1
|
Taylor HA, Finkel T, Gao Y, Ballinger SW, Campo R, Chen R, Chen SH, Davidson K, Iruela-Arispe ML, Jaquish C, LeBrasseur NK, Odden MC, Papanicolaou GJ, Picard M, Srinivas P, Tjurmina O, Wolz M, Galis ZS. Scientific opportunities in resilience research for cardiovascular health and wellness. Report from a National Heart, Lung, and Blood Institute workshop. FASEB J 2022; 36:e22639. [PMID: 36322029 PMCID: PMC9703084 DOI: 10.1096/fj.202201407r] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Revised: 10/14/2022] [Accepted: 10/21/2022] [Indexed: 11/07/2022]
Abstract
Exposure of biological systems to acute or chronic insults triggers a host of molecular and physiological responses to either tolerate, adapt, or fully restore homeostasis; these responses constitute the hallmarks of resilience. Given the many facets, dimensions, and discipline-specific focus, gaining a shared understanding of "resilience" has been identified as a priority for supporting advances in cardiovascular health. This report is based on the working definition: "Resilience is the ability of living systems to successfully maintain or return to homeostasis in response to physical, molecular, individual, social, societal, or environmental stressors or challenges," developed after considering many factors contributing to cardiovascular resilience through deliberations of multidisciplinary experts convened by the National Heart, Lung, and Blood Institute during a workshop entitled: "Enhancing Resilience for Cardiovascular Health and Wellness." Some of the main emerging themes that support the possibility of enhancing resilience for cardiovascular health include optimal energy management and substrate diversity, a robust immune system that safeguards tissue homeostasis, and social and community support. The report also highlights existing research challenges, along with immediate and long-term opportunities for resilience research. Certain immediate opportunities identified are based on leveraging existing high-dimensional data from longitudinal clinical studies to identify vascular resilience measures, create a 'resilience index,' and adopt a life-course approach. Long-term opportunities include developing quantitative cell/organ/system/community models to identify resilience factors and mechanisms at these various levels, designing experimental and clinical interventions that specifically assess resilience, adopting global sharing of resilience-related data, and cross-domain training of next-generation researchers in this field.
Collapse
Affiliation(s)
- Herman A. Taylor
- Cardiovascular Research Institute Morehouse School of Medicine, Atlanta, Georgia, USA
- Morehouse-Emory Cardiovascular Center for Health Equity, Atlanta, Georgia, USA
- Harvard Chan School of Public Health, Atlanta, Georgia, USA
- Emory School of Medicine, Atlanta, Georgia, USA
| | - Toren Finkel
- University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Yunling Gao
- Division of Cardiovascular Sciences, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Scott W. Ballinger
- University of Alabama Heersink School of Medicine, Birmingham, Alabama, USA
| | - Rebecca Campo
- Division of Cardiovascular Sciences, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Rong Chen
- Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Sema4, Stamford, Connecticut, USA
| | - Shu Hui Chen
- Division of Cardiovascular Sciences, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Karina Davidson
- Feinstein Institutes for Medical Research, Northwell Health, New York, New York, USA
| | | | - Cashell Jaquish
- Division of Cardiovascular Sciences, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | | | | | - George J. Papanicolaou
- Division of Cardiovascular Sciences, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Martin Picard
- Columbia University Irving Medical Center, New York, New York, USA
- New York State Psychiatric Institute, New York, New York, USA
| | - Pothur Srinivas
- Division of Cardiovascular Sciences, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Olga Tjurmina
- Division of Cardiovascular Sciences, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Michael Wolz
- Division of Cardiovascular Sciences, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Zorina S. Galis
- Division of Cardiovascular Sciences, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
2
|
Martini AC, Gross TJ, Head E, Mapstone M. Beyond amyloid: Immune, cerebrovascular, and metabolic contributions to Alzheimer disease in people with Down syndrome. Neuron 2022; 110:2063-2079. [PMID: 35472307 PMCID: PMC9262826 DOI: 10.1016/j.neuron.2022.04.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 03/08/2022] [Accepted: 03/31/2022] [Indexed: 12/16/2022]
Abstract
People with Down syndrome (DS) have increased risk of Alzheimer disease (AD), presumably conferred through genetic predispositions arising from trisomy 21. These predispositions necessarily include triplication of the amyloid precursor protein (APP), but also other Ch21 genes that confer risk directly or through interactions with genes on other chromosomes. We discuss evidence that multiple genes on chromosome 21 are associated with metabolic dysfunction in DS. The resulting dysregulated pathways involve the immune system, leading to chronic inflammation; the cerebrovascular system, leading to disruption of the blood brain barrier (BBB); and cellular energy metabolism, promoting increased oxidative stress. In combination, these disruptions may produce a precarious biological milieu that, in the presence of accumulating amyloid, drives the pathophysiological cascade of AD in people with DS. Critically, mechanistic drivers of this dysfunction may be targetable in future clinical trials of pharmaceutical and/or lifestyle interventions.
Collapse
Affiliation(s)
- Alessandra C Martini
- Department of Pathology and Laboratory Medicine, University of California, Irvine, Irvine, CA 92697, USA
| | - Thomas J Gross
- Department of Neurology, University of California, Irvine, Irvine, CA 92697, USA
| | - Elizabeth Head
- Department of Pathology and Laboratory Medicine, University of California, Irvine, Irvine, CA 92697, USA
| | - Mark Mapstone
- Department of Neurology, University of California, Irvine, Irvine, CA 92697, USA.
| |
Collapse
|
3
|
Arvanitakis Z, Wang HY, Capuano AW, Khan A, Taïb B, Anokye-Danso F, Schneider JA, Bennett DA, Ahima RS, Arnold SE. Brain Insulin Signaling, Alzheimer Disease Pathology, and Cognitive Function. Ann Neurol 2020; 88:513-525. [PMID: 32557841 DOI: 10.1002/ana.25826] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 06/10/2020] [Accepted: 06/14/2020] [Indexed: 12/15/2022]
Abstract
OBJECTIVE To examine associations of molecular markers of brain insulin signaling with Alzheimer disease (AD) and cognition among older persons with or without diabetes. METHODS This clinical-pathologic study was derived from a community-based cohort study, the Religious Orders Study. We studied 150 individuals (mean age at death =87 years, 48% women): 75 with and 75 without diabetes (matched by sex on age at death and education). Using enzyme-linked immunosorbent assay, immunohistochemistry, and ex vivo stimulation of brain tissue with insulin, we assessed insulin signaling in the postmortem middle frontal gyrus cortex. Postmortem data documented AD neuropathology. Clinical evaluations documented cognitive function proximate to death, based on 17 neuropsychological tests. In adjusted regression analyses, we examined associations of brain insulin signaling with diabetes, AD, and level of cognition. RESULTS Brain insulin receptor substrate-1 (IRS1) phosphorylation (pS307 IRS1/total IRS1) and serine/threonine-protein kinase (AKT) phosphorylation (pT308 AKT1/total AKT1) were similar in persons with or without diabetes. AKT phosphorylation was associated with the global AD pathology score (p = 0.001). In contrast, IRS1 phosphorylation was not associated with AD (p = 0.536). No other associations of insulin signaling were found with the global AD score, including when using the ex vivo brain insulin stimulation method. In secondary analyses, normalized pT308 AKT1 was positively correlated with both the amyloid burden and tau tangle density, and no other associations of brain insulin signaling with neuropathology were observed. Moreover, normalized pT308 AKT1 was associated with a lower level of global cognitive function (estimate = -0.212, standard error = 0.097; p = 0.031). INTERPRETATION Brain AKT phosphorylation, a critical node in the signaling of insulin and other growth factors, is associated with AD neuropathology and lower cognitive function. ANN NEUROL 2020;88:513-525.
Collapse
Affiliation(s)
- Zoe Arvanitakis
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, Illinois
| | - Hoau-Yan Wang
- Department of Molecular, Cellular, and Biomedical Science, City University of New York School of Medicine, New York, New York.,Department of Biology, Neuroscience Program, Graduate School of the City University of New York, New York, New York
| | - Ana W Capuano
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, Illinois
| | - Amber Khan
- Department of Molecular, Cellular, and Biomedical Science, City University of New York School of Medicine, New York, New York.,Department of Biology, Neuroscience Program, Graduate School of the City University of New York, New York, New York
| | - Bouchra Taïb
- Division of Endocrinology, Diabetes, and Metabolism, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Frederick Anokye-Danso
- Division of Endocrinology, Diabetes, and Metabolism, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Julie A Schneider
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, Illinois
| | - David A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, Illinois
| | - Rexford S Ahima
- Division of Endocrinology, Diabetes, and Metabolism, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Steven E Arnold
- Department of Neurology and Massachusetts Alzheimer's Disease Research Center, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts
| |
Collapse
|
4
|
Candesartan Neuroprotection in Rat Primary Neurons Negatively Correlates with Aging and Senescence: a Transcriptomic Analysis. Mol Neurobiol 2019; 57:1656-1673. [PMID: 31811565 DOI: 10.1007/s12035-019-01800-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Accepted: 09/22/2019] [Indexed: 12/11/2022]
Abstract
Preclinical experiments and clinical trials demonstrated that angiotensin II AT1 receptor overactivity associates with aging and cellular senescence and that AT1 receptor blockers (ARBs) protect from age-related brain disorders. In a primary neuronal culture submitted to glutamate excitotoxicity, gene set enrichment analysis (GSEA) revealed expression of several hundred genes altered by glutamate and normalized by candesartan correlated with changes in expression in Alzheimer's patient's hippocampus. To further establish whether our data correlated with gene expression alterations associated with aging and senescence, we compared our global transcriptional data with additional published datasets, including alterations in gene expression in the neocortex and cerebellum of old mice, human frontal cortex after age of 40, gene alterations in the Werner syndrome, rodent caloric restriction, Ras and oncogene-induced senescence in fibroblasts, and to tissues besides the brain such as the muscle and kidney. The most significant and enriched pathways associated with aging and senescence were positively correlated with alterations in gene expression in glutamate-injured neurons and, conversely, negatively correlated when the injured neurons were treated with candesartan. Our results involve multiple genes and pathways, including CAV1, CCND1, CDKN1A, CHEK1, ICAM1, IL-1B, IL-6, MAPK14, PTGS2, SERPINE1, and TP53, encoding proteins associated with aging and senescence hallmarks, such as inflammation, oxidative stress, cell cycle and mitochondrial function alterations, insulin resistance, genomic instability including telomere shortening and DNA damage, and the senescent-associated secretory phenotype. Our results demonstrate that AT1 receptor blockade ameliorates central mechanisms of aging and senescence. Using ARBs for prevention and treatment of age-related disorders has important translational value.
Collapse
|
5
|
Taormina G, Ferrante F, Vieni S, Grassi N, Russo A, Mirisola MG. Longevity: Lesson from Model Organisms. Genes (Basel) 2019; 10:genes10070518. [PMID: 31324014 PMCID: PMC6678192 DOI: 10.3390/genes10070518] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Revised: 07/01/2019] [Accepted: 07/02/2019] [Indexed: 12/31/2022] Open
Abstract
Research on longevity and healthy aging promises to increase our lifespan and decrease the burden of degenerative diseases with important social and economic effects. Many aging theories have been proposed, and important aging pathways have been discovered. Model organisms have had a crucial role in this process because of their short lifespan, cheap maintenance, and manipulation possibilities. Yeasts, worms, fruit flies, or mammalian models such as mice, monkeys, and recently, dogs, have helped shed light on aging processes. Genes and molecular mechanisms that were found to be critical in simple eukaryotic cells and species have been confirmed in humans mainly by the functional analysis of mammalian orthologues. Here, we review conserved aging mechanisms discovered in different model systems that are implicated in human longevity as well and that could be the target of anti-aging interventions in human.
Collapse
Affiliation(s)
- Giusi Taormina
- Dipartimento di Discipline Chirurgiche, Oncologiche e Stomatologiche, Università di Palermo, Via del Vespro 129, 90100 Palermo, Italy
| | - Federica Ferrante
- Dipartimento di Discipline Chirurgiche, Oncologiche e Stomatologiche, Università di Palermo, Via del Vespro 129, 90100 Palermo, Italy
| | - Salvatore Vieni
- Dipartimento di Discipline Chirurgiche, Oncologiche e Stomatologiche, Università di Palermo, Via del Vespro 129, 90100 Palermo, Italy
| | - Nello Grassi
- Dipartimento di Discipline Chirurgiche, Oncologiche e Stomatologiche, Università di Palermo, Via del Vespro 129, 90100 Palermo, Italy
| | - Antonio Russo
- Dipartimento di Discipline Chirurgiche, Oncologiche e Stomatologiche, Università di Palermo, Via del Vespro 129, 90100 Palermo, Italy
| | - Mario G Mirisola
- Dipartimento di Discipline Chirurgiche, Oncologiche e Stomatologiche, Università di Palermo, Via del Vespro 129, 90100 Palermo, Italy.
| |
Collapse
|
6
|
Wahl D, Solon-Biet SM, Cogger VC, Fontana L, Simpson SJ, Le Couteur DG, Ribeiro RV. Aging, lifestyle and dementia. Neurobiol Dis 2019; 130:104481. [PMID: 31136814 DOI: 10.1016/j.nbd.2019.104481] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2018] [Revised: 05/13/2019] [Accepted: 05/22/2019] [Indexed: 12/21/2022] Open
Abstract
Aging is the greatest risk factor for most diseases including cancer, cardiovascular disorders, and neurodegenerative disease. There is emerging evidence that interventions that improve metabolic health with aging may also be effective for brain health. The most robust interventions are non-pharmacological and include limiting calorie or protein intake, increasing aerobic exercise, or environmental enrichment. In humans, dietary patterns including the Mediterranean, Finnish Geriatric Intervention Study to Prevent Cognitive Impairment and Disability (FINGER) and Okinawan diets are associated with improved age-related health and may reduce neurodegenerative disease including dementia. Rapamycin, metformin and resveratrol act on nutrient sensing pathways that improve cardiometabolic health and decrease the risk for age-associated disease. There is some evidence that they may reduce the risk for dementia in rodents. There is a growing recognition that improving metabolic function may be an effective way to optimize brain health during aging.
Collapse
Affiliation(s)
- Devin Wahl
- Charles Perkins Centre, University of Sydney, Sydney 2006, Australia; Aging and Alzheimers Institute, ANZAC Research Institute, Concord Clinical School/Sydney Medical School, Concord 2139, Australia.
| | - Samantha M Solon-Biet
- Charles Perkins Centre, University of Sydney, Sydney 2006, Australia; Aging and Alzheimers Institute, ANZAC Research Institute, Concord Clinical School/Sydney Medical School, Concord 2139, Australia
| | - Victoria C Cogger
- Charles Perkins Centre, University of Sydney, Sydney 2006, Australia; Aging and Alzheimers Institute, ANZAC Research Institute, Concord Clinical School/Sydney Medical School, Concord 2139, Australia
| | - Luigi Fontana
- Charles Perkins Centre, University of Sydney, Sydney 2006, Australia
| | - Stephen J Simpson
- Charles Perkins Centre, University of Sydney, Sydney 2006, Australia; School of Life and Environmental Sciences, University of Sydney, Sydney 2006, Australia
| | - David G Le Couteur
- Charles Perkins Centre, University of Sydney, Sydney 2006, Australia; Aging and Alzheimers Institute, ANZAC Research Institute, Concord Clinical School/Sydney Medical School, Concord 2139, Australia
| | - Rosilene V Ribeiro
- Charles Perkins Centre, University of Sydney, Sydney 2006, Australia; School of Life and Environmental Sciences, University of Sydney, Sydney 2006, Australia
| |
Collapse
|
7
|
Andrews RN, Dugan GO, Peiffer AM, Hawkins GA, Hanbury DB, Bourland JD, Hampson RE, Deadwyler SA, Cline JM. White Matter is the Predilection Site of Late-Delayed Radiation-Induced Brain Injury in Non-Human Primates. Radiat Res 2019; 191:217-231. [PMID: 30694733 PMCID: PMC6422025 DOI: 10.1667/rr15263.1] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Fractionated whole-brain irradiation for the treatment of intracranial neoplasia causes progressive neurodegeneration and neuroinflammation. The long-term consequences of single-fraction high-dose irradiation to the brain are unknown. To assess the late effects of brain irradiation we compared transcriptomic gene expression profiles from nonhuman primates (NHP; rhesus macaques Macaca mulatta) receiving single-fraction total-body irradiation (TBI; n = 5, 6.75-8.05 Gy, 6-9 years prior to necropsy) to those receiving fractionated whole-brain irradiation (fWBI; n = 5, 40 Gy, 8 × 5 Gy fractions; 12 months prior to necropsy) and control comparators (n = 5). Gene expression profiles from the dorsolateral prefrontal cortex (DLPFC), hippocampus (HC) and deep white matter (WM; centrum semiovale) were compared. Stratified analyses by treatment and region revealed that radiation-induced transcriptomic alterations were most prominent in animals receiving fWBI, and primarily affected white matter in both TBI and fWBI groups. Unsupervised canonical and ontologic analysis revealed that TBI or fWBI animals demonstrated shared patterns of injury, including white matter neuroinflammation, increased expression of complement factors and T-cell activation. Both irradiated groups also showed evidence of impaired glutamatergic neurotransmission and signal transduction within white matter, but not within the dorsolateral prefrontal cortex or hippocampus. Signaling pathways and structural elements involved in extracellular matrix (ECM) deposition and remodeling were noted within the white matter of animals receiving fWBI, but not of those receiving TBI. These findings indicate that those animals receiving TBI are susceptible to neurological injury similar to that observed after fWBI, and these changes persist for years postirradiation. Transcriptomic profiling reaffirmed that macrophage/microglial-mediated neuroinflammation is present in radiation-induced brain injury (RIBI), and our data provide novel evidence that the complement system may contribute to the pathogenesis of RIBI. Finally, these data challenge the assumption that the hippocampus is the predilection site of injury in RIBI, and indicate that impaired glutamatergic neurotransmission may occur in white matter injury.
Collapse
Affiliation(s)
- Rachel N. Andrews
- Departments of Pathology, Section on Comparative Medicine, Wake Forest University School of Medicine, Medical Center Boulevard, Winston-Salem, North Carolina 27157
| | - Gregory O. Dugan
- Departments of Pathology, Section on Comparative Medicine, Wake Forest University School of Medicine, Medical Center Boulevard, Winston-Salem, North Carolina 27157
| | - Ann M. Peiffer
- Departments of Radiation Oncology, Wake Forest University School of Medicine, Medical Center Boulevard, Winston-Salem, North Carolina 27157
- Departments of Brain Tumor Center of Excellence, Wake Forest University School of Medicine, Medical Center Boulevard, Winston-Salem, North Carolina 27157
| | - Gregory A. Hawkins
- Departments of Biochemistry, Wake Forest University School of Medicine, Medical Center Boulevard, Winston-Salem, North Carolina 27157
- Departments of Wake Forest Baptist Comprehensive Cancer Center, Wake Forest University School of Medicine, Medical Center Boulevard, Winston-Salem, North Carolina 27157
| | - David B. Hanbury
- Department of Psychology, Averett University, Danville, Virginia 24541
| | - J. Daniel Bourland
- Departments of Radiation Oncology, Wake Forest University School of Medicine, Medical Center Boulevard, Winston-Salem, North Carolina 27157
- Departments of Brain Tumor Center of Excellence, Wake Forest University School of Medicine, Medical Center Boulevard, Winston-Salem, North Carolina 27157
| | - Robert E. Hampson
- Departments of Physiology and Pharmacology, Wake Forest University School of Medicine, Medical Center Boulevard, Winston-Salem, North Carolina 27157
| | - Samuel A. Deadwyler
- Departments of Physiology and Pharmacology, Wake Forest University School of Medicine, Medical Center Boulevard, Winston-Salem, North Carolina 27157
| | - J. Mark Cline
- Departments of Pathology, Section on Comparative Medicine, Wake Forest University School of Medicine, Medical Center Boulevard, Winston-Salem, North Carolina 27157
- Departments of Radiation Oncology, Wake Forest University School of Medicine, Medical Center Boulevard, Winston-Salem, North Carolina 27157
| |
Collapse
|
8
|
FK506-Binding Protein 12.6/1b, a Negative Regulator of [Ca 2+], Rescues Memory and Restores Genomic Regulation in the Hippocampus of Aging Rats. J Neurosci 2017; 38:1030-1041. [PMID: 29255009 PMCID: PMC5783960 DOI: 10.1523/jneurosci.2234-17.2017] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Revised: 10/10/2017] [Accepted: 11/24/2017] [Indexed: 12/20/2022] Open
Abstract
Hippocampal overexpression of FK506-binding protein 12.6/1b (FKBP1b), a negative regulator of ryanodine receptor Ca2+ release, reverses aging-induced memory impairment and neuronal Ca2+ dysregulation. Here, we tested the hypothesis that FKBP1b also can protect downstream transcriptional networks from aging-induced dysregulation. We gave hippocampal microinjections of FKBP1b-expressing viral vector to male rats at either 13 months of age (long-term, LT) or 19 months of age (short-term, ST) and tested memory performance in the Morris water maze at 21 months of age. Aged rats treated ST or LT with FKBP1b substantially outperformed age-matched vector controls and performed similarly to each other and young controls (YCs). Transcriptional profiling in the same animals identified 2342 genes with hippocampal expression that was upregulated/downregulated in aged controls (ACs) compared with YCs (the aging effect). Of these aging-dependent genes, 876 (37%) also showed altered expression in aged FKBP1b-treated rats compared with ACs, with FKBP1b restoring expression of essentially all such genes (872/876, 99.5%) in the direction opposite the aging effect and closer to levels in YCs. This inverse relationship between the aging and FKBP1b effects suggests that the aging effects arise from FKBP1b deficiency. Functional category analysis revealed that genes downregulated with aging and restored by FKBP1b were associated predominantly with diverse brain structure categories, including cytoskeleton, membrane channels, and extracellular region. Conversely, genes upregulated with aging but not restored by FKBP1b associated primarily with glial–neuroinflammatory, ribosomal, and lysosomal categories. Immunohistochemistry confirmed aging-induced rarefaction and FKBP1b-mediated restoration of neuronal microtubular structure. Therefore, a previously unrecognized genomic network modulating diverse brain structural processes is dysregulated by aging and restored by FKBP1b overexpression. SIGNIFICANCE STATEMENT Previously, we found that hippocampal overexpression of FK506-binding protein 12.6/1b (FKBP1b), a negative regulator of intracellular Ca2+ responses, reverses both aging-related Ca2+ dysregulation and cognitive impairment. Here, we tested whether hippocampal FKBP1b overexpression also counteracts aging changes in gene transcriptional networks. In addition to reducing memory deficits in aged rats, FKBP1b selectively counteracted aging-induced expression changes in 37% of aging-dependent genes, with cytoskeletal and extracellular structure categories highly associated with the FKBP1b-rescued genes. Our results indicate that, in parallel with cognitive processes, a novel transcriptional network coordinating brain structural organization is dysregulated with aging and restored by FKBP1b.
Collapse
|
9
|
Ianov L, De Both M, Chawla MK, Rani A, Kennedy AJ, Piras I, Day JJ, Siniard A, Kumar A, Sweatt JD, Barnes CA, Huentelman MJ, Foster TC. Hippocampal Transcriptomic Profiles: Subfield Vulnerability to Age and Cognitive Impairment. Front Aging Neurosci 2017; 9:383. [PMID: 29276487 PMCID: PMC5727020 DOI: 10.3389/fnagi.2017.00383] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Accepted: 11/07/2017] [Indexed: 01/11/2023] Open
Abstract
The current study employed next-generation RNA sequencing to examine gene expression differences related to brain aging, cognitive decline, and hippocampal subfields. Young and aged rats were trained on a spatial episodic memory task. Hippocampal regions CA1, CA3, and the dentate gyrus were isolated. Poly-A mRNA was examined using two different sequencing platforms, Illumina, and Ion Proton. The Illumina platform was used to generate seed lists of genes that were statistically differentially expressed across regions, ages, or in association with cognitive function. The gene lists were then retested using the data from the Ion Proton platform. The results indicate hippocampal subfield differences in gene expression and point to regional differences in vulnerability to aging. Aging was associated with increased expression of immune response-related genes, particularly in the dentate gyrus. For the memory task, impaired performance of aged animals was linked to the regulation of Ca2+ and synaptic function in region CA1. Finally, we provide a transcriptomic characterization of the three subfields regardless of age or cognitive status, highlighting and confirming a correspondence between cytoarchitectural boundaries and molecular profiling.
Collapse
Affiliation(s)
- Lara Ianov
- Departments of Neuroscience and Genetics and Genomics Program, Evelyn F. and William L. McKnight Brain Institute, University of Florida, Gainesville, FL, United States.,Civitan International Research Center, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Matt De Both
- Neurogenomics Division, Translational Genomics Research Institute, Phoenix, AZ, United States
| | - Monica K Chawla
- Evelyn F. McKnight Brain Institute, University of Arizona, Tucson, AZ, United States
| | - Asha Rani
- Departments of Neuroscience and Genetics and Genomics Program, Evelyn F. and William L. McKnight Brain Institute, University of Florida, Gainesville, FL, United States
| | - Andrew J Kennedy
- Evelyn F. McKnight Brain Institute, University of Alabama, Birmingham, AL, United States
| | - Ignazio Piras
- Neurogenomics Division, Translational Genomics Research Institute, Phoenix, AZ, United States
| | - Jeremy J Day
- Evelyn F. McKnight Brain Institute, University of Alabama, Birmingham, AL, United States
| | - Ashley Siniard
- Neurogenomics Division, Translational Genomics Research Institute, Phoenix, AZ, United States
| | - Ashok Kumar
- Departments of Neuroscience and Genetics and Genomics Program, Evelyn F. and William L. McKnight Brain Institute, University of Florida, Gainesville, FL, United States
| | - J David Sweatt
- Evelyn F. McKnight Brain Institute, University of Alabama, Birmingham, AL, United States.,Department of Pharmacology, Vanderbilt University, Nashville, TN, United States
| | - Carol A Barnes
- Evelyn F. McKnight Brain Institute, University of Arizona, Tucson, AZ, United States.,Departments of Psychology, Neurology and Neuroscience, University of Arizona, Tucson, AZ, United States
| | - Matthew J Huentelman
- Neurogenomics Division, Translational Genomics Research Institute, Phoenix, AZ, United States.,Evelyn F. McKnight Brain Institute, University of Arizona, Tucson, AZ, United States
| | - Thomas C Foster
- Departments of Neuroscience and Genetics and Genomics Program, Evelyn F. and William L. McKnight Brain Institute, University of Florida, Gainesville, FL, United States
| |
Collapse
|
10
|
Mangold CA, Wronowski B, Du M, Masser DR, Hadad N, Bixler GV, Brucklacher RM, Ford MM, Sonntag WE, Freeman WM. Sexually divergent induction of microglial-associated neuroinflammation with hippocampal aging. J Neuroinflammation 2017; 14:141. [PMID: 28732515 PMCID: PMC5521082 DOI: 10.1186/s12974-017-0920-8] [Citation(s) in RCA: 122] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Accepted: 07/13/2017] [Indexed: 01/11/2023] Open
Abstract
Background The necessity of including both males and females in molecular neuroscience research is now well understood. However, there is relatively limited basic biological data on brain sex differences across the lifespan despite the differences in age-related neurological dysfunction and disease between males and females. Methods Whole genome gene expression of young (3 months), adult (12 months), and old (24 months) male and female C57BL6 mice hippocampus was analyzed. Subsequent bioinformatic analyses and confirmations of age-related changes and sex differences in hippocampal gene and protein expression were performed. Results Males and females demonstrate both common expression changes with aging and marked sex differences in the nature and magnitude of the aging responses. Age-related hippocampal induction of neuroinflammatory gene expression was sexually divergent and enriched for microglia-specific genes such as complement pathway components. Sexually divergent C1q protein expression was confirmed by immunoblotting and immunohistochemistry. Similar patterns of cortical sexually divergent gene expression were also evident. Additionally, inter-animal gene expression variability increased with aging in males, but not females. Conclusions These findings demonstrate sexually divergent neuroinflammation with aging that may contribute to sex differences in age-related neurological diseases such as stroke and Alzheimer’s, specifically in the complement system. The increased expression variability in males suggests a loss of fidelity in gene expression regulation with aging. These findings reveal a central role of sex in the transcriptomic response of the hippocampus to aging that warrants further, in depth, investigations. Electronic supplementary material The online version of this article (doi:10.1186/s12974-017-0920-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Colleen A Mangold
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, State College, PA, USA
| | - Benjamin Wronowski
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Mei Du
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Dustin R Masser
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.,Reynolds Oklahoma Center on Aging & Nathan Shock Center of Excellence in the Biology of Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Niran Hadad
- Reynolds Oklahoma Center on Aging & Nathan Shock Center of Excellence in the Biology of Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.,Oklahoma Center for Neuroscience, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Georgina V Bixler
- Genome Sciences Facility, Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Robert M Brucklacher
- Genome Sciences Facility, Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Matthew M Ford
- Division of Neuroscience, Oregon National Primate Research Center, Beaverton, Oregon, USA
| | - William E Sonntag
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.,Reynolds Oklahoma Center on Aging & Nathan Shock Center of Excellence in the Biology of Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.,Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, USA
| | - Willard M Freeman
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA. .,Reynolds Oklahoma Center on Aging & Nathan Shock Center of Excellence in the Biology of Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA. .,Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, USA. .,, SLY-BRC 1370, 975 NE 10th St, Oklahoma City, OK, 73104, USA.
| |
Collapse
|
11
|
Omics analysis of mouse brain models of human diseases. Gene 2017; 600:90-100. [DOI: 10.1016/j.gene.2016.11.022] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Revised: 11/04/2016] [Accepted: 11/10/2016] [Indexed: 01/24/2023]
|
12
|
Frazier HN, Maimaiti S, Anderson KL, Brewer LD, Gant JC, Porter NM, Thibault O. Calcium's role as nuanced modulator of cellular physiology in the brain. Biochem Biophys Res Commun 2016; 483:981-987. [PMID: 27553276 DOI: 10.1016/j.bbrc.2016.08.105] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Revised: 08/04/2016] [Accepted: 08/18/2016] [Indexed: 12/22/2022]
Abstract
Neuroscientists studying normal brain aging, spinal cord injury, Alzheimer's disease (AD) and other neurodegenerative diseases have focused considerable effort on carefully characterizing intracellular perturbations in calcium dynamics or levels. At the cellular level, calcium is known for controlling life and death and orchestrating most events in between. For many years, intracellular calcium has been recognized as an essential ion associated with nearly all cellular functions from cell growth to degeneration. Often the emphasis is on the negative impact of calcium dysregulation and the typical worse-case-scenario leading inevitably to cell death. However, even high amplitude calcium transients, when executed acutely, can alter neuronal communication and synaptic strength in positive ways, without necessarily killing neurons. Here, we focus on the evidence that calcium has a subtle and distinctive role in shaping and controlling synaptic events that underpin neuronal communication and that these subtle changes in aging or AD may contribute to cognitive decline. We emphasize that calcium imaging in dendritic components is ultimately necessary to directly test for the presence of age- or disease-associated alterations during periods of synaptic activation.
Collapse
Affiliation(s)
- Hilaree N Frazier
- UKMC, MS-313, Department of Pharmacology and Nutritional Sciences, University of Kentucky Medical Center, 800 Rose Street, Lexington, KY 40536, USA
| | - Shaniya Maimaiti
- UKMC, MS-313, Department of Pharmacology and Nutritional Sciences, University of Kentucky Medical Center, 800 Rose Street, Lexington, KY 40536, USA
| | - Katie L Anderson
- UKMC, MS-313, Department of Pharmacology and Nutritional Sciences, University of Kentucky Medical Center, 800 Rose Street, Lexington, KY 40536, USA
| | - Lawrence D Brewer
- UKMC, MS-313, Department of Pharmacology and Nutritional Sciences, University of Kentucky Medical Center, 800 Rose Street, Lexington, KY 40536, USA
| | - John C Gant
- UKMC, MS-313, Department of Pharmacology and Nutritional Sciences, University of Kentucky Medical Center, 800 Rose Street, Lexington, KY 40536, USA
| | - Nada M Porter
- UKMC, MS-313, Department of Pharmacology and Nutritional Sciences, University of Kentucky Medical Center, 800 Rose Street, Lexington, KY 40536, USA
| | - Olivier Thibault
- UKMC, MS-313, Department of Pharmacology and Nutritional Sciences, University of Kentucky Medical Center, 800 Rose Street, Lexington, KY 40536, USA.
| |
Collapse
|
13
|
Daulatzai MA. Dysfunctional Sensory Modalities, Locus Coeruleus, and Basal Forebrain: Early Determinants that Promote Neuropathogenesis of Cognitive and Memory Decline and Alzheimer’s Disease. Neurotox Res 2016; 30:295-337. [DOI: 10.1007/s12640-016-9643-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Revised: 06/08/2016] [Accepted: 06/10/2016] [Indexed: 12/22/2022]
|
14
|
Pandya JD, Royland JE, MacPhail RC, Sullivan PG, Kodavanti PRS. Age- and brain region-specific differences in mitochondrial bioenergetics in Brown Norway rats. Neurobiol Aging 2016; 42:25-34. [PMID: 27143418 DOI: 10.1016/j.neurobiolaging.2016.02.027] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Revised: 02/09/2016] [Accepted: 02/25/2016] [Indexed: 02/03/2023]
Abstract
Mitochondria are central regulators of energy homeostasis and play a pivotal role in mechanisms of cellular senescence. The objective of the present study was to evaluate mitochondrial bioenergetic parameters in 5 brain regions (brain stem [BS], frontal cortex, cerebellum, striatum, hippocampus [HIP]) of 4 diverse age groups (1 month [young], 4 months [adult], 12 months [middle-aged], 24 months [old age]) to understand age-related differences in selected brain regions and their possible contribution to age-related chemical sensitivity. Mitochondrial bioenergetic parameters and enzyme activities were measured under identical conditions across multiple age groups and brain regions in Brown Norway rats (n = 5/group). The results indicate age- and brain region-specific patterns in mitochondrial functional endpoints. For example, an age-specific decline in ATP synthesis (State III respiration) was observed in BS and HIP. Similarly, the maximal respiratory capacities (State V1 and V2) showed age-specific declines in all brain regions examined (young > adult > middle-aged > old age). Amongst all regions, HIP had the greatest change in mitochondrial bioenergetics, showing declines in the 4, 12, and 24-months age groups. Activities of mitochondrial pyruvate dehydrogenase complex and electron transport chain complexes I, II, and IV enzymes were also age and brain region specific. In general, changes associated with age were more pronounced with enzyme activities declining as the animals aged (young > adult > middle-aged > old age). These age- and brain region-specific observations may aid in evaluating brain bioenergetic impact on the age-related susceptibility to environmental chemical stressors.
Collapse
Affiliation(s)
- Jignesh D Pandya
- Spinal Cord and Brain Injury Research Center, Department of Anatomy and Neurobiology, University of Kentucky, Lexington, KY, USA
| | - Joyce E Royland
- Genetic and Cellular Toxicology Branch, Integrated Systems Toxicology Division, NHEERL/ORD, U.S. Environmental Protection Agency, Research Triangle Park, NC, USA
| | - Robert C MacPhail
- Neurotoxicology Branch, Toxicity Assessment Division, NHEERL/ORD, U.S. Environmental Protection Agency, Research Triangle Park, NC, USA
| | - Patrick G Sullivan
- Spinal Cord and Brain Injury Research Center, Department of Anatomy and Neurobiology, University of Kentucky, Lexington, KY, USA
| | - Prasada Rao S Kodavanti
- Neurotoxicology Branch, Toxicity Assessment Division, NHEERL/ORD, U.S. Environmental Protection Agency, Research Triangle Park, NC, USA.
| |
Collapse
|
15
|
Didier ES, MacLean AG, Mohan M, Didier PJ, Lackner AA, Kuroda MJ. Contributions of Nonhuman Primates to Research on Aging. Vet Pathol 2016; 53:277-90. [PMID: 26869153 PMCID: PMC5027759 DOI: 10.1177/0300985815622974] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Aging is the biological process of declining physiologic function associated with increasing mortality rate during advancing age. Humans and higher nonhuman primates exhibit unusually longer average life spans as compared with mammals of similar body mass. Furthermore, the population of humans worldwide is growing older as a result of improvements in public health, social services, and health care systems. Comparative studies among a wide range of organisms that include nonhuman primates contribute greatly to our understanding about the basic mechanisms of aging. Based on their genetic and physiologic relatedness to humans, nonhuman primates are especially important for better understanding processes of aging unique to primates, as well as for testing intervention strategies to improve healthy aging and to treat diseases and disabilities in older people. Rhesus and cynomolgus macaques are the predominant monkeys used in studies on aging, but research with lower nonhuman primate species is increasing. One of the priority topics of research about aging in nonhuman primates involves neurologic changes associated with cognitive decline and neurodegenerative diseases. Additional areas of research include osteoporosis, reproductive decline, caloric restriction, and their mimetics, as well as immune senescence and chronic inflammation that affect vaccine efficacy and resistance to infections and cancer. The purpose of this review is to highlight the findings from nonhuman primate research that contribute to our understanding about aging and health span in humans.
Collapse
Affiliation(s)
- E S Didier
- Division of Microbiology, Tulane National Primate Research Center, Covington, LA, USA
| | - A G MacLean
- Division of Comparative Pathology, Tulane National Primate Research Center, Covington, LA, USA
| | - M Mohan
- Division of Comparative Pathology, Tulane National Primate Research Center, Covington, LA, USA
| | - P J Didier
- Division of Comparative Pathology, Tulane National Primate Research Center, Covington, LA, USA
| | - A A Lackner
- Division of Comparative Pathology, Tulane National Primate Research Center, Covington, LA, USA
| | - M J Kuroda
- Division of Immunology, Tulane National Primate Research Center, Covington, LA, USA
| |
Collapse
|
16
|
Klosinski LP, Yao J, Yin F, Fonteh AN, Harrington MG, Christensen TA, Trushina E, Brinton RD. White Matter Lipids as a Ketogenic Fuel Supply in Aging Female Brain: Implications for Alzheimer's Disease. EBioMedicine 2015; 2:1888-904. [PMID: 26844268 PMCID: PMC4703712 DOI: 10.1016/j.ebiom.2015.11.002] [Citation(s) in RCA: 104] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Revised: 10/24/2015] [Accepted: 11/02/2015] [Indexed: 01/28/2023] Open
Abstract
White matter degeneration is a pathological hallmark of neurodegenerative diseases including Alzheimer's. Age remains the greatest risk factor for Alzheimer's and the prevalence of age-related late onset Alzheimer's is greatest in females. We investigated mechanisms underlying white matter degeneration in an animal model consistent with the sex at greatest Alzheimer's risk. Results of these analyses demonstrated decline in mitochondrial respiration, increased mitochondrial hydrogen peroxide production and cytosolic-phospholipase-A2 sphingomyelinase pathway activation during female brain aging. Electron microscopic and lipidomic analyses confirmed myelin degeneration. An increase in fatty acids and mitochondrial fatty acid metabolism machinery was coincident with a rise in brain ketone bodies and decline in plasma ketone bodies. This mechanistic pathway and its chronologically phased activation, links mitochondrial dysfunction early in aging with later age development of white matter degeneration. The catabolism of myelin lipids to generate ketone bodies can be viewed as a systems level adaptive response to address brain fuel and energy demand. Elucidation of the initiating factors and the mechanistic pathway leading to white matter catabolism in the aging female brain provides potential therapeutic targets to prevent and treat demyelinating diseases such as Alzheimer's and multiple sclerosis. Targeting stages of disease and associated mechanisms will be critical. Mitochondrial dysfunction activates mechanisms for catabolism of myelin lipids to generate ketone bodies for ATP production. Mechanisms leading to ketone body driven energy production in brain coincide with stages of reproductive aging in females. Sequential activation of myelin catabolism pathway during aging provides multiple therapeutic targets and windows of efficacy.
The mechanisms underlying white matter degeneration, a hallmark of multiple neurodegenerative diseases including Alzheimer's, remain unclear. Herein we provide a mechanistic pathway, spanning multiple transitions of aging, that links mitochondrial dysfunction early in aging with later age white matter degeneration. Catabolism of myelin lipids to generate ketone bodies can be viewed as an adaptive survival response to address brain fuel and energy demand. Women are at greatest risk of late-onset-AD, thus, our analyses in female brain address mechanisms of AD pathology and therapeutic targets to prevent, delay and treat AD in the sex most affected with potential relevance to men.
Collapse
Key Words
- ABAD, Aβ-binding alcohol dehydrogenase
- ABAD, Aβ-binding-alcohol-dehydrogenase
- ACER3, alkaline ceramidase
- AD, Alzheimer's disease
- APO-ε4, apolipoprotein ε4
- APP, amyloid precursor protein
- Aging oxidative stress
- Alzheimer's disease
- BACE1, beta-secretase 1
- BBB, blood brain barrier
- CC, corpus callosum
- CMRglu, cerebral glucose metabolic rate
- COX, complex IV cytochrome c oxidase
- CPT1, carnitine palmitoyltransferase 1
- Cldn11, claudin 11
- Cyp2j6, arachidonic acid epoxygenase
- Cytosolic phospholipase A2
- DHA, docosahexaesnoic acid
- Erbb3, Erb-B2 receptor tyrosine kinase 3
- FDG-PET, 2-[18F]fluoro-2-deoxy-d-glucose
- GFAP, glial fibrillary acidic protein
- H2O2, hydrogen peroxide
- HADHA, hydroxyacyl-CoA dehydrogenase
- HK, hexokinase
- Ketone bodies
- LC MS, liquid chromatography mass spectrometer
- MAG, myelin associated glycoprotein
- MBP, myelin basic protein
- MCT1, monocarboxylate transporter 1
- MIB, mitochondrial isolation buffer
- MOG, myelin oligodendrocyte glycoprotein
- MTL, medial temporal lobe
- Mitochondria
- NEFA, nonesterified fatty acids
- Neurodegeneration
- OCR, oxygen consumption rate
- Olig2, oligodendrocyte transcription factor
- PB, phosphate buffer
- PCC, posterior cingulate
- PCR, polymerase chain reaction
- PDH, pyruvate dehydrogenase
- PEI, polyethyleneimine
- RCR, respiratory control ratio
- ROS, reactive oxygen species
- S1P, sphingosine
- TLDA, TaqMan low density array
- WM, white matter
- WT, wild type
- White matter
- cPLA2, cytosolic phospholipase A2
Collapse
Affiliation(s)
- Lauren P Klosinski
- Department of Neuroscience, Dornsife College of Letters, Arts and Sciences, University of Southern California, Los Angeles, CA, USA
| | - Jia Yao
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA, USA
| | - Fei Yin
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA, USA
| | | | | | | | - Eugenia Trushina
- Department of Neurology, Mayo Clinic Rochester, MN, USA; Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
| | - Roberta Diaz Brinton
- Department of Neuroscience, Dornsife College of Letters, Arts and Sciences, University of Southern California, Los Angeles, CA, USA; Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA, USA; Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
17
|
Park CS, Valomon A, Welzl H. Integrative Transcriptome Profiling of Cognitive Aging and Its Preservation through Ser/Thr Protein Phosphatase Regulation. PLoS One 2015; 10:e0130891. [PMID: 26102285 PMCID: PMC4478024 DOI: 10.1371/journal.pone.0130891] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Accepted: 05/26/2015] [Indexed: 01/28/2023] Open
Abstract
Environmental enrichment has been reported to delay or restore age-related cognitive deficits, however, a mechanism to account for the cause and progression of normal cognitive decline and its preservation by environmental enrichment is lacking. Using genome-wide SAGE-Seq, we provide a global assessment of differentially expressed genes altered with age and environmental enrichment in the hippocampus. Qualitative and quantitative proteomics in naïve young and aged mice was used to further identify phosphorylated proteins differentially expressed with age. We found that increased expression of endogenous protein phosphatase-1 inhibitors in aged mice may be characteristic of long-term environmental enrichment and improved cognitive status. As such, hippocampus-dependent performances in spatial, recognition, and associative memories, which are sensitive to aging, were preserved by environmental enrichment and accompanied by decreased protein phosphatase activity. Age-associated phosphorylated proteins were also found to correspond to the functional categories of age-associated genes identified through transcriptome analysis. Together, this study provides a comprehensive map of the transcriptome and proteome in the aging brain, and elucidates endogenous protein phosphatase-1 inhibition as a potential means through which environmental enrichment may ameliorate age-related cognitive deficits.
Collapse
Affiliation(s)
- C. Sehwan Park
- Brain Research Institute, Medical Faculty of the University of Zürich and Department of Health Science and Technology ETH Zürich, Zürich, Switzerland
| | - Amandine Valomon
- Brain Research Institute, Medical Faculty of the University of Zürich and Department of Health Science and Technology ETH Zürich, Zürich, Switzerland
- Institute of Pharmacology and Toxicology, Human Sleep Psychopharmacology, University of Zürich, Zürich, Switzerland
| | - Hans Welzl
- Institute of Anatomy, Neuroanatomy and Behavior, University of Zürich, Zürich, Switzerland
| |
Collapse
|
18
|
Dong W, Guo W, Zheng X, Wang F, Chen Y, Zhang W, Shi H. Electroacupuncture improves cognitive deficits associated with AMPK activation in SAMP8 mice. Metab Brain Dis 2015; 30:777-84. [PMID: 25502012 DOI: 10.1007/s11011-014-9641-1] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2014] [Accepted: 12/05/2014] [Indexed: 10/24/2022]
Abstract
Perturbations of brain energy metabolism are involved in Alzheimer's disease (AD). Adenosine monophosphate-activated kinase (AMPK) is a master energy sensor that monitors the levels of key energy metabolites. Electroacupuncture (EA) has demonstrated therapeutic potential for the treatment of AD. The effects of EA on cognitive functions and the changes of AMPK and its phosphorylated form (p-AMPK) expression were investigated in senescence-accelerated mouse prone 8 (SAMP8) mice. Cognitive function of SAMP8 mice was assessed using Morris water maze test after EA treatment. Then mice were sacrificed for immunohistochemistry and western blot analysis. EA stimulation significantly alleviated memory impairment of AD mice, and increased the levels of p-AMPK in the hippocampus. These results suggest that EA improved cognitive function associated with AMPK activation, AMPK may be a molecular target of EA in treating AD.
Collapse
Affiliation(s)
- Weiguo Dong
- Department of Integrated Traditional Chinese and Western Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, Peoples Republic of China,
| | | | | | | | | | | | | |
Collapse
|
19
|
Yin F, Yao J, Sancheti H, Feng T, Melcangi RC, Morgan TE, Finch CE, Pike CJ, Mack WJ, Cadenas E, Brinton RD. The perimenopausal aging transition in the female rat brain: decline in bioenergetic systems and synaptic plasticity. Neurobiol Aging 2015; 36:2282-2295. [PMID: 25921624 DOI: 10.1016/j.neurobiolaging.2015.03.013] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Revised: 02/20/2015] [Accepted: 03/25/2015] [Indexed: 01/03/2023]
Abstract
The perimenopause is an aging transition unique to the female that leads to reproductive senescence which can be characterized by multiple neurological symptoms. To better understand potential underlying mechanisms of neurological symptoms of perimenopause, the present study determined genomic, biochemical, brain metabolic, and electrophysiological transformations that occur during this transition using a rat model recapitulating fundamental characteristics of the human perimenopause. Gene expression analyses indicated two distinct aging programs: chronological and endocrine. A critical period emerged during the endocrine transition from regular to irregular cycling characterized by decline in bioenergetic gene expression, confirmed by deficits in fluorodeoxyglucose-positron emission tomography (FDG-PET) brain metabolism, mitochondrial function, and long-term potentiation. Bioinformatic analysis predicted insulin/insulin-like growth factor 1 and adenosine monophosphate-activated protein kinase/peroxisome proliferator-activated receptor gamma coactivator 1 alpha (AMPK/PGC1α) signaling pathways as upstream regulators. Onset of acyclicity was accompanied by a rise in genes required for fatty acid metabolism, inflammation, and mitochondrial function. Subsequent chronological aging resulted in decline of genes required for mitochondrial function and β-amyloid degradation. Emergence of glucose hypometabolism and impaired synaptic function in brain provide plausible mechanisms of neurological symptoms of perimenopause and may be predictive of later-life vulnerability to hypometabolic conditions such as Alzheimer's.
Collapse
Affiliation(s)
- Fei Yin
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA, USA
| | - Jia Yao
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA, USA
| | - Harsh Sancheti
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA, USA
| | - Tao Feng
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Roberto C Melcangi
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Todd E Morgan
- Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Caleb E Finch
- Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Christian J Pike
- Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Wendy J Mack
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Enrique Cadenas
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA, USA
| | - Roberta D Brinton
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA, USA; Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.
| |
Collapse
|
20
|
Maimaiti S, Anderson KL, DeMoll C, Brewer LD, Rauh BA, Gant JC, Blalock EM, Porter NM, Thibault O. Intranasal Insulin Improves Age-Related Cognitive Deficits and Reverses Electrophysiological Correlates of Brain Aging. J Gerontol A Biol Sci Med Sci 2015; 71:30-9. [PMID: 25659889 DOI: 10.1093/gerona/glu314] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2014] [Accepted: 12/23/2014] [Indexed: 12/25/2022] Open
Abstract
Peripheral insulin resistance is a key component of metabolic syndrome associated with obesity, dyslipidemia, hypertension, and type 2 diabetes. While the impact of insulin resistance is well recognized in the periphery, it is also becoming apparent in the brain. Recent studies suggest that insulin resistance may be a factor in brain aging and Alzheimer's disease (AD) whereby intranasal insulin therapy, which delivers insulin to the brain, improves cognition and memory in AD patients. Here, we tested a clinically relevant delivery method to determine the impact of two forms of insulin, short-acting insulin lispro (Humalog) or long-acting insulin detemir (Levemir), on cognitive functions in aged F344 rats. We also explored insulin effects on the Ca(2+)-dependent hippocampal afterhyperpolarization (AHP), a well-characterized neurophysiological marker of aging which is increased in the aged, memory impaired animal. Low-dose intranasal insulin improved memory recall in aged animals such that their performance was similar to that seen in younger animals. Further, because ex vivo insulin also reduced the AHP, our results suggest that the AHP may be a novel cellular target of insulin in the brain, and improved cognitive performance following intranasal insulin therapy may be the result of insulin actions on the AHP.
Collapse
Affiliation(s)
- Shaniya Maimaiti
- Department of Pharmacology and Nutritional Sciences, University of Kentucky Medical Center, Lexington, Kentucky
| | - Katie L Anderson
- Department of Pharmacology and Nutritional Sciences, University of Kentucky Medical Center, Lexington, Kentucky
| | - Chris DeMoll
- Department of Pharmacology and Nutritional Sciences, University of Kentucky Medical Center, Lexington, Kentucky
| | - Lawrence D Brewer
- Department of Pharmacology and Nutritional Sciences, University of Kentucky Medical Center, Lexington, Kentucky
| | - Benjamin A Rauh
- Department of Pharmacology and Nutritional Sciences, University of Kentucky Medical Center, Lexington, Kentucky
| | - John C Gant
- Department of Pharmacology and Nutritional Sciences, University of Kentucky Medical Center, Lexington, Kentucky
| | - Eric M Blalock
- Department of Pharmacology and Nutritional Sciences, University of Kentucky Medical Center, Lexington, Kentucky
| | - Nada M Porter
- Department of Pharmacology and Nutritional Sciences, University of Kentucky Medical Center, Lexington, Kentucky
| | - Olivier Thibault
- Department of Pharmacology and Nutritional Sciences, University of Kentucky Medical Center, Lexington, Kentucky.
| |
Collapse
|
21
|
Pandya JD, Grondin R, Yonutas HM, Haghnazar H, Gash DM, Zhang Z, Sullivan PG. Decreased mitochondrial bioenergetics and calcium buffering capacity in the basal ganglia correlates with motor deficits in a nonhuman primate model of aging. Neurobiol Aging 2015; 36:1903-13. [PMID: 25726361 DOI: 10.1016/j.neurobiolaging.2015.01.018] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2014] [Revised: 12/24/2014] [Accepted: 01/23/2015] [Indexed: 01/02/2023]
Abstract
Altered mitochondrial function in the basal ganglia has been hypothesized to underlie cellular senescence and promote age-related motor decline. We tested this hypothesis in a nonhuman primate model of human aging. Six young (6-8 years old) and 6 aged (20-25 years old) female Rhesus monkeys (Macaca mulatta) were behaviorally characterized from standardized video records. Additionally, we measured mitochondrial bioenergetics along with calcium buffering capacity in the substantia nigra and putamen (PUT) from both age groups. Our results demonstrate that the aged animals had significantly reduced locomotor activity and movement speed compared with younger animals. Moreover, aged monkeys had significantly reduced ATP synthesis capacity (in substantia nigra and PUT), reduced pyruvate dehydrogenase activity (in PUT), and reduced calcium buffering capacity (in PUT) compared with younger animals. Furthermore, this age-related decline in mitochondrial function in the basal ganglia correlated with decline in motor function. Overall, our results suggest that drug therapies designed to enhance altered mitochondrial function may help improve motor deficits in the elderly.
Collapse
Affiliation(s)
- Jignesh D Pandya
- Spinal Cord and Brain Injury Research Center, The University of Kentucky Chandler College of Medicine, Lexington, KY, USA; Department of Anatomy and Neurobiology, The University of Kentucky Chandler College of Medicine, Lexington, KY, USA
| | - Richard Grondin
- Department of Anatomy and Neurobiology, The University of Kentucky Chandler College of Medicine, Lexington, KY, USA
| | - Heather M Yonutas
- Spinal Cord and Brain Injury Research Center, The University of Kentucky Chandler College of Medicine, Lexington, KY, USA; Department of Anatomy and Neurobiology, The University of Kentucky Chandler College of Medicine, Lexington, KY, USA
| | - Hamed Haghnazar
- Department of Anatomy and Neurobiology, The University of Kentucky Chandler College of Medicine, Lexington, KY, USA
| | - Don M Gash
- Department of Anatomy and Neurobiology, The University of Kentucky Chandler College of Medicine, Lexington, KY, USA
| | - Zhiming Zhang
- Department of Anatomy and Neurobiology, The University of Kentucky Chandler College of Medicine, Lexington, KY, USA
| | - Patrick G Sullivan
- Spinal Cord and Brain Injury Research Center, The University of Kentucky Chandler College of Medicine, Lexington, KY, USA; Department of Anatomy and Neurobiology, The University of Kentucky Chandler College of Medicine, Lexington, KY, USA.
| |
Collapse
|
22
|
Ryan MM, Guévremont D, Luxmanan C, Abraham WC, Williams JM. Aging alters long-term potentiation--related gene networks and impairs synaptic protein synthesis in the rat hippocampus. Neurobiol Aging 2015; 36:1868-80. [PMID: 25716081 DOI: 10.1016/j.neurobiolaging.2015.01.012] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2014] [Revised: 01/12/2015] [Accepted: 01/14/2015] [Indexed: 10/24/2022]
Abstract
During aging, memory retention and persistence of long-term potentiation (LTP) are impaired, suggesting an aging-related deterioration in mechanisms regulating information storage. Late-phase LTP requires synthesis of proteins at synapses as well as integrated regulation of gene networks. Because aging diminishes the persistence of LTP, primarily by affecting the transition between early and late phases, we assessed whether this was reflected in perturbation of gene networks. Using DNA microarray analysis, we compared LTP-associated gene expression in young (5 months), middle-aged (15 months), and old (22 months) male Sprague-Dawley rats. As expected, we found no significant difference in LTP measured 20 minutes postinduction; however, we found that overall more genes were regulated in the young group. Bioinformatics predicted not only dysregulation of activator protein-1 and nuclear factor kB transcription factor activity and epigenetic modifications but also dysregulation of protein synthesis. Notably, we confirmed an age-related impairment in metabotropic and ionotropic receptor-mediated synaptic protein synthesis. Together, these results demonstrate that LTP-specific gene expression is altered with aging and suggest that dysregulation of synaptic protein synthesis also contributes to the age-dependent reduction in LTP persistence.
Collapse
Affiliation(s)
- Margaret M Ryan
- Department of Anatomy, University of Otago, Dunedin, New Zealand; Brain Health Research Centre, University of Otago, Dunedin, New Zealand
| | - Diane Guévremont
- Department of Anatomy, University of Otago, Dunedin, New Zealand; Brain Health Research Centre, University of Otago, Dunedin, New Zealand
| | - Carthika Luxmanan
- Department of Anatomy, University of Otago, Dunedin, New Zealand; Brain Health Research Centre, University of Otago, Dunedin, New Zealand
| | - Wickliffe C Abraham
- Brain Health Research Centre, University of Otago, Dunedin, New Zealand; Department of Psychology, University of Otago, Dunedin, New Zealand
| | - Joanna M Williams
- Department of Anatomy, University of Otago, Dunedin, New Zealand; Brain Health Research Centre, University of Otago, Dunedin, New Zealand.
| |
Collapse
|
23
|
Prenderville JA, Kennedy PJ, Dinan TG, Cryan JF. Adding fuel to the fire: the impact of stress on the ageing brain. Trends Neurosci 2015; 38:13-25. [DOI: 10.1016/j.tins.2014.11.001] [Citation(s) in RCA: 90] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
24
|
Li L, Liu Y. Aging-related gene signature regulated by Nlrp3 predicts glioma progression. Am J Cancer Res 2014; 5:442-449. [PMID: 25628952 PMCID: PMC4300695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2014] [Accepted: 11/15/2014] [Indexed: 06/04/2023] Open
Abstract
Aging is the strongest risk factor for glioma development, suggesting that molecular crosstalks between aging and tumorigenesis exist in many cellular pathways. Recently, Nlrp3 inflammasome have been shown to modulate several major cellular pathways such as inflammation and cell death and have been demonstrated to be an upstream target that controlled the process of brain aging. We proposed Nlrp3 inflammasome may serve as a possible molecular link between aging and glioma progression. In this study, we generated a aging-related gene signature that regulated by Nlrp3 in mouse hippocampus and demonstrated that this gene signature can distinguish subsets of glioma samples and predicts clinical outcome in radiotherapy-treated patients. In addition, using U87 and GL261 xenograft mouse glioblastoma model, we found that Nlrp3 inflammasome contributed to radiotherapy resistance in glioma. Ionizing radiation can induce Nlrp3 inflammasome expression; Nlrp3 inhibition reduced tumor growth and prolonged the survival of mouse following IR treatment; Nlrp3 inhibition reduced number of senescent cells induced by IR. These results above suggest that Nlrp3 inflammasome is an important molecular link between brain aging and glioma progression; the Nlrp3 gene signature may serve as a predictive biomarker for glioma patients.
Collapse
Affiliation(s)
- Lianling Li
- Department of Neurosurgery, Qilu Hospital of Shandong UniversityJinan 250012, China
- Brain Science Research Institute of Shandong UniversityJinan 250012, China
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Public Health, Qilu Hospital of Shandong UniversityJinan 250012, China
| | - Yuguang Liu
- Department of Neurosurgery, Qilu Hospital of Shandong UniversityJinan 250012, China
- Brain Science Research Institute of Shandong UniversityJinan 250012, China
| |
Collapse
|
25
|
Stilling RM, Benito E, Gertig M, Barth J, Capece V, Burkhardt S, Bonn S, Fischer A. De-regulation of gene expression and alternative splicing affects distinct cellular pathways in the aging hippocampus. Front Cell Neurosci 2014; 8:373. [PMID: 25431548 PMCID: PMC4230043 DOI: 10.3389/fncel.2014.00373] [Citation(s) in RCA: 83] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2014] [Accepted: 10/21/2014] [Indexed: 01/20/2023] Open
Abstract
Aging is accompanied by gradually increasing impairment of cognitive abilities and constitutes the main risk factor of neurodegenerative conditions like Alzheimer's disease (AD). The underlying mechanisms are however not well understood. Here we analyze the hippocampal transcriptome of young adult mice and two groups of mice at advanced age using RNA sequencing. This approach enabled us to test differential expression of coding and non-coding transcripts, as well as differential splicing and RNA editing. We report a specific age-associated gene expression signature that is associated with major genetic risk factors for late-onset AD (LOAD). This signature is dominated by neuroinflammatory processes, specifically activation of the complement system at the level of increased gene expression, while de-regulation of neuronal plasticity appears to be mediated by compromised RNA splicing.
Collapse
Affiliation(s)
- Roman M Stilling
- Department of Psychiatry and Psychotherapy, University Medical Center Göttingen Göttingen, Germany ; Research Group for Epigenetics in Neurodegenerative Diseases, German Center for Neurodegenerative Diseases (DZNE) Göttingen Göttingen, Germany
| | - Eva Benito
- Research Group for Epigenetics in Neurodegenerative Diseases, German Center for Neurodegenerative Diseases (DZNE) Göttingen Göttingen, Germany
| | - Michael Gertig
- Research Group for Epigenetics in Neurodegenerative Diseases, German Center for Neurodegenerative Diseases (DZNE) Göttingen Göttingen, Germany
| | - Jonas Barth
- Research Group for Epigenetics in Neurodegenerative Diseases, German Center for Neurodegenerative Diseases (DZNE) Göttingen Göttingen, Germany
| | - Vincenzo Capece
- Research Group for Computational Analysis of Biological Networks, German Center for Neurodegenerative Diseases (DZNE) Göttingen Göttingen, Germany
| | - Susanne Burkhardt
- Research Group for Epigenetics in Neurodegenerative Diseases, German Center for Neurodegenerative Diseases (DZNE) Göttingen Göttingen, Germany
| | - Stefan Bonn
- Research Group for Computational Analysis of Biological Networks, German Center for Neurodegenerative Diseases (DZNE) Göttingen Göttingen, Germany
| | - Andre Fischer
- Department of Psychiatry and Psychotherapy, University Medical Center Göttingen Göttingen, Germany ; Research Group for Epigenetics in Neurodegenerative Diseases, German Center for Neurodegenerative Diseases (DZNE) Göttingen Göttingen, Germany
| |
Collapse
|
26
|
Yamada J, Jinno S. Age-related differences in oligodendrogenesis across the dorsal-ventral axis of the mouse hippocampus. Hippocampus 2014; 24:1017-29. [PMID: 24753086 DOI: 10.1002/hipo.22287] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2013] [Revised: 03/12/2014] [Accepted: 04/11/2014] [Indexed: 11/07/2022]
Abstract
Oligodendrocyte precursor cells (OPCs) continue to divide and generate new oligodendrocytes (OLs) in the healthy adult brain. Although recent studies have indicated that adult oligodendrogenesis may be vital for the maintenance of normal brain function, the significance of adult oligodendrogenesis in brain aging remains unclear. In this study, we report a stereological estimation of age-related oligodendrogenesis changes in the mouse hippocampus: the dorsal subdivision is related to learning and memory, while the ventral subdivision is involved in emotional behaviors. To identify OPCs and OLs, we used a set of molecular markers, OL lineage transcription factor (Olig2) and platelet-derived growth factor receptor-alpha (PDGFαR). Intracellular dye injection shows that PDGFαR+/Olig2+ cells and PDGFαR-/Olig2+ cells can be defined as OPCs and OLs, respectively. In the dorsal Ammon's horn, the numbers of OPCs decreased with age, while those of OLs remained unchanged during aging. In the ventral Ammon's horn, the numbers of OPCs and OLs generally decreased with age. Bromodeoxyuridine (BrdU) fate-tracing analysis revealed that the numbers of BrdU+ mitotic OPCs in the Ammon's horn remained unchanged during aging in both the dorsal and ventral subdivisions. Unexpectedly, the numbers of BrdU+ newly generated OLs increased with age in the dorsal Ammon's horn, but remained unchanged in the ventral Ammon's horn. Together, the numbers of OLs in the dorsal Ammon's horn may be maintained during aging by increased survival of adult born OLs, while the numbers of OLs in the ventral Ammon's horn may be reduced with age due to the lack of such compensatory mechanisms. These observations provide new insight into the involvement of adult oligodendrogenesis in age-related changes in the structure and function of the hippocampus.
Collapse
Affiliation(s)
- Jun Yamada
- Department of Developmental Molecular Anatomy, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | | |
Collapse
|
27
|
Mapping NAD(+) metabolism in the brain of ageing Wistar rats: potential targets for influencing brain senescence. Biogerontology 2013; 15:177-98. [PMID: 24337988 DOI: 10.1007/s10522-013-9489-5] [Citation(s) in RCA: 80] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2013] [Accepted: 12/09/2013] [Indexed: 12/21/2022]
Abstract
Over the last decade, the importance of NAD(+) has expanded beyond its role as an essential cofactor for energy metabolism. NAD(+) has emerged as a major signalling molecule that serves as the sole substrate for several enzymatic reactions including the DNA repair enzyme, poly(ADP-ribose) polymerase (PARP), NAD-dependent protein deacetylases or CD38, and transcriptional factors by a new class of histone deacetylases known as sirtuins. NAD(+) levels are regulated by the metabolic status and cellular stress caused by oxidative stress and DNA damage. Since a detailed study of NAD(+) metabolism in the healthy ageing mammalian brain is nascent, we examined the effect of ageing on intracellular NAD(+) metabolism in different brain regions in female Wistar rats in young (3 months), middle aged (12 months) and older adults (24 months). Our results are the first to show a significant decline in intracellular NAD(+) levels and NAD:NADH ratio with ageing in the CNS, occurring in parallel to an increase in lipid peroxidation and protein oxidation (o- and m-tyrosine) and a decline in total antioxidant capacity. Hyperphosphorylation of H2AX levels was also observed together with increased PARP-1 and PARP-2 expression, and CD38 activity, concomitantly with reduced NAD(+) and ATP levels and SIRT1 function in the cortex, brainstem, hippocampus and cerebellum. Reduced activity of mitochondrial complex I-IV and impaired maximum mitochondrial respiration rate were also observed in the ageing rat brain. Among the multiple physiological pathways associated with NAD(+) catabolism, our discovery of CD38 as the major regulator of cellular NAD(+) levels in rat neurons indicates that CD38 is a promising therapeutic target for the treatment of age-related neurodegenerative diseases.
Collapse
|
28
|
Gant JC, Blalock EM, Chen KC, Kadish I, Porter NM, Norris CM, Thibault O, Landfield PW. FK506-binding protein 1b/12.6: a key to aging-related hippocampal Ca2+ dysregulation? Eur J Pharmacol 2013; 739:74-82. [PMID: 24291098 DOI: 10.1016/j.ejphar.2013.10.070] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2013] [Revised: 10/16/2013] [Accepted: 10/17/2013] [Indexed: 12/25/2022]
Abstract
It has been recognized for some time that the Ca(2+)-dependent slow afterhyperpolarization (sAHP) is larger in hippocampal neurons of aged compared with young animals. In addition, extensive studies since have shown that other Ca(2+)-mediated electrophysiological responses are increased in hippocampus with aging, including Ca(2+) transients, L-type voltage-gated Ca(2+) channel activity, Ca(2+) spike duration and action potential accommodation. Elevated Ca(2+)-induced Ca(2+) release from ryanodine receptors (RyRs) appears to drive amplification of the Ca(2+) responses. Components of this Ca(2+) dysregulation phenotype correlate with deficits in cognitive function and plasticity, indicating they may play critical roles in aging-related impairment of brain function. However, the molecular mechanisms underlying aging-related Ca(2+) dysregulation are not well understood. FK506-binding proteins 1a and 1b (FKBP1a/1b, also known as FKBP12/12.6) are immunophilin proteins that bind the immunosuppressant drugs FK506 and rapamycin. In muscle cells, FKBP1a/1b also bind RyRs and inhibits Ca(2+)-induced Ca(2+) release, but it is not clear whether FKBPs act similarly in brain cells. Recently, we found that selectively disrupting hippocampal FKBP1b function in young rats, either by microinjecting adeno-associated viral vectors expressing siRNA, or by treatment with rapamycin, increases the sAHP and recapitulates much of the hippocampal Ca(2+) dysregulation phenotype. Moreover, in microarray studies, we found FKBP1b gene expression was downregulated in hippocampus of aging rats and early-stage Alzheimer's disease subjects. These results suggest the novel hypothesis that declining FKBP function is a key factor in aging-related Ca(2+) dysregulation in the brain and point to potential new therapeutic targets for counteracting unhealthy brain aging.
Collapse
Affiliation(s)
- J C Gant
- Department of Molecular and Biomedical Pharmacology, University of Kentucky College of Medicine, 800 Rose St., UKMC Lexington, KY 40536, United States
| | - E M Blalock
- Department of Molecular and Biomedical Pharmacology, University of Kentucky College of Medicine, 800 Rose St., UKMC Lexington, KY 40536, United States
| | - K-C Chen
- Department of Molecular and Biomedical Pharmacology, University of Kentucky College of Medicine, 800 Rose St., UKMC Lexington, KY 40536, United States
| | - I Kadish
- Department of Molecular and Biomedical Pharmacology, University of Kentucky College of Medicine, 800 Rose St., UKMC Lexington, KY 40536, United States
| | - N M Porter
- Department of Molecular and Biomedical Pharmacology, University of Kentucky College of Medicine, 800 Rose St., UKMC Lexington, KY 40536, United States
| | - C M Norris
- Department of Molecular and Biomedical Pharmacology, University of Kentucky College of Medicine, 800 Rose St., UKMC Lexington, KY 40536, United States
| | - O Thibault
- Department of Molecular and Biomedical Pharmacology, University of Kentucky College of Medicine, 800 Rose St., UKMC Lexington, KY 40536, United States
| | - P W Landfield
- Department of Molecular and Biomedical Pharmacology, University of Kentucky College of Medicine, 800 Rose St., UKMC Lexington, KY 40536, United States.
| |
Collapse
|
29
|
Chen KC, Blalock EM, Curran-Rauhut MA, Kadish I, Blalock SJ, Brewer L, Porter NM, Landfield PW. Glucocorticoid-dependent hippocampal transcriptome in male rats: pathway-specific alterations with aging. Endocrinology 2013; 154:2807-20. [PMID: 23736296 PMCID: PMC3713214 DOI: 10.1210/en.2013-1139] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Although glucocorticoids (GCs) are known to exert numerous effects in the hippocampus, their chronic regulatory functions remain poorly understood. Moreover, evidence is inconsistent regarding the long-standing hypothesis that chronic GC exposure promotes brain aging/Alzheimer disease. Here, we adrenalectomized male F344 rats at 15 months of age, maintained them for 3 months with implanted corticosterone (CORT) pellets producing low or intermediate (glucocorticoid receptor-activating) blood levels of CORT, and performed microarray/pathway analyses in hippocampal CA1. We defined the chronic GC-dependent transcriptome as 393 genes that exhibited differential expression between intermediate and low CORT groups. Short-term CORT (4 days) did not recapitulate this transcriptome. Functional processes/pathways overrepresented by chronic CORT-up-regulated genes included learning/plasticity, differentiation, glucose metabolism, and cholesterol biosynthesis, whereas processes overrepresented by CORT-down-regulated genes included inflammatory/immune/glial responses and extracellular structure. These profiles indicate that GCs chronically activate neuronal/metabolic processes while coordinately repressing a glial axis of reactivity/inflammation. We then compared the GC transcriptome with a previously defined hippocampal aging transcriptome, revealing a high proportion of common genes. Although CORT and aging moved expression of some common genes in the same direction, the majority were shifted in opposite directions by CORT and aging (eg, glial inflammatory genes down-regulated by CORT are up-regulated with aging). These results contradict the hypothesis that GCs simply promote brain aging and also suggest that the opposite direction shifts during aging reflect resistance to CORT regulation. Therefore, we propose a new model in which aging-related GC resistance develops in some target pathways, whereas GC overstimulation develops in others, together generating much of the brain aging phenotype.
Collapse
Affiliation(s)
- Kuey-Chu Chen
- Department of Molecular and Biomedical Pharmacology, University of Kentucky College of Medicine, Lexington, KY 40536, USA
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Willette AA, Xu G, Johnson SC, Birdsill AC, Jonaitis EM, Sager MA, Hermann BP, La Rue A, Asthana S, Bendlin BB. Insulin resistance, brain atrophy, and cognitive performance in late middle-aged adults. Diabetes Care 2013; 36:443-9. [PMID: 23069842 PMCID: PMC3554303 DOI: 10.2337/dc12-0922] [Citation(s) in RCA: 157] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
OBJECTIVE Insulin resistance dysregulates glucose uptake and other functions in brain areas affected by Alzheimer disease. Insulin resistance may play a role in Alzheimer disease etiopathogenesis. This longitudinal study examined whether insulin resistance among late middle-aged, cognitively healthy individuals was associated with 1) less gray matter in Alzheimer disease-sensitive brain regions and 2) worse cognitive performance. RESEARCH DESIGN AND METHODS Homeostasis model assessment of insulin resistance, gray matter volume, and the Rey Auditory Verbal Learning Test (RAVLT) were acquired in 372 participants at baseline and a consecutive subset of 121 individuals ~4 years later. Voxel-based morphometry and tensor-based morphometry were used, respectively, to test the association of insulin resistance with baseline brain volume and progressive gray matter atrophy. RESULTS Higher insulin resistance predicted less gray matter at baseline and 4 years later in medial temporal lobe, prefrontal cortices, precuneus, and other parietal gyri. A region-of-interest analysis, independent of the voxel-wise analyses, confirmed that higher insulin resistance was related to medial temporal lobe atrophy. Atrophy itself corresponded to cognitive deficits in the RAVLT. Temporal lobe atrophy that was predicted by higher insulin resistance significantly mediated worse RAVLT encoding performance. CONCLUSIONS These results suggest that insulin resistance in an asymptomatic, late middle-aged cohort is associated with progressive atrophy in regions affected by early Alzheimer disease. Insulin resistance may also affect the ability to encode episodic information by negatively influencing gray matter volume in medial temporal lobe.
Collapse
Affiliation(s)
- Auriel A Willette
- Geriatric Research Education and Clinical Center, William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Paban V, Billard JM, Bouet V, Freret T, Boulouard M, Chambon C, Loriod B, Alescio-Lautier B. Genomic transcriptional profiling in LOU/C/Jall rats identifies genes for successful aging. Brain Struct Funct 2012; 218:1501-12. [DOI: 10.1007/s00429-012-0472-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2012] [Accepted: 10/25/2012] [Indexed: 02/04/2023]
|
32
|
Finch CE, Austad SN. Primate aging in the mammalian scheme: the puzzle of extreme variation in brain aging. AGE (DORDRECHT, NETHERLANDS) 2012; 34:1075-91. [PMID: 22218781 PMCID: PMC3448989 DOI: 10.1007/s11357-011-9355-9] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/27/2011] [Accepted: 12/01/2011] [Indexed: 05/31/2023]
Abstract
At later ages, humans have high risk of developing Alzheimer disease (AD) which may afflict up to 50% by 90 years. While prosimians and monkeys show more substantial changes, the great apes brains examined show mild neurodegenerative changes. Compared with rodents, primates develop and reproduce slowly and are long lived. The New World primates contain some of the shortest as well as some of the longest-lived monkey species, while the prosimians develop the most rapidly and are the shortest lived. Great apes have the largest brains, slowest development, and longest lives among the primates. All primates share some level of slowly progressive, age-related neurodegenerative changes. However, no species besides humans has yet shown regular drastic neuron loss or cognitive decline approaching clinical grade AD. Several primates accumulate extensive deposits of diffuse amyloid-beta protein (Aβ) but only a prosimian-the gray mouse lemur-regularly develops a tauopathy approaching the neurofibrillary tangles of AD. Compared with monkeys, nonhuman great apes display even milder brain-aging changes, a deeply puzzling observation. The genetic basis for these major species differences in brain aging remains obscure but does not involve the Aβ coding sequence which is identical in nonhuman primates and humans. While chimpanzees merit more study, we note the value of smaller, shorter-lived species such as marmosets and small lemurs for aging studies. A continuing concern for all aging studies employing primates is that relative to laboratory rodents, primate husbandry is in a relatively primitive state, and better husbandry to control infections and obesity is needed for brain aging research.
Collapse
Affiliation(s)
- Caleb E Finch
- Ethel Percy Andrus Gerontology Center, University of Southern California, 3715 McClintock Ave, Los Angeles, CA 90089-0191,
| | | |
Collapse
|
33
|
Porter NM, Bohannon JH, Curran-Rauhut M, Buechel HM, Dowling ALS, Brewer LD, Popovic J, Thibault V, Kraner SD, Chen KC, Blalock EM. Hippocampal CA1 transcriptional profile of sleep deprivation: relation to aging and stress. PLoS One 2012; 7:e40128. [PMID: 22792227 PMCID: PMC3390348 DOI: 10.1371/journal.pone.0040128] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2012] [Accepted: 06/01/2012] [Indexed: 11/18/2022] Open
Abstract
Background Many aging changes seem similar to those elicited by sleep-deprivation and psychosocial stress. Further, sleep architecture changes with age suggest an age-related loss of sleep. Here, we hypothesized that sleep deprivation in young subjects would elicit both stress and aging-like transcriptional responses. Methodology/Principal Findings F344 rats were divided into control and sleep deprivation groups. Body weight, adrenal weight, corticosterone level and hippocampal CA1 transcriptional profiles were measured. A second group of animals was exposed to novel environment stress (NES), and their hippocampal transcriptional profiles measured. A third cohort exposed to control or SD was used to validate transcriptional results with Western blots. Microarray results were statistically contrasted with prior transcriptional studies. Microarray results pointed to sleep pressure signaling and macromolecular synthesis disruptions in the hippocampal CA1 region. Animals exposed to NES recapitulated nearly one third of the SD transcriptional profile. However, the SD -aging relationship was more complex. Compared to aging, SD profiles influenced a significant subset of genes. mRNA associated with neurogenesis and energy pathways showed agreement between aging and SD, while immune, glial, and macromolecular synthesis pathways showed SD profiles that opposed those seen in aging. Conclusions/Significance We conclude that although NES and SD exert similar transcriptional changes, selective presynaptic release machinery and Homer1 expression changes are seen in SD. Among other changes, the marked decrease in Homer1 expression with age may represent an important divergence between young and aged brain response to SD. Based on this, it seems reasonable to conclude that therapeutic strategies designed to promote sleep in young subjects may have off-target effects in the aged. Finally, this work identifies presynaptic vesicular release and intercellular adhesion molecular signatures as novel therapeutic targets to counter effects of SD in young subjects.
Collapse
Affiliation(s)
- Nada M. Porter
- Department of Molecular and Biomedical Pharmacology, University of Kentucky College of Medicine, Lexington, Kentucky, United States of America
| | - Julia H. Bohannon
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Meredith Curran-Rauhut
- Department of Biology, Dickinson College, Carlisle, Pennsylvania, United States of America
| | - Heather M. Buechel
- Department of Molecular and Biomedical Pharmacology, University of Kentucky College of Medicine, Lexington, Kentucky, United States of America
| | - Amy L. S. Dowling
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky, United States of America
| | - Lawrence D. Brewer
- Department of Molecular and Biomedical Pharmacology, University of Kentucky College of Medicine, Lexington, Kentucky, United States of America
| | - Jelena Popovic
- Department of Molecular and Biomedical Pharmacology, University of Kentucky College of Medicine, Lexington, Kentucky, United States of America
| | - Veronique Thibault
- Department of Molecular and Biomedical Pharmacology, University of Kentucky College of Medicine, Lexington, Kentucky, United States of America
| | - Susan D. Kraner
- Department of Neuroscience Cell Biology Physiology, Wright State University, Dayton, Ohio, United States of America
| | - Kuey Chu Chen
- Department of Molecular and Biomedical Pharmacology, University of Kentucky College of Medicine, Lexington, Kentucky, United States of America
| | - Eric M. Blalock
- Department of Molecular and Biomedical Pharmacology, University of Kentucky College of Medicine, Lexington, Kentucky, United States of America
- * E-mail:
| |
Collapse
|
34
|
Peng ZF, Chen MJ, Manikandan J, Melendez AJ, Shui G, Russo-Marie F, Whiteman M, Beart PM, Moore PK, Cheung NS. Multifaceted role of nitric oxide in an in vitro mouse neuronal injury model: transcriptomic profiling defines the temporal recruitment of death signalling cascades. J Cell Mol Med 2012; 16:41-58. [PMID: 21352476 PMCID: PMC3823092 DOI: 10.1111/j.1582-4934.2011.01288.x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Abstract
Nitric oxide is implicated in the pathogenesis of various neuropathologies characterized by oxidative stress. Although nitric oxide has been reported to be involved in the exacerbation of oxidative stress observed in several neuropathologies, existent data fail to provide a holistic description of how nitrergic pathobiology elicits neuronal injury. Here we provide a comprehensive description of mechanisms contributing to nitric oxide induced neuronal injury by global transcriptomic profiling. Microarray analyses were undertaken on RNA from murine primary cortical neurons treated with the nitric oxide generator DETA-NONOate (NOC-18, 0.5 mM) for 8–24 hrs. Biological pathway analysis focused upon 3672 gene probes which demonstrated at least a ±1.5-fold expression in a minimum of one out of three time-points and passed statistical analysis (one-way anova, P < 0.05). Numerous enriched processes potentially determining nitric oxide mediated neuronal injury were identified from the transcriptomic profile: cell death, developmental growth and survival, cell cycle, calcium ion homeostasis, endoplasmic reticulum stress, oxidative stress, mitochondrial homeostasis, ubiquitin-mediated proteolysis, and GSH and nitric oxide metabolism. Our detailed time-course study of nitric oxide induced neuronal injury allowed us to provide the first time a holistic description of the temporal sequence of cellular events contributing to nitrergic injury. These data form a foundation for the development of screening platforms and define targets for intervention in nitric oxide neuropathologies where nitric oxide mediated injury is causative.
Collapse
Affiliation(s)
- Zhao Feng Peng
- Key Laboratory of Biogeology and Environmental Geology of the Ministry of Education, China University of Geosciences, Wuhan, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Craft S, Foster TC, Landfield PW, Maier SF, Resnick SM, Yaffe K. Session III: Mechanisms of age-related cognitive change and targets for intervention: inflammatory, oxidative, and metabolic processes. J Gerontol A Biol Sci Med Sci 2012; 67:754-9. [PMID: 22570133 DOI: 10.1093/gerona/gls112] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
There is increasing evidence from basic science and human epidemiological studies that inflammation, oxidative stress, and metabolic abnormalities are associated with age-related cognitive decline and impairment. This article summarizes selected research on these topics presented at the Cognitive Aging Summit II. Speakers in this session presented evidence highlighting the roles of these processes and pathways on age-related cognitive decline, pointing to possible targets for intervention in nondemented older adults. Specific areas discussed included age differences in the production of cytokines following injury or infection, mechanisms underlying oxidative stress-induced changes in memory consolidation, insulin effects on brain signaling and memory, and the association between metabolic syndrome and cognitive decline in older adults. These presentations emphasize advances in our understanding of mechanisms and modifiers of age-related cognitive decline and provide insights into potential targets to promote cognitive health in older adults.
Collapse
Affiliation(s)
- Suzanne Craft
- Department of Psychiatry and Behavioral Sciences, University of Washington, Geriatric Research, Education, and Clinical Center, Veterans Affairs Puget Sound, Seattle, USA
| | | | | | | | | | | |
Collapse
|
36
|
VanGuilder Starkey HD, Van Kirk CA, Bixler GV, Imperio CG, Kale VP, Serfass JM, Farley JA, Yan H, Warrington JP, Han S, Mitschelen M, Sonntag WE, Freeman WM. Neuroglial expression of the MHCI pathway and PirB receptor is upregulated in the hippocampus with advanced aging. J Mol Neurosci 2012; 48:111-26. [PMID: 22562814 DOI: 10.1007/s12031-012-9783-8] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2012] [Accepted: 04/16/2012] [Indexed: 12/31/2022]
Abstract
The hippocampus undergoes changes with aging that impact neuronal function, such as synapse loss and altered neurotransmitter release. Nearly half of the aged population also develops deficits in spatial learning and memory. To identify age-related hippocampal changes that may contribute to cognitive decline, transcriptomic analysis of synaptosome preparations from adult (12 months) and aged (28 months) Fischer 344-Brown Norway rats assessed for spatial learning and memory was performed. Bioinformatic analysis identified the MHCI pathway as significantly upregulated with aging. Age-related increases in mRNAs encoding the MHCI genes RT1-A1, RT1-A2, and RT1-A3 were confirmed by qPCR in synaptosomes and in CA1 and CA3 dissections. Elevated levels of the MHCI cofactor (B2m), antigen-loading components (Tap1, Tap2, Tapbp), and two known MHCI receptors (PirB, Klra2) were also confirmed. Protein expression of MHCI was elevated with aging in synaptosomes, CA1, and DG, while PirB protein expression was induced in both CA1 and DG. MHCI expression was localized to microglia and neuronal excitatory postsynaptic densities, and PirB was localized to neuronal somata, axons, and dendrites. Induction of the MHCI antigen processing and presentation pathway in hippocampal neurons and glia may contribute to age-related hippocampal dysfunction by increasing neuroimmune signaling or altering synaptic homeostasis.
Collapse
|
37
|
Willette AA, Bendlin BB, Colman RJ, Kastman EK, Field AS, Alexander AL, Sridharan A, Allison DB, Anderson R, Voytko ML, Kemnitz JW, Weindruch RH, Johnson SC. Calorie restriction reduces the influence of glucoregulatory dysfunction on regional brain volume in aged rhesus monkeys. Diabetes 2012; 61:1036-42. [PMID: 22415875 PMCID: PMC3331743 DOI: 10.2337/db11-1187] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
Insulin signaling dysregulation is related to neural atrophy in hippocampus and other areas affected by neurovascular and neurodegenerative disorders. It is not known if long-term calorie restriction (CR) can ameliorate this relationship through improved insulin signaling or if such an effect might influence task learning and performance. To model this hypothesis, magnetic resonance imaging was conducted on 27 CR and 17 control rhesus monkeys aged 19-31 years from a longitudinal study. Voxel-based regression analyses were used to associate insulin sensitivity with brain volume and microstructure cross-sectionally. Monkey motor assessment panel (mMAP) performance was used as a measure of task performance. CR improved glucoregulation parameters and related indices. Higher insulin sensitivity predicted more gray matter in parietal and frontal cortices across groups. An insulin sensitivity × dietary condition interaction indicated that CR animals had more gray matter in hippocampus and other areas per unit increase relative to controls, suggesting a beneficial effect. Finally, bilateral hippocampal volume adjusted by insulin sensitivity, but not volume itself, was significantly associated with mMAP learning and performance. These results suggest that CR improves glucose regulation and may positively influence specific brain regions and at least motor task performance. Additional studies are warranted to validate these relationships.
Collapse
Affiliation(s)
- Auriel A. Willette
- Geriatric Research Education and Clinical Center, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin
- Waisman Laboratory for Brain Imaging and Behavior, University of Wisconsin-Madison, Madison, Wisconsin
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Barbara B. Bendlin
- Geriatric Research Education and Clinical Center, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Ricki J. Colman
- Wisconsin National Primate Research Center, Madison, Wisconsin
| | - Erik K. Kastman
- Geriatric Research Education and Clinical Center, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Aaron S. Field
- Department of Radiology, University of Wisconsin-Madison, Madison, Wisconsin
| | - Andrew L. Alexander
- Waisman Laboratory for Brain Imaging and Behavior, University of Wisconsin-Madison, Madison, Wisconsin
| | - Aadhavi Sridharan
- Geriatric Research Education and Clinical Center, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
- Neuroscience Training Program, University of Wisconsin-Madison, Madison, Wisconsin
| | - David B. Allison
- Department of Biostatistics, University of Alabama at Birmingham, Birmingham, Alabama
| | - Rozalyn Anderson
- Geriatric Research Education and Clinical Center, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin
- Wisconsin National Primate Research Center, Madison, Wisconsin
| | - Mary-Lou Voytko
- Department of Neurobiology and Anatomy Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, North Carolina
| | - Joseph W. Kemnitz
- Wisconsin National Primate Research Center, Madison, Wisconsin
- Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
- Institute for Clinical and Translational Research, University of Wisconsin-Madison, Madison, Wisconsin
| | - Richard H. Weindruch
- Geriatric Research Education and Clinical Center, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin
| | - Sterling C. Johnson
- Geriatric Research Education and Clinical Center, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin
- Waisman Laboratory for Brain Imaging and Behavior, University of Wisconsin-Madison, Madison, Wisconsin
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
- Wisconsin National Primate Research Center, Madison, Wisconsin
- Neuroscience Training Program, University of Wisconsin-Madison, Madison, Wisconsin
- Corresponding author: Sterling C. Johnson,
| |
Collapse
|
38
|
Zhao L, Morgan TE, Mao Z, Lin S, Cadenas E, Finch CE, Pike CJ, Mack WJ, Brinton RD. Continuous versus cyclic progesterone exposure differentially regulates hippocampal gene expression and functional profiles. PLoS One 2012; 7:e31267. [PMID: 22393359 PMCID: PMC3290616 DOI: 10.1371/journal.pone.0031267] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2011] [Accepted: 01/05/2012] [Indexed: 12/27/2022] Open
Abstract
This study investigated the impact of chronic exposure to continuous (CoP4) versus cyclic progesterone (CyP4) alone or in combination with 17β-estradiol (E2) on gene expression profiles targeting bioenergetics, metabolism and inflammation in the adult female rat hippocampus. High-throughput qRT-PCR analyses revealed that ovarian hormonal depletion induced by ovariectomy (OVX) led to multiple significant gene expression alterations, which were to a great extent reversed by co-administration of E2 and CyP4. In contrast, co-administration of E2 and CoP4 induced a pattern highly resembling OVX. Bioinformatics analyses further revealed clear disparities in functional profiles associated with E2+CoP4 and E2+CyP4. Genes involved in mitochondrial energy (ATP synthase α subunit; Atp5a1), redox homeostasis (peroxiredoxin 5; Prdx5), insulin signaling (insulin-like growth factor I; Igf1), and cholesterol trafficking (liver X receptor α subtype; Nr1h3), differed in direction of regulation by E2+CoP4 (down-regulation relative to OVX) and E2+CyP4 (up-regulation relative to OVX). In contrast, genes involved in amyloid metabolism (β-secretase; Bace1) differed only in degree of regulation, as both E2+CoP4 and E2+CyP4 induced down-regulation at different efficacy. E2+CyP4-induced changes could be associated with regulation of progesterone receptor membrane component 1(Pgrmc1). In summary, results from this study provide evidence at the molecular level that differing regimens of hormone therapy (HT) can induce disparate gene expression profiles in brain. From a translational perspective, confirmation of these results in a model of natural menopause, would imply that the common regimen of continuous combined HT may have adverse consequences whereas a cyclic combined regimen, which is more physiological, could be an effective strategy to maintain neurological health and function throughout menopausal aging.
Collapse
Affiliation(s)
- Liqin Zhao
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, California, United States of America
- * E-mail: (LZ); (RDB)
| | - Todd E. Morgan
- Davis School of Gerontology, University of Southern California, Los Angeles, California, United States of America
| | - Zisu Mao
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, California, United States of America
| | - Sharon Lin
- Davis School of Gerontology, University of Southern California, Los Angeles, California, United States of America
| | - Enrique Cadenas
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, California, United States of America
| | - Caleb E. Finch
- Davis School of Gerontology, University of Southern California, Los Angeles, California, United States of America
| | - Christian J. Pike
- Davis School of Gerontology, University of Southern California, Los Angeles, California, United States of America
| | - Wendy J. Mack
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - Roberta D. Brinton
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, California, United States of America
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
- * E-mail: (LZ); (RDB)
| |
Collapse
|
39
|
|
40
|
Madrigano J, Baccarelli A, Mittleman MA, Sparrow D, Vokonas PS, Tarantini L, Schwartz J. Aging and epigenetics: longitudinal changes in gene-specific DNA methylation. Epigenetics 2012; 7:63-70. [PMID: 22207354 DOI: 10.4161/epi.7.1.18749] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
DNA methylation has been associated with age-related disease. Intra-individual changes in gene-specific DNA methylation over time in a community-based cohort has not been well described. We estimated the change in DNA methylation due to aging for nine genes in an elderly, community-dwelling cohort of men. Seven hundred and eighty four men from the Veterans Administration Normative Aging Study who were living in metropolitan Boston from 1999-2009 donated a blood sample for DNA methylation analysis at clinical examinations repeated at approximately 3-5 year intervals. We used mixed effects regression models. Aging was significantly associated with decreased methylation of GCR, iNOS and TLR2 and with increased methylation of IFNγ, F3, CRAT and OGG. Obstructive pulmonary disease at baseline modified the effect of aging on methylation of IFNγ (interaction p = 0.04). For participants who had obstructive pulmonary disease at their baseline visit, the rate of change of methylation of IFNγ was -0.05% 5-methyl-cytosine (5-mC) per year (95% CI: -0.22, 0.13), but was 0.14% 5-mC per year (95% CI: 0.05, 0.24) for those without this condition. Models with random slopes indicated significant heterogeneity in the effect of aging on methylation of GCR, iNOS and OGG. These findings suggest that DNA methylation may reflect differential biological aging.
Collapse
Affiliation(s)
- Jaime Madrigano
- The Earth Institute and Mailman School of Public Health, Columbia University, New York, NY, USA.
| | | | | | | | | | | | | |
Collapse
|
41
|
Xie C, Mao X, Huang J, Ding Y, Wu J, Dong S, Kong L, Gao G, Li CY, Wei L. KOBAS 2.0: a web server for annotation and identification of enriched pathways and diseases. Nucleic Acids Res 2011; 39:W316-22. [PMID: 21715386 PMCID: PMC3125809 DOI: 10.1093/nar/gkr483] [Citation(s) in RCA: 3226] [Impact Index Per Article: 248.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
High-throughput experimental technologies often identify dozens to hundreds of genes related to, or changed in, a biological or pathological process. From these genes one wants to identify biological pathways that may be involved and diseases that may be implicated. Here, we report a web server, KOBAS 2.0, which annotates an input set of genes with putative pathways and disease relationships based on mapping to genes with known annotations. It allows for both ID mapping and cross-species sequence similarity mapping. It then performs statistical tests to identify statistically significantly enriched pathways and diseases. KOBAS 2.0 incorporates knowledge across 1327 species from 5 pathway databases (KEGG PATHWAY, PID, BioCyc, Reactome and Panther) and 5 human disease databases (OMIM, KEGG DISEASE, FunDO, GAD and NHGRI GWAS Catalog). KOBAS 2.0 can be accessed at http://kobas.cbi.pku.edu.cn.
Collapse
Affiliation(s)
- Chen Xie
- Center for Bioinformatics, State Key Laboratory of Protein and Plant Gene Research, College of Life Sciences, Peking University, Beijing, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
VanGuilder HD, Bixler GV, Brucklacher RM, Farley JA, Yan H, Warrington JP, Sonntag WE, Freeman WM. Concurrent hippocampal induction of MHC II pathway components and glial activation with advanced aging is not correlated with cognitive impairment. J Neuroinflammation 2011; 8:138. [PMID: 21989322 PMCID: PMC3216278 DOI: 10.1186/1742-2094-8-138] [Citation(s) in RCA: 99] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2011] [Accepted: 10/11/2011] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Age-related cognitive dysfunction, including impairment of hippocampus-dependent spatial learning and memory, affects approximately half of the aged population. Induction of a variety of neuroinflammatory measures has been reported with brain aging but the relationship between neuroinflammation and cognitive decline with non-neurodegenerative, normative aging remains largely unexplored. This study sought to comprehensively investigate expression of the MHC II immune response pathway and glial activation in the hippocampus in the context of both aging and age-related cognitive decline. METHODS Three independent cohorts of adult (12-13 months) and aged (26-28 months) F344xBN rats were behaviorally characterized by Morris water maze testing. Expression of MHC II pathway-associated genes identified by transcriptomic analysis as upregulated with advanced aging was quantified by qPCR in synaptosomal fractions derived from whole hippocampus and in hippocampal subregion dissections (CA1, CA3, and DG). Activation of astrocytes and microglia was assessed by GFAP and Iba1 protein expression, and by immunohistochemical visualization of GFAP and both CD74 (Ox6) and Iba1. RESULTS We report a marked age-related induction of neuroinflammatory signaling transcripts (i.e., MHC II components, toll-like receptors, complement, and downstream signaling factors) throughout the hippocampus in all aged rats regardless of cognitive status. Astrocyte and microglial activation was evident in CA1, CA3 and DG of intact and impaired aged rat groups, in the absence of differences in total numbers of GFAP+ astrocytes or Iba1+ microglia. Both mild and moderate microglial activation was significantly increased in all three hippocampal subregions in aged cognitively intact and cognitively impaired rats compared to adults. Neither induction of MHCII pathway gene expression nor glial activation correlated to cognitive performance. CONCLUSIONS These data demonstrate a novel, coordinated age-related induction of the MHC II immune response pathway and glial activation in the hippocampus, indicating an allostatic shift toward a para-inflammatory phenotype with advancing age. Our findings demonstrate that age-related induction of these aspects of hippocampal neuroinflammation, while a potential contributing factor, is not sufficient by itself to elicit impairment of spatial learning and memory in models of normative aging. Future efforts are needed to understand how neuroinflammation may act synergistically with cognitive-decline specific alterations to cause cognitive impairment.
Collapse
Affiliation(s)
- Heather D VanGuilder
- Department of Pharmacology, Pennsylvania State University College of Medicine, 500 University Drive, Hershey, Pennsylvania 17057, USA
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Dinel AL, André C, Aubert A, Ferreira G, Layé S, Castanon N. Cognitive and emotional alterations are related to hippocampal inflammation in a mouse model of metabolic syndrome. PLoS One 2011; 6:e24325. [PMID: 21949705 PMCID: PMC3174932 DOI: 10.1371/journal.pone.0024325] [Citation(s) in RCA: 188] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2011] [Accepted: 08/06/2011] [Indexed: 01/01/2023] Open
Abstract
Converging clinical data suggest that peripheral inflammation is likely involved in the pathogenesis of the neuropsychiatric symptoms associated with metabolic syndrome (MetS). However, the question arises as to whether the increased prevalence of behavioral alterations in MetS is also associated with central inflammation, i.e. cytokine activation, in brain areas particularly involved in controlling behavior. To answer this question, we measured in a mouse model of MetS, namely the diabetic and obese db/db mice, and in their healthy db/+ littermates emotional behaviors and memory performances, as well as plasma levels and brain expression (hippocampus; hypothalamus) of inflammatory cytokines. Our results shows that db/db mice displayed increased anxiety-like behaviors in the open-field and the elevated plus-maze (i.e. reduced percent of time spent in anxiogenic areas of each device), but not depressive-like behaviors as assessed by immobility time in the forced swim and tail suspension tests. Moreover, db/db mice displayed impaired spatial recognition memory (hippocampus-dependent task), but unaltered object recognition memory (hippocampus-independent task). In agreement with the well-established role of the hippocampus in anxiety-like behavior and spatial memory, behavioral alterations of db/db mice were associated with increased inflammatory cytokines (interleukin-1β, tumor necrosis factor-α and interleukin-6) and reduced expression of brain-derived neurotrophic factor (BDNF) in the hippocampus but not the hypothalamus. These results strongly point to interactions between cytokines and central processes involving the hippocampus as important contributing factor to the behavioral alterations of db/db mice. These findings may prove valuable for introducing novel approaches to treat neuropsychiatric complications associated with MetS.
Collapse
Affiliation(s)
- Anne-Laure Dinel
- Nutrition et Neurobiologie Intégrée, INRA UMR 1286, Bordeaux, France
- University of Bordeaux, Bordeaux, France
| | - Caroline André
- Nutrition et Neurobiologie Intégrée, INRA UMR 1286, Bordeaux, France
- University of Bordeaux, Bordeaux, France
| | - Agnès Aubert
- Nutrition et Neurobiologie Intégrée, INRA UMR 1286, Bordeaux, France
- University of Bordeaux, Bordeaux, France
| | - Guillaume Ferreira
- Nutrition et Neurobiologie Intégrée, INRA UMR 1286, Bordeaux, France
- University of Bordeaux, Bordeaux, France
| | - Sophie Layé
- Nutrition et Neurobiologie Intégrée, INRA UMR 1286, Bordeaux, France
- University of Bordeaux, Bordeaux, France
| | - Nathalie Castanon
- Nutrition et Neurobiologie Intégrée, INRA UMR 1286, Bordeaux, France
- University of Bordeaux, Bordeaux, France
- * E-mail:
| |
Collapse
|
44
|
Blalock EM, Buechel HM, Popovic J, Geddes JW, Landfield PW. Microarray analyses of laser-captured hippocampus reveal distinct gray and white matter signatures associated with incipient Alzheimer's disease. J Chem Neuroanat 2011; 42:118-26. [PMID: 21756998 DOI: 10.1016/j.jchemneu.2011.06.007] [Citation(s) in RCA: 146] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2011] [Revised: 06/27/2011] [Accepted: 06/27/2011] [Indexed: 01/03/2023]
Abstract
Alzheimer's disease (AD) is a devastating neurodegenerative disorder that threatens to reach epidemic proportions as our population ages. Although much research has examined molecular pathways associated with AD, relatively few such studies have focused on the disease's critical early stages. In a prior microarray study we correlated gene expression in hippocampus with degree of Alzheimer's disease and found close associations between upregulation of apparent glial transcription factor/epigenetic/tumor suppressor genes and incipient AD. The results suggested a new model in which AD pathology spreads along myelinated axons (Blalock et al., 2004). However, the microarray analyses were performed on RNA extracted from frozen hand-dissected hippocampal CA1 tissue blocks containing both gray and white matter, limiting the confidence with which transcriptional changes in gray matter could be distinguished from those in white matter. Here, we used laser capture microdissection (LCM) to exclude major white matter tracts while selectively collecting CA1 hippocampal gray matter from formalin-fixed, paraffin-embedded (FFPE) hippocampal sections of the same subjects assessed in our prior study. Microarray analyses of this gray matter-enriched tissue revealed many transcriptional changes similar to those seen in our past study and in studies by others, particularly for downregulated neuron-related genes. Additionally, the present analyses identified several previously undetected pathway alterations, including downregulation of molecules that stabilize ryanodine receptor Ca2+ release and upregulation of vasculature development. Conversely, we found a striking paucity of the upregulated changes in the putative glial and growth-related genes that had been strongly overrepresented in the prior mixed-tissue study. We conclude that FFPE tissue can be a reliable resource for microarray studies of brain tissue, that upregulation of growth-related epigenetic/transcription factors during incipient AD is predominantly localized in and around white matter (supporting our prior findings and model), and that novel alterations in vascular and ryanodine receptor-related pathways in gray matter are closely associated with incipient AD.
Collapse
Affiliation(s)
- Eric M Blalock
- Department of Molecular and Biomedical Pharmacology, University of Kentucky College of Medicine, Lexington, KY 40536, USA.
| | | | | | | | | |
Collapse
|
45
|
Vanguilder HD, Freeman WM. The hippocampal neuroproteome with aging and cognitive decline: past progress and future directions. Front Aging Neurosci 2011; 3:8. [PMID: 21647399 PMCID: PMC3102218 DOI: 10.3389/fnagi.2011.00008] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2011] [Accepted: 05/12/2011] [Indexed: 01/12/2023] Open
Abstract
Although steady progress on understanding brain aging has been made over recent decades through standard anatomical, immunohistochemical, and biochemical techniques, the biological basis of non-neurodegenerative cognitive decline with aging remains to be determined. This is due in part to technical limitations of traditional approaches, in which only a small fraction of neurobiologically relevant proteins, mRNAs or metabolites can be assessed at a time. With the development and refinement of proteomic technologies that enable simultaneous quantitative assessment of hundreds to thousands of proteins, neuroproteomic studies of brain aging and cognitive decline are becoming more widespread. This review focuses on the contributions of neuroproteomic investigations to advances in our understanding of age-related deficits of hippocampus-dependent spatial learning and memory. Accumulating neuroproteomic data demonstrate that hippocampal aging involves common themes of dysregulated metabolism, increased oxidative stress, altered protein processing, and decreased synaptic function. Additionally, growing evidence suggests that cognitive decline does not represent a "more aged" phenotype, but rather is associated with specific neuroproteomic changes that occur in addition to age-related alterations. Understanding if and how age-related changes in the hippocampal neuroproteome contribute to cognitive decline and elucidating the pathways and processes that lead to cognitive decline are critical objectives that remain to be achieved. Progress in the field and challenges that remain to be addressed with regard to animal models, behavioral testing, and proteomic reporting are also discussed.
Collapse
Affiliation(s)
- Heather D Vanguilder
- Department of Pharmacology, Penn State College of Medicine, Milton S. Hershey Medical Center Hershey, PA, USA
| | | |
Collapse
|
46
|
Buechel HM, Popovic J, Searcy JL, Porter NM, Thibault O, Blalock EM. Deep sleep and parietal cortex gene expression changes are related to cognitive deficits with age. PLoS One 2011; 6:e18387. [PMID: 21483696 PMCID: PMC3070733 DOI: 10.1371/journal.pone.0018387] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2010] [Accepted: 03/06/2011] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Age-related cognitive deficits negatively affect quality of life and can presage serious neurodegenerative disorders. Despite sleep disruption's well-recognized negative influence on cognition, and its prevalence with age, surprisingly few studies have tested sleep's relationship to cognitive aging. METHODOLOGY We measured sleep stages in young adult and aged F344 rats during inactive (enhanced sleep) and active (enhanced wake) periods. Animals were behaviorally characterized on the Morris water maze and gene expression profiles of their parietal cortices were taken. PRINCIPAL FINDINGS Water maze performance was impaired, and inactive period deep sleep was decreased with age. However, increased deep sleep during the active period was most strongly correlated to maze performance. Transcriptional profiles were strongly associated with behavior and age, and were validated against prior studies. Bioinformatic analysis revealed increased translation and decreased myelin/neuronal pathways. CONCLUSIONS The F344 rat appears to serve as a reasonable model for some common sleep architecture and cognitive changes seen with age in humans, including the cognitively disrupting influence of active period deep sleep. Microarray analysis suggests that the processes engaged by this sleep are consistent with its function. Thus, active period deep sleep appears temporally misaligned but mechanistically intact, leading to the following: first, aged brain tissue appears capable of generating the slow waves necessary for deep sleep, albeit at a weaker intensity than in young. Second, this activity, presented during the active period, seems disruptive rather than beneficial to cognition. Third, this active period deep sleep may be a cognitively pathologic attempt to recover age-related loss of inactive period deep sleep. Finally, therapeutic strategies aimed at reducing active period deep sleep (e.g., by promoting active period wakefulness and/or inactive period deep sleep) may be highly relevant to cognitive function in the aging community.
Collapse
Affiliation(s)
- Heather M. Buechel
- Department of Molecular and Biomedical Pharmacology, University of Kentucky College of Medicine, Lexington, Kentucky, United States of America
| | - Jelena Popovic
- Department of Molecular and Biomedical Pharmacology, University of Kentucky College of Medicine, Lexington, Kentucky, United States of America
| | - James L. Searcy
- Department of Molecular and Biomedical Pharmacology, University of Kentucky College of Medicine, Lexington, Kentucky, United States of America
| | - Nada M. Porter
- Department of Molecular and Biomedical Pharmacology, University of Kentucky College of Medicine, Lexington, Kentucky, United States of America
| | - Olivier Thibault
- Department of Molecular and Biomedical Pharmacology, University of Kentucky College of Medicine, Lexington, Kentucky, United States of America
| | - Eric M. Blalock
- Department of Molecular and Biomedical Pharmacology, University of Kentucky College of Medicine, Lexington, Kentucky, United States of America
- * E-mail:
| |
Collapse
|
47
|
Disrupting function of FK506-binding protein 1b/12.6 induces the Ca²+-dysregulation aging phenotype in hippocampal neurons. J Neurosci 2011; 31:1693-703. [PMID: 21289178 DOI: 10.1523/jneurosci.4805-10.2011] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
With aging, multiple Ca(2+)-associated electrophysiological processes exhibit increased magnitude in hippocampal pyramidal neurons, including the Ca(2+)-dependent slow afterhyperpolarization (sAHP), L-type voltage-gated Ca(2+) channel (L-VGCC) activity, Ca(2+)-induced Ca(2+) release (CICR) from ryanodine receptors (RyRs), and Ca(2+) transients. This pattern of Ca(2+) dysregulation correlates with reduced neuronal excitability/plasticity and impaired learning/memory and has been proposed to contribute to unhealthy brain aging and Alzheimer's disease. However, little is known about the underlying molecular mechanisms. In cardiomyocytes, FK506-binding protein 1b/12.6 (FKBP1b) binds and stabilizes RyR2 in the closed state, inhibiting RyR-mediated Ca(2+) release. Moreover, we recently found that hippocampal Fkbp1b expression is downregulated, whereas Ryr2 and Frap1/Mtor (mammalian target of rapamycin) expression is upregulated with aging in rats. Here, we tested the hypothesis that disrupting FKBP1b function also destabilizes Ca(2+) homeostasis in hippocampal neurons and is sufficient to induce the aging phenotype of Ca(2+) dysregulation in young animals. Selective knockdown of Fkbp1b with interfering RNA in vitro (96 h) enhanced voltage-gated Ca(2+) current in cultured neurons, whereas in vivo Fkbp1b knockdown by microinjection of viral vector (3-4 weeks) dramatically increased the sAHP in hippocampal slice neurons from young-adult rats. Rapamycin, which displaces FKBP1b from RyRs in myocytes, similarly enhanced VGCC current and the sAHP and also increased CICR. Moreover, FKBP1b knockdown in vivo was associated with upregulation of RyR2 and mTOR protein expression. Thus, disruption of FKBP1b recapitulated much of the Ca(2+)-dysregulation aging phenotype in young rat hippocampus, supporting a novel hypothesis that declining FKBP function plays a major role in unhealthy brain aging.
Collapse
|
48
|
Aguilera G. HPA axis responsiveness to stress: implications for healthy aging. Exp Gerontol 2011; 46:90-5. [PMID: 20833240 PMCID: PMC3026863 DOI: 10.1016/j.exger.2010.08.023] [Citation(s) in RCA: 159] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2010] [Revised: 08/19/2010] [Accepted: 08/27/2010] [Indexed: 02/06/2023]
Abstract
The major neuroendocrine response mediating stress adaptation is activation of the hypothalamic pituitary adrenal axis, with stimulation of corticotropin releasing hormone (CRH) and vasopressin (VP) from parvocellular neurons of the hypothalamic paraventricular nucleus, leading to stimulation of pituitary ACTH secretion and increases in glucocorticoid secretion from the adrenal cortex. Basal production and transient increases during stress of glucocorticoids and its hypothalamic regulators are essential for neuronal plasticity and normal brain function. While activation of the HPA axis is essential for survival during stress, chronic exposure to stress hormones can predispose to psychological, metabolic and immune alterations. Thus, prompt termination of the stress response is essential to prevent negative effects of inappropriate levels of CRH and glucocorticoids. This review addresses the regulation of HPA axis activity with emphasis on the mechanisms of termination of CRH transcription, which is a critical step in this process. In addition, the actions by which glucocorticoids, CRH and VP can affect the aging process will be discussed.
Collapse
Affiliation(s)
- Greti Aguilera
- Section on Endocrine Physiology, Program on Developmental Endocrinology and Genetics, Eunice Kennedy Shiver Institute of Child Health and Human Development, NIH, Bethesda, MD 20892, USA.
| |
Collapse
|
49
|
Disrupted energy metabolism and neuronal circuit dysfunction in cognitive impairment and Alzheimer's disease. Lancet Neurol 2010; 10:187-98. [PMID: 21147038 DOI: 10.1016/s1474-4422(10)70277-5] [Citation(s) in RCA: 408] [Impact Index Per Article: 29.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Epidemiological, neuropathological, and functional neuroimaging evidence implicates global and regional disruptions in brain metabolism and energetics in the pathogenesis of cognitive impairment. Nerve cell microcircuits are modified by excitatory and inhibitory synaptic activity and neurotrophic factors. Ageing and Alzheimer's disease cause perturbations in cellular energy metabolism, level of excitation or inhibition, and neurotrophic factor release, which overwhelm compensatory mechanisms and result in dysfunction of neuronal microcircuits and brain networks. A prolonged positive energy balance impairs the ability of neurons to adapt to oxidative and metabolic stress. Results from experimental studies in animals show how disruptions caused by chronic positive energy balance, such as diabetes, lead to accelerated cognitive ageing and Alzheimer's disease. Therapeutic interventions to allay cognitive dysfunction that target energy metabolism and adaptive stress responses (such as neurotrophin signalling) have been effective in animal models and in preliminary studies in humans.
Collapse
|
50
|
C825T polymorphism of the GNB3 gene on valproate-related metabolic abnormalities in bipolar disorder patients. J Clin Psychopharmacol 2010; 30:512-7. [PMID: 20814328 DOI: 10.1097/jcp.0b013e3181f03f50] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
BACKGROUND Valproate (VPA) is a mood stabilizer for treating patients with bipolar disorder (BD). It may cause metabolic abnormalities in certain bipolar patients. However, the genetic factors that influence the susceptibility remain unclear. Genetic polymorphism of the G-protein β3 subunit (GNB3) is reported to be associated with metabolic phenotypes. In the current study, we investigated the possible associations between the GNB3 variation and VPA-induced metabolic abnormalities. METHODS Subjects (n = 96) who met the Diagnostic and Statistical Manual of Mental Disorders, Fourth Edition criteria for BD were recruited from the National Cheng Kung University Hospital. Their metabolic indices were measured. RESULTS The variation of GNB3 C825T showed an association with higher plasma total cholesterol (P = 0.037), triglyceride (P = 0.014), and leptin (P < 0.001) levels in BD patients treated with VPA. After adjusting for age, sex, types of BDs, and serum concentration of VPA, the variation of GNB3 C825T remained significantly associated with the levels of serum leptin and body mass index (BMI; P < 0.001 and P = 0.030, respectively). In addition, the GNB3 C825T showed significant drug-single-nucleotide polymorphism interactions with insulin levels (P = 0.033), triglyceride levels (P = 0.013), leptin levels (P = 0.013), and BMI (P = 0.018). These results indicated that the T allele may be associated with lower serum leptin levels and BMI in BD patients treated with VPA. CONCLUSIONS The current study provides evidence that BD patients who are T allele carriers of the GNB3 C825T polymorphism have a lower risk for VPA-induced metabolic abnormalities. Further studies about the underlying mechanisms of G protein in VPA-induced metabolic abnormalities are warranted.
Collapse
|