201
|
Bar-Shira O, Maor R, Chechik G. Gene Expression Switching of Receptor Subunits in Human Brain Development. PLoS Comput Biol 2015; 11:e1004559. [PMID: 26636753 PMCID: PMC4670163 DOI: 10.1371/journal.pcbi.1004559] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Accepted: 09/15/2015] [Indexed: 01/09/2023] Open
Abstract
Synaptic receptors in the human brain consist of multiple protein subunits, many of which have multiple variants, coded by different genes, and are differentially expressed across brain regions and developmental stages. The brain can tune the electrophysiological properties of synapses to regulate plasticity and information processing by switching from one protein variant to another. Such condition-dependent variant switch during development has been demonstrated in several neurotransmitter systems including NMDA and GABA. Here we systematically detect pairs of receptor-subunit variants that switch during the lifetime of the human brain by analyzing postmortem expression data collected in a population of donors at various ages and brain regions measured using microarray and RNA-seq. To further detect variant pairs that co-vary across subjects, we present a method to quantify age-corrected expression correlation in face of strong temporal trends. This is achieved by computing the correlations in the residual expression beyond a cubic-spline model of the population temporal trend, and can be seen as a nonlinear version of partial correlations. Using these methods, we detect multiple new pairs of context dependent variants. For instance, we find a switch from GLRA2 to GLRA3 that differs from the known switch in the rat. We also detect an early switch from HTR1A to HTR5A whose trends are negatively correlated and find that their age-corrected expression is strongly positively correlated. Finally, we observe that GRIN2B switch to GRIN2A occurs mostly during embryonic development, presumably earlier than observed in rodents. These results provide a systematic map of developmental switching in the neurotransmitter systems of the human brain. Synapses change their properties during development affecting information processing and learning. Most synaptic receptors consist of several proteins, each having several variants coded by closely related genes. These protein variants are similar in structure, yet often differ slightly in their biophysical attributes. Switching a synapse from using one variant to another provides the brain with a way to fine-tune electrophysiological properties of synapses and has been described in NMDA and GABA receptors. Here we describe a systematic approach to detect pairs of context-dependent variants at a genome-wide scale based on a set of post-mortem expression measurements taken from brains at multiple ages. We take into account both the profile of expression as it changes along life and also the detrended age-corrected correlation among genes. This method characterizes the landscape of developmental switches in brain transcriptome, putting forward new candidates pairs for deeper analysis. The abundance of switching between context-dependent variants through life suggests that it is a major mechanism by which the brain tunes its plasticity and information processing.
Collapse
Affiliation(s)
- Ossnat Bar-Shira
- Gonda Brain Research Center, Bar-Ilan University, Ramat Gan, Israel
| | - Ronnie Maor
- Gonda Brain Research Center, Bar-Ilan University, Ramat Gan, Israel
| | - Gal Chechik
- Gonda Brain Research Center, Bar-Ilan University, Ramat Gan, Israel
- * E-mail:
| |
Collapse
|
202
|
Weinberg RB, Mufson EJ, Counts SE. Evidence for a neuroprotective microRNA pathway in amnestic mild cognitive impairment. Front Neurosci 2015; 9:430. [PMID: 26594146 PMCID: PMC4633499 DOI: 10.3389/fnins.2015.00430] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Accepted: 10/22/2015] [Indexed: 12/22/2022] Open
Abstract
MicroRNAs (miRNAs) that regulate mRNA stability have been linked to amyloid production, tau phosphorylation, and inflammation in Alzheimer's disease (AD). However, whether cerebral miRNA networks are dysregulated during the earliest stages of AD remains underexplored. We performed miRNA expression analysis using frontal cortex tissue harvested from subjects who died with a clinical diagnosis of no cognitive impairment (NCI), amnestic mild cognitive impairment (aMCI, a putative prodromal AD stage), or mild AD. Analysis revealed that the miRNA clusters miR-212/132 and miR-23a/23b were down-regulated in the frontal cortex of aMCI subjects. Both miR-212/132 and miR23a/b are predicted to destabilize the message for sirtuin 1 (sirt1); hence, down-regulation of either miR-212/132 or miR-23a/b in frontal cortex should promote sirt1 mRNA expression in this region. qPCR studies revealed that frontal cortex levels of sirt1 were increased in aMCI. Given the ability of frontal cortex to respond to the onset of dementia by neuronal reorganization, these data suggest that miRNA-mediated up-regulation of the sirt1 pathway represents a compensatory response to the onset of the disease. By contrast, qPCR analysis of inferior temporal cortex, an area affected early in the progression of AD, showed no changes in miR-212/132, miR-23a/b, or sirt1 transcripts in the same aMCI subjects. In vitro mechanistic studies showed that coordinated down-regulation of miR-212 and miR-23a increased sirt1 protein expression and provided neuroprotection from β-amyloid toxicity in human neuronal cells. Taken together, these data suggest a novel miRNA-mediated neuroprotective pathway activated during the progression of AD that may be amenable to therapeutic manipulation.
Collapse
Affiliation(s)
- Rebecca B. Weinberg
- Department of Translational Science and Molecular Medicine, Michigan State UniversityGrand Rapids, MI, USA
| | - Elliott J. Mufson
- Department of Neurobiology, Barrow Neurological InstitutePhoenix, AZ, USA
| | - Scott E. Counts
- Department of Translational Science and Molecular Medicine, Michigan State UniversityGrand Rapids, MI, USA
- Department of Family Medicine, Michigan State UniversityGrand Rapids, MI, USA
- Hauenstein Neuroscience Center, Mercy Health Saint Mary's HospitalGrand Rapids, MI, USA
| |
Collapse
|
203
|
López-Avalos MD, Fernández-Llebrez Zayas R, Cifuentes M, De Andrés MV, Fernández-Llebrez Del Rey P, Grondona JM, Pérez-Martín M, Pedraza C. Mente Activa® Improves Impaired Spatial Memory in Aging Rats. J Nutr Health Aging 2015; 19:819-27. [PMID: 26412286 DOI: 10.1007/s12603-015-0546-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
OBJECTIVES Aging is accompanied by a decline in several aspects of the cognitive function, having negative personal and socioeconomic impacts. Dietary supplements could be beneficial for preventing age-related cognitive decline. In this context, we examined whether the nutritional supplement Mente Activa® has beneficial effects on aging-related cognitive deficits without inducing side effects. METHODS Mente Activa® was administered to old rats (n= 30 treated rats and n= 30 control rats) during 5 months, and the Morris water maze was used to test the learning capacities of the animals. The first assessment was conducted before the nutritional intervention (age of 18-19 months), to determine the baseline of the performance of animals on this test, and the second assessment was performed at the end of the treatment (23-24 moths). In order to examine possible secondary effects of this nutritional supplement, plasma, heart anatomy and liver parameters were evaluated. RESULTS Our data indicate that supplemented rats showed less escape latency, distance swum, higher use of spatial search strategies, and crossed the former platform location with higher frequency than control rats. These effects were specific of the treatment, indicating that this nutritional supplement has a beneficial effect on spatial memory. On the other hand, the regular intake of Mente Activa® did not induce any negative effects in plasma parameters and heart size. CONCLUSIONS Aged rats under a sustained dietary intake of the nutritional supplement Mente Activa® displayed improved learning and memory abilities compared to the non-treated rats. These results suggest the therapeutic potential and safety of use of Mente Activa® for age-related cognitive deficits, particularly, in the onset of the first cognitive dysfunction symptoms.
Collapse
Affiliation(s)
- M D López-Avalos
- C. Pedraza, Dpto. Psicobiología y Metodología de las CC. Facultad de Psicología. Universidad de Málaga, Campus de Teatinos s/n., Málaga, 29071. Spain, Tel: +34 952 132 510; Fax: +34 952 134 142, E-mail:
| | | | | | | | | | | | | | | |
Collapse
|
204
|
Mastroeni D, Delvaux E, Nolz J, Tan Y, Grover A, Oddo S, Coleman PD. Aberrant intracellular localization of H3k4me3 demonstrates an early epigenetic phenomenon in Alzheimer's disease. Neurobiol Aging 2015; 36:3121-3129. [PMID: 26553823 DOI: 10.1016/j.neurobiolaging.2015.08.017] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Revised: 07/27/2015] [Accepted: 08/14/2015] [Indexed: 11/19/2022]
Abstract
We have previously reported in Alzheimer's disease (AD) the mislocalization of epigenetic molecules between the cell nucleus and the cytoplasm. We have extended our finding to include the aberrant localization of histone 3 trimethylation on lysine 4 (H3k4me3), an epigenetic mark associated with actively transcribing genes as well as those poised for transcription. These findings raise the question of where the ectopic localization of H3k4me3 fits within the cascade of cell biological events in the progression of AD. We, therefore, examined the expression and intracellular location of H3k4me3 as a function of Braak stage and also in relation to a series of tau markers that are indicative of disease state. Both lines of evidence showed that ectopic localization of H3k4me3 is early in the course of disease. Because of the known role of H3k4me3 in the expression of synaptic genes, our data suggest an epigenetic role in synaptic deficits early in the course of AD.
Collapse
Affiliation(s)
- Diego Mastroeni
- ASU-Banner Neurodegenerative Disease Research Center, Biodesign Institute and School of Life Sciences, Arizona State University, Tempe, AZ, USA; L.J. Roberts Alzheimer's Disease Center, Banner Sun Health Research Institute, Sun City, AZ, USA; Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNS), Faculty of Health, Medicine and Life Sciences, European Graduate School of Neuroscience (EURON), Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Elaine Delvaux
- ASU-Banner Neurodegenerative Disease Research Center, Biodesign Institute and School of Life Sciences, Arizona State University, Tempe, AZ, USA; L.J. Roberts Alzheimer's Disease Center, Banner Sun Health Research Institute, Sun City, AZ, USA
| | - Jennifer Nolz
- ASU-Banner Neurodegenerative Disease Research Center, Biodesign Institute and School of Life Sciences, Arizona State University, Tempe, AZ, USA; L.J. Roberts Alzheimer's Disease Center, Banner Sun Health Research Institute, Sun City, AZ, USA
| | - Yuyan Tan
- Department of Neurology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Andrew Grover
- L.J. Roberts Alzheimer's Disease Center, Banner Sun Health Research Institute, Sun City, AZ, USA
| | - Salvatore Oddo
- Oddo Laboratory-Neurobiology of Aging and Dementia, Banner Sun Health Research Institute, Sun City, AZ, USA; Department of Basic Medical Sciences, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, USA
| | - Paul D Coleman
- ASU-Banner Neurodegenerative Disease Research Center, Biodesign Institute and School of Life Sciences, Arizona State University, Tempe, AZ, USA; L.J. Roberts Alzheimer's Disease Center, Banner Sun Health Research Institute, Sun City, AZ, USA.
| |
Collapse
|
205
|
Saura CA, Parra-Damas A, Enriquez-Barreto L. Gene expression parallels synaptic excitability and plasticity changes in Alzheimer's disease. Front Cell Neurosci 2015; 9:318. [PMID: 26379494 PMCID: PMC4548151 DOI: 10.3389/fncel.2015.00318] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2015] [Accepted: 08/03/2015] [Indexed: 11/14/2022] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder characterized by abnormal accumulation of β-amyloid and tau and synapse dysfunction in memory-related neural circuits. Pathological and functional changes in the medial temporal lobe, a region essential for explicit memory encoding, contribute to cognitive decline in AD. Surprisingly, functional imaging studies show increased activity of the hippocampus and associated cortical regions during memory tasks in presymptomatic and early AD stages, whereas brain activity declines as the disease progresses. These findings suggest an emerging scenario where early pathogenic events might increase neuronal excitability leading to enhanced brain activity before clinical manifestations of the disease, a stage that is followed by decreased brain activity as neurodegeneration progresses. The mechanisms linking pathology with synaptic excitability and plasticity changes leading to memory loss in AD remain largely unclear. Recent studies suggest that increased brain activity parallels enhanced expression of genes involved in synaptic transmission and plasticity in preclinical stages, whereas expression of synaptic and activity-dependent genes are reduced by the onset of pathological and cognitive symptoms. Here, we review recent evidences indicating a relationship between transcriptional deregulation of synaptic genes and neuronal activity and memory loss in AD and mouse models. These findings provide the basis for potential clinical applications of memory-related transcriptional programs and their regulatory mechanisms as novel biomarkers and therapeutic targets to restore brain function in AD and other cognitive disorders.
Collapse
Affiliation(s)
- Carlos A. Saura
- Institut de Neurociències, Departament de Bioquímica i Biologia Molecular, Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CIBERNED), Universitat Autònoma de BarcelonaBarcelona, Spain
| | | | | |
Collapse
|
206
|
Climbing experience in glass eels: A cognitive task or a matter of physical capacities? Physiol Behav 2015; 151:448-55. [PMID: 26260433 DOI: 10.1016/j.physbeh.2015.08.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Revised: 07/28/2015] [Accepted: 08/01/2015] [Indexed: 12/29/2022]
Abstract
The European eel is a panmictic species, whose decline has been recorded since the last 30 years. Among human-induced environmental factors of decline, the impact of water dams during species migration is questioned. Indeed, water impoundments can be a severe obstacle for young eels trying to reach the upstream freshwater zones, even if they are equipped with fish-friendly passes. The passage by such devices could be an important event shaping the outcome of the future life and life history traits of eels. We studied what phenotypic traits were associated with the event of experience of passage by water obstacles. We analyzed specific enzyme activities and/or gene transcription levels in the muscle and brain to test whether the obstacle passage is rather a physical or cognitive task. We found that after a long period of maintenance under homogenous conditions, transcription levels of several genes linked to synaptic plasticity, neurogenesis and thyroid activity differed among the field-experience groups. In contrast, muscle gene transcription levels or enzymatic activities did not show any differences among fish groups. We suggest that cognitive processes such as learning and memory acquisition rather than swimming-related metabolic capacities are involved in passage of water obstacles by young eels.
Collapse
|
207
|
Parikshak NN, Gandal MJ, Geschwind DH. Systems biology and gene networks in neurodevelopmental and neurodegenerative disorders. Nat Rev Genet 2015; 16:441-58. [PMID: 26149713 PMCID: PMC4699316 DOI: 10.1038/nrg3934] [Citation(s) in RCA: 304] [Impact Index Per Article: 30.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Genetic and genomic approaches have implicated hundreds of genetic loci in neurodevelopmental disorders and neurodegeneration, but mechanistic understanding continues to lag behind the pace of gene discovery. Understanding the role of specific genetic variants in the brain involves dissecting a functional hierarchy that encompasses molecular pathways, diverse cell types, neural circuits and, ultimately, cognition and behaviour. With a focus on transcriptomics, this Review discusses how high-throughput molecular, integrative and network approaches inform disease biology by placing human genetics in a molecular systems and neurobiological context. We provide a framework for interpreting network biology studies and leveraging big genomics data sets in neurobiology.
Collapse
Affiliation(s)
- Neelroop N Parikshak
- 1] Program in Neurobehavioral Genetics, Semel Institute, and Program in Neurogenetics, Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, California 90095, USA. [2] Interdepartmental Program in Neuroscience, University of California, Los Angeles, California 90095, USA
| | - Michael J Gandal
- 1] Program in Neurobehavioral Genetics, Semel Institute, and Program in Neurogenetics, Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, California 90095, USA. [2] Center for Autism Treatment and Research, Semel Institute, David Geffen School of Medicine, University of California, Los Angeles, California 90095, USA
| | - Daniel H Geschwind
- 1] Program in Neurobehavioral Genetics, Semel Institute, and Program in Neurogenetics, Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, California 90095, USA. [2] Interdepartmental Program in Neuroscience, University of California, Los Angeles, California 90095, USA. [3] Center for Autism Treatment and Research, Semel Institute, David Geffen School of Medicine, University of California, Los Angeles, California 90095, USA. [4] Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, California 90095, USA
| |
Collapse
|
208
|
Jung M, Jin SG, Zhang X, Xiong W, Gogoshin G, Rodin AS, Pfeifer GP. Longitudinal epigenetic and gene expression profiles analyzed by three-component analysis reveal down-regulation of genes involved in protein translation in human aging. Nucleic Acids Res 2015; 43:e100. [PMID: 25977295 PMCID: PMC4551908 DOI: 10.1093/nar/gkv473] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Accepted: 04/29/2015] [Indexed: 12/28/2022] Open
Abstract
Data on biological mechanisms of aging are mostly obtained from cross-sectional study designs. An inherent disadvantage of this design is that inter-individual differences can mask small but biologically significant age-dependent changes. A serially sampled design (same individual at different time points) would overcome this problem but is often limited by the relatively small numbers of available paired samples and the statistics being used. To overcome these limitations, we have developed a new vector-based approach, termed three-component analysis, which incorporates temporal distance, signal intensity and variance into one single score for gene ranking and is combined with gene set enrichment analysis. We tested our method on a unique age-based sample set of human skin fibroblasts and combined genome-wide transcription, DNA methylation and histone methylation (H3K4me3 and H3K27me3) data. Importantly, our method can now for the first time demonstrate a clear age-dependent decrease in expression of genes coding for proteins involved in translation and ribosome function. Using analogies with data from lower organisms, we propose a model where age-dependent down-regulation of protein translation-related components contributes to extend human lifespan.
Collapse
Affiliation(s)
- Marc Jung
- Department of Cancer Biology, Beckman Research Institute, City of Hope, Duarte, CA 91010, USA
| | - Seung-Gi Jin
- Department of Cancer Biology, Beckman Research Institute, City of Hope, Duarte, CA 91010, USA Department of Diabetes and Metabolic Diseases Research, Beckman Research Institute, City of Hope, Duarte, CA 91010, USA
| | - Xiaoying Zhang
- Department of Cancer Biology, Beckman Research Institute, City of Hope, Duarte, CA 91010, USA
| | - Wenying Xiong
- Department of Cancer Biology, Beckman Research Institute, City of Hope, Duarte, CA 91010, USA
| | - Grigoriy Gogoshin
- Center for Epigenetics, Van Andel Research Institute, Grand Rapids, MI 49503, USA
| | - Andrei S Rodin
- Center for Epigenetics, Van Andel Research Institute, Grand Rapids, MI 49503, USA
| | - Gerd P Pfeifer
- Department of Cancer Biology, Beckman Research Institute, City of Hope, Duarte, CA 91010, USA Department of Diabetes and Metabolic Diseases Research, Beckman Research Institute, City of Hope, Duarte, CA 91010, USA
| |
Collapse
|
209
|
Li JM, Liu C, Hu X, Cai Y, Ma C, Luo XG, Yan XX. Inverse correlation between Alzheimer's disease and cancer: implication for a strong impact of regenerative propensity on neurodegeneration? BMC Neurol 2014; 14:211. [PMID: 25394409 PMCID: PMC4232711 DOI: 10.1186/s12883-014-0211-2] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2014] [Accepted: 10/24/2014] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Recent studies have revealed an inverse epidemiological correlation between Alzheimer's disease (AD) and cancer - patients with AD show a reduced risk of cancer, while cancer survivors are less likely to develop AD. These late discoveries in human subjects call for explorative studies to unlock the underlying biological mechanism, but also may shed new light on conceptual interrogation of the principal pathogenic players in AD etiology. DISCUSSION Here we hypothesize that this negative correlation reflects a rebalance of biosynthetic propensity between body systems under the two disease statuses. In normal condition the body cellular systems are maintained homeostatically under a balanced cell degenerative vs. surviving/regenerative propensities, determined by biosynthetic resources for anabolic processing. AD pathogenesis involves neurodegeneration but also aberrant regenerative, or reactive anabolic, burden, while cancer development is driving by uncontrolled proliferation inherent with excessive anabolic activity. The aberrant neural regenerative propensity in AD pathogenesis and the uncontrolled cellular proliferative propensity in cancer pathogeneses can manifest as competitive processes, which could result in the inverse epidemiological correlation seen among the elderly. SUMMARY The reduced prevalence of AD in cancer survivors may implicate a strong impact of aberrant neural regenerative burden in neurodegeneration. Further explorative studies into the inverse correlation between AD and cancer should include examinations of the proliferative propensity of tumor cells in AD models, and the development of AD-like neuropathology in cancer models as well as following anti-proliferative drug treatment.
Collapse
Affiliation(s)
- Jian-Ming Li
- />Department of Anatomy and Neurobiology, Central South University School of Basic Medical Science, Changsha, 410013 Hunan China
- />Neuroscience Research Center, Changsha Medical University, Changsha, 410219 Hunan China
| | - Chao Liu
- />Department of Anatomy and Neurobiology, Central South University School of Basic Medical Science, Changsha, 410013 Hunan China
- />Department of Neurology, The First Hospital of Changsha, Changsha, 410005 Hunan China
| | - Xia Hu
- />Department of Anatomy and Neurobiology, Central South University School of Basic Medical Science, Changsha, 410013 Hunan China
| | - Yan Cai
- />Department of Anatomy and Neurobiology, Central South University School of Basic Medical Science, Changsha, 410013 Hunan China
| | - Chao Ma
- />Department of Human Anatomy, Histology & Embryology, Institute of Basic Medical Sciences, Neuroscience Center, Chinese Academy of Medical Sciences; School of Basic Medicine, Peking Union Medical College, Beijing, 100730 China
| | - Xue-Gang Luo
- />Department of Anatomy and Neurobiology, Central South University School of Basic Medical Science, Changsha, 410013 Hunan China
| | - Xiao-Xin Yan
- />Department of Anatomy and Neurobiology, Central South University School of Basic Medical Science, Changsha, 410013 Hunan China
| |
Collapse
|
210
|
Vitamin D prevents cognitive decline and enhances hippocampal synaptic function in aging rats. Proc Natl Acad Sci U S A 2014; 111:E4359-66. [PMID: 25267625 DOI: 10.1073/pnas.1404477111] [Citation(s) in RCA: 159] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Vitamin D is an important calcium-regulating hormone with diverse functions in numerous tissues, including the brain. Increasing evidence suggests that vitamin D may play a role in maintaining cognitive function and that vitamin D deficiency may accelerate age-related cognitive decline. Using aging rodents, we attempted to model the range of human serum vitamin D levels, from deficient to sufficient, to test whether vitamin D could preserve or improve cognitive function with aging. For 5-6 mo, middle-aged F344 rats were fed diets containing low, medium (typical amount), or high (100, 1,000, or 10,000 international units/kg diet, respectively) vitamin D3, and hippocampal-dependent learning and memory were then tested in the Morris water maze. Rats on high vitamin D achieved the highest blood levels (in the sufficient range) and significantly outperformed low and medium groups on maze reversal, a particularly challenging task that detects more subtle changes in memory. In addition to calcium-related processes, hippocampal gene expression microarrays identified pathways pertaining to synaptic transmission, cell communication, and G protein function as being up-regulated with high vitamin D. Basal synaptic transmission also was enhanced, corroborating observed effects on gene expression and learning and memory. Our studies demonstrate a causal relationship between vitamin D status and cognitive function, and they suggest that vitamin D-mediated changes in hippocampal gene expression may improve the likelihood of successful brain aging.
Collapse
|
211
|
Abstract
Loss of memory is among the first symptoms reported by patients suffering from Alzheimer's disease (AD) and by their caretakers. Working memory and long-term declarative memory are affected early during the course of the disease. The individual pattern of impaired memory functions correlates with parameters of structural or functional brain integrity. AD pathology interferes with the formation of memories from the molecular level to the framework of neural networks. The investigation of AD memory loss helps to identify the involved neural structures, such as the default mode network, the influence of epigenetic and genetic factors, such as ApoE4 status, and evolutionary aspects of human cognition. Clinically, the analysis of memory assists the definition of AD subtypes, disease grading, and prognostic predictions. Despite new AD criteria that allow the earlier diagnosis of the disease by inclusion of biomarkers derived from cerebrospinal fluid or hippocampal volume analysis, neuropsychological testing remains at the core of AD diagnosis.
Collapse
Affiliation(s)
- Holger Jahn
- University Hospital Hamburg-Eppendorf, Dept of Psychiatry and Psychotherapy, Hamburg, Germany
| |
Collapse
|
212
|
Berchtold NC, Sabbagh MN, Beach TG, Kim RC, Cribbs DH, Cotman CW. Brain gene expression patterns differentiate mild cognitive impairment from normal aged and Alzheimer's disease. Neurobiol Aging 2014; 35:1961-72. [PMID: 24786631 PMCID: PMC4067010 DOI: 10.1016/j.neurobiolaging.2014.03.031] [Citation(s) in RCA: 82] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2013] [Revised: 03/25/2014] [Accepted: 03/28/2014] [Indexed: 02/08/2023]
Abstract
Mild cognitive impairment (MCI) represents a cognitive state intermediate between normal aging and early Alzheimer's disease (AD). To investigate if the molecular signature of MCI parallels the clinical picture, we use microarrays to extensively profile gene expression in 4 cortical brain regions (entorhinal cortex, hippocampus, superior frontal gyrus, post-central gyrus) using the postmortem tissue from cognitively normal aged controls, MCI, and AD cases. Our data reveal that gene expression patterns in MCI are not an extension of aging, and for the most part, are not intermediate between aged controls and AD. Functional enrichment analysis of significant genes revealed prominent upregulation in MCI brains of genes associated with anabolic and biosynthetic pathways (notably transcription, protein biosynthesis, protein trafficking, and turnover) as well as mitochondrial energy generation. In addition, many synaptic genes showed altered expression in MCI, predominantly upregulation, including genes for central components of the vesicle fusion machinery at the synapse, synaptic vesicle trafficking, neurotransmitter receptors, and synaptic structure and stabilization. These data suggest that there is a rebalancing of synaptic transmission in the MCI brain. To investigate if synaptic gene expression levels in MCI were related to cognitive function, Pearson correlation coefficient between the Mini Mental State Examination (MMSE) and region-specific messenger RNA expression were computed for MCI cases. A number of synaptic genes showed strong significant correlations (r > 0.8, p < 0.01) most notably in the entorhinal cortex, with fewer in the hippocampus, and very few in neocortical regions. The synaptic genes with highly significant correlations were predominantly related to synaptic transmission and plasticity, and myelin composition. Unexpectedly, we found that gene expression changes that facilitate synaptic excitability and plasticity were overwhelmingly associated with poorer MMSE, and conversely that gene expression changes that inhibit plasticity were positively associated with MMSE. These data suggest that there are excessive excitability and apparent plasticity in limbic brain regions in MCI, that is associated with impaired synaptic and cognitive function. Such changes would be predicted to contribute to increased excitability, in turn leading to greater metabolic demand and ultimately progressive degeneration and AD, if not controlled.
Collapse
Affiliation(s)
- Nicole C Berchtold
- Institute for Mental Impairments and Neurological Disorders (MIND), University of California Irvine, Irvine, CA, USA.
| | | | | | - Ronald C Kim
- Institute for Mental Impairments and Neurological Disorders (MIND), University of California Irvine, Irvine, CA, USA
| | - David H Cribbs
- Institute for Mental Impairments and Neurological Disorders (MIND), University of California Irvine, Irvine, CA, USA; Departments of Neurology and Neurobiology and Behavior, University of California Irvine, Irvine, CA, USA
| | - Carl W Cotman
- Institute for Mental Impairments and Neurological Disorders (MIND), University of California Irvine, Irvine, CA, USA; Departments of Neurology and Neurobiology and Behavior, University of California Irvine, Irvine, CA, USA
| |
Collapse
|
213
|
Mariga A, Zavadil J, Ginsberg SD, Chao MV. Withdrawal of BDNF from hippocampal cultures leads to changes in genes involved in synaptic function. Dev Neurobiol 2014; 75:173-92. [PMID: 25059794 DOI: 10.1002/dneu.22216] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2014] [Revised: 06/26/2014] [Accepted: 07/23/2014] [Indexed: 11/07/2022]
Abstract
Neurotrophins play a crucial role in mediating neuronal survival and synaptic plasticity. A lack of trophic factor support in the peripheral nervous system (PNS) is associated with a transcription-dependent programmed cell death process in developing sympathetic neurons. While most of the attention has been on events culminating in cell death in the PNS, the earliest events that occur after trophic factor withdrawal in the central nervous system (CNS) have not been investigated. In the CNS, brain-derived neurotrophic factor (BDNF) is widely expressed and is released in an activity-dependent manner to shape the structure and function of neuronal populations. Reduced neurotrophic factor support has been proposed as a mechanism to account for changes in synaptic plasticity during neurodevelopment to aging and neurodegenerative disorders. To this end, we performed transcriptional profiling in cultured rat hippocampal neurons. We used a TrkB ligand scavenger (TrkB-FC ) to sequester endogenous neurotrophic factor activity from hippocampal neurons in culture. Using a high-density microarray platform, we identified a significant decrease in genes that are associated with vesicular trafficking and synaptic function, as well as selective increases in MAP kinase phosphatases. A comparison of these changes with recent studies of Alzheimer's disease and cognitive impairment in postmortem brain tissue revealed striking similarities in gene expression changes for genes involved in synaptic function. These changes are relevant to a wide number of conditions in which levels of BDNF are compromised.
Collapse
Affiliation(s)
- Abigail Mariga
- Cell and Molecular Biology Program, New York University Langone Medical Center, New York, New York, 10016
| | | | | | | |
Collapse
|
214
|
Neuman KM, Molina-Campos E, Musial TF, Price AL, Oh KJ, Wolke ML, Buss EW, Scheff SW, Mufson EJ, Nicholson DA. Evidence for Alzheimer's disease-linked synapse loss and compensation in mouse and human hippocampal CA1 pyramidal neurons. Brain Struct Funct 2014; 220:3143-65. [PMID: 25031178 DOI: 10.1007/s00429-014-0848-z] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2013] [Accepted: 07/09/2014] [Indexed: 12/17/2022]
Abstract
Alzheimer's disease (AD) is associated with alterations in the distribution, number, and size of inputs to hippocampal neurons. Some of these changes are thought to be neurodegenerative, whereas others are conceptualized as compensatory, plasticity-like responses, wherein the remaining inputs reactively innervate vulnerable dendritic regions. Here, we provide evidence that the axospinous synapses of human AD cases and mice harboring AD-linked genetic mutations (the 5XFAD line) exhibit both, in the form of synapse loss and compensatory changes in the synapses that remain. Using array tomography, quantitative conventional electron microscopy, immunogold electron microscopy for AMPARs, and whole-cell patch-clamp physiology, we find that hippocampal CA1 pyramidal neurons in transgenic mice are host to an age-related synapse loss in their distal dendrites, and that the remaining synapses express more AMPA-type glutamate receptors. Moreover, the number of axonal boutons that synapse with multiple spines is significantly reduced in the transgenic mice. Through serial section electron microscopic analyses of human hippocampal tissue, we further show that putative compensatory changes in synapse strength are also detectable in axospinous synapses of proximal and distal dendrites in human AD cases, and that their multiple synapse boutons may be more powerful than those in non-cognitively impaired human cases. Such findings are consistent with the notion that the pathophysiology of AD is a multivariate product of both neurodegenerative and neuroplastic processes, which may produce adaptive and/or maladaptive responses in hippocampal synaptic strength and plasticity.
Collapse
Affiliation(s)
- Krystina M Neuman
- Department of Neurological Sciences, Rush University Medical Center, 1653 West Harrison Street, Chicago, IL, 60612, USA
| | - Elizabeth Molina-Campos
- Department of Neurological Sciences, Rush University Medical Center, 1653 West Harrison Street, Chicago, IL, 60612, USA
| | - Timothy F Musial
- Department of Neurological Sciences, Rush University Medical Center, 1653 West Harrison Street, Chicago, IL, 60612, USA
| | - Andrea L Price
- Department of Neurological Sciences, Rush University Medical Center, 1653 West Harrison Street, Chicago, IL, 60612, USA
| | - Kwang-Jin Oh
- Department of Neurological Sciences, Rush University Medical Center, 1653 West Harrison Street, Chicago, IL, 60612, USA
| | - Malerie L Wolke
- Department of Neurological Sciences, Rush University Medical Center, 1653 West Harrison Street, Chicago, IL, 60612, USA
| | - Eric W Buss
- Department of Neurological Sciences, Rush University Medical Center, 1653 West Harrison Street, Chicago, IL, 60612, USA
| | - Stephen W Scheff
- Sanders Brown Center on Aging, University of Kentucky, Lexington, KY, 40536, USA
| | - Elliott J Mufson
- Department of Neurological Sciences, Rush University Medical Center, 1653 West Harrison Street, Chicago, IL, 60612, USA
| | - Daniel A Nicholson
- Department of Neurological Sciences, Rush University Medical Center, 1653 West Harrison Street, Chicago, IL, 60612, USA.
| |
Collapse
|
215
|
Carlessi L, Poli EF, Bechi G, Mantegazza M, Pascucci B, Narciso L, Dogliotti E, Sala C, Verpelli C, Lecis D, Delia D. Functional and molecular defects of hiPSC-derived neurons from patients with ATM deficiency. Cell Death Dis 2014; 5:e1342. [PMID: 25032865 PMCID: PMC4123100 DOI: 10.1038/cddis.2014.310] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2014] [Revised: 05/30/2014] [Accepted: 06/16/2014] [Indexed: 11/21/2022]
Abstract
Loss of ataxia telangiectasia mutated (ATM) kinase, a key factor of the DNA damage response (DDR) pathway, causes the cancer predisposing and neurodegenerative syndrome ataxia-telangiectasia (A-T). To investigate the mechanisms of neurodegeneration, we have reprogrammed fibroblasts from ATM-null A-T patients and normal controls to pluripotency (human-induced pluripotent stem cells), and derived from these neural precursor cells able to terminally differentiate into post-mitotic neurons positive to >90% for β-tubulin III+/microtubule-associated protein 2+. We show that A-T neurons display similar voltage-gated potassium and sodium currents and discharges of action potentials as control neurons, but defective expression of the maturation and synaptic markers SCG10, SYP and PSD95 (postsynaptic density protein 95). A-T neurons exhibited defective repair of DNA double-strand breaks (DSBs) and repressed phosphorylation of ATM substrates (e.g., γH2AX, Smc1-S966, Kap1-S824, Chk2-T68, p53-S15), but normal repair of single-strand breaks, and normal short- and long-patch base excision repair activities. Moreover, A-T neurons were resistant to apoptosis induced by the genotoxic agents camptothecin and trabectedin, but as sensitive as controls to the oxidative agents. Most notably, A-T neurons exhibited abnormal accumulation of topoisomerase 1-DNA covalent complexes (Top1-ccs). These findings reveal that ATM deficiency impairs neuronal maturation, suppresses the response and repair of DNA DSBs, and enhances Top1-cc accumulation. Top1-cc could be a risk factor for neurodegeneration as they may interfere with transcription elongation and promote transcriptional decline.
Collapse
Affiliation(s)
- L Carlessi
- Department of Experimental Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Amadeo 42, 20133 Milano, Italy
| | - E Fusar Poli
- Department of Experimental Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Amadeo 42, 20133 Milano, Italy
| | - G Bechi
- Department of Neurophysiopathology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Via Amadeo 42, 20133 Milano, Italy
| | - M Mantegazza
- Department of Neurophysiopathology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Via Amadeo 42, 20133 Milano, Italy
- Institute of Molecular and Cellular Pharmacology (IPMC) CNRS UMR7275 and University of Nice-Sophia Antipolis, 660 Route des Lucioles, 06560 Valbonne, France
| | - B Pascucci
- CNR Institute of Crystallography, Via Salaria, Km. 29.300, 00016 Monterotondo Scalo, Roma, Italy
| | - L Narciso
- Department of Food Safety and Veterinary Public Health, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Roma, Italy
| | - E Dogliotti
- Department of Environment and Primary Prevention, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Roma, Italy
| | - C Sala
- CNR Institute of Neuroscience and Department of Biotechnology and Translational Medicine, Via Vanvitelli 32, 20129 Milano, Italy
| | - C Verpelli
- CNR Institute of Neuroscience and Department of Biotechnology and Translational Medicine, Via Vanvitelli 32, 20129 Milano, Italy
| | - D Lecis
- Department of Experimental Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Amadeo 42, 20133 Milano, Italy
| | - D Delia
- Department of Experimental Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Amadeo 42, 20133 Milano, Italy
| |
Collapse
|
216
|
Deng X, Li M, Ai W, He L, Lu D, Patrylo PR, Cai H, Luo X, Li Z, Yan X. Lipolysaccharide-Induced Neuroinflammation Is Associated with Alzheimer-Like Amyloidogenic Axonal Pathology and Dendritic Degeneration in Rats. ADVANCES IN ALZHEIMER'S DISEASE 2014; 3:78-93. [PMID: 25360394 PMCID: PMC4211261 DOI: 10.4236/aad.2014.32009] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Chronic neuroinflammation is thought to play an etiological role in Alzheimer's disease (AD), which is characterized pathologically by amyloid and tau formation, as well as neuritic dystrophy and synaptic degeneration. The causal relationship between these pathological events is a topic of ongoing research and discussion. Recent data from transgenic AD models point to a tight spatiotemporal link between neuritic and amyloid pathology, with the obligatory enzyme for β-amyloid (Aβ) production, namely β-secretase-1 (BACE1), is overexpressed in axon terminals undergoing dystrophic change. However, the axonal pathology inherent with BACE1 elevation seen in transgenic AD mice may be secondary to increased soluble Aβ in these genetically modified animals. Here we explored the occurrence of the AD-like axonal and dendritic pathology in adult rat brain affected by LPS-induced chronic neuroinflammation. Unilateral intracerebral LPS injection induced prominent inflammatory response in glial cells in the ipsilateral cortex and hippocampal formation. BACE1 protein levels were elevated the ipsilateral hippocampal lysates in the LPS treated animals relative to controls. BACE1 immunoreactive dystrophic axons appeared in the LPS-treated ipsilateral cortex and hippocampal formation, colocalizing with increased β-amyloid precursor protein and Aβ antibody (4G8) immunolabeling. Quantitative Golgi studies revealed reduction of dendritic branching points and spine density on cortical layer III and hippocampal CA3 pyramidal neurons in the LPS-treated ipsilateral cerebrum. These findings suggest that Alzheimer-like amyloidogenic axonal pathology and dendritic degeneration occur in wildtype mammalian brain in partnership with neuroinflammation following LPS injection.
Collapse
Affiliation(s)
- Xiaohua Deng
- Department of Anatomy and Neurobiology, Central South University School of Basic Medical Science, Changsha, China
| | - Meili Li
- Department of Anatomy and Neurobiology, Central South University School of Basic Medical Science, Changsha, China
| | - Weiming Ai
- Department of Anatomy and Neurobiology, Central South University School of Basic Medical Science, Changsha, China
- Department of Nursing in Internal Medicine, School of Nursing, Xiangtan Vocational and Technical College, Xiangtan, China
| | - Lixin He
- Department of Anatomy and Neurobiology, Central South University School of Basic Medical Science, Changsha, China
- Department of Anatomy and Physiology, School of Nursing, Xiangtan Vocational and technical College, Xiangtan, China
| | - Dahua Lu
- Department of Anatomy and Neurobiology, Central South University School of Basic Medical Science, Changsha, China
| | - Peter R. Patrylo
- Departments of Physiology, Anatomy and Center for Integrated Research in Cognitive and Neural Sciences, Southern Illinois University Carbondale, Carbondale, USA
| | - Huaibin Cai
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, USA
| | - Xuegang Luo
- Department of Anatomy and Neurobiology, Central South University School of Basic Medical Science, Changsha, China
| | - Zhiyuan Li
- Department of Anatomy and Neurobiology, Central South University School of Basic Medical Science, Changsha, China
| | - Xiaoxin Yan
- Department of Anatomy and Neurobiology, Central South University School of Basic Medical Science, Changsha, China
| |
Collapse
|
217
|
Overk CR, Masliah E. Pathogenesis of synaptic degeneration in Alzheimer's disease and Lewy body disease. Biochem Pharmacol 2014; 88:508-16. [PMID: 24462903 PMCID: PMC3973539 DOI: 10.1016/j.bcp.2014.01.015] [Citation(s) in RCA: 173] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2013] [Revised: 01/10/2014] [Accepted: 01/14/2014] [Indexed: 12/15/2022]
Abstract
Considerable progress has been made in the past few years in the fight against Alzheimer's disease (AD) and Parkinson's disease (PD). Neuropathological studies in human brains and experimental in vivo and in vitro models support the notion that synapses are affected even at the earliest stages of the neurodegenerative process. The objective of this manuscript is to review some of the mechanisms of synaptic damage in AD and PD. Some lines of evidence support the notion that oligomeric neurotoxic species of amyloid β, α-synuclein, and Tau might contribute to the pathogenesis of synaptic failure at early stages of the diseases. The mechanisms leading to synaptic damage by oligomers might involve dysregulation of glutamate receptors and scaffold molecules that results in alterations in the axonal transport of synaptic vesicles and mitochondria that later on lead to dendritic and spine alterations, axonal dystrophy, and eventually neuronal loss. However, while some studies support a role of oligomers, there is an ongoing debate as to the exact nature of the toxic species. Given the efforts toward earlier clinical and preclinical diagnosis of these disorders, understanding the molecular and cellular mechanisms of synaptic degeneration is crucial toward developing specific biomarkers and new therapies targeting the synaptic apparatus of vulnerable neurons.
Collapse
Affiliation(s)
- Cassia R Overk
- Department of Neurosciences, University of California, San Diego, La Jolla, CA 92039, USA
| | - Eliezer Masliah
- Department of Neurosciences, University of California, San Diego, La Jolla, CA 92039, USA; Department of Pathology, University of California, San Diego, La Jolla, CA 92039, USA.
| |
Collapse
|
218
|
Synaptic gene dysregulation within hippocampal CA1 pyramidal neurons in mild cognitive impairment. Neuropharmacology 2013; 79:172-9. [PMID: 24445080 DOI: 10.1016/j.neuropharm.2013.10.018] [Citation(s) in RCA: 105] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2013] [Revised: 09/25/2013] [Accepted: 10/14/2013] [Indexed: 12/16/2022]
Abstract
Clinical neuropathologic studies suggest that the selective vulnerability of hippocampal CA1 pyramidal projection neurons plays a key role in the onset of cognitive impairment during the early phases of Alzheimer's disease (AD). Disruption of this neuronal population likely affects hippocampal pre- and postsynaptic efficacy underlying episodic memory circuits. Therefore, identifying perturbations in the expression of synaptic gene products within CA1 neurons prior to frank AD is crucial for the development of disease modifying therapies. Here we used custom-designed microarrays to examine progressive alterations in synaptic gene expression within CA1 neurons in cases harvested from the Rush Religious Orders Study who died with a clinical diagnosis of no cognitive impairment (NCI), mild cognitive impairment (MCI, a putative prodromal AD stage), or mild/moderate AD. Quantitative analysis revealed that 21 out of 28 different transcripts encoding regulators of synaptic function were significantly downregulated (1.4-1.8 fold) in CA1 neurons in MCI and AD compared to NCI, whereas synaptic transcript levels were not significantly different between MCI and AD. The downregulated transcripts encoded regulators of presynaptic vesicle trafficking, including synaptophysin and synaptogyrin, regulators of vesicle docking and fusion/release, such as synaptotagmin and syntaxin 1, and regulators of glutamatergic postsynaptic function, including PSD-95 and synaptopodin. Clinical pathologic correlation analysis revealed that downregulation of these synaptic markers was strongly associated with poorer antemortem cognitive status and postmortem AD pathological criteria such as Braak stage, NIA-Reagan, and CERAD diagnosis. In contrast to the widespread loss of synaptic gene expression observed in CA1 neurons in MCI, transcripts encoding β-amyloid precursor protein (APP), APP family members, and regulators of APP metabolism were not differentially regulated in CA1 neurons across the clinical diagnostic groups. Taken together, these data suggest that CA1 synaptic gene dysregulation occurs early in the cascade of pathogenic molecular events prior to the onset of AD, which may form the basis for novel pharmacological treatment approaches for this dementing disorder. This article is part of a Special Issue entitled 'Neurodegenerative Disorders'.
Collapse
|
219
|
Valdes AM, Glass D, Spector TD. Omics technologies and the study of human ageing. Nat Rev Genet 2013; 14:601-7. [PMID: 23938363 DOI: 10.1038/nrg3553] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Normal ageing is associated with diverse physiological changes in all organ systems but the rate and extent of these changes vary markedly among individuals. One aspect of ageing research focuses on the molecular profiling of the changes that occur with increasing age in humans. Such profiling has implications for disease prevention and treatment. New high-throughput 'omics' technologies (such as genomics, metabolomics, metagenomics and transcriptomics) are enabling detailed studies of these molecular changes and are thus revealing information about the biological pathways that change with age.
Collapse
Affiliation(s)
- Ana M Valdes
- 1] Department of Twin Research and Genetic Epidemiology, Kings College London, London SE1 7EH, UK. [2] Academic Rheumatology, University of Nottingham, Nottingham NG5 1PB, UK
| | | | | |
Collapse
|
220
|
Ingiosi AM, Opp MR, Krueger JM. Sleep and immune function: glial contributions and consequences of aging. Curr Opin Neurobiol 2013; 23:806-11. [PMID: 23452941 DOI: 10.1016/j.conb.2013.02.003] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2013] [Revised: 02/05/2013] [Accepted: 02/05/2013] [Indexed: 11/18/2022]
Abstract
The reciprocal interactions between sleep and immune function are well-studied. Insufficient sleep induces innate immune responses as evidenced by increased expression of pro-inflammatory mediators in the brain and periphery. Conversely, immune challenges upregulate immunomodulator expression, which alters central nervous system-mediated processes and behaviors, including sleep. Recent studies indicate that glial cells, namely microglia and astrocytes, are active contributors to sleep and immune system interactions. Evidence suggests glial regulation of these interactions is mediated, in part, by adenosine and adenosine 5'-triphosphate actions at purinergic type 1 and type 2 receptors. Furthermore, microglia and astrocytes may modulate declines in sleep-wake behavior and immunity observed in aging.
Collapse
Affiliation(s)
- Ashley M Ingiosi
- Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI, United States; Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA, United States
| | | | | |
Collapse
|