in the neonatal neocortex. Glia 2015; 63:1784-96. [PMID: 25914127 DOI: 10.1002/glia.22844] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Revised: 03/30/2015] [Accepted: 04/07/2015] [Indexed: 01/20/2023]
Abstract
Glutamate uptake by astrocytes controls the time course of glutamate in the extracellular space and affects neurotransmission, synaptogenesis, and circuit development. Astrocytic glutamate uptake has been shown to undergo post-natal maturation in the hippocampus, but has been largely unexplored in other brain regions. Notably, glutamate uptake has never been examined in the developing neocortex. In these studies, we investigated the development of astrocytic glutamate transport, intrinsic membrane properties, and control of neuronal NMDA receptor activation in the developing neocortex. Using astrocytic and neuronal electrophysiology, immunofluorescence, and Western blot analysis we show that: (1) glutamate uptake in the neonatal neocortex is slow relative to neonatal hippocampus; (2) astrocytes in the neonatal neocortex undergo a significant maturation of intrinsic membrane properties; (3) slow glutamate uptake is accompanied by lower expression of both GLT-1 and GLAST; (4) glutamate uptake is less dependent on GLT-1 in neonatal neocortex than in neonatal hippocampus; and (5) the slow glutamate uptake we report in the neonatal neocortex corresponds to minimal astrocytic control of neuronal NMDA receptor activation. Taken together, our results clearly show fundamental differences between astrocytic maturation in the developing neocortex and hippocampus, and corresponding changes in how astrocytes control glutamate signaling.
Collapse
Affiliation(s)
- Elizabeth Hanson
- Department of Neuroscience, Tufts University School of Medicine, Boston, Massachusetts.,Neuroscience Program, Tufts Sackler School of Biomedical Sciences, Boston, Massachusetts
| | - Moritz Armbruster
- Department of Neuroscience, Tufts University School of Medicine, Boston, Massachusetts
| | - David Cantu
- Department of Neuroscience, Tufts University School of Medicine, Boston, Massachusetts
| | - Lauren Andresen
- Department of Neuroscience, Tufts University School of Medicine, Boston, Massachusetts.,Neuroscience Program, Tufts Sackler School of Biomedical Sciences, Boston, Massachusetts
| | - Amaro Taylor
- Department of Neuroscience, Tufts University School of Medicine, Boston, Massachusetts
| | - Niels Christian Danbolt
- Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Chris G Dulla
- Department of Neuroscience, Tufts University School of Medicine, Boston, Massachusetts.,Neuroscience Program, Tufts Sackler School of Biomedical Sciences, Boston, Massachusetts
| |
Collapse
|
29
|
Rutkovskiy A, Mariero LH, Vaage J. Deletion of the aquaporin-4 gene alters expression and phosphorylation of protective kinases in the mouse heart. Scandinavian Journal of Clinical and Laboratory Investigation 2014; 74:500-5. [PMID: 24792367 DOI: 10.3109/00365513.2014.905698] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
AIM Aquaporins are channel-forming proteins highly permeable to water and some small molecular solutes. We have previously shown that aquaporin-4 knockout mice have increased tolerance to ischemia. However, the mechanism of cardioprotection was unclear. The aim of the current study was to investigate the effects of aquaporin-4 deletion on baseline expression and phosphorylation of some cardioprotective protein kinases. METHODS Proteins were extracted from hearts of aquaporin-4 knockout mice and littermate wild-type controls and analyzed with Western blot. Samples were taken from young (≤ 6 months of age), and old (≥ 1 year) mice. RESULTS Western blots showed three different isoforms of aquaporin-4 in wild types, likely representing M1, M23, and Mz. Total AMP-dependent kinase expression was decreased in aquaporin-4 knockout hearts by 18 ± 13% (p = 0.02), while the expression of Akt kinase, extracellular signal regulated kinase 1/2, protein kinase C-epsilon, mitogen-associated kinase P38 and C-Jun N-terminal kinase was unchanged. The phosphorylation of Akt kinase was reduced in hearts from knockout mice by 41 ± 16% (p = 0.01). No other alterations in phosphorylation were found. These effects were only detected in young mice. CONCLUSION Deletion of the aquaporin-4 gene decreased AMP-dependent kinase expression and Akt kinase phosphorylation in the heart. These changes may influence energy metabolism and endogenous cardioprotection.
Collapse
Affiliation(s)
- Arkady Rutkovskiy
- Department of Emergency and Intensive Care at the Institute of Clinical Medicine , Norway
| | | | | |
Collapse
|
30
|
Verma P, Singh P, Gandhi BS. Neuromodulatory role of Bacopa monnieri on oxidative stress induced by postnatal exposure to decabromodiphenyl ether (PBDE -209) in neonate and young female mice. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2014; 17:307-11. [PMID: 24904725 PMCID: PMC4046233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/15/2013] [Accepted: 10/07/2013] [Indexed: 12/01/2022]
Abstract
OBJECTIVES Bacopa monnieri (BM), a traditional ayurvedic medicine, is a well-known memory enhancer. We have explored the role of BM against decabrominated diphenyl ether (PBDE-209)-induced alterations in neonate and young female mice. MATERIALS AND METHODS Mice were orally administered with B. monnieri at the doses of 40, 80 or 120 mg/kg body weight along with PBDE-209 (20 mg/kg body weight) from postnatal day (PND) 3-10. Levels of malondialdehyde, protein carbonyl and activities of superoxide dismutase and glutathione peroxidase were measured at both ages. The correct choices and reference/working memory errors of young mice were evaluated by Morris water and radial arm maze. RESULTS The results showed that BM at the dose of 120 mg/kg significantly (P<0.05) restored the levels of oxidants and the activities of antioxidant enzymes in frontal cortex and hippocampus of neonates against PBDE-209-induced toxicity. CONCLUSION BM plays a neuroprotective role against PBDE-209-induced alterations in oxidative status.
Collapse
Affiliation(s)
- Priya Verma
- Department of Zoology, Mahila Mahavidyalaya, Banaras Hindu University, Varanasi 221005, India,Corresponding author: Behrose S. Gandhi, Department of Zoology, Mahila Mahavidyalaya, Banaras Hindu University, Varanasi 221005, India; Tel:+91- 9839185458;
| | - Poonam Singh
- Department of Zoology, Mahila Mahavidyalaya, Banaras Hindu University, Varanasi 221005, India
| | - Behrose S. Gandhi
- Department of Zoology, Mahila Mahavidyalaya, Banaras Hindu University, Varanasi 221005, India,Corresponding author: Behrose S. Gandhi, Department of Zoology, Mahila Mahavidyalaya, Banaras Hindu University, Varanasi 221005, India; Tel:+91- 9839185458;
| |
Collapse
|
31
|
Sfera A, Osorio C. Water for thought: is there a role for aquaporin channels in delirium? Front Psychiatry 2014; 5:57. [PMID: 24904440 PMCID: PMC4033263 DOI: 10.3389/fpsyt.2014.00057] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2014] [Accepted: 05/12/2014] [Indexed: 12/14/2022] Open
Affiliation(s)
- Adonis Sfera
- Psychiatry, Patton State Hospital , Patton, CA , USA
| | | |
Collapse
|
32
|
Gupta RK, Prasad S. Differential regulation of GLT-1/EAAT2 gene expression by NF-κB and N-myc in male mouse brain during postnatal development. Neurochem Res 2013; 39:150-60. [PMID: 24277080 DOI: 10.1007/s11064-013-1200-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2013] [Revised: 11/06/2013] [Accepted: 11/13/2013] [Indexed: 12/14/2022]
Abstract
The synaptic glutamate level homeostasis is mainly maintained by the astrocytes membrane bound glutamate transporter type-1 (GLT-1/EAAT2). Alterations in its expression during development and aging and the underlying mechanisms are not well studied. Here, we report that NF-κB interaction was highest in both cerebral and cerebellar cortices at day 15 when compared with that at day 0 during development, and it further declined significantly in day 45, and remained unchanged in 20 and 70 weeks mice. On the other hand, N-myc interaction was highest at 0 day which significantly declined at 15-day and interestingly remained unaltered at later ages in both the cortices. This age dependent reciprocal pattern of NF-κB and N-myc interactions with their cognate GLT-1 promoter sequences was further correlated with GLT-1 protein and transcript levels. We found that higher NF-κB interaction with its cognate GLT-1 promoter sequences correlates with up-regulation whereas the higher N-myc interaction correlates with down-regulation of GLT-1 expression during postnatal developmental age up to 15 day, however, such phenomenon was not found in the higher ages from day 45 to 70 weeks. Thus our data suggests a postnatal development- and age dependent differential interaction of transcription factors NF-κB and N-myc to their respective sequences and they act as positive and negative regulator, respectively of GLT-1 gene expression in the brain during early developmental period in both cerebral and cerebellar cortices which might be different in aging of mice.
Collapse
Affiliation(s)
- Rajaneesh Kumar Gupta
- Molecular Biology and Biochemistry Lab., Centre of Advanced Study in Zoology, Banaras Hindu University, Varanasi, 221005, Uttar Pradesh, India
| | | |
Collapse
|
33
|
Gupta RK, Prasad S. Early down regulation of the glial Kir4.1 and GLT-1 expression in pericontusional cortex of the old male mice subjected to traumatic brain injury. Biogerontology 2013; 14:531-41. [PMID: 24026668 DOI: 10.1007/s10522-013-9459-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2013] [Accepted: 08/27/2013] [Indexed: 10/26/2022]
Abstract
Astroglia play multiple roles in brain function by providing matrix to neurons, secreting neurotrophic factors, maintaining K(+) and glutamate homeostasis and thereby controlling synaptic plasticity which undergoes alterations during aging. K(+) and glutamate homeostasis is maintained by astrocytes membrane bound inwardly rectifying K(+) channel (Kir4.1) and glutamate transporter-1 (GLT-1 or EAAT-2) proteins, respectively in the synapse and their expression may be altered due to traumatic brain injury (TBI). Also, it is not well understood whether this change is age dependent. To find out this, TBI was experimentally induced in adult and old male AKR strain mice using CHI technique, and expression of the Kir4.1 and GLT-1 in the pericontusional cortex at various time intervals was studied by Western blotting and semi quantitative RT-PCR techniques. Here, we report that expression of both Kir4.1 and GLT-1 genes at transcript and protein levels is significantly down regulated in the pericontusional ipsi-lateral cortex of old TBI mice as compared to that in the adult TBI mice as function of time after injury. Further, expression of both the genes starts decreasing early in old mice i.e., from the first hour after TBI as compared to that starts from fourth hour in adult TBI mice. Thus TBI affects expression of Kir4.1 and GLT-1 genes in age- and time dependent manner and it may lead to accumulations of more K(+) and glutamate early in the synapse of old mice as compared to adult. This may be implicated in the TBI induced early and severe neuronal depolarization and excito-neurotoxicity in old age.
Collapse
Affiliation(s)
- R K Gupta
- Biochemistry and Molecular Biology Laboratory, Department of Zoology, Banaras Hindu University, Varanasi, 221005, Uttar Pradesh, India
| | | |
Collapse
|
34
|
Bhattacharya P, Pandey AK, Paul S, Patnaik R, Yavagal DR. Aquaporin-4 inhibition mediates piroxicam-induced neuroprotection against focal cerebral ischemia/reperfusion injury in rodents. PLoS One 2013; 8:e73481. [PMID: 24023878 PMCID: PMC3762750 DOI: 10.1371/journal.pone.0073481] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2013] [Accepted: 07/22/2013] [Indexed: 02/02/2023] Open
Abstract
BACKGROUND AND PURPOSE Aquaporin-4(AQP4) is an abundant water channel protein in brain that regulates water transport to maintain homeostasis. Cerebral edema resulting from AQP4 over expression is considered to be one of the major determinants for progressive neuronal insult during cerebral ischemia. Although, both upregulation and downregulation of AQP4 expression is associated with brain pathology, over expression of AQP4 is one of the chief contributors of water imbalance in brain during ischemic pathology. We have found that Piroxicam binds to AQP4 with optimal binding energy value. Thus, we hypothesized that Piroxicam is neuroprotective in the rodent cerebral ischemic model by mitigating cerebral edema via AQP4 regulation. METHODS Rats were treated with Piroxicam OR placebo at 30 min prior, 2 h post and 4 h post 60 minutes of MCAO followed by 24 hour reperfusion. Rats were evaluated for neurological deficits and motor function just before sacrifice. Brains were harvested for infarct size estimation, water content measurement, biochemical analysis, RT-PCR and western blot experiments. RESULTS Piroxicam pretreatment thirty minutes prior to ischemia and four hour post reperfusion afforded neuroprotection as evident through significant reduction in cerebral infarct volume, improvement in motor behavior, neurological deficit and reduction in brain edema. Furthermore, ischemia induced surge in levels of nitrite and malondialdehyde were also found to be significantly reduced in ischemic brain regions in treated animals. This neuroprotection was found to be associated with inhibition of acid mediated rise in intracellular calcium levels and also downregulated AQP4 expression. CONCLUSIONS Findings of the present study provide significant evidence that Piroxicam acts as a potent AQP4 regulator and renders neuroprotection in focal cerebral ischemia. Piroxicam could be clinically exploited for the treatment of brain stroke along with other anti-stroke therapeutics in future.
Collapse
Affiliation(s)
- Pallab Bhattacharya
- School of Biomedical Engineering, Indian Institute of Technology, Banaras Hindu University, Varanasi, (U.P.), India
- Department of Neurology, Leonard M. Miller School of Medicine, University of Miami, Miami, Florida, United States of America
| | - Anand Kumar Pandey
- School of Biomedical Engineering, Indian Institute of Technology, Banaras Hindu University, Varanasi, (U.P.), India
| | - Sudip Paul
- School of Biomedical Engineering, Indian Institute of Technology, Banaras Hindu University, Varanasi, (U.P.), India
- Department of Biomedical Engineering, North Eastern Hill University (NEHU), Shillong, Meghalaya, India
| | - Ranjana Patnaik
- School of Biomedical Engineering, Indian Institute of Technology, Banaras Hindu University, Varanasi, (U.P.), India
| | - Dileep R. Yavagal
- Department of Neurology, Leonard M. Miller School of Medicine, University of Miami, Miami, Florida, United States of America
| |
Collapse
|
35
|
Human stem cell-derived spinal cord astrocytes with defined mature or reactive phenotypes. Cell Rep 2013; 4:1035-1048. [PMID: 23994478 DOI: 10.1016/j.celrep.2013.06.021] [Citation(s) in RCA: 158] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2012] [Revised: 05/15/2013] [Accepted: 06/18/2013] [Indexed: 12/23/2022] Open
Abstract
Differentiation of astrocytes from human stem cells has significant potential for analysis of their role in normal brain function and disease, but existing protocols generate only immature astrocytes. Using early neuralization, we generated spinal cord astrocytes from mouse or human embryonic or induced pluripotent stem cells with high efficiency. Remarkably, short exposure to fibroblast growth factor 1 (FGF1) or FGF2 was sufficient to direct these astrocytes selectively toward a mature quiescent phenotype, as judged by both marker expression and functional analysis. In contrast, tumor necrosis factor alpha and interleukin-1β, but not FGFs, induced multiple elements of a reactive inflammatory phenotype but did not affect maturation. These phenotypically defined, scalable populations of spinal cord astrocytes will be important both for studying normal astrocyte function and for modeling human pathological processes in vitro.
Collapse
|
36
|
Hawkes M, Elphinstone RE, Conroy AL, Kain KC. Contrasting pediatric and adult cerebral malaria: the role of the endothelial barrier. Virulence 2013; 4:543-55. [PMID: 23924893 PMCID: PMC5359751 DOI: 10.4161/viru.25949] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Malaria affects millions of people around the world and a small subset of those infected develop cerebral malaria. The clinical presentation of cerebral malaria differs between children and adults, and it has been suggested that age-related changes in the endothelial response may account for some of these differences. During cerebral malaria, parasites sequester within the brain microvasculature but do not penetrate into the brain parenchyma and yet, the infection causes severe neurological symptoms. Endothelial dysfunction is thought to play an important role in mediating these adverse clinical outcomes. During infection, the endothelium becomes activated and more permeable, which leads to increased inflammation, hemorrhages, and edema in the surrounding tissue. We hypothesize that post-natal developmental changes, occurring in both endothelial response and the neurovascular unit, account for the differences observed in the clinical presentations of cerebral malaria in children compared with adults.
Collapse
|